UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

WASHINGTON,Washington, D.C. 20549

FORM 10-K

(Mark One)

[X]ANNUAL REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

For the fiscal year endedNovember 30, 2018December 31, 2020

or

[ ]TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934

For the transition period from __________________________ to __________________________

Commission file number000-54329001-38416

ORGENESIS INC.


(Exact(Exact name of registrant as specified in its charter)

Nevada98-0583166
State or Other Jurisdictionother jurisdiction(I.R.S. Employer
of Incorporationincorporation or OrganizationorganizationIdentification No.)

20271 Goldenrod Lane, Germantown, MD20876


(Address(Address of Principal Executive Offices) (Zip Code)

Registrant’s telephone number, including area code:(480)659-6404

Securities registered pursuant to Section 12(b) of the Act:

Common stock, par value $0.0001 per share

Title of each classTrading Symbol(s)Name of each exchange on which registered
Common Stock, par value $0.0001 per shareORGSThe Nasdaq Capital Market

Securities registered pursuant to Section 12(g) of the Act:None

Indicate by check mark if the registrant is a well-known seasoned issuer, as defined in Rule 405 of the Securities Act. Yes [ ] No [X]

Indicate by check mark if the registrant is not required to file reports pursuant to Section 13 or Section 15(d) of the Act. Yes [ ] No [X]

Indicate by check mark whether the registrant (1) has filed all reports required to be filed by Section 13 or 15(d) of the Securities Exchange Act of 1934 during the preceding 12 months (or for such shorter period that the registrant was required to file such reports), and (2) has been subject to such filing requirements for the past 90 days. Yes [X] No [ ]

Indicate by check mark whether the registrant has submitted electronically every Interactive Data File required to be submitted pursuant to Rule 405 of Regulation S-T (§ 232.405 of this chapter) during the preceding 12 months (or for such shorter period that the registrant was required to submit such files). Yes [X] No [ ]

Indicate by check mark if disclosure of delinquent filers pursuant to Item 405 of Regulation S-K (§ 229.405 of this chapter) is not contained herein, and will not be contained, to the best of registrant’s knowledge, in definitive proxy or information statements incorporated by reference in Part III of this Form 10-K or any amendment to this Form 10-K.[ ]

Indicate by check mark whether the registrant is a large accelerated filer, an accelerated filer, a non-accelerated filer, a smaller reporting company, or an emerging growth company. See the definitions of “large accelerated filer,” “accelerated filer” “smaller reporting company,” and “emerging growth company” in Rule 12b-2 of the Exchange Act.

Large accelerated filer [   ]Accelerated filer[X]
Non-accelerated filer [   ]Smaller reporting company[X]
Emerging growth company [   ]

If an emerging growth company, indicate by checkmark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13(a) of the Exchange Act. [ ]

Indicate by check mark whether the registrant has filed a report on and attestation to its management’s assessment of the effectiveness of its internal control over financial reporting under Section 404(b) of the Sarbanes-Oxley Act (15 U.S.C. 7262(b)) by the registered public accounting firm that prepared or issued its audit report. ☐

Indicate by check mark whether the registrant is a shell company (as defined in Rule 12b-2 of the Act). Yes[ ] No [X]

The registrant had 15,620,97124,199,674 shares of common stock outstanding as of February 13, 2019.March 9, 2021. The aggregate market value of the common stock held by non-affiliates of the registrant as of May 31, 2018the last business day of the registrant’s most recently completed second fiscal quarter (June 30, 2020) was $92,456,008,$105,399,427, as computed by reference to the closing price of such common stock on The Nasdaq Capital Market on such date.

 


 

ORGENESIS INC.

20182020 FORM 10-K ANNUAL REPORT

TABLE OF CONTENTSsCONTENTS

Page

PART I

ITEM 1. BUSINESS4
  
ITEM 1. BUSINESS5
ITEM 1A. RISK FACTORS3221
ITEM 1B. UNRESOLVED STAFF COMMENTS5342
ITEM 2. PROPERTIES5342
ITEM 3. LEGAL PROCEEDINGS5443
ITEM 4. MINE SAFETY DISCLOSURES5443

PART II

PART II
ITEM 5. MARKET FOR REGISTRANT’S COMMON EQUITY, RELATED STOCKHOLDER MATTERS AND ISSUER PURCHASES OF EQUITY SECURITIES5443
ITEM 6. SELECTED FINANCIAL DATA5645
ITEM 7. MANAGEMENT’S DISCUSSION AND ANALYSIS OF FINANCIAL CONDITION AND RESULTS OF OPERATIONS5645
ITEM 7A. QUANTITATIVE AND QUALITATIVE DISCLOSURES ABOUT MARKET RISK7261
ITEM 8. FINANCIAL STATEMENTS AND SUPPLEMENTARY DATA7261
ITEM 9. CHANGES IN AND DISAGREEMENTS WITH ACCOUNTANTS ON ACCOUNTING AND FINANCIAL DISCLOSURE7261
ITEM 9A. CONTROLS AND PROCEDURES61
ITEM 9B. OTHER INFORMATION62
  
ITEM 9A. CONTROLS AND PROCEDURES

PART III

72
  
ITEM 9B. OTHER INFORMATION73

PART III

ITEM 10. DIRECTORS, EXECUTIVE OFFICERS AND CORPORATE GOVERNANCE7462
ITEM 11. EXECUTIVE COMPENSATION7967
ITEM 12. SECURITY OWNERSHIP OF CERTAIN BENEFICIAL OWNERS AND MANAGEMENT AND RELATED STOCKHOLDER MATTERS8573
ITEM 13. CERTAIN RELATIONSHIPS AND RELATED TRANSACTIONS, AND DIRECTOR INDEPENDENCE8876

ITEM 14. PRINCIPAL ACCOUNTANT FEES AND SERVICES

8977

PART IV

ITEM 15. EXHIBITS, FINANCIAL STATEMENT SCHEDULES90
  
ITEM 15. EXHIBITS AND FINANCIAL STATEMENT SCHEDULES78
ITEM 16. FORM 10-K SUMMARY9381
SIGNATURES82

 2 
SIGNATURES94

-1-

FORWARD-LOOKING STATEMENTS

CAUTIONARY STATEMENT FOR PURPOSES OF THE "SAFE HARBOR"“SAFE HARBOR” PROVISIONS OF THE PRIVATE SECURITIES LITIGATION REFORM ACT OF 1995

The following discussion should be read in conjunction with the financial statements and related notes contained elsewhere in this Annual Report on Form 10-K. Certain statements made in this discussion are “forward-looking statements” within the meaning of 27A of the Securities Act of 1933, as amended (the “Securities Act”), and Section 21E of the Securities Exchange Act of 1934, as amended. These statements are based upon beliefs of, and information currently available to, the Company’s management as well as estimates and assumptions made by the Company’s management. Readers are cautioned not to place undue reliance on these forward-looking statements, which are only predictions and speak only as of the date hereof. When used herein, the words “anticipate,” “believe,” “estimate,” “expect,” “forecast,” “future,” “intend,” “plan,” “predict,” “project,” “target,” “potential,” “will,” “would,” “could,” “should,” “continue” or the negative of these terms and similar expressions as they relate to the Company or the Company’s management identify forward-looking statements. Such statements reflect the current view of the Company with respect to future events and are subject to risks, uncertainties, assumptions, and other factors, including the risks relating to the Company’s business, industry, and the Company’s operations and results of operations. Should one or more of these risks or uncertainties materialize, or should the underlying assumptions prove incorrect, actual results may differ significantly from those anticipated, believed, estimated, expected, intended, or planned.

Although the Company believes that the expectations reflected in the forward-looking statements are reasonable, the Company cannot guarantee future results, levels of activity, performance, or achievements. Except as required by applicable law, including the securities laws of the United States, the Company does not intend to update any of the forward-looking statements to conform these statements to actual results.

Our financial statements are prepared in accordance with accounting principles generally accepted in the United States (“GAAP”). These accounting principles require us to make certain estimates, judgments and assumptions. We believe that the estimates, judgments and assumptions upon which we rely are reasonable based upon information available to us at the time that these estimates, judgments and assumptions are made. These estimates, judgments and assumptions can affect the reported amounts of assets and liabilities as of the date of the financial statements as well as the reported amounts of revenues and expenses during the periods presented. Our financial statements would be affected to the extent there are material differences between these estimates and actual results. The following discussion should be read in conjunction with our financial statements and notes thereto appearing elsewhere in this report.

Unless otherwise indicated or the context requires otherwise, the words “we,” “us,” “our,” the “Company” or“Company,” “our Company” or “Orgenesis” refer to Orgenesis Inc., a Nevada corporation, and its majority-owned subsidiary, Masthercell Global Inc., a Delaware corporation (“Masthercell Global”our majority or wholly-owned subsidiaries, Orgenesis Korea Co. Ltd. (the “Korean Subsidiary”), and(formerly known as CureCell); Orgenesis SPRL,Belgium SRL, a Belgian-based entity which is engaged in development and manufacturing activities, together with clinical development studies in Europe (the “Belgian Subsidiary”), and its wholly-owned subsidiaries; Orgenesis Ltd., an Israeli corporation (the “Israeli Subsidiary”),; Orgenesis Maryland Inc., a Maryland corporation (the “Maryland“U.S. Subsidiary”); Orgenesis Switzerland Sarl, which was incorporated in October 2020 (the “Swiss Subsidiary”); Orgenesis Biotech Israel Ltd. (formerly known as Atvio Biotech Ltd.) (“OBI”); Koligo Therapeutics Inc., a Kentucky corporation, purchased in 2020 (“Koligo”); Masthercell Global Inc. (“Masthercell”) and its wholly owned subsidiaries Cell Therapy Holdings S.A.  Masthercell Global’s wholly-owned subsidiaries include, MaSTherCell, S.AS.A. (“MaSTherCell”), a Belgian-based subsidiary and a Contract Development and Manufacturing Organization (“CDMO”) specialized in cell therapy development and manufacturing for advanced medicinal products, and Masthercell U.S., LLC (“Masthercell U.S.”), a U.S.-based CDMO Atvio Biotech Ltd. (“Atvio”(collectively, “Masthercell”), an Israeli-based CDMO,. The Company sold all of its equity interests in Masthercell and CureCell Co. Ltd. (“CureCell”), a Korea-based CDMO.its subsidiaries on February 20, 2020.

3

Forward-looking statements made in this annual reportAnnual Report on Form 10-K include statements about:

Corporate and Financial

·our ability to increase revenues;
our ability to achieve profitability;
·our ability to increase revenuesmanage our research and raise sufficient capital or strategic business arrangements to expand our CDMO global network;development programs that are based on novel technologies;
·our ability to grow the size and capabilities of our organization through further collaboration and strategic alliances;alliances to expand our point-of-care cell therapy business;

-2-

·our ability to control key elements relating to the development and commercialization of therapeutic product candidates with third parties;
our ability to manage potential disruptions as a result of the coronavirus outbreak;
our ability to manage the growth of our CDMO business;company;
·our ability to attract and retain key scientific or management personnel and to expand our management team;
·the accuracy of estimates regarding expenses, future revenue, capital requirements, profitability, and needs for additional financing; and
·our belief that our therapeutic related developments have competitive advantages such as our cell trans-differentiation technology being developed by our Israeli Subsidiary and being able tocan compete favorably and profitably in the cell and gene therapy industry;industry.

CDMOCell & Gene Therapy Business (“CGT”)

·our ability to grow the business of Masthercell Global (including each of its subsidiaries, MaSTherCell, Atvio and CureCell, which we consolidated into Masthercell Global in 2018);
·our ability to attract and retain customers;
·our ability to expand and maintain our CDMO business through strategic alliances, joint ventures and internal growth;
·our ability to fund the operational and capital requirements of the global expansion of Masthercell Global and our CDMO business;
·our expectations regarding Masthercell Global’s expenses and revenue, including our expectations that our research and development expenses and selling, general and administrative expenses may increase in absolute dollars;
·the successful integration of our clinical and CDMO strategy through Masthercell Global and its subsidiaries;
·our ability to contract (through Masthercell Global and its subsidiaries) with third-party suppliers and manufacturers and their ability to perform adequately;
·extensive industry regulation, and how that will continue to have a significant impact on our business, especially our product development, manufacturing and distribution capabilities;
·the ability of Masthercell Global to receive future payments pursuant to that certain Stock Purchase Agreement dated June 28, 2018 by and between the Company, Masthercell Global and GPP-II Masthercell, LLC (“GPP-II”);
·our and our shareholders’ ability to receive value on par with GPP-II upon its forced sale of Masthercell Global;
·our ability to control, direct the activities of or hold any shares in Masthercell Global if GPP-II were to assume control of the Board of Directors of Masthercell Global or if it were to purchase our shares in Masthercell Global;
·our ability to receive benefits or value from Masthercell Global in the event of a spin-off effectuated by GPP-II; and
·the significant potential dilution to our existing shareholders due to GPP-II’s option to exchange its Masthercell Global Preferred Stock for shares of our common stock.

PT Business

·our ability to adequately fund and scale our various collaboration, license, partnership and joint venture agreements for the development of therapeutic products and technologies;
·our ability to develop, through our Israeli Subsidiary and Belgian Subsidiary, to the clinical stage a new technology to transdifferentiate liver cells into functional insulin-producing cells, thus enabling normal glucose regulated insulin secretion, via cell therapy;
·our ability to advance our therapeutic collaborations in terms of industrial development, clinical development, regulatory challenges, commercial partners and manufacturing availability;
·our ability to implement our point-of-care (“POCare”) cell therapy (“PT”POC”) strategy in order to further develop and advance autologous therapies to reach patients;
·expectations regarding theour ability of our Maryland Subsidiary, Israeli Subsidiary and Belgian Subsidiary to obtain additional and maintain existing intellectual property protection for our technologies and therapies;
·our ability to commercialize products in light of the intellectual property rights of others;

-3-

·our ability to obtain funding necessary to start and complete such clinical trials;
·our ability to further our CGT development projects, either directly or through our JV partner agreements, and to fulfill our obligations under such agreements;
our belief that Diabetes Mellitus will be one of the most challenging health problems in the 21st century and will have staggering health, societal and economic impact;
·our belief that our diabetes-related treatment seems to be safer thansystems and therapies are as at least as safe and as effective as other options;
·our Subsidiary’s relationship with Tel Hashomer Medical Research Infrastructure and Services Ltd. (“THM”) and the risk that THM may cancel or, at the very least continue to challenge, the License Agreement; andAgreement with Orgenesis Ltd. as we continue to expand our focus to other therapies;
·our license agreements with other institutions;
expenditures not resulting in commercially successful products.products;
our dependence on the financial results of our POC business; and
our ability to grow our POC business and to develop additional joint venture relationships in order to produce demonstrable revenues.

These statements are only predictions and involve known and unknown risks, uncertainties and other factors, including the risks in the section entitled “Risk Factors” set forth in this Annual Report on Form 10-K for the year ended November 30, 2018,December 31, 2020, any of which may cause our Company’s or our industry’s actual results, levels of activity, performance or achievements to be materially different from any future results, levels of activity, performance or achievements expressed or implied by these forward-looking statements. These risks may cause the Company’s or its industry’s actual results, levels of activity or performance to be materially different from any future results, levels of activity or performance expressed or implied by these forward looking statements.

Although we believe that the expectations reflected in the forward-looking statements are reasonable, we cannot guarantee future results, levels of activity or performance. Moreover, neither we nor any other person assumes responsibility for the accuracy and completeness of these forward-looking statements. The Company is under no duty to update any forward-looking statements after the date of this report to conform these statements to actual results.

4

PART I

ITEM 1. BUSINESS

Business Overview

We areOrgenesis Inc., a biotechnologyNevada corporation, is a global biotech company specializing inworking to unlock the development, manufacturing and provisionpotential of technologies and services in the cell and gene therapies in an affordable and accessible format (“CGTs”).

CGTs can be centered on autologous (using the patient’s own cells) or allogenic (using master banked donor cells) and are part of a class of medicines referred to as advanced therapy industry.medicinal products (ATMPs). We operatemostly focus on autologous therapies, with processes and systems that are developed for each therapy using a closed and automated processing system approach that is validated for compliant production near the patient at their point of care for treatment of the patient. This approach has the potential to overcome the limitations of traditional commercial manufacturing methods that do not translate well to commercial production of advanced therapies due to their cost prohibitive nature and complex logistics to deliver the treatments to patients (ultimately limiting the number of patients that can have access to, or can afford, these therapies).

To achieve these goals, we have developed a Point of Care Platform comprised of three enabling components: a pipeline of licensed POCare Therapies that are designed to be processed and produced in closed, automated POCare Technology systems across a collaborative POCare Network. Via a combination of science, technology, engineering, and networking, we are working to provide a more efficient and scalable pathway for advanced therapies to reach patients more rapidly at lowered costs. We also draw on extensive medical expertise to identify promising new autologous therapies to leverage within the POCare Platform either via ownership or licensing.

The POCare Network brings together patients, doctors, industry partners, research institutes and hospitals worldwide with a goal of achieving harmonized, regulated clinical development and production of the therapies.

POCare Platform Operations via Subsidiaries

We currently conduct our core business operations ourselves and through our subsidiaries which are all wholly-owned except as otherwise stated below (collectively, the “Subsidiaries”). The Subsidiaries are as follows:

United States

Orgenesis Maryland Inc. (the “U.S. Subsidiary”) is the center of activity in North America and is currently focused on setting up the POCare Network.
Koligo Therapeutics Inc. (“Koligo”) is a Kentucky corporation that we acquired in 2020 and is currently focused on developing the POCare network and therapies. .

5

Europe

Orgenesis Belgium SRL (the “Belgian Subsidiary”) is the center of activity in Europe and is currently focused on process development and the preparation of European clinical trials.
Orgenesis Switzerland Sarl (the “Swiss Subsidiary”), was incorporated in October 2020, and is currently focused on providing management services to us.

Asia

Orgenesis Ltd. in Israel (the “Israeli Subsidiary”) is a provider of regulatory, clinical and pre-clinical services.
Orgenesis Biotech Israel Ltd. (“OBI”), is a provider of cell-processing services in Israel.
Korea: Orgenesis Korea Co. Ltd. (the “Korean Subsidiary”), is a provider of processing and pre-clinical services in Korea. We own 94.12% of the Korean Subsidiary.

Discontinued Operations

Until December 31, 2019, we operated the POCare Platform as one of two platforms: (i) a point-of-care (“POCare”) cell therapy platform (“PT”) and (ii)business separate business segments.

Historically, the second separate business segment was operated as a Contract Development and Manufacturing Organization (“CDMO”) platform, conducted through our subsidiary, Masthercell Global. Through our PT business, our aim is to further the development of Advanced Therapy Medicinal Products (“ATMPs”) through collaborations and in-licensing with other pre-clinical and clinical-stage biopharmaceutical companies and research and healthcare institutes to bring such ATMPs to patients. We out-license these ATMPs through regional partners to whom we also provide regulatory, pre-clinical and training services to support their activity in order to reach patients in a point-of-care hospital setting. Through our CDMO platform, we are focused on providing contract manufacturing and development services for biopharmaceutical companies.companies (the “CDMO Business”). The CDMO platform was historically operated mainly through majority owned Masthercell Global (which consisted of the following two subsidiaries: MaSTherCell S.A. in Belgium (“MaSTherCell”), and Masthercell U.S., LLC in the United States (“Masthercell U.S.”) (collectively, the “Masthercell Global Subsidiaries”)).

ActivitiesIn February 2020, we and GPP-II Masthercell LLC (“GPP”) sold 100% of the outstanding equity interests of Masthercell (the “Masthercell Business”), which comprised the majority of our CDMO Business, to Catalent Pharma Solutions, Inc. for an aggregate nominal purchase price of $315 million, subject to customary adjustments (the “Masthercell Sale”). After accounting for GPP’s liquidation preference and equity stake in our PT business include a multitude of cell therapies, including autoimmune, oncologic, neurologic and metabolic diseases and other indications. We provide services for our joint venture (“JV”) partners, pharmaceutical and biotech companiesMasthercell as well as research institutionsother investor interests in our Belgian subsidiary MaSTherCell, distributions to Masthercell option holders and hospitalstransaction costs, we received approximately $126.7 million. We incurred an additional approximately $5.6 million in transaction costs.

We determined that the Masthercell Business (“Discontinued Operation”) meets the criteria to be classified as a discontinued operation as of the first quarter of 2020. The Discontinued Operation includes the vast majority of the previous CDMO Business, including majority-owned Masthercell, including MaSTherCell, Masthercell U.S. and all of the Masthercell Global Subsidiaries.

Since the Masthercell Sale, we entered into new joint venture agreements with new partners in various jurisdictions. This has allowed us to grow our infrastructure and expand our processing sites into new markets and jurisdictions. In addition, we have cell therapies in clinical development. Eachengaged some of these customersjoint venture partners to perform research and collaborations representsdevelopment services to further develop and adapt our systems and devices for specific purposes. We have been investing manpower and financial resources to focus on developing, manufacturing and rolling out several types of OMPULs to be used and/or distributed through our POCare Network of partners, collaborators, and joint ventures.

The Chief Executive Officer (“CEO”) is the Company’s chief operating decision-maker who reviews financial information prepared on a revenueconsolidated basis. Effective from the first quarter of 2020, all of our continuing operations are in the point-of-care business via our POCare Platform. Therefore, no segment report has been presented.

6

Advanced Therapy Medicinal Products (ATMPs) Overview

Advanced Therapy Medicinal Product (“ATMP”) means one of any of the following medicinal products that are developed and growth opportunity upon regulatory approval. Furthermore,commercialized for human use:

A somatic cell therapy medicinal product (“STMP”) that contains cells or tissues that have been manipulated to change their biological characteristics or cells or tissues not intended to be used for the same essential functions in the body.
A tissue engineered product (“TEP”) that contains cells or tissues that have been modified so that they can be used to repair, regenerate, or replace human tissue.
A gene therapy medicinal product (“GTMP”) that engineers genes that lead to a therapeutic, prophylactic, or diagnostic effect and, in many cases, work by inserting “recombinant” genes into the body, usually to treat a variety of diseases, including genetic disorders, cancer, or long-term diseases. In this case, a recombinant gene is a stretch of DNA that is created in the laboratory, bringing together DNA from different sources.

It is important to note that although STMPs and GTMPs currently dominate the market, in order to access the market potential and trends in the future, other cell products are likely to be essential in all of these categories.

We believe that autologous therapies represent a substantial segment of the ATMP market. Autologous therapies are produced from a patient’s own cells versus allogeneic therapies that are mass-cultivated from donor cells via the construction of master and working cell banks, are then produced on a large scale. Developers and manufacturers of ATMPs (both autologous and allogeneic) currently rely heavily on production using traditional centralized supply chains and manufacturing sites.

CGTs are costly and complex to produce. We also refer to CGTs as “living” drugs since they are based on maintaining the cells vitality. Therefore, there is no possibility to sterilize the products, since such a process involves killing any living organism. Many of these therapies require sourcing of the patient’s cells, engineering them in a sterile environment and then transplanting them back to the patient (so-called “autologous” CGT). This presents multiple logistic challenges as each patient requires its own production batch, and the current processes involve complex laboratory-based types of manipulations requiring highly trained lab technicians. We are leveraging a unique approach to therapy production using the POCare Platform to potentially overcome some of the development and supply chain challenges of affordably bringing autologous therapies to patients.

Allogeneic therapies are costly and complex to produce because autologous therapies are derived from the treated patient and manufactured through a defined protocol before re-administration. We are leveraging a unique approach to therapy production using the POCare Platform to potentially overcome some of the development and supply chain challenges of bringing autologous therapies to patients affordably.

Our Therapies

Products in Clinical Use

KYSLECEL® (autologous Pancreatic Islets)

KYSLECEL is made from a patient’s own pancreatic islets – the cells that make insulin to regulate blood sugar. KYSLECEL is intended to preserve insulin secretory capacity in chronic or acute recurrent pancreatitis patients after total pancreatectomy (TP-IAT). KYSLECEL is a minimally manipulated autologous cell-based product available in the United States and regulated by the Food and Drug Administration (“FDA”). KYSLECEL is produced according to current good tissue practices (cGTP) and in compliance with federal and state regulations. Prior to being acquired by us, Koligo treated approximately 40 patients with KYSLECEL at six U.S. hospitals through a commercial pilot program.

Tissue Genesis Icellator® for Cell Assisted Lipotransfer

The Tissue Genesis Icellator is a point-of-care medical device that isolates stromal and vascular fraction cells (“SVF”) from a patient’s own (autologous) adipose tissue (fat). The Icellator is commercially available in Korea through a medical device distributor. The SVF obtained from the Icellator is for use in cell-assisted lipotransfer, a plastic surgery procedure intended to improve fat engraftments.

7

It is expected that the Icellator may also become commercially available in Japan in 2021 for use in cell assisted lipotransfer, pending review and approval by the Japanese Pharmaceuticals and Medical Devices Agency.

Cartil-S Autologous Products for the Treatment of Osteoarthritis

Cartil-S is a cell therapy for Osteoarthritis. This product is produced by performing a minimally invasive biopsy of adipose (fat) tissue from a patient, followed by isolation and expansion of adipose-derived stem cells (ADSCs), to be injected arthroscopically. The autologous injectable product helps delay/stop the progression of osteoarthritis, involving the patient’s own stem cells.

Chondroseal Autologous Products for the Treatment of Cartilage Defects (Osteoarthritis)

Chondroseal is a cell therapy for cartilage defects. Following collection of adipose tissue by minimally invasive biopsy that is composed of ADSCs, the cells are combined with a natural gel serving as a scaffold for local cartilage regeneration in the joint.

Products in Clinical Development

The following chart depicts our therapeutic development pipeline.


Products in Clinical Trials

RanTop, Ranpirnase Topical Formulation

We are currently developing a novel topical formulation of an active RNA-degrading enzyme, called Ranpirnase. Ranpirnase combats viral infections by targeting double-stranded RNA including miRNA precursors, via RNA degradation catalysis. It acts through a dual mechanism: 1) Inhibition of viral replication; and 2) induction of host cell apoptosis. Ranpirnase was previously developed for the treatment of human papillomavirus (HPV)-related pathologies such as external genital warts (EGW) and anal dysplasia. It has demonstrated clinical efficacy and good tolerability in a Phase IIa clinical study for the treatment of HPV-associated EGW. The initiation of a clinical Phase IIb for EGW is planned for 2021.

Tissue Geneseis Icellator® for Erectile Dysfunction and COVID-19 (SVF-CLI-ED)

8

The safety of the Tissue Genesis Icellator, and use of SVF produced by the Icellator, has previously been tested in a number of pilot clinical trials in the United States. Orgenesis has prioritized the clinical development of the Icellator for potential use in the treatment of erectile dysfunction and COVID-19 related respiratory complications. Pending review and approval of the FDA of the clinical trials, we expect to start a phase 2 trial in erectile dysfunction and a phase 1 trial for COVID -19 in 2021.

The Tissue Genesis Icellator is also being used by research collaborators in FDA-regulated clinical trials to test the use of SVF during rotator cuff surgery. These trials are investigator sponsored initiatives that Orgenesis will continue to support.

Products in IND Enabling Studies

We are engaged in the following IND-enabling studies:

Generation of Autologous Insulin-Producing Cells (AIPs) from Adult Liver Cells (“Trans-differentiation technology”)

Orgenesis Ltd. has trans-differentiation in-vitro technology demonstratesthat has demonstrated in animal models the capacity to induce a shift in the developmental fate of cells from the liver or other tissues, and transdifferentiating them into “pancreatic beta cell-like” Autologous Insulin Producing (“AIP”)AIP cells for patients with Type 1 Diabetes (“T1D”), acute pancreatitis and other insulin deficient diseases. For the treatment of diabetes, cells are derived from the liver or other adult tissue and are trans-differentiated to become AIP cells. This technology, which has yet to be proven in human clinical trials, has shown in pre-clinicalrelevant animal models that the human derived AIP cells produce insulin in a glucose-sensitive manner. No adverse effects were observed in any of the animal studies. This trans-differentiation technology is licensed by ourthe Israeli Subsidiary and is based on the work of Prof. Sarah Ferber, our Chief Scientific Officer and a researcher at Tel Hashomer Medical Research Infrastructure and Services Ltd. (“THM”) in Israel. OurThe development plan calls for conducting additional pre-clinical safety and efficacy studies with respect to diabetes and other potential indications prior to initiating human clinical trials.

With respect to thisthe trans-differentiation technology, we own or have exclusive rights to ten (10)seven (7) United States and nineteen (19)twelve (12) foreign issued patents, nine (9)five (5) pending patent applications in the United States, thirty-two (32)twenty four (24) pending patent applications in foreign jurisdictions, including, Europe, Australia, Brazil, Canada, China, Eurasia,Europe, India, Israel, Japan,Panama, South Korea, Mexico, and Singapore, and four (4) international Patent Cooperation Treaty (“PCT”) patent applications.Singapore. These patents and patent applications relate, among others, to (1) the trans-differentiation of cells (including hepatic cells) to cells having pancreatic β-cell-like phenotype and function and to their use in the treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis, and (2) scaffolds, including alginate and sulfated alginate scaffolds, polysaccharides thereof, and scaffoldspancreatitis.

On June 11, 2019, the FDA granted Orphan Drug Designation for useour AIP cells as a cell replacement therapy for cell propagation, transdifferentiation, and transplantation in the treatment of autoimmune diseases, including diabetes.

-4-

Our CDMO platform operates through Masthercell Global, which currently consistssevere hypoglycemia-prone diabetes resulting from total pancreatectomy (“TP”) due to chronic pancreatitis. The incidence of diabetes following TP is 100%, resulting in immediate and lifelong insulin-dependence with the loss of both endogenous insulin secretion and that of the following subsidiaries: MaSTherCellcounter-regulatory hormone, glucagon. Glycemic control after TP is notoriously difficult with conventional insulin therapy due to complete insulin dependence and loss of glucagon-dependent counter-regulation. Patients with this condition experience both severe hyperglycemic and hypoglycemic episodes.

On April 29, 2019, we received Institutional Review Board (“IRB”) approval to collect liver biopsies from patients at Rambam Medical Center located in Belgium, AtvioHaifa, Israel for a planned study to confirm the suitability of liver cells for personalized cell replacement therapy for patients with insulin-dependent diabetes resulting from total or partial pancreatectomy. The first patients were enrolled during 2020. The goal of the proposed study, entitled “Collection of Human Liver Biopsy and Whole Blood Samples from Type 1 Diabetes Mellitus (T1DM), Total or Partial Pancreatectomy Patients for Potential use as an Autologous Source for Insulin Producing Cells in IsraelFuture Clinical Studies,” is to confirm the suitability of the liver cells for personalized cell replacement therapy, as well as eligibility of patients to participate in a future clinical study, as defined by successful AIP cell production from their own liver biopsy. The secondary objective of the study is to evaluate patients’ immune response to AIPs based on the patient’s blood samples and CureCellfollowed by subcutaneous implantation into the patients’ arm which would represent the first human trial.

9

The trans-differentiation technology is from a licensing agreement entered into as of February 2, 2012 by the Israeli Subsidiary and THM pursuant to which the Israeli Subsidiary, Orgenesis Ltd, was granted a worldwide royalty bearing and exclusive license (the “THM License Agreement”). By using therapeutic agents that efficiently convert a sub-population of liver cells into pancreatic islets phenotype and function, this approach allows the diabetic patient to be the donor of his own therapeutic tissue. While we believe that this provides a major competitive advantage to the cell transformation technology of the Israeli Subsidiary, we also believe that our expanded focus to other therapies and business activities may continue to prompt THM to inquire of such activities as they may relate to our compliance with the terms or direction in South Korearegards to the THM License Agreement. While we have not received any notice of cancellation of the THM License Agreement, we have received an allegation regarding the scope of the rights by THM that may present future challenges for our Israeli Subsidiary to continue to develop, manufacture, sell and Masthercell U.S., LLCmarket the products pursuant to the milestones and time schedule specified in the United States, each having unique know-how and expertise for manufacturing in a multitude of cell types. As part of our United States (“U.S.”) activity, we intend to also set up a CDMO facility in the United States. We believe that, in-order to provide the optimal service to our customers, we need to have a global presence. We target the international market as a key priority through our network of facilities that provide development manufacturing and logistics services, utilizing our advanced quality management system and experienced staff. All of these capabilities offered to third-parties are utilized for our internal development projects, with the goal of allowing us to be able to bring new products to patients faster and in a more cost-effective way. Masthercell Global strives to provide services that are all compliant with Good Manufacturing Practice, or GMP, requirements, ensuring identity, purity, stability, potency and robustness of cell therapy products for clinical phase I, II, III and through commercialization.

We operate our CDMO and the PT platforms as two separate business segments.

Overview for Advanced Therapy Medicinal Products (ATMPs)

Advanced Therapy Medicinal Product (“ATMP”) means anyplan of the following medicinal products for human use:THM License Agreement.

·a somatic cell therapy medicinal product (“STMP”);
·a tissue engineered product (“TEP”);
·a gene therapy medicinal product (“GTMP”); or
·a combined ATMP.

An STMP containsORG-CAR19, Autologous CD-19 CAR-T

Chimeric antigen receptor T cells or tissues(also known as CAR-T cells) are T cells that have been manipulatedgenetically engineered to change their biological characteristics orproduce an artificial T-cell receptor for use in immunotherapy. CAR-T cell therapy uses T cells or tissues not intendedengineered with CARs for cancer therapy. The premise of CAR-T immunotherapy is to be used for the same essential functions in the body. A TEP containsmodify T cells or tissues that have been modified so they can be used to repair, regenerate or replace human tissue. A GTMP contains genes that lead to a therapeutic, prophylactic or diagnostic effect and work by inserting “recombinant” genes into the body, usually to treat a variety of diseases, including genetic disorders,recognize cancer or long-term diseases. A recombinant gene is a stretch of DNA that is created in the laboratory, bringing together DNA from different sources. Combined ATMPs contain one or more medical devices as an integral part of the medicine, such as cells embedded in a biodegradable matrix or scaffold. Although STMPs and GTMPs currently dominate the market, in order to accessmore effectively target and destroy them. Physicians harvest T cells from patients, genetically alter them, then infuse the market potential and trends in the future, other cell products are likelyresulting CAR-T cells into patients to attack their tumors. CAR-T cells can be essential in all these categories.

Furthermore, we believe that autologous therapies will be a substantial segment of the ATMP market. Autologous therapies are produced from a patients’ own cells, instead of mass-cultivated donor-cells, or allogeneic cells. Allogeneic therapies areeither derived from donorT cells and, through the construction of master and working cell banks, are produced onin a large scale. Autologous therapies arepatient’s own blood (autologous) or derived from the treated patient and manufactured throughT cells of another healthy donor (allogeneic). Once isolated from a defined protocol before re-administration and generally demand a more complex supply chain. Currently with the ATMP network relying heavily on production and supply chain of manufacturing sites, we believe our POCare model may help overcome some of the development and supply challenges with bringingperson, these therapies to patients.

-5-

CDMO Business

Companies developing cell therapies need to make a decision early on in their approach to the transition from the lab to the clinic regarding the process development and manufacturing of theT cells necessary for their respective therapeutic treatments. Of the companies active in this market, only a small number have developed their own GMP manufacturing facilities due to the high costs and expertise required to develop these processes. In addition to the limitations imposed by a limited number of trained personnel and high infrastructure/operational costs, the industry faces a need for custom innovative process development and manufacturing solutions. Due to the complexity and diversity of the industry, such solutions are often customized to the particular needs of a company and, accordingly, a multidisciplinary team of engineers, cell therapy experts, cGMP and regulatory experts is required. Such a unique group of experts can exist only in an organization that both specializes in developing characterization assays and solutions and manufactures cell therapies.

Companies can establish their own process and GMP manufacturing facility or engage a contract manufacturing organization for each step. A CDMO is an entity that serves other companies in the pharmaceutical industry on a contract basis to provide comprehensive services from cell therapy development through cell therapy manufacturing for and end-to-end solution. Due to the complexity, global outreach needs, redundancy and operational costs of manufacturing biologics and cell therapies, the CDMO business is expanding. With more than 861 companies in the field of cell therapy worldwide (versus 580 in 2015) and 959 clinical trials underway by the end of the first quarter of 2018 (versus 486 in first quarter of 2015), we believe that the industry shows a rapid growth rate accompanied by a lack of sufficient GMP manufacturing capacities (Source:Informa, 2015 and 2018). Over recent years, advances in the field of cell therapy, including the growth of autologous CAR T-cell therapies, led to a significant increase in investment in the industry. T-cells, the backbone of CAR T-cell therapy, are often called the workhorses of the immune system because of their critical role in orchestrating the immune response and killing cells infected by pathogens. The therapy requires drawing blood from patients and separating out the T-cells. Next, using a disarmed virus, the T-cells are genetically engineered to produce receptorsexpress a specific CAR, which programs them to target an antigen that is present on the surface of tumors. After CAR-T cells are infused into a patient, they act as a “living drug” against cancer cells. When they come in contact with their targeted antigen on a cell, CAR-T cells bind to it and become activated, then proceed to proliferate and become cytotoxic.

We are developing a new and advanced anti-CD19 CAR-T therapy for treating B-cell Acute lymphoblastic leukemia (B-ALL) and lymphoma patients, based on a clinically used CAR-T therapy licensed from Kecellitics Biotech. B-ALL is driven by malignant B-cell, expressing the B-cell surface called Chimeric Antigen Receptors, or CARs. protein, CD19. Orgenesis is also working on combining the CD19 with CD22 CAR on a single, bi-specific CD19/22 CAR-T that can target both antigens simultaneously for the treatment of blood cancers.

Dual Cellular vaccine (DUVAC), Therapy for Pancreatic Cancer

The genetically modified T-cells thatDUVAC cell-based immunotherapy, licensed from Columbia University, is based on autologous dendritic cells and macrophages. These cells are re-injected intokey coordinators of the innate and adaptive immune system and have critical roles in the induction of antitumor immunity. The cells are exposed to whole cancer cells constitute the most comprehensive source of cancer antigens and by so boosting the patient are then much more effective at targetingimmune system and killing tumors.

Two landmark U.S. Food Drug Administration (the “FDA”) approvals in CAR T-cell therapy significantly impacteddirect it against the cell therapy industry. In August 2017, Novartis’s CAR T-cell therapy, Kymriah, was approvedtumor. The DUVAC vaccine can be a developed for relapsed/refractory acute lymphoblastic leukemia for pediatric and young adult patients, making it the first cell-base immunotherapy to move across the finish line in the United States. Kymriah received a second FDA approval to treat appropriate relapsed/refractory patients with large B-cell lymphoma in May 2018. Europe has also followed this path as, in August 2018, the European Commission approved Kymriah based on the first global CAR-T registration trials, which included patients from eight European countries and demonstrated durable responses and a consistent safety profile in relapsed/refractory pediatric B-cell ALL and r/r DLBCL. Furthermore, after Gilead's acquisition of Kite Pharma, Inc. for $12 billion in 2017, Kite Pharma’s CAR T-cell therapy, Yescarta, was approved for adult patients with relapsed/refractory large B cell lymphoma after two or more lines of systemic therapy (Source: Alliance for Regenerative Medicine). We believe that these approvals are indicative of the future potential of many more cellular therapies that address a wide range of diseases. Celgene Corporation (“Celgene”) acquired Junosolid tumor, but our initial focus is on pancreatic cancer.

Metabolically Optimized T-Cells (MOTC): Therapy for melanoma and lung cancer

In the early stages of cancer, some lymphocytes successfully attack and infiltrate the tumor microenvironment, surround the tumor cells, and mount an anti-tumor response. TIL therapy is a clinically validated personalized cancer treatment based on infusion of autologous TILs expanded ex vivo from tumors. Once expanded, the TILs are infused back into the patient where they attack the cancer cells with a high degree of specificity. Orgenesis is developing an advanced cellular biomanufacturing platform integrated with metabolic control. The expanded TILs possess an optimized metabolic state referred to as MOTC (Metabolically Optimized T-Cells), which can potentially lead to a more robust therapeutic response, especially for solid tumors such as lung cancer and melanoma.

10

Products in Pre-Clinical Studies

CAR-NK, Therapy for Solid Tumors

We have licensed from Caerus Therapeutics Inc., another pioneer(a related party) a unique CAR platform that contains additional immunological effectors that aim to address significant challenges emerged in the CAR-T space, in January 2018development process of CAR technologies such as: safety, viability, immunosuppression by tumor microenvironment and dense desmoplastic stroma. Orgenesis aims target this CAR platform Natural Killer Cells (CAR-NK) platform for approximately $9 billion. Then, Bristol-Myers Squibb Company, in their pursuitthe treatment of this new spacesolid tumors.

Autologous Cell-Based Vaccine for protecting against SARS-CoV-2

We are working on developing a cell-based vaccine platform for the prevention of cancer treatments, acquired Celgene in January 2019viral diseases. The initial target for approximately $74 billion.

the platform is SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2, the causative agent of COVID-19). This cell-based vaccine platform utilizes an autologous approach. The complexitygoal is to enable the COVID-19 engineered cells will have the ability to activate an endogenous immune response and induce the production of manufacturing individual cell therapy treatments poses a fundamental challenge for cell therapy-based companies as they enter the field. This complexity potentially casts a spotlight on improved cGMP, large-scale manufacturing processes, such as the services provided by Masthercell Global. Manufacturing and delivery can be more complex in cell therapy products than for a typical drug. In the U.S., only a few dozen specialized hospitals are currently qualified to provide CAR T treatments, which require retrieving, processing and then returning immune cells to the patient, all done under strict cGMP,neutralizing antibodies as well as monitoringcellular response.

Bioxomes

Exosomes are small, membrane-enclosed extracellular vesicles implicated in cell-to-cell communication. Exosomes may serve as a valuable therapeutic modality because of their ability to transfer a wide variety of therapeutic payloads among cells that can influence a cell in multiple ways, and they can be designed to reach specific cell types. We are developing a proprietary manufacturing process for exosome like structures, termed Bioxomes. Bioxomes can carry specific therapeutic cargo into target cells. Orgenesis is developing this platform technology to treat liver fibrosis, dermatology, and other indications.

MSCP

Orgenesis is developing a personalized cell-based therapy product for wound healing and psoriasis. The product is based on Adipose-Derived Stem Cells (ADSCs). Following expansion, the ADSCs are formulated with Topiramate, a well-known substrate used in other indications, and a commercially available hyaluronic acid (HA), a well-known dermal filler, for topical treatment.

Muscle-derived Mesenchymal Stem Cells for Human Regenerative Medicine

An innovative and patented technology licensed by Revatis that enables the isolation of pluripotent adult Mesenchymal Stem Cells (MSCs) from a minimally invasive muscle micro-biopsy. The isolated autologously undifferentiated muscle-derived MSCs are developed to explore autologous therapeutics fields in humans such as Urine Incontinence

Kidney Disease

We are also developing multiple Proprietary cell and cell derived products therapies for treating side effects. These factorskidney failure and End-Stage Renal Disease (ESRD).

KT-DM-103 and KT-CP-203 (3D-Printed Pancreatic Islets)

Koligo had exclusively licensed patents and technology from the University of Louisville Research Foundation related to the revascularization and 3D printing of cell and tissue for transplant (“3D-V” technology platform). Orgenesis is developing this technology for potential autologous and allogeneic pancreatic islet transplant to treat type 1 diabetes (KT-DM-103) and chronic pancreatitis (KT-CP-203). The 3D-V technology platform may also support improved transplantation of other cell and tissue types in additional to pancreatic islets.

11

POCare Platform Strategy

Our aim is to provide real incentivesa pathway to bring ATMPs in the cell and gene therapy industry from research to patients worldwide through our POCare Platform. We define point of care as a process of collecting, processing, and administering cells within the patient care environment, namely through academic partnerships in a hospital setting. We believe that this approach is an attractive proposition for personalized medicine because of our strategic partnerships with suppliers that help us to customize closed systems into effective mobile clean room facilities. This will potentially help to minimize or eliminate the need for cell therapy companiestransportation, which is a high-risk and costly aspect of the supply chain.

We aim to seek third-party partners, or contract manufacturers, who possess technical, manufacturing,build value in various aspects of our company ranging from supply related processes including development and distribution systems, clinical and regulatory expertise in cell therapy developmentservices, engineering and manufacturingdevices such as cell therapy CDMOs like MaSTherCell. Additionally, establishing a manufacturing facility for cell therapy requires specific expertise and significant capital which can delay the clinical trials by at least 2 years. As companies are looking to expedite their market approval, utilization of a CDMO can result in faster time to market and overall lower expenditure.

-6-

Integration of development and manufacturing and logistics services through Masthercell Global (and its subsidiaries) provide the basis for generating a recurring revenue stream,OMPULs discussed below, delivery systems, therapies including immuno-oncology, anti-aging, anti-viral, metabolic, nephrology, dermatology, orthopedic, as well as carefully managing our fixed cost structureregenerative technologies.

Over time, we have worked to maximize optionalitydevelop and drive downvalidate POCare Technologies that can be combined within mobile production cost. We believe that Masthercell Global is also beneficialunits for our own manufacturing needs and provides us, and our customers, with enhanced control of material supply for both clinical trials and the commercial market.

Consolidation of CDMO Entities and Strategic Funding

On June 28, 2018, the Company, Masthercell Global, Great Point Partners, LLC, a manager of private equity funds focused on growing small to medium sized heath care companies (“Great Point”), and certain of Great Point’s affiliates, entered into a series of definitive strategic agreements intended to finance, strengthen and expand Orgenesis’ CDMO business.advanced therapies. In connection therewith, the Company, Masthercell Global and GPP-II Masthercell, LLC, a Delaware limited liability company (“GPP-II”) and an affiliate of Great Point, entered into a Stock Purchase Agreement (the “SPA”) pursuant to which GPP-II purchased 378,000 shares of newly designated Series A Preferred Stock of Masthercell Global (the “Masthercell Global Preferred Stock”), representing 37.8% of the issued and outstanding share capital of Masthercell Global, for cash consideration to be paid into Masthercell Global of up to $25 million, subject to certain adjustments (the “Consideration”). Orgenesis holds 622,000 shares of Masthercell Global’s Common Stock, representing 62.2% of the issued and outstanding equity share capital of Masthercell Global. An initial cash payment of $11.8 million of the Consideration was remitted at closing by GPP-II, with a follow up payment of $6,600,000 to be2020, we made in each of years 2018 and 2019 (the “Future Payments”), or an aggregate of $13.2 million, if (a) Masthercell Global achieves specified EBITDA and revenues targets during each of these years, and (b) the Orgenesis’ shareholders approve certain provisions of the Stockholders’ Agreement referred to below on or before December 31, 2019. None of the future Consideration amounts, if any, will result in an increase in GPP-II’s equity holdings in Masthercell Global beyond the 378,000 shares of Series A Preferred Stock issued to GPP-II at closing. The proceeds of the investment will be used to fund the activities of Masthercell Global and its consolidated subsidiaries. Notwithstanding the foregoing, GPP-II may, in its sole discretion, elect to pay all or a portion of the future Consideration amounts even if the financial targets described above have not been achieved and the Orgenesis Stockholder Approval has not been obtained. In satisfaction of the two conditions described above, Masthercell Global achieved the specified EBITDA and revenues targets in 2018 as describedsignificant investments in the SPAdevelopment of several types of Orgenesis Mobile Processing Units and obtained the approval from its requisite shareholders on October 23, 2018. As such, Masthercell Global received the First Future Payment of $6,600,000 on January 16, 2019.

In connectionLabs (OMPULs) with the entry into the SPA,expectation of use and/or distribution through our POCare Network of partners, collaborators, and pursuant to the terms hereof, described above, each of the Company, Masthercell Global and GPP-II entered into the Masthercell Global Inc. Stockholders’ Agreement (the “Stockholders’ Agreement”) providing for certain restrictions on the disposition of Masthercell Global securities, the provisions of certain options and rights with respect to the management and operations of Masthercell Global, certain favorable, preferential rights to GPP-II (including, without limitation, a tag right, drag right and certain protective provisions), a right to exchange the Masthercell Global Preferred Stock for shares of Orgenesis common stock and certain other rights and obligations. joint ventures.

In addition, after the earlier of the second anniversary of the closing or certain enumerated circumstances, GPP-II is entitled to effectuate a spinoff of Masthercell Global and the Masthercell Global Subsidiaries (the “Spinoff”). The Spinoff is required to reflect a market value determined by one of the top ten independent accounting firms2020 we made significant investments in the U.S. selected by GPP, provided that under certain conditions, such market valuation shall reflect a valuationdevelopment of Masthercell Globalseveral types of OMPULs and the Masthercell Global Subsidiaries of at least $50 million. In addition, upon certain enumerated events as described below, GPP-II is entitled, at its option, to put to the Company (or, at Company’s discretion, to Masthercell Global if Masthercell Global shall then have the funds available to consummate the transaction) its shares in Masthercell Global or, alternatively, purchase from the Company its share capital in Masthercell Global at a purchase price equal to the fair market value of such equity holdings as determined by one of the top ten independent accounting firmsmade significant progress in the U.S. selected by GPP-II, provided that the purchase price shall not be greater than three times the price per share of Masthercell Global Preferred Stock paid by GPP-IIvalidation, risk analysis, regulatory and shall not be less than the price per share of Masthercell Global Preferred Stock paid by GPP-II. GPP-II may exercise its put or call option upon the occurrence of any of the following: (i) there is an Activist Shareholder of the Company; (ii) the Chief Executive Officer and/or Chairman of the board of directors of the Company resigns or is replaced, removed, or terminated for any reason prior to June 28, 2023; (iii) there is a Change of Control event of the Company; or (iv) the industry expert director appointed to the board of directors of Masthercell Global is removed or replaced (or a new such director is appointed) without the prior written consent of GPP-II. For the purposes of the foregoing, the following definitions shall apply: (A) “Activist Shareholder” shall mean any Person who acquires shares of capital stock of the Company who either: (x) acquires more than a majority of the voting power of the Company, (y) actively takes over and controls a majority of the board of directors of the Company, or (z) is required to file a Schedule 13D with respect to such Person’s ownership of the Company and has described a plan, proposal or intent to take action with respect to exerting significant pressure on the management of or directors of, the Company; and (B) “Change of Control” shall mean any of: (a) the acquisition, directly or indirectly (in a single transaction or a series ofother related transactions) by a Person or group of Persons of either (I) a majority of the common stock of the Company (whether by merger, consolidation, stock purchase, tender offer, reorganization, recapitalization or otherwise), or (II) all or substantially all of the assets of the Company and its Subsidiaries (but only if such transaction includes the transfer of Securities held by the Company), (b) if any four (4) of the directors of the Company as of June 28, 2018 are removed or replaced or for any other reason cease to serve as directors of the Company, (c) the filing of a petition in bankruptcy or the commencement of any proceedings under bankruptcy laws by or against the Company, provided that such filing or commencement shall be deemed a Change of Control immediately if filed or commenced by the Company or after sixty (60) days if such filing is initiated by a creditor of the Company and is not dismissed; (d) insolvency of the Company that is not cured by the Company within thirty (30) days; (e) the appointment of a receiver for the Company, provided that such appointment shall constitute an Change of Control immediately if the appointment was consented to by the Company or after sixty (60) days if not consented to by the Company and such appointment is not terminated; or (f) or dissolution of the Company.

-7-

The Stockholders’ Agreement further provides that GPP-II is entitled, at any time, to convert its share capital in Masthercell Global for the Company’s common stock in an amount equal to the lesser of (a)(i) the fair market value of GPP-II’s shares of Masthercell Global Preferred Stock to be exchanged, as determined by one of the top ten independent accounting firms in the U.S. selected by GPP-II and the Company, divided by (ii) the average closing price per share of Orgenesis Common Stock during the thirty (30) day period ending on the date that GPP-II provides the exchange notice (the “Exchange Price”) and (b)(i) the fair market value of GPP-II’s shares of Masthercell Global Preferred Stock to be exchanged assuming a value of Masthercell Global equal to three and a half (3.5) times the revenue of Masthercell Global during the last twelve (12) complete calendar months immediately prior to the exchange divided by (ii) the Exchange Price; provided, that in no event will (A) the Exchange Price be less than a price per share that would result in Orgenesis having an enterprise value of less than $250,000,000 and (B) the maximum number of shares of Orgenesis Common Stock to be issued shall not exceed 2,704,247 shares of outstanding Orgenesis Common Stock (representing approximately 19.99% of then outstanding Orgenesis Common Stock), unless Orgenesis obtains shareholder approval for the issuance of such greater amount of shares of Orgenesis Common Stock in accordance with the rules and regulations of the Nasdaq Stock Market. Such shareholder approval for a greater number was obtained on October 23, 2018.

Great Point and Masthercell Global entered into an advisory services agreement pursuant to which Great Point is to provide management services to Masthercell Global for which Great Point will be compensated at an annual base compensation equal to the greater of (i) $250,000 per each 12 month period or (ii) 5% of the EBITDA for such 12 month period, payable in arrears in quarterly installments; provided, that these payments will (A) begin to accrue immediately, but shall not be paid in cash to Great Point until such time as Masthercell Global generates EBITDA of at least $2,000,000 for any 12 month period or the sale of or change in control of Masthercell Global, and (B) shall not exceed an aggregate annual amount of $500,000. Such compensation accrues but is not owed to Great Point until the earlier of (i) Masthercell Global generating EBITDA of at least $2 million for any 12 months period following the date of the agreement or (ii) a Sale of the Company or Change of Control of the Company (as both terms are defined therein).

GPP Securities, LLC, a Delaware limited liability company and an affiliate of Great Point and Masthercell Global entered into a transaction services agreement pursuant to which GPP Securities, LLC is to provide certain brokerage services to Masthercell Global for which GPP Securities LLC will be entitled to a certain Exit Fee and Transaction Fee (as both terms are defined in the agreement), such fees not to be less than 2% of the applicable transaction value.

Corporate Reorganization

Contemporaneous with the execution of the SPA and the Stockholders’ Agreement, Orgenesis and Masthercell Global entered into a Contribution, Assignment and Assumption Agreement pursuant to which Orgenesis contributed to Masthercell Global the Orgenesis’ assetstasks relating to the CDMO Business (as defined below), includingOMPULS. We anticipate distributing and using the CDMO subsidiaries (the “Corporate Reorganization”). In furtherance thereof, Masthercell Global, as Orgenesis’ assignee, acquired allOMPULS through our POCare Network of the issuedpartners, collaborators, and outstanding share capital of Atvio, the Company’s Israel based CDMO partner since May 2016, and 94.12% of the share capital of CureCell, the Company’s Korea based CDMO partner since March 2016. Orgenesis exercised the "call option" to which it was entitled under the joint venture agreements with each of these entities to purchase from the former shareholders their equity holding. The considerationventures. OMPULs are designed for the outstanding share equity in eachpurpose of Atvio and CureCell consisted solelyvalidation, development, performance of Orgenesis common stock. In respectclinical trials, manufacturing and/or processing of the acquisition of Atvio, Orgenesis issued to the former Atvio shareholders an aggregate of 83,965 shares of Orgenesis common stock. In respect of the acquisition of CureCell, Orgenesis issued to the former CureCell shareholders an aggregate of 202,846 shares of Orgenesis common stock subject to a third-party valuation. Together with MaSTherCell S.A., Atvio and CureCell are directly held subsidiaries under Masthercell Global (collectively, the “Masthercell Global Subsidiaries”).

-8-

Masthercell Global, through the Masthercell Global Subsidiaries, will be engaged in the business of providing manufacturing and development services to third parties related topotential or approved cell and gene therapy products processes and solutions and providing related manufacturing or development services, and the creation and development of technology, intellectual property, tools and optimizations in connection with such manufacturing and development services for third parties (the “CDMO Business”). Under the terms of the Stockholders’ Agreement and SPA, Orgenesis has agreed that so long as it owns equity in Masthercell Global and for two years thereafter it will not engage in the CDMO Business, except through Masthercell Global. Notwithstanding the foregoing, nothing in the Stockholders’ Agreement or the SPA prohibits or restricts the Company’s ability to conduct any business outside the CDMO Business and the Company retained the right to research, manufacture, develop and conduct all other activities related to the development, discovery, manufacturing and commercialization of therapeutic products, and the process, methods and services thereof (including, without limitation, such therapeutic products for itself and in which the Company has an economic interest or any relationship with any Third Party in which the Company has an economic interest or that are created, developed, manufactured or sold by a joint venture, partnership or collaboration between the Company and a Third Party) with a Third Party. For purposes hereof, the term “Third Party” shall mean any entity (other than our Company or our subsidiaries) with whom we (or our subsidiaries) has a collaboration, joint venture, partnership or similar economic relationship for the development of a product with therapeutic use where the primary purpose of such collaboration, joint venture, partnership or relationship is not the manufacturing related to such product. We intend, through our direct subsidiaries, to continue engaging in such research, marketing, development, selling and commercialization of such therapeutic products either for our own internal purposes or with Third Parties.

Masthercell Global’s Business

Our subsidiary, Masthercell Global, is a CDMO specialized in cell therapy development for advanced therapeutically products. In the last decade, cell therapy medicinal products have gained significant importance, particularly in the fields of ex-vivo gene therapy and immunotherapy. While academic and industrial research has led scientific development in the sector, industrialization and manufacturing expertise remains insufficient. Masthercell Global plans to fill this gap by providing three types of services to its customers: (i) process and assay development services and (ii) current Good Manufacturing Practices (cGMP) contract manufacturing services and (iii) technology innovation and engineering services. These services offer a double advantage to Masthercell Global’s customers. First, customers can continue allocating their financial and human resources on their product/therapy, while relying on a long-termsafe, reliable, and trusted partner for their process development/production. Second, through its subsidiaries, it allows customers to benefit from Masthercell Global’s expertise in cell therapy manufacturing and all related aspects.

Masthercell Global continues to invest in its manufacturing capabilities and services to offer a “one-stop-shop” service to its customers from pre-clinical up to commercial development. MaSTherCell S.A., Masthercell Global’s Belgian subsidiary, has recently inaugurated a new production center in Gosselies, doubling its manufacturing capacity from 600 sqm GMP area to 1,200 sqm. This new facility can also accommodate commercial manufacturing. Masthercell Global’s Israeli-based CDMO, Atvio, has relocated its process engineering activities into a new and larger facility locatedcost-effective manner at Bar-Lev industrial park. These subsidiaries, including Masthercell Global’s Korea-based CDMO, have started to offer viral vector CDMO services. Our target customers are primarily cell therapy companies that are in clinical trials with the aimpoint of accompanying them as their manufacturing and logistic partner once their product candidates reach commercial stage.

-9-

We devote significant resources to process development and manufacturing in order to optimize the safety and efficacy of our future product candidates for our customers,care, as well as the manufacturing of such CGTs in a consistent and standardized manner in all locations. The design delivers a potential industrial solution for us to deliver CGTs to most clinical institutions at the point of care.

A picture containing graphical user interface

Description automatically generated

*For illustrative purposes only

Currently, we are finalizing the development over 30 OMPUL-based POC processing locations worldwide and, with the assistance of our partners, we are adapting the local requirements of each partner with the target of achieving a capacity to process and supply CGTs to as many as 2,000 patients annually. The responsibility for setting up the OMPULS falls on the joint venture partners, who are also responsible for marketing and distribution worldwide. As we expand operations, the OMPUL setup cost is expected to decline proportionately. Most of goodsour POC revenue to date is in support of the implementation of our technologies and time to market. This integration of development, manufacturing and logistics services through Masthercell Global aims to providetherapies in our partners’ POC activities, which will be the basis for generatingfuture royalties and supply revenues.

We have embarked on a recurring revenue stream, as well as carefully managing our fixed cost structure, maximize optionality, and drive long-term coststrategy of goods as low as possible.collaborative arrangements with strategically situated regional joint venture partners around the world. We believe that operatingthese parties have the expertise, experience and strategic location to advance our own manufacturing facility provides us with enhanced controlPOCare Platform.

12

Strategic CGT Therapeutics Collaborations

Collaborations, partnerships, joint ventures and license agreements are a key component of material supply for both clinical trialsour POC strategy.

Our POC technology collaborators and the commercial market, will enable the more rapid implementation of process changes, and will allow for better long-term margins.

Masthercell Global continues to invest resources to maintain best practices in quality service, quality control, quality assurance and permanent staff training to uphold the highest standards to serve its customers. Masthercell Global (through itself and its subsidiaries) has built-up a team of more than 140 industry experts in Belgium, 30 experts in Korea, 19 experts in Israel and 3 peoplepartners include Mircod, , Cure Therapeutics, Columbia University in the US.City of New York, Caerus Therapeutics Corporation, Sescom Ltd., UC Davis, SBH Sciences, Inc., The entire team is dedicated to support process developmentJohns Hopkins University and manufacturing effortsothers.

In addition, we have collaborations and joint ventures for setting up POCare Platform operations facilities in a fast, safejurisdictions throughout the world, including various countries in North America, Europe, Latin America, Asia, the Middle East, and cost-effective way. Masthercell Global’s strategy is to build long term relationships with its customers in order to help them bring highly potent cell therapy products fasterAustralia.

For more information, see Note 11, “Collaboration and Licensing Agreements” of the “Notes to the market andFinancial Statements” included in cost-effective ways. To provide these services,Item 8.

CDMO Business

Regarding the Masthercell Global relies on a teamSale, see Note 3 to Item 8 of dedicated experts both from academic and industry backgrounds. It operates through state-of-the-art facilities organized as a global network in Belgium, Israel, Korea and soon in the U.S. This network of facilities operates on a common Quality Management System backbone enabling for streamlined technology transfers among the different sites.this Annual Report.

Our Growth StrategyCurrent Development Facilities

In light of the globalization of the industry in general and the therapeutics industry in particular, adding to that the high cost of reaching the market, developers of cell therapies see themselves as global organizations and build their models on global markets. As cell therapies are based on living cells, they are limited in their ability to be centrally manufactured. An additional challenge for globalization is the fact that the regulatory requirements for the therapies is not harmonized between jurisdictions, presenting additional operational challenges.OBI

We have leveraged the recognized quality expertise and experience in cell process development and manufacturing of our Belgian subsidiary, MaSTherCell S.A., to first-class entities in Israel and Korea and to build a global CDMO in the cell therapy development and manufacturing area. We believe that cell therapy companies need to be global in order to truly succeed. We target the international manufacturing market as a key priority through joint-venture agreements that provide development capabilities, along with manufacturing facilities and experienced staff.

The main revenue drivers of our growth strategy on a global reach are the number of batches and the number of patients per manufacturing batch. These parameters vary along the development cycle of the new treatments (starting from as few as 20 patients in Phase I to thousands of patients when reaching commercialization). When a client reaches the commercial stage, their demand for manufacturing substantially increases, while barriers preventing the client from switching to another manufacturing organization remain extremely high. The difficulty in transferring CDMOs is a function of the tech transfer of such complex manufacturing processes being extremely lengthy, requiring many months of training highly specialized employees, while also possibly requiring new regulatory approvals. Therefore, we believe we are well positioned to continue expanding our revenue for the following reasons:

-10-

(1)A higher number of companies in later phases of clinical trials and soon potentially in commercial phases;
(2)Therapy companies requiring higher manufacturing abilities concurrent with a global reach; and
(3)An increasing need for the manufacturing scalability provided by a CDMO.

Current CDMO Facilities

MaSTherCell S.A.

MaSTherCell S.A. in Belgium is in the European hub for the continental activities of the global CDMO network and the globally-recognized center of excellence of the GMP manufacturing activities of the group. At the heart of MaSTherCell is a team of more than 140 highly dedicated experts combining strong experience in cGMP cell therapy manufacturing with a technology-focused approach and a substantial knowledge of the industry. As a one-stop shop, they provide services from technology selection, business modeling to GMP manufacturing, process development, quality management and assay development. MaSTherCell's teams are fully committed to helping their clients fulfill their objective of providing sustainable and affordable therapies to their patients. The company operates in a validated and flexible facility located in Biowin, the strategic center of Europe within the Walloon healthcare cluster. The facility in Belgium has received the final cGMP manufacturing authorization from the Belgian Drug Agency (AFMPS) in September 2013 and a renewal in October 2017 for cell-based therapies manufacturing. It spreads over 2,000 sqm including 1,200 sqm of GMP area.

Atvio Biotech Ltd.

Atvio Biotech Ltd. in IsraelOBI is a specialized process and technology development firmwholly owned subsidiary focused on custom-made process development, upscaling design from lab to industry innovation and automation procedures, which are extremely essential in the cell therapy industry. AtvioOBI is located in Bar-Lev Industrial Park utilizing the exclusive Israeli innovative ecosystem and highly experienced and talented associates including Ph.D. holders and biotechnology engineers. The center provides end to end solutions to cell therapy industries,industrialization, process development capabilities and proficiency, custom-made engineering and a unique platform for creative design and process optimization. The company spreads overOBI occupies 1300 sqm2square meters of labs and offices resulting in an efficient and unique environment for cell therapy development. In connection with the Masthercell Sale, for a period of 3 years in the European Union and five years in the United States and the rest of the world from the closing date of the Masthercell Sale, we agreed that OBI will not manufacture products on a contract basis for third-party customers in any jurisdiction other than the State of Israel, but it may conduct such CDMO business in the State of Israel, solely for customers located within the State of Israel or with respect to therapies intended for distribution solely within the State of Israel.

CureCell Co. Ltd.The Korean Subsidiary

CureCell Co. Ltd. in KoreaThe Korean Subsidiary has a particular focus on developing innovative cell therapies for both the Korean and international markets.therapies. Together, with promising in-house research programs, the foreign technologies are currently under development for the rapidly growing Asian market well beyond the Korean market offering the most favorable environment for the cell therapy industry in the world. Through close collaboration with leading medical and academic facilities, CureCellthe Korean Subsidiary is accelerating the development of foreign technologies in KoreaKorea. In connection with the Masthercell Sale, for a period of 3 years in the European Union and is well positioned to expand international markets for Korean technologies.

Planned CDMO Facilities

We are currently preparing to launch a new production center in Houston, Texasfive years in the United States which we expect will become operational during fiscal 2020. We will continue to engage in discussions with other strategic partners throughoutand the rest of the world from the closing date of the Masthercell Sale, we agreed that the Korean Subsidiary will not manufacture cell and gene products on a contract basis for third-party customers in any jurisdiction other than South Korea, but it may conduct CDMO business in South Korea, solely for customers located within South Korea and with respect to set up CDMOtherapies intended for distribution solely within South Korea.

Koligo

Koligo maintains commercial production facilities for KYSLECEL at an FDA-registered establishment in other geographic locations.Indiana. The Tissue Genesis Icellators, and associated reagents and kits, are made by contract manufacturers and warehoused at our facility in Texas. Koligo also maintains development labs at the Indiana and Texas locations to support continued development.

13

Our Competitive AdvantagesThe Belgian subsidiary

We believe that we offerThe Belgian subsidiary specializes in developing and validating proprietary and licensed advanced cell and gene therapies. The subsidiary benefits both from its central position in Europe and its being in the following benefitsleading Walloon biotech cluster. Located near Namur, at Novalis Science Park, the Belgian subsidiary collaborates with leading medical and academic facilities which enables it to our CDMO clients:cover the drug product life cycle from research to clinical stage through pre-clinical and quality control. It occupies innovative facilities for the development and quality control of therapies in R&D and GMP grades.

We enable our clients to go fasterIts talented and highly experienced staff and collaborators, including Ph.D. holders, quality assurance experts and biotechnology manufacturing engineers, contribute to the market in a cost-effective way. We continue to invest in our manufacturing capabilitiesPOCare platform development and expertise to offer a “one-stop-shop” service to our customers from pre-clinical up to commercial development. This can also include preferred access to critical raw materials supplies. This stems from the finding that these companies' processes have to be set up right from the start in order for them to obtain approved products that have the simplest possible process and with the lowest possible cost of goods sold (COGS).

-11-

Quality. We work alongside our customers to transform the promises of their cell-based therapies into a robust and scalable process, compliant with GMP requirements. Our stringent quality system is applied throughout the process and ensures identity, purity, stability, potency and robustness of cell therapy products from clinical phase I, II, III to commercialization. We continue to invest resources to maintain best practices in quality service, quality control,roll-out. The subsidiary provides quality assurance and permanent staff trainingsupply activities for the global POCare network. It has developed smart and agile methodologies to upholdensure compliant and harmonized decentralization operations at POCare.

Notable 2020 Activities

On April 7, 2020, we entered into an Asset Purchase Agreement (the “Tamir Purchase Agreement”) with Tamir Biotechnology, Inc. (“Tamir” or “Seller”), pursuant to which we agreed to acquire certain assets and liabilities of Tamir related to the highest standards.

Transforming academic technologydiscovery, development and testing of therapeutic products for the treatment of diseases and conditions in humans, including all rights to clinicalranpirnase and commercial manufacturinguse for antiviral therapy (collectively, the “Purchased Assets and Assumed Liabilities” and such acquisition, the “Tamir Transaction”). OneThe Tamir Transaction closed on April 23, 2020. We paid $2.5 million in cash and issued an aggregate of 3,400,000 shares (the “Shares”) of our common stock to Tamir resulting in a total consideration of $20.2 million. In November 2020, we filed a registration statement on Form S-3 to register the resale of the major issuesShares as required by the Tamir Purchase Agreement.

On September 26, 2020, we entered into an Agreement and Plan of Merger and Reorganization (the “Merger Agreement”) with movingOrgenesis Merger Sub, Inc., a Delaware corporation and our wholly-owned subsidiary (“Merger Sub”), Koligo Therapeutics Inc., a Kentucky corporation (“Koligo”), the shareholders of Koligo (collectively, the “Shareholders”) and Long Hill Capital V, LLC, solely in its capacity as the representative, agent and attorney-in-fact of the Shareholders. The Merger Agreement provided for the acquisition of Koligo by us through the merger of Merger Sub with and into Koligo, with Koligo surviving as our wholly-owned subsidiary (the “Merger”). The Merger closed on October 15, 2020.

Koligo’s operations include (a) the manufacture and sale of KYSLECEL® (autologous pancreatic islets) for chronic and acute recurrent pancreatitis diseases in the United States; (b) development and commercialization of the Tissue Genesis Icellator® platform, a cell therapy productsisolation system acquired by Koligo from “benchTissue Genesis, LLC prior to manufacturing bedside” has been manufacturing bottlenecks. The heterogeneous natureour acquisition of Koligo; and (c) preclinical development of the “3D-V” technology platform, a system exclusively licensed by Koligo from the University of Louisville Research Foundation intended for the revascularization and 3D printing of cell therapy products has introduced manufacturing complexity and regulatory concerns, as well as scale-up complexities that are not present within traditional pharmaceutical manufacturing. Overtissue for transplant applications. Koligo maintains facilities in Indiana and Texas.

The Tissue Genesis assets acquired by Koligo included the years, MaSTherCell has developed experienceentire inventory of Tissue Genesis Icellator® devices, related kits and expertise necessaryreagents, a broad patent portfolio to protect the technology, registered trademarks, clinical data, and existing business relationships for transforming academic concepts into a clinical manufacturing program to support all phases of clinical trials. This includes assessing the clinical efficiencycommercial and development stage use of the laboratory concept but alsoIcellator technology

Pursuant to the developmentterms of efficient, robust and scalable manufacturing processes, including technology engineering service, when needed.

Access to a global network. Many companies developing autologous cell therapies envision using multiple manufacturing sites and processing centers to distribute the workload and minimize the shipping distances for such time- sensitive products. Many cell therapy products are fragile preparations that must be shipped and applied to a patient rapidly. This time pressure means that standard product-release testing procedures are not feasible. In particular, sterility testing often cannot be completed before patient treatment. This unique challenge in cell-therapy manufacturing requires tighter environmental and handling controls to greatly reduce any riskMerger Agreement, an aggregate of sterility failure. Biotechnology companies have to anticipate their success and the logistics to cure at point of care. Therefore, the setup of a global CDMO meets this requirement and is the strategy behind our establishment2,061,713 shares of our CDMO facilitiescommon stock were issued to Koligo’s Shareholders who were accredited investors (with certain Shareholders who were not accredited investors being paid solely in cash in the amount of approximately $20 thousand) in accordance with the terms of the Merger Agreement. In connection with the Merger, we assumed an aggregate of approximately $1.9 million of Koligo’s liabilities, which were substantially all of Koligo’s liabilities at the closing of the Merger. In addition, we issued 66,910 shares to Maxim Group LLC for advisory services in connection with the Merger. In November 2020, we filed a registration statement on Form S-3 to register the resale of 1,425,962 shares of our common stock as required by the Merger Agreement.

14

In addition, according to the agreement between the parties, we also funded an additional cash consideration of $500 thousand (with $100 thousand of such reducing the ultimate consideration payable to Koligo) for the acquisition of the assets of Tissue Genesis, LLC (“Tissue Genesis”) by Koligo that was consummated on October 14, 2020. The Tissue Genesis assets include the entire inventory of Tissue Genesis Icellator® devices, related kits and reagents, a broad patent portfolio to protect the technology, registered trademarks, clinical data, and existing business relationships for commercial and development stage use of the Icellator technology. The Icellator device is already commercially available in Korea and Israel,the Bahamas, and is expected to gain regulatory approval in Japan during the first quarter of 2021, subject to completion of manufacturing tests requested by the Japanese Pharmaceutics and Medical Devices Agency. Tissue Genesis already initiated U.S. FDA IDE Phase 1 pilot trials SVF cells in the treatment of erectile dysfunction, critical limb ischemia, tissue repair, and other therapeutic indications.

Revenue Model, Business Development and Licenses

The Orgenesis Point of Care (POCare) Platform is comprised of three enabling components: a multitude of licensed cell based POCare Therapeutics that are produced in closed, automated POCare Technology systems across a collaborative POCare Network. Our therapies include, but are not limited to, autologous, cell-based immunotherapies, therapeutics for metabolic diseases, anti-viral diseases, and tissue regeneration. We are establishing and positioning the business to bring point-of-care therapies to patients in a scalable way working directly with hospitals and through regional JV partners and JVs active in autologous cell therapy product development, including facilities in various countries in North America, Europe, Latin America, Asia, the Middle East, and Australia. The POCare Platform’s goal is to enable a rapid, globally harmonized pathway for these therapies to reach large numbers of patients at lowered costs through efficient, and decentralized production. The Network brings together industry partners, research institutes and hospitals worldwide to achieve harmonized, regulated clinical development and production of the therapies.

We are focused on technology in licensing and therapeutic collaborations, and we out license therapies marketing rights and manufacturing rights to partners and / or to the JVs. In many cases, the JVs are responsible for the preparation of clinical trials, local regulatory approvals and regional marketing activities. Such licensing includes exclusive or nonexclusive, sublicensable, royalty bearing rights and license to the Orgenesis Background IP as required solely to manufacture, distribute and market and sell Orgenesis Products within the relevant territories. In consideration of the rights and the licenses so granted, we receive a royalty in the range of ten percent of the net sales generated by the JV Entity and/or its sublicensees (as applicable) with respect to the Orgenesis Products.

In addition, in many cases, once the JV entities become profitable, we are entitled (in addition to any of its rights as holder of the JV Entity and prior to any other distributions of dividends by the JV Entity to shareholders of the JV Entity) and in addition to any royalties to which we may be entitled pursuant to a Orgenesis License Agreement, to receive from the U.S. inJV entity royalties at a range of 10 to 15 percent of the future. To complyJV entity’s audited US GAAP profit after tax.

Further to revenues generated from out licenses we generate revenues from POCare services and sales which is comprised of:

R&D services provided to out licensing partners

The Company has signed POCare Master Services Agreements (“MSAs”) with anticipatedits JV partners. In terms of the MSAs, we provide certain broadly defined development services that relate to our licensed therapies designed to develop or enhance the therapy with the objective of preparing it for clinical use. Such services, per therapy, include regulatory harmonization, we have also invested inservices, pre-clinical studies, intellectual property services, development services, and GMP process translation.

Hospital supply

Hospital services includes the sale or lease of products and the performance of processing services to our Quality and Management Systems (QMS) and to structure them in a way they could be sharedPOCare hospitals or other medical providers. We either work directly with either affiliated companieshospitals or business partners, and even with customers or prospects. South Korea, Israeli and European requirements are essentially the same, allowing Masthercell Global to implement its QMS model in a quick and efficient way. This truly international footprint will give us a unique competitive advantage, thereby filling the gap of biotechnology companies’ requirement of “quality comparability” between the respective regional sites.

Central continental locations to deal with key logistics challenges. With respect to this challenge,receive payments through our subsidiaries, Masthercell Global has built up the following:regional JV partnerships.

15

·Team of dedicated experts both from academic and industry backgrounds with a strong experience in cGMP dealing with not yet harmonized regulatory requirements (European Medicines Agency (the “EMA”), FDA);
·State-of-the-art facilities located next to airports; and
·Multi-continental footprints to deal with therapies administration at or nearby point of care as many cell therapy products have a short shelf life.Cell process development revenue

Providing value-added manufacturing capacity. OneWe provide cell process development services in some regions to third party customers. Those services are unique to the customers who retain the ownership of the biggest challengesintellectual property created through the process.

Our POCare therapy revenue is developing reliable (quality)as follows:

  Year Ended December 31, 
Revenue stream: 2020  2019 
  (in thousands) 
POC and hospital services $6,068  $3,109 
Cell process development services  1,584   790 
Total $7,652  $3,899 

Cost of Research and robust manufacturing processes for cell-based therapy products that ensure adequate product safety, potency,Development and consistency at an economically viable cost. Additionally, manufacturing qualityResearch and comparability is at the heartDevelopment Services

We incurred $83,986 and $14,014 thousand in cost of biotechnology companies’ challenges. MaSTherCell has built-up a strong expertise to customize the production and manufacturing process to suit the particular needs of a given client. This process facilitates a deep understanding of clients’ needs and facilitates a long term revenue generating relationship.

Competition in the CDMO Field

We compete with a number of companies both directly and indirectly. Key competitors include the following CMOs and CDMOs: Lonza Group Ltd, Progenitor Cell Therapy (PCT) LLC (acquired by Hitachi), WuxiAppTec (WuXi PharmaTech (Cayman) Inc.), Cognate Bioservices Inc., Apceth GmbH & Co. KG, Eufets GmbH, Fraunhofer Gesellschaft, Cellforcure SASU, Cell Therapy Catapult Limited and Molmed S.p.A. Some of these companies are large, well-established manufacturers with financial, technical,sales, research and development and salesresearch and marketing resources that are significantly greater than those that we currently possess.

-12-

More generally, we face competition inherent in any third-party manufacturer’s business - namely, that potential customers may instead elect to invest in their own facilities and infrastructure, affording them greater control over their products and the hope of long-term cost savings compared to a third party contract manufacturer. To be successful, we will need to convince potential customers that our current and expanding capabilities are more innovative, of higher-quality and more cost-effective than could be achieved through internal manufacturing and that our experience and quality manufacturing and process development expertise are uniqueservices in the industry.fiscal years ended December 31, 2020 and December 31, 2019, respectively, of which $196 and $812 thousand was covered by grant funding. Part of the expense was funded by share issues. Our abilityresearch and development scope was expanded to achieve thisthe evaluation and to successfully compete against other manufacturers will depend,development of new cell therapies related technologies in large part, on our successthe field of immuno-oncology, liver pathologies and tissue regeneration.

Competition in developing technologiesthe Cell Therapy Field

The biopharmaceutical industry is intensely competitive. There is continuous demand for innovation and speed, and as the cell-based therapies market evolves, there is always the risk that improve both the quality and profitability associated with cell therapy manufacturing. If we are unable to successfully compete against other manufacturers, wea competitor may not be able to develop other compounds or drugs that are able to achieve similar or better results for indications. Potential competition includes major multinational pharmaceutical companies, established biotechnology companies, specialty pharmaceutical companies, universities, and other research institutions. Many of these competitors have substantially greater financial, technical, and other resources, such as larger research and development staff and experienced marketing and manufacturing organizations with established sales forces. Smaller or early-stage companies may also prove to be significant competitors, particularly through collaborative arrangements with large, established companies.

Currently, we are not aware of any other companies pursuing a business model similar to what we are developing under our CDMO business plans whichPOCare Platform. However, our competitors in the CGT field who are significantly larger and better capitalized than us could undertake strategies similar to what we are pursuing and even develop them at a much more rapid rate. These potential competitors include the same multinational pharmaceutical companies, established biotechnology companies, specialty pharmaceutical companies, universities, and other research institutions that are operating in the CGT field. In that respect, smaller or early-stage companies may harmalso prove to be significant competitors, particularly through collaborative arrangements with large, established companies.

Intellectual Property

We will be able to protect our business, financial conditiontechnology and resultsproducts from unauthorized use by third parties only to the extent it is covered by valid and enforceable claims of operations.our patents or is effectively maintained as trade secrets. Patents and other proprietary rights are thus an essential element of our business.

Our success will depend in part on our ability to obtain and maintain proprietary protection for our product candidates, technology, and know-how, to operate without infringing on the proprietary rights of others, and to prevent others from infringing our proprietary rights. Our policy is to seek to protect our proprietary position by, among other methods, filing U.S. and foreign patent applications related to our proprietary technology, inventions, and improvements that are important to the development of our business. We also rely on trade secrets, know-how, continuing technological innovation, and in-licensing opportunities to develop and maintain our proprietary position.

16

In addition, we own or have exclusive rights to twenty eight (28) United States patents, thirty six (36) foreign-issued patents, twenty five (25) pending patent applications in the United States, forty five (45) pending patent applications in foreign jurisdictions, including Australia, Brazil, Canada, China, Europe, Hong Kong, India, Israel, Japan, Mexico, New Zealand, North Korea, Russia, Singapore, South Africa, and South Korea, and two (2) international Patent Cooperation Treaty (“PCT”) patent applications. These patents and patent applications relate, among others, to (1) dendritic and macrophages based vaccines, and their use for treating cancer and viral diseases; (2) compositions comprising ranpirnase and other ribonucleases for treating viral diseases; (3) tumor infiltrating lymphocytes (TILs) and their use for treating cancer; (4) compositions comprising immune cells, ribonucleases, or antibodies for treating COVID-19; (5) whole-cell antiviral vaccines; (6) therapeutic compositions comprising exosomes, bioxomes, and redoxomes; (7) bioreactors for cell cultureand automated devices for supporting cell therapies: and (8) scaffolds, including alginate and sulfated alginate scaffolds, polysaccharides thereof, and scaffolds for use for cell propagation, transplantations, and in the treatment of autoimmune diseases.

We have a pending U.S. patent applications directed, among others, to dendritic and macrophages based vaccines, and their use for treating cancer and viral diseases. If issued, this application would expire in 2038.

We have pending U.S. patent applications directed, among others, to compositions comprising ranpirnase and other ribonucleases for treating viral diseases. If issued, these applications would expire between 2039 and 2040. Counterpart patents applications were filed in Australia, Canada, China, Europe, Hong Kong, Japan, Mexico, New Zealand, North Korea, Russian Federation, Singapore, South Africa, and were also filed as International (“PCT”) applications. If issued, these applications would expire between 2035 and 2037. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

We have pending U.S. patent applications directed, among others, to therapeutic compositions comprising exosomes, bioxomes, and redoxomes. If issued, these applications would expire in 2040. Counterpart patents applications were filed in Australia, Brazil, Canada, China, Europe, India, Israel, India, Japan and South Korea. If issued, these applications would expire in 2039. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

We have pending U.S. patent applications directed, among others, to automated devices for supporting cell therapies. If issued, these applications would expire between 2035 and 2038.

We have a pending U.S. provisional patent application directed, among others, to tumor infiltrating lymphocytes (TILs) and their use for treating cancer. If converted into a non-provisional application and issued, this application would expire in 2041, without including any patent term extensions that might be available following the grant of marketing authorizations.

17

We believehave pending U.S. provisional patent applications directed, among others, to compositions comprising immune cells, ribonucleases, or antibodies for treating COVID-19. If converted into a non-provisional application and issued, this application would expire in 2041, without including any patent term extensions that Masthercell Global’s services differ from our competitionmight be available following the grant of marketing authorizations.

Granted U.S. patents, which are directed among others to scaffolds, including alginate and sulfated alginate scaffolds, polysaccharides thereof, and scaffolds for use for cell propagation, transplantations, and in two major aspects:the treatment of autoimmune diseases, will expire between 2025 and 2036. Counterpart patents granted in Australia, France, Germany, Israel, Switzerland, and the United Kingdom, will expire between 2026 and 2035. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

·Quality and expertise of its services: Clients identify the excellence of its facility, quality system, and people as a major differentiating point compared to competitors; and
·Agile and tailored approach: Our philosophy is to build a true partnership with our clients and adapt ourselves to clients’ needs, which entails no “off-the-shelf process” nor in-house technology platform, but a dedicated person for each client, joint steering committees on each project and dedicated project managers.

* Diagram above signifies “one-stop-shop service offering” from process development through quality manufacturing and logistics to point of care.

We strengthen our position by our “one-stop-shop” service offering, from pre-clinicalhave pending U.S. patent applications directed, among others, to commercial, with a clear focus on COGSbioconjugates comprising sulfated polysaccharides and diverse bioactive peptides, and their use in the treatment of manufacturing processes. This differentiation resultsinflammatory conditions. If issued, these applications would expire in a price premium compared2038. Counterpart patents applications were filed in China, Europe, Israel, Japan, and South Korea. If issued, these applications would expire between 2026 and 2038. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations

Orgenesis Ltd, has exclusive rights to other CMO’s as we operate with a lean organization focused solely on cell therapy. Quality is a critical aspectsix (6) United States patents, fourteen (14) foreign-issued patents, five (5) pending patent applications in the United States, twenty six (26) pending patent applications in foreign jurisdictions, including Australia, Brazil, Canada, China, Europe, India, Israel, Japan, Mexico, Panama, Singapore, and South Korea. These patents and patent applications relate, among others, to the trans-differentiation of our industry,cells (including hepatic cells) to cells having pancreatic β-cell-like phenotype and we believe we have developed unique expertisefunction and to their use in this field. We devote significant resourcesthe treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis. Granted U.S. patents, which are directed among others to process developmenttrans-differentiation to pancreatic β-cell-like phenotype and manufacturingfunction cells and to their use in order to optimize the safetytreatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and efficacy of our future product candidates for our customers, as well as our cost of goodspancreatitis, will expire between 2024 and time to market. Our goal is to carefully manage our fixed cost structure, maximize optionality, and drive long-term cost of goods as low as possible. We believe that operating our own manufacturing facility, which provides us with enhanced control of material supply for both clinical trials2035. Counterpart patents granted in Australia, France, Germany, Israel, Switzerland, and the commercial market,United Kingdom, will enable a more rapid implementationexpire between 2024 and 2035. These expiration dates do not include any patent term extensions that might be available following the grant of process changes,marketing authorizations.

Orgenesis Ltd, has pending U.S. patent applications directed, among others, to the trans-differentiation of cells, to cells having pancreatic β-cell-like phenotype and will allow for better long-term margins.function and to their use in the treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis. If issued, these applications would expire between 2038 and 2040. Counterpart patents applications were filed in Australia, Brazil, Canada, China, Europe, India, Israel, Mexico, Panama, Singapore, South Korea, and were also filed as International (“PCT”) applications. If issued, these applications would expire between 2034 and 2039. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

Finally, we have sought to establish manufacturing centers in regions which logistically offers an ideal location given the high concentration of companies active in cell therapy, including potential clients and companies with complementary know-how, products and services.

-13-

CDMO Government Regulation

Development Business

We are required to comply with the regulatory requirements of various local, state, national and international regulatory bodies having jurisdiction in the countries or localities where we manufacture products or where our customers’ products are distributed. In particular, we are subject to laws and regulations concerning research and development, testing, manufacturing processes, equipment and facilities, including compliance with cGMPs, labeling and distribution, import and export, facility registration or licensing, and product registration and listing. As a result, our facilities are subject to regulation by the FDA, as well as regulatory bodies of other jurisdictions, such as the EMA, Health Canada,in Israel and the Australian Department of Health, depending on the countries in which our customers market and sell the products we manufacture and/or package on their behalf.South Korea. We are also required to comply with environmental, health and safety laws and regulations, as discussed below. These regulatory requirements impact many aspects of our operations, including manufacturing, developing, labeling, packaging, storage, distribution, import and export and record keeping related to customers'customers’ products. Noncompliance with any applicable regulatory requirements can result in government refusal to approve facilities for manufacturing products or products for commercialization.

Manufacturing facilities that produce cellular therapies are subject to extensive regulation by the FDA. In particular, FDA regulations set forth requirements pertaining to establishments that manufacture human cells, tissues, and cellular and tissue-based products (“HCT/Ps”). Title 21, Code of Federal Regulations, Part 1271 provides for a unified registration and listing system, donor-eligibility, current Good Tissue Practice ("cGTP"), and other requirements that are intended to prevent the introduction, transmission, and spread of communicable diseases by HCT/Ps. More specifically, key elements of Part 1271 include:

·Registration and listing requirements for establishments that manufacture HCT/Ps;
·Requirements for determining donor eligibility, including donor screening and testing;
·cGTP requirements, which include requirements pertaining to the manufacturer's quality program, personnel, procedures, manufacturing facilities, environmental controls, equipment, supplies and reagents, recovery, processing and process controls, labeling, storage, record-keeping, tracking, complaint files, receipt, pre-distribution shipment, distribution, and donor eligibility determinations, donor screening, and donor testing;
·Adverse reaction reporting;
·Labeling of HCT/Ps;
·Specific rules for importing HCT/Ps; and
·FDA inspection, retention, recall, destruction, and cessation of manufacturing operations.

Masthercell Global currently collects, processes, stores and manufactures HCT/Ps, including the manufacture of cellular therapy products. Therefore, Masthercell Global must comply with cGTP and with the current Good Manufacturing Practices (“cGMP”) requirements that apply to biological products. Cell and tissue-based products may also be subject to the same approval standards, including demonstration of safety and efficacy, as other biologic and drug products if they fail to meet all HCT/P criteria set forth in Title 21, Code of Federal Regulations, Section 1271.10.

The U.S. Federal Food, Drug, and Cosmetic Act (the “FD&C Act”) and FDA regulations govern the quality control, manufacture, packaging, and labeling procedures of products regulated as a drug or biological products, including cellular therapies comprising HCT/Ps. These laws and regulations include requirements for regulated entities to comply with cGTPs applicable to the specific product(s). The cGTPs are designed to ensure that a facility's processes - and products resulting from those processes - meet defined safety requirements. The FDA's objective in requiring compliance with cGTP standards is to protect the public health and safety by ensuring that regulated products (i) have the identity, strength, quality and purity that they purport or are represented to possess; (ii) meet their specifications; and (iii) are free of objectionable microorganisms and contamination. As a central focus of the cGTP requirements, regulated entities must design and build quality assurance safeguards into the manufacturing processes and the production facilities for regulated products and must ensure the consistency, product integrity, and reproducibility of results and product characteristics. This is done by implementing quality systems and processes including appropriate, controlled procedures, specifications and documentation. In addition, drug manufacturers and certain of their subcontractors are required to register their establishments with FDA and certain state agencies. Registration with the FDA subjects entities to periodic unannounced inspections by the FDA, during which the agency inspects manufacturing facilities to assess compliance with applicable cGTPs. The FDA may also initiate for-cause investigations of manufacturing facilities if it learns of possible serious regulatory violations at such facilities. Accordingly, manufacturers must continue to expend time, money, and effort in the areas of production and quality control to maintain compliance with cGTPs. Failure to comply with applicable FDA requirements can result in regulatory inspections and associated observations, warning letters, other enforcement measures requiring remedial action, and, in the case of failures that are more serious, suspension of manufacturing operations, seizure of product, injunctions, product recalls, fines, and other penalties. We believe that our facilities are in material compliance with applicable, existing FDA requirements. Additionally, FDA, other regulatory agencies, or the U.S. Congress may be considering, and may enact laws or regulations regarding the use and marketing of stem cells, cell therapy products, or products derived from human cells or tissue. These laws and regulations may directly affect us or the business of some of Masthercell’s Global’s clients and, therefore, the amount of business Masthercell Global receives from these clients.

-14-

The Clinical Laboratory Improvement Amendments (“CLIA”) extends U.S. federal oversight to clinical laboratories that examine or conduct testing on materials derived from the human body for the purpose of providing information for the diagnosis, prevention, or treatment of disease or for the assessment of the health of human beings. CLIA requirements apply to those laboratories that handle biological matter. CLIA requires that these laboratories be certified by the government, satisfy governmental quality and personnel standards, undergo proficiency testing, be subject to biennial inspections and remit fees. The sanctions for failure to comply with CLIA include suspension, revocation, or limitation of a laboratory's CLIA certificate necessary to conduct business, fines, or criminal penalties. Additionally, CLIA certification may sometimes be needed when an entity, such as Masthercell Global, desires to obtain accreditation, certification, or license from non-government entities for cord blood collection, storage and processing.

Our customers’ products must undergo pre-clinical and clinical evaluations relating to product safety and efficacy before they are approved as commercial therapeutic products. The regulatory authorities havingthat have jurisdiction in the countries in which our customers intend to market their products may delay or put on hold clinical trials, delay approval of a product or determine that the product is not approvable. The FDA or other regulatory agencies can delay approval of a drug if our manufacturing facilities are not able to demonstrate compliance with cGTPs, pass other aspects of pre-approval inspections (i.e., compliance with filed submissions) or properly scale up to produce commercial supplies. The FDA and comparable government authorities having jurisdiction in the countries in which our customers intend to market their products have the authority to withdraw product approval or suspend manufacture if there are significant problems with raw materials or supplies, quality control and assurance or the product is deemed adulterated or misbranded. In addition, if new legislation or regulations are enacted or existing legislation or regulations are amended or are interpreted or enforced differently, we may be required to obtain additional approvals or operate according to different manufacturing or operating standards or pay additional fees. This may require a change in our manufacturing techniques or additional capital investments in our facilities.

18

Certain products manufactured by us involve the use, storage and transportation of toxic and hazardous materials. Our operations are subject to extensive laws and regulations relating to the storage, handling, emission, transportation and discharge of materials into the environment and the maintenance of safe working conditions. We maintain environmental and industrial safety and health compliance programs and training at our facilities.

Prevailing legislation tends to hold companies primarily responsible for the proper disposal of their waste even after transfer to third party waste disposal facilities. Other future developments, such as increasingly strict environmental, health and safety laws and regulations, and enforcement policies, could result in substantial costs and liabilities to us and could subject the handling, manufacture, use, reuse or disposal of substances or pollutants at our facilities to more rigorous scrutiny than at present.

Our CDMOdevelopment operations involve the controlled use of hazardous materials and chemicals. We are subject to federal, state and local laws and regulations in the U.S. governing the use, manufacture, storage, handling and disposal of hazardous materials and chemicals. Although we believe that our procedures for using, handling, storing and disposing of these materials comply with legally prescribed standards, we may incur significant additional costs to comply with applicable laws in the future. Also, even if we are in compliance with applicable laws, we cannot completely eliminate the risk of contamination or injury resulting from hazardous materials or chemicals. As a result of any such contamination or injury, we may incur liability or local, city, state or federal authorities may curtail the use of these materials and interrupt our business operations. In the event of an accident, we could be held liable for damages or penalized with fines, and the liability could exceed our resources. Compliance with applicable environmental laws and regulations is expensive, and current or future environmental regulations may impair our contract manufacturing operations, which could materially harm our business, financial condition and results of operations.

-15-

The costs associated with complying with the various applicable local, state, national and international regulations could be significant and the failure to comply with such legal requirements could have an adverse effect on our results of operations and financial condition. See “Risk Factors—Factors — Risks Related to Development and Regulatory Approval of Our CDMO BusinessTherapies and Product Candidates — Extensive industry regulation has had, and will continue to have, a significant impact on our CDMO business, and it may require us to substantially invest inespecially our product development, manufacturing and distribution capabilities and may negatively impact our ability to generate and meet future demand for our products and improve profitability”capabilities.” for additional discussion of the costs associated with complying with the various regulations.

PT BusinessPOCare Therapies Portfolio

Our therapeutic development effortsportfolio pipeline is diverse and addresses various unmet clinical needs. It is predominantly comprised of novel autologous cell therapies, implying that patients receive cells that originate from their own body, virtually eliminating the risk of an immune response and rejection and thus easing various regulatory hurdles. In addition, by leveraging Orgenesis’ vast experience and proven track record in ourdeveloping and optimizing cell therapy businessprocessing, these selective therapies are focused on advancing breakthrough scientific achievementsadapted to be produced in closed, automated technology systems, reducing the fieldneed for high grade cleanroom environments. The systems enable each stage of autologous therapies which have a curative potential. We base our development on therapeutic collaborations and in-licensing with other pre-clinical and clinical-stage biopharma companies as well as direct collaboration with research and healthcare institutes. We are engaging in therapeutic collaborations and in-licensing with other academic centers and research centersthe manufacturing process (cell sorting, expansion, genetic modifications, quality control) to be optimized in order to pursue emerging technologiessubstantially reduce the cost burden for patients and making the therapies widely accessible. Notably, our therapeutic pipeline is developed by researchers from our network and are subsequently outlicensed and validated in multi-center clinical trials conducted across point of other ATMPs in cell and gene therapy in such areas as cell-based immunotherapies, metabolic diseases, neurodegenerative diseases and tissue regeneration. Each of these customers and collaborations represents a growth opportunity and future revenue potential as we out-license these ATMPs through regional partners to whom we also provide regulatory, pre-clinical and training services to support their activity in order to reach patients in a point-of-care hospital setting.

PT Subsidiaries and Collaboration Agreements

We intend to devote significant resources to process development and manufacturing in order to optimizecare partner sites leveraging the safety and efficacy of our future product candidates, as well as our cost of goods and time to market. Our goal is to carefully manage our fixed cost structure, maximize optionality, and drive long-term cost of goods as low as possible.

We carry out our PT business through three wholly-owned and separate subsidiaries. This corporate structure allows us to simplify the accounting treatment, minimize taxation and optimize local grant support. The subsidiaries related to this business are as follows:

·United States: Orgenesis Maryland Inc. – This is the center of activity for North America currently focused on technology licensing, therapeutic collaborations and preparation for U.S. clinical trials.

·European Union: Orgenesis SPRL – This is the center of activity for Europe, currently focused on process development and preparation of European clinical trials.

·Israel: Orgenesis Ltd. – This is a research and technology center, as well as a provider of regulatory, clinical and pre-clinical services.

We have embarked on a strategy of collaborative arrangements with strategically situated third parties around the world. We believe that these parties have the expertise, experience and strategic location to advance our PT therapy business. Activities in our PT platform include:

-16-

·Trans-differentiation Technology - Our trans-differentiation technology demonstrates the capacity to induce a shift in the developmental fate of cells from the liver or other tissues and transdifferentiating them into “pancreatic beta cell-like” Autologous Insulin Producing (“AIP”) cells for patients with Type 1 Diabetes (“T1D”), acute pancreatitis and other insulin deficient diseases. This technology, which has yet to be proven in human clinical trials, has shown in relevant animal models that the human derived AIP cells produce insulin in a glucose-sensitive manner. This trans-differentiation technology is licensed by our Israeli Subsidiary and is based on the work of Prof. Sarah Ferber, our Chief Scientific Officer and a researcher at Tel Hashomer Medical Research Infrastructure and Services Ltd. (“THM”) in Israel. Our development plan calls for conducting additional pre-clinical safety and efficacy studies with respect to diabetes and other potential indications prior to initiating human clinical trials. With respect to our trans-differentiation technology, we own or have exclusive rights to ten (10) United States and nineteen (19) foreign issued patents, nine (9) pending applications in the United States, thirty-two (32) pending applications in foreign jurisdictions, including Europe, Australia, Brazil, Canada, China, Eurasia, Israel, Japan, South Korea, Mexico, and Singapore, and four (4) international PCT patent applications. These patents and applications relate, among others, to (1) the trans-differentiation of cells (including hepatic cells) to cells having pancreatic β-cell-like phenotype and function and to their use in the treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis, and (2) to scaffolds, including alginate and sulfated alginate scaffolds, polysaccharides thereof, and scaffolds for use for cell propagation, transdifferentiation, and transplantation in the treatment of autoimmune diseases, including diabetes.

·Collaboration Agreement with Hemogenyx Pharmaceuticals Plc - On October 18, 2018, we entered into a collaboration agreement and other ancillary agreements with Hemogenyx Pharmaceuticals Plc (“Hemogenyx”) to collaborate on the development and commercialization of Hemogenyx’s Human Postnatal Hemogenic Endothelial (Hu-PHEC) technology. Hu-PHEC is a cell replacement product candidate that is being designed to generate cancer-free, patient-matched blood stem cells after transplantation into the patient. Pursuant to the terms of the agreements, we shall exclusively manufacture and supply to Hemogenyx, its affiliates and licensees all Hu-PHEC related products both during and following completion of clinical trials. We shall also receive the worldwide -exclusive rights to market such products and shall serve as a global distributor of Hemogenyx’s Hu-PHEC related products. In consideration for such rights, we agreed to advance to Hemogenyx a convertible loan in an amount of no less than $1.0 million for furthering the development of the Hu-PHEC technology. As of November 30, 2018, we have funded $0.5 million under this convertible loan. We also agreed to pay a royalty of 12% of our net revenues resulting from the sale or licensing of products covered by Hemogenyx’s Hu-PHEC technology.

·Collaboration Agreement with Immugenyx, LLC - On October 16, 2018, we entered into a collaboration agreement with Immugenyx, LLC (“Immugenyx”), a wholly owned subsidiary of Hemogenyx Pharmaceuticals Plc. Immugenyx will collaborate with us to further the development and commercialization of its advanced hematopoietic chimeras (“AHC”). AHC, a new type of humanized mouse with a functional human immune system, is being developed by Immugenyx as an in vivo platform for disease modelling, drug and cell therapy development. Pursuant to the terms of the agreement, we shall receive non-exclusive worldwide rights to market the products and shall serve as a global distributor of Immugenyx’s products. Immugenyx will retain exclusive rights to manufacture, make and supply us with all the Immugenyx technology and/or licensed products that are marketed, sold or otherwise commercialized by us. In consideration for the license, we agreed to advance to Immugenyx a convertible loan in an amount of no less than $1.0 million for furthering the development of humanized mice models and related antibody development. As of November 30, 2018, we have funded $0.5 million under this convertible loan. We also agreed to pay a royalty of 12% of our net revenues resulting from the sale or licensing of products covered by Immugenyx’s AHC technology.

·Collaboration and License Agreement with Mircod Limited - On June 18, 2018, we and Mircod Limited, a company formed under the laws of Cyprus (“Mircod”) entered into a collaboration and license agreement for the research, development and commercialization of potential key technologies related to biological sensing for our clinical development and manufacturing projects. Within 45 days of the execution of the agreement, we agreed to approve a written project development plan outlining each party’s responsibilities with respect to the project and said project development plan was duly approved. We also agreed to fund the projected development costs as outlined in the development plan. Under the terms of the agreement, we agreed to, and remitted, an advance payment of $50,000. Under the agreement, all project results of such collaboration shall be jointly owned by Mircod and the Company. We were also granted an exclusive, worldwide sublicensable license under Mircod’s right in such project results to use and commercialize such project results in consideration for a royalty of 5% of Net Sales (as defined in the collaboration agreement) of products incorporating project results. We will be solely responsible for the commercialization of any resulting products. Subject to completion of the development project, Mircod and the Company are to negotiate and enter into a manufacturing and supply agreement under which Mircod is to manufacture and supply products incorporating the project results and, at our request, to provide support and maintenance service for such products. If for whatever reason Mircod and the Company fail to enter into such manufacturing and supply agreement within 90 days of the completion of the development project or if Mircod is unable to perform such services, we are entitled to manufacture the products, in which event Mircod will be entitled to a payment of $80,000 and royalties on net sales are to increase to 8% of net sales.

-17-

·Research and License Agreement with B.G. Negev Technologies and Applications Ltd. and The National Institute of Biotechnology in the Negev Ltd.- On August 2, 2018 and November 25, 2018, respectively, we entered into research and license agreements (the “Agreements”) with B.G. Negev Technologies and Applications Ltd. (“BGN”) and/or The National Institute of Biotechnology in the Negev Ltd. (both herein, the “BG Entities”). Under the terms of the Agreements, we shall collaborate on the research and development of BGN’s dissolvable carriers for cell culturing and for developing and commercializing technology directed to RAFT modification of polysaccharides and use of a bioreactor for supporting cell constructs. We have received the exclusive, worldwide rights to make, develop and commercialize technologies utilizing the dissolvable carriers for cell culturing, with an initial focus on autoimmune diseases. This unique technology has the potential to allow us to reduce the cost and complexity of manufacturing of our cell therapy programs.

·Joint Venture Agreement with HekaBio K.K. -On July 10, 2018, we and HekaBio K.K. (“HB”), a corporation organized under the laws of Japan, entered into a joint venture agreement pursuant to which we agreed to collaborate in the clinical development and commercialization of cell and gene therapeutic products in Japan (the “JVA”). The parties intend to pursue the joint venture through a newly established Japanese company which we, or we together with a designee, will hold a 49% participating interest therein, with the remaining 51% participating interest being held by HB (the “JV Company”). HB will fund, at its sole expense, all costs associated with obtaining the requisite regulatory approvals for conducting clinical trials, as well as performing all clinical and other testing required for market authorization of the products in Japan. Under the joint venture agreement, each party may invest up to $10 million, which may take the form of a loan, if required, as determined by the steering committee. The terms of such investment, if any, will be on terms mutually agreeable to the parties, provided that the minimum pre-money valuation for any such investment shall not be less than $10 million. Additionally, HB was granted an option to affect an equity investment in us of up to $15 million within the next 12 months on mutually agreeable terms. If such investment is in fact consummated, we agreed to invest in the JV Company by way of a convertible loan an amount to HB’s pro-rata participating interest in the JV Company, which initially will be at 51%. Such loan may then be converted by us into share capital of the JV Company at an agreed upon formula for determining the JV Company valuation which in no event shall be less than $10 million. Under the joint venture agreement, we can require HB to sell to us its participating (including equity) interest in the JV Company in consideration for the issuance of our common stock based on an agreed upon formula for determining the JV Company’s valuation which in no event shall be less than $10 million. In addition, under the joint venture agreement, we shall grant the JV Company an exclusive license to certain intellectual property as may be required for the JV Company to develop and commercialize the products in Japan. In consideration of such license, the JV Company shall pay us, in addition to other payments, royalties at the rate of 10% of the JV Company’s net sales of the Products. On October 3, 2018, we entered into a license agreement with the JV Company pursuant to the joint venture agreement pertaining to the licenses described above.

-18-

·Joint Venture Agreement with Image Securities Ltd. -On July 11, 2018, we and Image Securities Ltd., a corporation with its registered office in Grand Cayman, Grand Cayman Islands (“India Partner”) entered into a joint venture agreement pursuant to which we agreed to collaborate in the development and/or marketing, clinical development and commercialization of cell therapy products in India. The India Partner will collaborate with a network of healthcare facilities and a healthcare infrastructure as well as financial partners to advance the development and commercialization of the cell therapy products in India. The joint venture agreement becomes effective upon the consummation of an equity investment by the India Partner in the Company of $5 million within 15 days of the execution of the joint venture agreement through the purchase of units of our securities at a per unit purchase price payable into the Company of $6.24, with each unit comprised of one share of our common stock and a three-year warrant for the acquisition of an additional common share at a per share exercise price of $6.24. Subject to the consummation of such equity investment in the Company, we are to advance to the joint venture company a convertible loan in the amount of $5 million. The loan is convertible into equity capital of the joint venture company at an agreed upon formula for determining joint venture company valuation. The investment in us by the India Partner was consummated by way of the previously disclosed private placement subscription agreement entered into in December 2016 between us and an affiliate of the India Partner. We advanced $1 million under our obligation under the convertible loan on October 18, 2018. Under the joint venture agreement, the India Partner agreed to invest in the joint venture company $10 million within 12 months of the incorporation of the joint venture company. If for whatever reason such investment is not made by the India Partner within such time, then we are authorized to convert our above-referenced loan into 50% of the equity capital of the joint venture company on a fully diluted basis, provided that if the pre- money valuation of the joint venture company is then independently determined to be less than $5 million, then such conversion to be effected in the basis of such valuation.

Background on Our Transdifferentiation Technology

Diabetes Mellitus (“DM”), or simply diabetes, is a metabolic disorder usually caused by a combination of hereditary and environmental factors, and results in abnormally high blood sugar levels (hyperglycemia). Diabetes occurs as a result of impaired insulin production by the pancreatic islet cells. The most common typesrobustness of the diseaseOrgenesis network. Once approved these therapies are Type-1 Diabetes (“T1D”) and Type-2 Diabetes (“T2D”). In T1D,distributed to leading medical institutions globally within the onset of the disease follows an autoimmune attack of β-cells that severely reduces β-cell mass. T1D usually has an early onset and is sometimes also called juvenile diabetes. In T2D, the pathogenesis involves insulin resistance, insulin deficiency and enhanced gluconeogenesis, while late progression stages eventually lead to β-cell failure and a significant reduction in β-cell function and mass. T2D often occurs later in life and is sometimes called adult onset diabetes. Both T1D and late-stage T2D result in marked hypoinsulinemia, reduction in β-cell function and mass and lead to severe secondary complications, such as myocardial infarcts, limb amputations, neuropathies and nephropathies and even death. In both cases, patients become insulin-dependent, requiring either multiple insulin injections per day or reliance on an insulin pump.

Diabetes is one of the most challenging health problems in the 21st Century, resulting in staggering health, social, and economic impacts. Diabetes is currently the fourth or fifth leading cause of death in most developed countries and has been declared an epidemic in many developing and newly industrialized nations.

Within the field of cell therapy, research and development using stem cells to treat a host of diseases and conditions has greatly expanded. All living complex organisms start as a single cell that replicates, differentiates (matures) and perpetuates in an adult organism throughout its lifetime. Stem cells (in either embryonic or adult forms) are primitive and undifferentiated cells that have the unique ability to transform into or otherwise affect many different cells, such as white blood cells, nerve cells or heart muscle cells. Our technology employs a molecular and cellular approach directed at converting liver cells into functional insulin-producing cells as a treatment for diabetes. This new therapeutic approach does not use stem cells, but rather is focused on the use of autologous, fully mature, adult cells.

There are two general classes of cell therapies: allogeneic and autologous. In allogeneic procedures, cells collected from a person (the donor) are transplanted into or used to develop a treatment for another patient (the recipient) with or without modification. In cases where the donor and the recipient are the same individual, these procedures are referred to as “autologous”.

Our treatment for diabetes focuses on autologous cells that offer a low likelihood of rejection by the patient. We believe the long-term benefits of this treatment can best be achieved with an autologous product. For our purposes in the treatment of diabetes, our cells are derived from the liver or other adult tissue and are transdifferentiated to become adult Autologous Insulin Producing (“AIP”) cells.

-19-

Through our Israeli and Belgian subsidiaries, our goal is to advance our AIP cell-based therapy into clinical development. AIP cells utilize the technology of ‘cellular trans-differentiation’ to transform an autologous adult liver cell into a fully functional and physiologically glucose-responsive insulin-producing cell. Treatment with AIP cells is expected to provide Type 1 Diabetes patients with long-term insulin independence. Because AIP cells are autologous, this benefit should be achieved and maintained without the need for concomitant immunosuppressive therapy.

Threats from Pancreas Islet Transplantation and Cell Therapies

To date, a significant portion of the amount invested in diabetes related research and development activities has been directed toward prevention and lifestyle management rather than toward the development of a cure. For some patients with severe and difficult to control diabetes (hypoglycemic unawareness), islet transplants are considered. Pancreatic islets are the cells in the pancreas that produce insulin. Scientists use enzymes to isolate the islets from the pancreas of a deceased donor. Because the islets are fragile, transplantation must occur soon after they are removed. Typically, a patient receives at least 10,000 islet “equivalents” per kilogram of body weight, extracted from pancreases obtained from different donors. Patients often require two separate transplants to achieve insulin independence.

Transplants are often performed by an interventional radiologist, who uses x-rays and ultrasound to guide placement of a catheter - a small plastic tube - through the upper abdomen and into the portal vein of the liver. The islets are then infused slowly through the catheter into the liver. The patient receives a local anesthetic and a sedative. In some cases, a surgeon may perform the transplant through a small incision, using general anesthesia. Because the islets are obtained from cadavers that are unrelated to the patient, the patient needs to be treated with drugs that inhibit the immune response so that the patient doesn’t reject the transplant. In the early days of islet transplantation, the drugs were so powerful that they actually were toxic to the islets; improvements in the procedure are widely used and are now referred to as the Edmonton Protocol.

Pancreatic islet transplantation (cadaver donors) is an allogeneic transplant, and, as in all allogeneic transplantations, there is a risk for graft rejection and patients must receive lifelong immune suppressants. Though this technology has shown good results clinically, there are several setbacks, such as patients being sensitive to recurrent T1D autoimmune attacks and a shortage in tissues available for islet cells transplantation.

Pancreatic islet auto transplantation is a means of reducing the risk of brittle diabetes following total pancreatectomy. In 1977, researchers at the University of Minnesota School of Medicine pioneered the first Total Pancreatectomy with Islet Autologous Transplant (“TP-IAT”) for the treatment for induced diabetes post-surgery. At that time, islet cell isolation techniques, which had been pursued to treat insulin-dependent diabetes via allotransplant, yielded variable results and raised uncertainty regarding the future efficacy of TP-IAT. Since then, advances in isolation and purification have improved islet transplant outcomes, and the practice of TP-IAT has expanded. In the United States, there are currently approximately 12 centers performing TP-IAT, with 1 to 2 centers annually establishing programs; there is no available information on the worldwide use of this procedure.

TP-IAT has the distinct advantage of allowing patients the ability to avoid the significant postoperative complication of surgically induced brittle diabetes. The severity of brittle diabetes, a condition in which a patient experiences both severe hyper and hypoglycemic episodes, should not be underestimated; in one early series, 50% of late deaths after TP were secondary to iatrogenic hypoglycemic episodes. Although total pancreatectomy in the era of modern endocrine and exocrine replacement therapy has witnessed improvements in long-term morbidity and mortality, it remains one of the most morbid abdominal operations performed today.

Our Solution

We are developing and bringing to clinical stage a technology that is based on the published work of Prof. Sarah Ferber, our Chief Scientific Officer and a researcher at THM, that demonstrates the capacity to induce a shift in the developmental fate of cells from the liver into “pancreatic beta cell-like” insulin-producing cells. Furthermore, those cells were found resistant to the autoimmune attack and able to produce insulin in a glucose-sensitive manner. Our cell therapy business derives from a licensing agreement entered into as of February 2, 2012 by Orgenesis Ltd., our Israeli Subsidiary, and THM pursuant to which our Israeli Subsidiary was granted a worldwide royalty bearing an exclusive license to certain information regarding a molecular and cellular approach directed at converting liver cells into functional insulin-producing cells as a treatment for diabetes (the “THM License Agreement”).

-20-

Toward this goal, we are working to advance a unique product into clinical development. AIP cells utilize the technology of ‘cellular trans-differentiation’ to transform an autologous adult liver cell into an adult, fully functional and physiologically glucose-responsive pancreatic-like insulin producing cell. Treatment with AIP cells is expected to provide diabetes patients with long-term insulin independence. Our aim is to develop our AIP cell therapy in the treatment of diabetes by essentially correcting malfunctioning organs with new functional tissues created from the patient’s own existing organs.

Because the AIP cells are autologous, this benefit should be achieved and maintained without the need for concomitant immunosuppressive therapy. The procedure to generate AIP cells begins with liver tissue accessed via needle biopsy from a patient. The liver tissue is then sent to a CDMO, such as MaSTherCell, where biopsied liver cells are isolated, expanded and trans-differentiated into AIP cells. The final product is a solution of AIP cells, which are packaged in an infusion bag and sent back to the patient’s treating physician where the cells are transplanted back into the patient’s liver via portal vein infusion. The entire process, from biopsy to transplantation, is expected to take 5-6 weeks.

Unique Benefits of AIP Cells

-21-

We believe that our singular focus on the acquisition, development, and commercialization of AIP cells may have many and meaningful benefits over other technologies, including:

·Physiologically glucose-responsive insulin production within one week of AIP cell transplantation;
·Insulin-independence within one month;
·Single course of therapy (~10-year insulin-independence);
·No need for concomitant immunosuppressive therapy;
·Return to (near) normal quality of life for patients;
·Single liver biopsy supplies unlimited source of therapeutic tissue (bio-banking for future use if needed);
·Highly controlled and tightly closed GMP systems; and
·Quality control of final product upon release and distribution.

We are aware of no other company focused on development of AIP cells based on trans-differentiation. The pharmaceutical industry is fragmented, and it is a competitive market. We compete with many pharmaceutical companies, both large and small and there may be technologies in development of which we are not aware.

We believe our ability to further develop our AIP cells is augmented by the following:

IP Strength - Orgenesis has broad patent claims on its process and has both issued and pending patents in the U.S. and internationally. The patent portfolio includes granted patent US 8119405, entitled “Methods of inducing regulated pancreatic hormone production in non-pancreatic islet tissues,” which includes broad claims on trans-differentiating any mature, non-pancreatic cell type into an islet cell phenotype. Importantly, the company’s IP portfolio is not dependent on processes owned by other companies, such as embryonic stem cell technologies, production of endodermal intermediates or reprogramming (iPS) technologies. As a result, the company has both freedom to operate and ability to obstruct competitors in developing autologous cells for treatment of diabetes.

Simplicity - There is no need for anti-rejection treatment or encapsulation. Using liver as pancreatic progenitor tissue allows the diabetic patient to be the donor of his own insulin-producing tissue, thus allowing autologous implantations with no need for anti-rejection therapy, which restricts the target population only to adult, severe diabetic patients. Moreover, drugs used for preventing the allo-transplanted tissue rejection are deleterious to insulin producing cell function and to the patient.

Safety - the generated cells do not regress to pluripotency, and no adverse effects of uncontrolled cells proliferation occur. The cells are already mature and can be inserted in to the patient following extensive quality assurance testing. Moreover, our cells transplanted in rodents do not cause any adverse effects even following many weeks in the animals.

Availability - Sufficient liver cells to treat a patient as well to store cells for additional future treatments may be generated. The cells can be frozen and thawed, without losing the trans-differentiation capacity for up to 20 passages in culture. It is anticipated that a biopsy from the diabetic patient's own liver is sufficient to generate enough insulin-producing cells to replace the entire cell function and control blood glucose level. As opposed to islets that are non-dividing (i.e., post-mitotic), it is necessary to use stem cells to generate sufficient numbers of cells that are then differentiated.

Future Product Candidates-Currently, liver cells are best suited for generating AIP cells. Future products may involve the use of cell types other than liver that are more easily accessible from the diabetic patient or from unrelated donors. Additionally, other adult cells (i.e. fibroblasts) may be studied for trans-differentiation into functional cells in diseases other than insulin-dependent disorders (i.e. neurodegenerative).

The THM License Agreement

Our cell therapy business derives from a licensing agreement entered into as of February 2, 2012 by Orgenesis Ltd., our Israeli Subsidiary, and THM pursuant to which our Israeli Subsidiary was granted a worldwide royalty bearing and exclusive license to certain information regarding a molecular and cellular approach directed at converting liver cells into functional insulin producing cells as a treatment for diabetes (the “THM License Agreement”). By using therapeutic agents (i.e., PDX-1, and additional pancreatic transcription factors in an adenovirus-vector) that efficiently convert a sub-population of liver cells into pancreatic islets phenotype and function, this approach allows the diabetic patient to be the donor of his own therapeutic tissue. We believe that this provides major competitive advantage to the cell transformation technology of our Israeli Subsidiary.

-22-

As consideration for the license, our Israeli Subsidiary has agreed to pay the following to THM:

1)A royalty of 3.5% of net sales;
2)16% of all sublicensing fees received;
3)An annual license fee of $15,000, which commenced on January 1, 2012 and is due once every year thereafter (the “Annual Fee”). The Annual Fee is non-refundable, but it shall be credited each year due, against the royalty noted above, to the extent that such are payable, during that year; and
4)Milestone payments as follows:

a)$50,000 on the date of initiation of phase I clinical trials in human subjects;
b)$50,000 on the date of initiation of phase II clinical trials in human subjects;
c)$150,000 on the date of initiation of phase III clinical trials in human subjects;
d)$750,000 on the date of initiation of issuance of an approval for marketing of the first product by the FDA; and
e)$2,000,000, when worldwide net sales of products have reached the amount of $150,000,000 for the first time (the “Sales Milestone”).

As of November 30, 2018, our Israeli Subsidiary has not reached any of these milestones.

In the event of an acquisition of all of the issued and outstanding share capital of the Israeli Subsidiary or of us and/or consolidation of the Israeli Subsidiary or us into or with another corporation (“Exit”), under the THM License Agreement, THM is entitled to elect, at its sole option, whether to receive from us a one-time payment based, as applicable, on the value at the time of the Exit of either 463,651 shares of our common stock or the value of 1,000 ordinary shares of the Israeli Subsidiary at the time of the Exit. If THM elects to receive the consideration as a result of an Exit, the royalty payments will cease.

If THM elects to not receive any consideration as a result of an Exit, THM is entitled under the THM License Agreement to continue to receive all the rights and consideration it is entitled to pursuant to the THM License Agreement (including, without limitation, the exercise of the rights pursuant to future Exit events), and any agreement relating to an Exit event shall be subject to the surviving entity’s and/or the purchaser’s undertaking towards THM to perform all of the Israeli Subsidiary's obligations pursuant to the THM License Agreement.

The Israeli Subsidiary agreed to submit to THM a commercially reasonable plan which shall include all research and development activities as required for the development and manufacture of the products, including preclinical and clinical activities until an FDA or any other equivalent regulatory authority’s approval for marketing and including all regulatory procedures required to obtain such approval for each product candidate (a “Development Plan”), within 18 months from the date of the THM License Agreement. Under the THM License Agreement, the Israeli Subsidiary undertook to develop, manufacture, sell and market the products pursuant to the milestones and time-frame schedule specified in the Development Plan. The Israeli Subsidiary submitted the Development Plan in May 2014.

Under the THM License Agreement, THM is entitled to terminate the THM License Agreement under certain conditions relating to a material change in the business of our Israeli Subsidiary or a breach of any material obligation thereunder or to a bankruptcy event of our Israeli Subsidiary. Under certain conditions, our Israeli Subsidiary may terminate the THM License Agreement and return the licensed information to THM.

-23-

Competition in the Cell Therapy Field

The current treatment for T1D, and some T2D, is constant monitoring of blood glucose and a highly controlled diet, coupled with multiple daily insulin injections. Despite the use of insulin and advances in its delivery, pharmaceutical insulin injections cannot replicate the level of feedback control afforded by naturally occurring intact beta cells. Even with the most diligent insulin use, the adverse short- and long-term effects of diabetes include life-threatening episodes of low blood sugar, nerve damage, blindness, kidney damage, erectile dysfunction, foot ulcers leading to amputations, and cardiovascular disease. Research has shown that, on average, the life expectancy of a person with T1D is reduced by approximately 12 years when compared to the general population.

T1D inflicts a significant economic cost on the U.S. healthcare system, estimated at $14.4 billion annually, and it is expected that a therapy that can modify the course of T1D could potentially achieve significant cost savings,network and thus command high market penetration and premium pricing. Ingranting the near future, the market for T1D is expected to continue to be dominated by insulin replacement therapies.inventors a royalty-based commercialization horizon.

Currently, there are no approved therapies for new onset T1D with potential curative effect but only regimens such as insulin or adjuvants to insulin that address the disease when the pancreas can no longer produce insulin. While not a direct competitor, in a more advanced population of T1D, sotagliflozin, an oral adjunctive therapy to insulin, is expected to receive FDA approval following positive results from a pivotal Phase 3 trial conducted by Lexicon Pharmaceuticals in collaboration with Sanofi SA and JDRF. There are multiple agents in development targeting the modification of the course of the disease. Current approaches in development can be broadly divided into immune modulatory agents that attempt to improve metabolic function by rescuing insulin producing beta cells, or regenerative agents that attempt to replace beta cells. From a broad review of these agents and approaches, no other autologous therapy for T1D is expected to be in advanced clinical trials or provide direct competition to our AIP cells in the near future. Other allogeneic approaches, such as Viactye’s PEC-01 technology, may enter clinical trials in the near future.

Insulin therapy is used for Insulin-Dependent Diabetes Mellitus (IDDM) patients who are not controlled with oral medications, although this therapy has well-known and well-characterized disadvantages. Weight gain is a common side effect of insulin therapy, which is a risk factor for cardiovascular disease. Injection of insulin causes pain and inconvenience for patients. Patient compliance and inconvenience of self-administering multiple daily insulin injections is also considered a disadvantage of this therapy. The most serious adverse effect of insulin therapy is hypoglycemia.

The biopharmaceutical industry, and the rapidly evolving market for developing cell-based therapies is characterized by intense competition and rapid innovation. Our competitors may be able to develop other compounds or drugs that are able to achieve similar or better results. Our potential competitors include major multinational pharmaceutical companies, established biotechnology companies, specialty pharmaceutical companies, universities, and other research institutions. Many of our competitors have substantially greater financial, technical and other resources, such as larger research and development staff and experienced marketing and manufacturing organizations as well as established sales forces. Smaller or early-stage companies may also prove to be significant competitors, particularly through collaborative arrangements with large, established companies.

Specifically, we face significant competition from companies in the insulin therapy market. Insulin therapy is widely used for Insulin-Dependent Diabetes Mellitus (IDDM) patients who are not controlled with oral medications. The global diabetes market comprising the insulin, insulin analogues and other anti-diabetic drugs has been evolving rapidly. A look at the diabetes market reveals that it is dominated by a handful of participants such as Novo Nordisk A/S, Eli Lilly and Company, Sanofi-Aventis, Takeda Pharmaceutical Company Limited, Pfizer Inc., Merck KgaA, and Bayer AG.

PT Revenue Model

Through analysis of the cell therapy landscape, we are introducing a novel POCare therapy business model with our goal of bringing autologous therapies in a cost-effective, high-quality and scalable manner to patients. We are establishing and positioning our PT business in order to bring POCare therapies to patients in a scalable way via a network of leading healthcare facilities active in autologous cell therapy product development, including facilities in Germany, Austria, Greece, the U.S., Korea and Japan.

-24-

Our unique understanding of industry needs allows us to offer our clients a range of technologies and processes that potentially generate revenues. This may include:

·Development Services – Industrial manufacturing know-how to the cell and gene therapy arena, thus reducing cost of goods and facilitating regulatory scrutiny, higher automation level required to increase process robustness and reduce attrition rates, biological assay development, assay validation and assay optimization;

·Sub-Licensing Fees – Innovative technologies such as scaffolds and IoT sensors and closed system bioreactors that allow autologous cell manufacturing in lower grade clean rooms; and

·POCare Services – Regulatory assistance and joint ventures with local partners who bring strong regional networks through (1) joint venture partnerships with local hospitals utilizing hospital networks for clinical development of therapies, (2) a global network of supply, (3) harmonized quality systems, (4) the provision of a comprehensive portfolio of ATMPs to hospitals via continuous in-licensing of autologous therapies from academia and research institutes, and (4) out-licensing hospital and academic-based therapies.

PT Business Strategy

Our aim is to provide a pathway to bring ATMPs in the cell and gene therapy industry from research to patients worldwide through our POCare network. We define POCare cell and gene therapy as a process of collecting, processing and administering cells within the patient care environment, namely through academic partnerships in a hospital setting. We believe this approach is an attractive proposition for personalized medicine because POCare therapy facilitates the development of technologies through our strategic partnerships and utilizes closed systems that have the potential of reducing the required grade of clean room facilities, thus substantially reducing manufacturing costs. Furthermore, cell transportation, which is a high-risk and costly aspect of the supply chain, could be minimized or eliminated.

While our PT business strategy is currently limited to early stage development to overcome certain industry challenges, we intend to continue developing a global POCare network, with the goal of developing ATMPs, and namely autologous cell therapies, via joint ventures with partners who bring strong regional networks. Such networks include partnerships with local hospitals which allows us to engage in continuous in-licensing of, namely, autologous therapies from academia and research institutes, co-development of hospital and academic-based therapies, and utilization of hospital networks for clinical development of therapies.

We consider the following to be the four pillars in order to advance our PT business strategy:

·Innovation – This leverages our unique know-how and expertise for industrial processes, operational excellence, process development and optimization, quality control assays development, quality management systems and regulatory expertise.
·Systems – We are developing cell production cGMP systems utilizing sensor technology and AI-based systems for biological production, closed system devices for processing cells, proprietary virus/ media technologies and partnerships with key system providers.
·Cell and Gene Products – We intend to grow our internal asset pipeline consisting of our unique portfolio of immuno-oncology related technologies, MSC and liver-based therapies and secretome-based therapies.
·Distribution – Our plan is to enable the industrialization, commercialization and distribution of POCare systems in major hospitals and key geographies, including Europe, Asia, North America, and South America.

-25-

Grant Funding

Walloon Region, Belgium, Direction Générale Opérationnelle de l'Economie, de l'Emploi & de la Recherche (“DGO6”)

i.                       On March 20, 2012, MaSTherCell was awarded an investment grant of Euro 1.2 million from the DGO6. This grant is related to the investment in the production facility with a coverage of 32% of the investment planned. As of November 30, 2018, the DGO6 transferred to MaSTherCell the entire amount.

ii.                       On November 17, 2014, the Belgian Subsidiary received the formal approval from the DGO6 for a Euro 2 million ($2.4 million) support program for the research and development of a potential cure for Type 1 Diabetes. The financial support was composed of a Euro 1,085 thousand (70% of budgeted costs) grant for the industrial research part of the research program and a further recoverable advance of Euro 930 thousand (60% of budgeted costs) of the experimental development part of the research program. In December 2014, the Belgian Subsidiary received advance payment of Euro 1,209 thousand under the grant. The grants are partially refundable subject to certain conditions and are also subject to conditions and restrictions with respect to the Belgian Subsidiary’s work in the Walloon Region and ownership of results of the research program. In addition, the DGO6 is also entitled to a royalty upon revenue being generated from any commercial application of the technology. In 2017, the Company received final approval from the DGO6 for Euro 1.8 million costs invested in the project, out of which Euro 1.2 million founded by the DGO6. As of November 30, 2018, the Company repaid $34 thousand (Euro 30 thousand) to the DGO6 and $152 thousand was recorded in other payables in the financial statements.

iii.                       In April 2016, the Belgian Subsidiary received formal approval from DGO6 for a Euro 1.3 million ($1.5 million) support program for the development of a potential cure for Type 1 Diabetes. The financial support was awarded to the Belgium Subsidiary as a recoverable advance payment at 55% of budgeted costs, or for a total of Euro 717 thousand ($800 thousand). The grant will be paid over the project period. On December 19, 2016, the Belgian Subsidiary received advance payment of Euro 359 thousand ($374 thousand). Up through November 30, 2018, Euro 303 thousand was recorded as a deduction of research and development expenses and $64 thousand was recorded as advance payments on account of the grant. The grants are partially refundable subject to certain conditions and are also subject to conditions and restrictions with respect to the Belgian Subsidiary’s work in the Walloon Region and ownership of results of the research program.

iv.                       On October 8, 2016, the Belgian Subsidiary received formal approval from the DGO6 for a Euro 12.3 million ($12.8 million) support program for the GMP production of AIP cells for two clinical trials that will be performed in Germany and Belgium. The project will be held during a period of three years that commenced on January 1, 2017. The financial support is awarded to the Belgium Subsidiary at 55% of budgeted costs, or for a total of Euro 6.8 million ($7 million). The grant will be paid over the project period. On December 19, 2016, the Belgian Subsidiary received a first payment of Euro 1.7 million ($1.8 million). Up through November 30, 2018, $1.1 million was recorded as a deduction of research and development expenses and $847 thousand was recorded as advance payments on account of the grant. The grants are partially refundable subject to certain conditions and are also subject to conditions and restrictions with respect to the Belgian Subsidiary’s work in the Walloon Region and ownership of results of the research program.

Israel-U.S. Binational Industrial Research and Development Foundation (“BIRD”)

On September 9, 2015, the Israeli Subsidiary entered into a pharma Cooperation and Project Funding Agreement (“CPFA”) with BIRD and Pall Corporation, a U.S. company. BIRD will give a conditional grant of $400 thousand each (according to terms defined in the agreement), for a joint research and development project for the use of Autologous Insulin Producing (AIP) Cells for the Treatment of Diabetes (the “BIRD Project”). The BIRD Project started on March 1, 2015. Upon the conclusion of product development, the grant shall be repaid at the yearly rate of 5% of gross sales. The grant will be used solely to finance the costs to conduct the research of the project during a period of 18 months starting on March 1, 2015. On July 28, 2016, BIRD approved an extension for the project period until May 31, 2017 and the final report was submitted to BIRD. To date, the Israeli Subsidiary received $200 thousand under the grant. Up through November 30, 2018, $359 thousand was recorded as a deduction of research and development expenses and $159 thousand was recorded as a receivable on account of the grant.

-26-

Korea Israel Industrial R&D Foundation (“KORIL”)

On March 14, 2016, the Israel Subsidiary entered into a collaboration agreement with CureCell, initially for the purpose of applying a grant from KORIL for pre-clinical and clinical activities related to the commercialization of the Israel Subsidiary AIP cell therapy product in Korea. The parties agreed to carry out at their own expenses and their respective commitments under the work plan approved by KORIL and any additional work plan to be agreed upon between the Israeli Subsidiary and CureCell. The Israeli Subsidiary will own sole rights to any intellectual property developed from the collaboration which is derived under the Israeli Subsidiary’s AIP cell therapy product, information licensed from THM. Subject to obtaining the requisite approval needed to commence commercialization in Korea, the Israel subsidiary has agreed to grant to CureCell, or a fully owned subsidiary thereof, under a separate sub-license agreement an exclusive sub-license to the intellectual property underlying the Company’s API product solely for commercialization of the Israel Subsidiary’s products in Korea. As part of any such license, CureCell has agreed to pay annual license fees, ongoing royalties based on net sales generated by CureCell and its sublicensees, milestone payments and sublicense fees. Under the agreement, CureCell is entitled to share in the net profits derived by the Israeli Subsidiary from world-wide sales (except for sales in Korea) of any product developed as a result of the collaboration with CureCell. Additionally, CureCell was given the first right to obtain exclusive commercialization rights in Japan of the AIP product, subject to CureCell procuring all the regulatory approvals required for commercialization in Japan. As of November 30, 2018, none of the requisite regulatory approvals for conducting clinical trials had been obtained.

On May 26, 2016, the Israeli Subsidiary and CureCell entered into a pharma CPFA with KORIL. KORIL will give a conditional grant of up to $400 thousand each (according to terms defined in the agreement), for a joint research and development project for the use of AIP Cells for the Treatment of Diabetes (the “KORIL Project”). The KORIL Project started on June 1, 2016. Upon the conclusion of product development, the grant shall be repaid at the yearly rate of 2.5% of gross sales. The grant will be used solely to finance the costs to conduct the research of the project during a period of 18 months starting on June 1, 2016. On July 26, 2018, KORIL approved an extension for the project period until May 31, 2019. As of November 30, 2018, the Israeli Subsidiary and CureCell received $440 thousand under the grant.

Maryland Technology Development Corporation

On June 30, 2014, the Company’s U.S. Subsidiary entered into a grant agreement with Maryland Technology Development Corporation (“TEDCO”). TEDCO was created by the Maryland State Legislature in 1998 to facilitate the transfer and commercialization of technology from Maryland’s research universities and federal labs into the marketplace and to assist in the creation and growth of technology based businesses in all regions of the State. TEDCO is an independent organization that strives to be Maryland’s lead source for entrepreneurial business assistance and seed funding for the development of startup companies in Maryland’s innovation economy. TEDCO administers the Maryland Stem Cell Research Fund to promote State funded stem cell research and cures through financial assistance to public and private entities within the State. Under the agreement, TEDCO has agreed to give the U.S. Subsidiary an amount not to exceed approximately $406 thousand (the “Grant”). The Grant will be used solely to finance the costs to conduct the research project entitled “Autologous Insulin Producing (AIP) Cells for Diabetes” during a period of two years. On June 21, 2016, TEDCO approved an extension for the project period until June 30, 2017.

On July 22, 2014 and September 21, 2015, the U.S. Subsidiary received an advance payment of $406 thousand on account of the Grant. Through November 30, 2018, the Company utilized $356 thousand from the grant and recorded it as a deduction of research and development expenses in the statement of comprehensive loss.

Research and Development

We incurred $7,386 and $3,326 thousand in research and development expenditures in the fiscal years ended November 30, 2018 and 2017, respectively, of which $922 thousand and $848 thousand was covered by grant funding. The increase in research and development expenses was due to an increase in salaries and related expenses for the year ended November 30, 2018, as compared to 2017 and reflects management’s plan to move our transdifferentiation technology to the next the stage towards clinical studies. In the fiscal year ended 2018, we focused mainly on setting up infrastructure and regulatory approvals for sourcing of liver tissue and biopsies and combining the in-vitro research to increase insulin production and secretion with our pre-clinical studies’ aim to evaluate the efficacy and safety of the product in animal models. In this respect, new transdifferentiation methods are being evaluated. Sourcing of the starting material (liver sampling and cell collection) and upscaling of virus production and cell propagation using advance technologies complement this effort with the target to establish start to end production capabilities. Our research and development scope was also expanded to the evaluation and development of new cell therapies related technologies in the field of immunoncology, liver pathologies and tissue regeneration.

-27-

Intellectual Property

We will be able to protect our technology and products from unauthorized use by third parties only to the extent it is covered by valid and enforceable claims of our patents or is effectively maintained as trade secrets. Patents and other proprietary rights are thus an essential element of our business.

Our success will depend in part on our ability to obtain and maintain proprietary protection for our product candidates, technology, and know-how, to operate without infringing on the proprietary rights of others, and to prevent others from infringing it proprietary rights. Our policy is to seek to protect our proprietary position by, among other methods, filing U.S. and foreign patent applications related to our proprietary technology, inventions, and improvements that are important to the development of our business. We also rely on trade secrets, know-how, continuing technological innovation, and in-licensing opportunities to develop and maintain our proprietary position.

We own or have exclusive rights to ten (10) United States and nineteen (19) foreign issued patents, nine (9) pending applications in the United States, thirty-two (32) pending applications in foreign jurisdictions, including Europe, Australia, Brazil, Canada, China, Eurasia, Israel, Japan, South Korea, Mexico, and Singapore, and four (4) international Patent Cooperation Treaty (“PCT”) patent applications. These patents and applications relate, among others, to (1) the trans-differentiation of cells (including hepatic cells) to cells having pancreatic β-cell-like phenotype and function and to their use in the treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis, and (2) scaffolds, including alginate and sulfated alginate scaffolds, polysaccharides thereof, and scaffolds for use for cell propagation, transdifferentiation, and transplantation in the treatment of autoimmune diseases, including diabetes.

Granted U.S. patents, which are directed among others to compositions comprising sulfated polysaccharide bioconjugates, modified polysaccharides, and epithelial organoids having liver phenotype, will expire between 2025 and 2027, excluding any patent term extensions that might be available following the grant of marketing authorizations. Patents granted in Australia, France, Germany, Israel, Switzerland, and the United Kingdom, which are directed among others to compositions comprising sulfated polysaccharide bioconjugate, and to epithelial organoids having liver phenotype, will expire between 2025 and 2027, excluding any patent term extensions that might be available following the grant of marketing authorizations. Granted U.S. patents, which are directed, among others, to methods of inducing pancreatic hormone expression, methods of inducing a beta cell phenotype, methods for transdifferentiating cells, and methods of producing hydrogels, will expire between 2020 and 2035, excluding any patent term extensions that might be available following the grant of marketing authorizations. Patents granted in Australia, Canada, France, Germany, Israel, Italy, and the United Kingdom, which are directed, among others, to methods of inducing pancreatic hormone expression, methods of inducing a beta cell phenotype, and methods of producing hydrogels, will expire between 2020 and 2024, excluding any patent term extensions that might be available following the grant of marketing authorizations. A granted U.S. patent which is directed, among others, to components of a bioreactor will expire in 2024, excluding any patent term extensions that might be available following the grant of marketing authorizations. Patents granted in Austria, France, Germany, Israel, and the United Kingdom, which are directed, among others, to components of a bioreactor, will expire in 2024, excluding any patent term extensions that might be available following the grant of marketing authorizations.

-28-

We have pending U.S. patent applications directed, among others, to compositions comprising clusters of transdifferentiated cells, modified polysaccharides, and dermatological compositions. If issued, these applications would expire between 2036 and 2038, excluding any patent term adjustment that might be available following the grant of the patent and any patent term extensions that might be available following the grant of marketing authorizations. We have pending patent applications in Australia, Brazil, Canada, China, Eurasia, Europe, Israel, Japan, the Republic of Korea, Mexico and Singapore directed, among others, to compositions comprising sulfated polysaccharide bioconjugates, modified polysaccharides, and multi-compartment hydrogel. If issued, these applications would expire between 2026 and 2036, excluding any patent term adjustment that might be available following the grant of the patent and any patent term extensions that might be available following the grant of marketing authorizations. We have pending U.S. patent applications directed, among others, to methods of isolating cells predisposed to transdifferentiation, methods of manufacturing insulin producing cells, and methods for treating autoimmune diseases. If issued, these applications would expire between 2035 and 2037, excluding any patent term adjustment that might be available following the grant of the patent and any patent term extensions that might be available following the grant of marketing authorizations. We have pending patent applications in Australia, Brazil, Canada, China, Eurasia, Europe, Israel, Japan, the Republic of Korea, Mexico and Singapore directed, among others, to methods of producing transdifferentiated cells having beta cell phenotype, methods for treating a liver disease, and methods for treating autoimmune disorders. If issued, these applications would expire between 2035 and 2038, excluding any patent term adjustment that might be available following the grant of the patent and any patent term extensions that might be available following the grant of marketing authorizations. We have PCT applications directed, among others, to compositions comprising clusters of transdifferentiated cells, compositions comprising vascular secretome components, methods of producing thereof, and methods for treating liver diseases with the compositions thereof. If issued, National Phase applications claiming benefit of those PCT applications would expire in 2038, excluding any patent term adjustment that might be available following the grant of the patent and any patent term extensions that might be available following the grant of marketing authorizations.

Government Regulation

We have not sought approval from the FDA for the AIP cells. Among all forms of cell therapy modalities, we believe that autologous cell replacement therapy is of the highest benefit. We believe that it is safer than other options as it does not alter the host genome but only alters the set of expressed epigenetic information that seems to be highly specific to the reprogramming protocol. It provides an abundant source of therapeutic tissue, which is not rejected by the patient and does not have to be treated by immune suppressants. It is highly ethical because no human organ donations or embryo-derived cells are needed. The proposed therapeutic approach does not require cell bio-banking at birth, which is both expensive and cannot be used for patients born prior to 2000.

Over the past decade, many studies published in leading scientific journals confirmed the capacity of reprogramming adult cells from many of our mature organs to either alternate organs or to “stem like cells”. Most widely used autologous cell replacement protocols are used for autologous implantation of bone marrow stem cells. This protocol is widely used in patients undergoing a massive chemotherapy session that destroys their bone marrow cells. However, the stem cells used for cancer patients delineated above do not require extensive manipulation and is regarded by the FDA as “minimally manipulated.”

An additional autologous cell therapy approach already used in man is autologous chondrocyte implantation (“ACI”). In the United States, Genzyme Corporation provides the only FDA approved ACI treatment called Carticel. The Carticel treatment is designated for young, healthy patients with medium to large sized damage to cartilage. During an initial procedure, the patient’s own chondrocytes are removed arthroscopically from a non-load-bearing area from either the intercondylar notch or the superior ridge of the medial or lateral femoral condyles.

To aid us in our efforts to achieve the highest level of compliance with FDA requirements, we have looked to hire experts in the field of pharmaceutical compliance.

Regulatory Process in the United States

Our potential product iscandidates are subject to regulation as a biological product under the Public Health Service Act and the Food, Drug and Cosmetic Act. The FDA generally requires the following steps for pre-market approval or licensure of a new biological product:

·Pre-clinical laboratory and animal tests conducted in compliance with the Good Laboratory Practice, or GLP, requirements to assess a drug’s biological activity and to identify potential safety problems, and to characterize and document the product’s chemistry, manufacturing controls, formulation, and stability;

-29-
19

·Submission to the FDA of an Investigational New Drug, or IND, application, which must become effective before clinical testing in humans can start;
·Obtaining approval of Institutional Review Boards, or IRBs, of research institutions or other clinical sites to introduce a first human biologic drug candidate into humans in clinical trials;
·Conducting adequate and well-controlled human clinical trials to establish the safety and efficacy of the product for its intended indication conducted in compliance with Good Clinical Practice, or GCP, requirements;
·Compliance with current Good Manufacturing Practices (“cGMP”) regulations and standards;
·Submission to the FDA of a Biologics License Application (“BLA”) for marketing that includes adequate results of pre-clinical testing and clinical trials;
·The FDA reviews the marketing application in order to determine, among other things, whether the product is safe, effective and potent for its intended uses; and
·Obtaining FDA approval of the BLA, including inspection and approval of the product manufacturing facility as compliant with cGMP requirements, prior to any commercial sale or shipment of the pharmaceutical agent. The FDA may also require post marketing testing and surveillance of approved products or place other conditions on the approvals.

Regulatory Process in Europe

The European Union (“EU”) has approved a regulation specific to cell and tissue therapy product,products, the Advanced Therapy Medicinal Product (“ATMP”) regulation. For products such as our AIP cells that are regulated as an ATMP, the EU directive requires:

·Compliance with current cGMP regulations and standards, pre-clinical laboratory and animal testing;
·Filing a Clinical Trial Application (“CTA”) with the various member states or a centralized procedure;
·Voluntary Harmonization Procedure (“VHP”), a procedure which makes it possible to obtain a coordinated assessment of an application for a clinical trial that is to take place in several European countries;
·Obtaining approval of ethic committees of research institutions or other clinical sites to introduce the AIP into humans in clinical trials;
·Adequate and well-controlled clinical trials to establish the safety and efficacy of the product for its intended use;
·Submission to EMEA for a Marketing Authorization (“MA”); and
·Review and approval of the MAA (“Marketing Authorization Application”).

As in the U.S., prior to the general regulatory process of a new biologic products, we will prosecute an Orphan Drug Designation for treatment of Patients with Established Diabetes Mellitus (“DM”) Induced by Total pancreatectomy. In the EU, in order to be qualified, the prevalence must be below 5 per 10,000 of the EU population, except where the expected return on investment is insufficient to justify the investment.

Authorized orphan medicines benefit from 10 years of protection from market competition with similar medicines with similar indications once they are approved. Companies applying for designated orphan medicines pay reduced fees for regulatory activities. This includes reduced fees for protocol assistance, marketing-authorization applications, inspections before authorization, applications for changes to marketing authorizations made after approval, and reduced annual fees.

Clinical Trials

Typically, both in the U.S. and the EU, clinical testing involves a three-phase process, although the phases may overlap. In Phase I, clinical trials are conducted with a small number of healthy volunteers or patients and are designed to provide information about product safety and to evaluate the pattern of drug distribution and metabolism within the body. In Phase II, clinical trials are conducted with groups of patients afflicted with a specific disease in order to determine preliminary efficacy, optimal dosages and expanded evidence of safety. In some cases, an initial trial is conducted in diseased patients to assess both preliminary efficacy and preliminary safety and patterns of drug metabolism and distribution, in which case it is referred to as a Phase I/II trial. Phase III clinical trials are generally large-scale, multi-center, comparative trials conducted with patients afflicted with a target disease in order to provide statistically valid proof of efficacy, as well as safety and potency. In some circumstances, the FDA or EMA may require Phase IV or post-marketing trials if it feels that additional information needs to be collected about the drug after it is on the market. During all phases of clinical development, regulatory agencies require extensive monitoring and auditing of all clinical activities, clinical data, as well as clinical trial investigators. An agency may, at its discretion, re-evaluate, alter, suspend, or terminate the testing based upon the data that have been accumulated to that point and its assessment of the risk/benefit ratio to the patient. Monitoring all aspects of the study to minimize risks is a continuing process. All adverse events must be reported to the FDA or EMAEMA.

20

The FDA has granted Orphan Drug designation for our AIP cells as a cell replacement therapy for the treatment of severe hypoglycemia-prone diabetes resulting from TP due to chronic pancreatitis. The FDA’s Orphan Drug Designation Program provides orphan status to drugs and biologics which are defined as those intended for the safe and effective treatment, diagnosis or prevention of rare diseases/disorders that affect fewer than 200,000 people in the United States. Orphan designation qualifies the sponsor of the drug for various development incentives, including eligibility for seven years of market exclusivity upon regulatory approval, exemption from FDA application fees, tax credits for qualified clinical trials, and other potential assistance in the drug development process.

-30-

Employees

As of November 30, 2018,December 31, 2020, we had an aggregate of 231111 employees working at our Companycompany and subsidiaries. In addition, we retain the services of outside consultants for various functions including clinical work, finance, accounting and business development services. Most of our senior management and professional employees have had prior experience in pharmaceutical or biotechnology companies. None of our employees are covered by collective bargaining agreements. We believe that we have good relations with our employees.

Subsidiaries

Orgenesis Inc. is a Nevada corporation, and our subsidiaries currently consist of Masthercell Global Inc., a Delaware corporation (“Masthercell Global”), Orgenesis SPRL, a Belgian-based entity (the “Belgian Subsidiary ”), Orgenesis Ltd., an Israeli corporation (the “Israeli Subsidiary”), and Orgenesis Maryland Inc., a Maryland corporation.  Masthercell Global’s wholly-owned subsidiaries include MaSTherCell S.A. (“MaSTherCell”), a Belgian-based entity, Cell Therapy Holdings S.A., a Belgian-based entity, Masthercell U.S., LLC, a U.S.-based entity, Atvio Biotech Ltd. (“Atvio”), an Israeli-based CDMO, and CureCell Co. Ltd. (“CureCell”), a Korea-based CDMO (Orgenesis owned 94.12% of CureCell which was consolidated into Masthercell Global).

The corporate organization diagram below shows how we classify each subsidiary and each joint venture partner between its two business units:

Corporate and Available Information

Our annual reportAnnual Report on Form 10-K, quarterly reportsQuarterly Reports on Form 10-Q, current reportsCurrent Reports on Form 8-K, and all amendments to those reports are available free of charge though our website (http://www.orgenesis.com) as soon as practicable after such material is electronically filed with, or furnished to, the Securities and Exchange Commission (the “SEC”). Except as otherwise stated in these documents, the information contained on our website or available by hyperlink from our website is not incorporated by reference into this report or any other documents we file, with or furnish to, the SEC.

-31-

Our common stock is listed and traded on the Nasdaq Capital Market under the symbol “ORGS.”

As used in this Annual Report on Form 10-K and unless otherwise indicated, the term “Company” refers to Orgenesis Inc. and its Subsidiaries. Unless otherwise specified, all amounts are expressed in United States Dollars.

ITEM 1A. RISK FACTORS

Summary of Risk Factors

Below is a summary of the principal factors that make an investment in our common stock speculative or risky. This summary does not address all of the risks that we face. Additional discussion of the risks summarized in this risk factor summary, and other risks that we face, can be found below under the heading “Risk Factors” and should be carefully considered, together with other information in this Annual Report on Form 10-K and our other filings with the SEC, before making an investment decision regarding our common stock.

The failure to effectively utilize the proceeds from the sale of Masthercell to scale our POC business to show demonstrable revenue may adversely affect our business.
Our research and development efforts on novel technology using cell-based therapy and our future success is highly dependent on the successful development of that technology.

21

We have entered into collaborations and may form or seek collaborations or strategic alliances or enter into additional licensing arrangements in the future, and we may not realize the benefits of such alliances or licensing arrangements.
Our success will depend on strategic collaborations with third parties to develop and commercialize therapeutic product candidates, and we may not have control over a number of key elements relating to the development and commercialization of any such product candidate.
The coronavirus outbreak has the potential to cause disruptions in our business, including our clinical development activities.
Our success depends on our ability to protect our intellectual property and our proprietary technologies.
If product liability lawsuits are brought against us, we may incur substantial liabilities and may be required to limit commercialization of our product candidates.
We are increasingly dependent on information technology and our systems and infrastructure face certain risks, including cybersecurity and data storage risks.
There can be no assurance that we will be able to develop in-house sales and commercial distribution capabilities or establish or maintain relationships with third-party collaborators to successfully commercialize any product in the United States or overseas, and as a result, we may not be able to generate product revenue.
Our product candidates may cause undesirable side effects or have other properties that could halt their clinical development, prevent their regulatory approval, limit their commercial potential, or result in significant negative consequences.
Our product candidates are biologics and the manufacture of our product candidates is complex and we may encounter difficulties in production, particularly with respect to process development or scaling-out of our manufacturing capabilities.
Cell-based therapies rely on the availability of reagents, specialized equipment, and other specialty materials, which may not be available to us on acceptable terms or at all. For some of these reagents, equipment, and materials, we rely or may rely on sole source vendors or a limited number of vendors, which could impair our ability to manufacture and supply our products.
We currently have no marketing and sales organization and have no experience in marketing therapeutic products. If we are unable to establish marketing and sales capabilities or enter into agreements with third parties to market and sell our product candidates, we may not be able to generate product revenue.
There can be no assurance that we will be able to develop in-house sales and commercial distribution capabilities or establish or maintain relationships with third-party collaborators to successfully commercialize any product in the United States or overseas, and as a result, we may not be able to generate product revenue.
We face significant competition from other biotechnology and pharmaceutical companies, many of which have substantially greater financial, technical and other resources, and our operating results will suffer if we fail to compete effectively.
We are highly dependent on key personnel who would be difficult to replace, and our business plans will likely be harmed if we lose their services or cannot hire additional qualified personnel.
Extensive industry regulation has had, and will continue to have, a significant impact on our business, especially our product development, manufacturing and distribution capabilities.
Third parties to whom we may license or transfer development and commercialization rights for products covered by intellectual property rights may not be successful in their efforts and, as a result, we may not receive future royalty or other milestone payments relating to those products or rights.

22

Risk Factors

An investment in our common stock involves a number of very significant risks. You should carefully consider the following risks and uncertainties in addition to other information in this report in evaluating our company and its business before purchasing shares of our company’s common stock. Our business, operating results and financial condition could be seriously harmed due to any of the following risks. You could lose all or part of your investment due to any of these risks.

Risks Related to Our Company and POC Business

We will need to raisedeploy our capital from the sale of Masthercell in ordera manner to realizescale our POC business plan,to show demonstrable revenue and market value for our shareholders, the failure of which could adversely impact our operations.operations and the price of our stock.

On February 2, 2020, we entered into a Stock Purchase Agreement (the “Purchase Agreement”) with GPP-II Masthercell LLC (“GPP” and together with Orgenesis, the “Sellers”), Masthercell Global Inc. (“Masthercell”) and Catalent Pharma Solutions, Inc. (the “Buyer”). Pursuant to the terms and conditions of the Purchase Agreement, on February 10, 2020, the Sellers sold 100% of the outstanding equity interests of Masthercell to Buyer (the “Sale”) for an aggregate nominal purchase price of $315 million, subject to customary adjustments. After accounting for GPP’s liquidation preference and equity stake in Masthercell as well as SFPI - FPIM’s interest in MaSTherCell S.A., distributions to Masthercell option holders and transaction costs, we received approximately $126.7 million at the closing of the Sale transaction, of which $7.2 million was used for the repayment of intercompany loans and payables.

The proceeds from the sale of Masthercell were received by us and not our shareholders. We used or will use a portion of the proceeds to repay certain outstanding indebtedness and to pay for certain additional transaction costs associated with the sale. We will also be paying taxes on the proceeds.

We expect to use the remainder of net proceeds from the sale of Masthercell, at the discretion of our Board of Directors, for working capital and other general corporate purposes, including to continue to grow our POC cell therapy business and to further the development of ATMPs. Although we currentlynow have sufficient capital resources for the next 12 months without adequate funding or a significant increase in revenues,and the foreseeable future, we may not be able to expand our global CDMO network, establish additional CDMO facilities in the United States or other parts of the world, implement our POCare therapyPOC business seek out strategic CDMO acquisitions,and commence clinical trials for our diabetes solution or respond to competitive pressures. Aspressures due to other non-financial factors beyond our control. Our failure to effectively utilize the proceeds from the sale of November 30, 2018, we had available cash resources of $16.1 million.

Overall, we have funded our cash needs from inception through the date hereof with a series of debt and equity transactions, grants from governmental agencies and, more recently, through cash flow from our revenue generating operations from Masthercell Global.

We expect to continue to finance our operations, acquisitions and develop strategic relationships, primarily by issuing equity or convertible debt securities, which could significantly reduce the percentage ownership of our existing stockholders. Furthermore, any newly issued securities could have rights, preferences and privileges senior to those of our existing common stock. Moreover, any issuances by us of equity securities may be at or below the prevailing market price of our common stock and in any event may have a dilutive impact on your ownership interest, which could cause the market price of our common stock to decline. We may also issue securities in one or more of our subsidiaries, and these securities may have rights or privileges senior to those of our common stock.

We may have difficulty obtaining additional funds as and when needed, and we may have to accept terms that would adversely affect our stockholders. In addition, any adverse conditions in the credit and equity markets may adversely affect our ability to raise funds when needed. Any failuresuccessfully grow our POC business and develop cell therapy product candidates, which could cause the value of your investment in Orgenesis to achieve adequate funding will delay our development programs and product launches and could lead to abandonment of one or more of our development initiatives, as well as prevent us from responding to competitive pressures or take advantage of unanticipated acquisition opportunities. Any additional equity financing will likely be dilutive to stockholders, and certain types of equity financing, if available, may involve restrictive covenants or other provisions that would limit how we conduct our business or finance our operations.decline.

We are not profitable as of November 30, 2018, December 31, 2020, have limited cash flow and, unless we increase revenues and cash flow or raise additional capital, we may be unable to take advantage of any commercial opportunities that arise orto expand CDMO operations, allour POC business, the perceived value of whichour company may decrease and our stock price could adversely impact us.be affected accordingly.

For the fiscal year ended November 30, 2018December 31, 2020 and as of the date of this report, we assessed our financial condition and concluded that we have sufficient resources for the next 12 months from the date of the report.report as a result of the receipt of the net proceeds from the sale of Masthercell. Our auditor’s report for the year ended November 30, 2018December 31, 2020 does not include a going concern opinion on the matter. However, management is still required to assess our ability to continue as a going concern. We had a net loss of $19.1 million for the year ended November 30, 2018. During the same period, cash used in operations was $15.7 million, the working capital surplus and accumulated deficit as of November 30, 2018 were $13.2 million and $62.4 million, respectively. Management is unable to predict if and when we will be able to generate significant positive cash flowrevenues or achieve profitability. Our plan regarding these matters is to strengthen our revenues and continue improving the net results in the CDMO segment and to raise additional equity financing to allow us the ability to cover our cash flow requirementsPOC business into fiscal year 2019.2021. There can be no assurancesassurance that we will be successful in increasing revenues, improving CDMO segmentour POC results or that financingthe perceived value of our company will be available or, if available, that such financing will be available under favorable terms.increase. In the event that we are unable to generate adequatesignificant revenues in our POC business, our stock price could be adversely affected.

23

Our research and development programs are based on novel technologies and are inherently risky.

We are subject to cover expensesthe risks of failure inherent in the development of products based on new technologies. The novel nature of our cell therapy technology creates significant challenges with respect to product development and cannotoptimization, manufacturing, government regulation and approval, third-party reimbursement and market acceptance. For example, the FDA and EMA have relatively limited experience with the development and regulation of cell therapy products and, therefore, the pathway to marketing approval for our cell therapy product candidates may accordingly be more complex, lengthy and uncertain than for a more conventional product candidate. The indications of use for which we choose to pursue development may have clinical effectiveness endpoints that have not previously been reviewed or validated by the FDA or EMA, which may complicate or delay our effort to ultimately obtain additional financing into fiscal year 2019,FDA or EMA approval. Because this is a new approach to treating diseases, developing and commercializing our product candidates subjects us to a number of challenges, including:

obtaining regulatory approval from the FDA, EMA and other regulatory authorities that have very limited experience with the commercial development of our technology for treating different diseases;
developing and deploying consistent and reliable processes for removing the cells from the patient engineering cells ex vivo and infusing the engineered cells back into the patient;
developing processes for the safe administration of these products, including long-term follow-up for all patients who receive our products;
sourcing clinical and, if approved, commercial supplies for the materials used to manufacture and process our products;
developing a manufacturing process and distribution network with a cost of goods that allows for an attractive return on investment;
establishing sales and marketing capabilities after obtaining any regulatory approval to gain market acceptance; and
maintaining a system of post marketing surveillance and risk assessment programs to identify adverse events that did not appear during the drug approval process.

Our efforts to overcome these challenges may not prove successful, and any product candidate we seek to develop may not be successfully developed or commercialized.

Kyslecel may not achieve patient or market acceptance, which could have a material adverse effect on our business.

Our commercialization strategy for Kyslecel relies on medical specialists, medical facilities and patients adopting TP-IAT with Kyslecel as an accepted treatment for chronic pancreatitis. However, medical specialists are historically slow to adopt new treatments, regardless of perceived merits, when older treatments continue to be supported by established providers. Overcoming such resistance often requires significant marketing expenditure or definitive product performance and/or pricing superiority. The cost of allocating resources for such requirements might severely impact the potential for profitability of Kyslecel.

There is no guarantee that physician or patient acceptance of TP-IAT with Kyslecel will be substantial. Further, there is no guarantee that Koligo will be able to achieve patient acceptance or obtain enough customers (clinical providers) to meet its sales objectives. If we do not meet our sales objectives, our business prospects and financial performance will be materially and adversely affected.

Further, we are partially reliant on published clinical trials and scientific research conducted by third parties to justify the patient benefit and safety of TP-IAT with Kyslecel and, as such, we rely, in part, on the accuracy and integrity of those third-parties to have reported the results and correctly collected and interpreted the data from all clinical trials conducted to date. If published data turn out to later be incorrect or incomplete, our business prospects and financial performance may be materially and adversely affected.

24

The therapeutic efficacy of ranpirnase and our other product candidates is unproven in humans, and we may not be able to successfully develop and commercialize ranpirnase or any of our other product candidates.

Ranpirnase and our other product candidates are novel compounds and their potential benefit as antiviral drugs or immunotherapies is unproven. Ranpirnase and our other product candidates may not prove to be effective against the indications for which they are being designed to act and may not demonstrate in clinical trials any or all of the pharmacological effects that have been observed in preclinical studies. As a result, our clinical trial results may not be indicative of the results of future clinical trials.

Ranpirnase and our other product candidates may interact with human biological systems in unforeseen, ineffective or harmful ways. If ranpirnase or any of our other product candidates is associated with undesirable side effects or have characteristics that are unexpected, we may need to cut backabandon the development of such product candidate or curtaillimit development to certain uses or subpopulations in which the undesirable side effects or other characteristics are less prevalent, less severe or more acceptable from a risk-benefit perspective. Because of these and other risks described herein that are inherent in the development of novel therapeutic agents, we may never successfully develop or commercialize ranpirnase or any of our expansion plans.other product candidates, in which case our business will be harmed.

-32-

We will need to grow the size and capabilities of our organization, and we may experience difficulties in managing this growth.

As of November 30, 2018,December 31, 2020, we had 231111 employees. As our development and commercialization plans and strategies develop, we must add a significant number of additional managerial, operational, sales, marketing, financial, and other personnel. Future growth will impose significant added responsibilities on members of management, including:

·identifying, recruiting, integrating, maintaining, and motivating additional employees;
·managing our internal development efforts effectively, including the clinical and FDA review process for our product candidates, while complying with our contractual obligations to contractors and other third parties; and
·improving our operational, financial and management controls, reporting systems, and procedures.

Our future financial performance and our ability to commercialize our product candidates will depend, in part, on our ability to effectively manage any future growth, and our management may also have to divert a disproportionate amount of its attention away from day-to-day activities in order to devote a substantial amount of time to managing these growth activities. This lack of long-term experience working together may adversely impact our senior management team’s ability to effectively manage our business and growth.

We currently rely, and for the foreseeable future will continue to rely, in substantial part on certain independent organizations, advisors and consultants to provide certain services. There can be no assurance that the services of these independent organizations, advisors and consultants will continue to be available to us on a timely basis when needed, or that we can find qualified replacements. In addition, if we are unable to effectively manage our outsourced activities or if the quality or accuracy of the services provided by consultants is compromised for any reason, our clinical trials may be extended, delayed, or terminated, and we may not be able to obtain regulatory approval of our product candidates or otherwise advance our business. There can be no assurance that we will be able to manage our existing consultants or find other competent outside contractors and consultants on economically reasonable terms, if at all. If we are not able to effectively expand our organization by hiring new employees and expanding our groups of consultants and contractors, we may not be able to successfully implement the tasks necessary to further develop and commercialize our product candidates and, accordingly, may not achieve our research, development, and commercialization goals.

We depend on key personnel who would be difficult to replace, and our business plans will likely be harmed if we lose their services or cannot hire additional qualified personnel.

Our success depends substantially on the efforts and abilities of our senior management and certain key personnel. The competition for qualified management and key personnel, especially engineers, is intense. The loss of services of one or more of our key employees, or the inability to hire, train, and retain key personnel, especially engineers and technical support personnel, could delay the development and sale of our products, disrupt our business, and interfere with our ability to execute our business plan.

Currency exchange fluctuations may impact the results of our operations.

The provision of services by our former subsidiary, Masthercell Global, arewere usually transacted in U.S. dollars and European currencies.currencies during the year ended December 31, 2020. Our results of operations are affected by fluctuations in currency exchange rates in both sourcing and selling locations. Although we enter into foreign currency exchange forward contracts from time to time to reduce our risk related to currency exchange fluctuation, ourOur results of operations may still be impacted by foreign currency exchange rates, primarily, the euro-to-U.S. dollar exchange rate. In recent years, the euro-to-U.S. dollar exchange rate has been subject to substantial volatility which may continue, particularly in light of recent political events regarding the European Union, or EU. Because we do not hedge against all of our foreign currency exposure, our business will continue to be susceptible to foreign currency fluctuations.

-33-
25

We have entered into collaborations and may form or seek collaborations or strategic alliances or enter into additional licensing arrangements in the future, and we may not realize the benefits of such alliances or licensing arrangements.

We have entered into collaborations and joint ventures and may form or seek strategic alliances, create joint ventures or collaborations, or enter into additional licensing arrangements with third parties that we believe will complement or augment our development and commercialization efforts with respect to our product candidates and any future product candidates that we may develop. Any of these relationships may require us to incur non-recurring and other charges, increase our near and long-term expenditures, issue securities that dilute our existing stockholders, or disrupt our management and business. In addition, we face significant competition in seeking appropriate strategic partners for which the negotiation process is time-consuming and complex. Moreover, we may not be successful in our efforts to establish a strategic partnership or other alternative arrangements for our product candidates because they may be deemed to be at too early of a stage of development for collaborative effort and third parties may not view our product candidates as having the requisite potential to demonstrate safety and efficacy. Further, collaborations involving our product candidates, such as our collaborations with third-party research institutions, are subject to numerous risks, which may include the following:

·collaborators have significant discretion in determining the efforts and resources that they will apply to a collaboration;
·collaborators may not perform their obligations as expected;
collaborators may not pursue development and commercialization of our product candidates or may elect not to continue or renew development or commercialization programs based on clinical trial results, changes in their strategic focus due to the acquisition of competitive products, availability of funding, or other external factors, such as a business combination that diverts resources or creates competing priorities;
·collaborators may delay clinical trials, provide insufficient funding for a clinical trial, stop a clinical trial, abandon a product candidate, repeat or conduct new clinical trials, or require a new formulation of a product candidate for clinical testing;
·collaborators could fail to make timely regulatory submissions for a product candidate;
collaborators may not comply with all applicable regulatory requirements or may fail to report safety data in accordance with all applicable regulatory requirements;
collaborators could independently develop, or develop with third parties, products that compete directly or indirectly with our products or product candidates;
·product candidates developed in collaboration with us may be viewed by our collaborators as competitive with their own product candidates or products, which may cause collaborators to cease to devote resources to the commercialization of our product candidates;
a collaborator with marketing and distribution rights to one or more products may not commit sufficient resources to their marketing and distribution;
·collaborators may not properly maintain or defend our intellectual property rights or may use our intellectual property or proprietary information in a way that gives rise to actual or threatened litigation that could jeopardize or invalidate our intellectual property or proprietary information or expose us to potential liability;
·disputes may arise between us and a collaborator that cause the delay or termination of the research, development or commercialization of our product candidates, or that result in costly litigation or arbitration that diverts management attention and resources;
·collaborations may be terminated and, if terminated, may result in a need for additional capital to pursue further development or commercialization of the applicable product candidates; and
·collaborators may own or co-own intellectual property covering our products that results from our collaborating with them and, in such cases, we would not have the exclusive right to commercialize such intellectual property.

As a result, if we enter into collaboration agreements and strategic partnerships or license our products or businesses, we may not be able to realize the benefit of such transactions if we are unable to successfully integrate them with our existing operations and company culture, which could delay our timelines or otherwise adversely affect our business. The success of our existing and future collaboration arrangements and strategic partnerships, which include research and development services by our collaborators to improve our intellectual property, will depend heavily on the efforts and activities of our collaborators and may not be successful. We also cannot be certain that, following a strategic transaction or license, we will achieve the revenue or specific net income that justifies such transaction. Any delays in entering into new collaborations or strategic partnership agreements related to our product candidates could delay the development and commercialization of our product candidates in certain geographies for certain indications, which would harm our business prospects, financial condition, and results of operations.

-34-
26

Our success will depend on strategic collaborations with third parties to develop and commercialize therapeutic product candidates, and we may not have control over a number of key elements relating to the development and commercialization of any such product candidate.

A key aspect of our strategy is to seek collaborations with partners, such as a large pharmaceutical organization, that are willing to further develop and commercialize a selected product candidate. To date, we have entered into a number of collaborative arrangements with cell therapy organizations. By entering into any such strategic collaborations, we may rely on our partner for financial resources and for development, regulatory and commercialization expertise. Our partner may fail to develop or effectively commercialize our product candidate because they:

do not have sufficient resources or decide not to devote the necessary resources due to internal constraints such as limited cash or human resources;
decide to pursue a competitive potential product developed outside of the collaboration;
cannot obtain the necessary regulatory approvals;
determine that the market opportunity is not attractive; or
cannot manufacture or obtain the necessary materials in sufficient quantities from multiple sources or at a reasonable cost.

We may not be able to enter into additional collaborations on acceptable terms, if at all. We face competition in our search for partners from other organizations worldwide, many of whom are larger and are able to offer more attractive deals in terms of financial commitments, contribution of human resources, or development, manufacturing, regulatory or commercial expertise and support. If we are not successful in attracting a partner and entering into a collaboration on acceptable terms, we may not be able to complete development of or commercialize any product candidate. In such event, our ability to generate revenues and achieve or sustain profitability would be significantly hindered and we may not be able to continue operations as proposed, requiring us to modify our business plan, curtail various aspects of our operations or cease operations.

The coronavirus outbreak has the potential to cause disruptions in our business, including our clinical development activities.

The outbreak of the novel strain of coronavirus, or COVID-19, has currently impacted and may continue to impact our business, including our preclinical studies and clinical trials. COVID-19 has spread to multiple countries, including the United States and Israel, where we conduct most of our operations.

Efforts to contain the spread of COVID-19 have intensified and the United States and Israel, among other countries, have implemented and may continue to implement severe travel restrictions, shelter in place orders, social distancing and delays or cancellations of elective surgeries. These and other disruptions have caused, and may continue to cause, a delay in the supply of consumable goods, which could result in further delays, increased costs to source alternative suppliers and affect our ability to commercialize and develop our product candidates.

The spread of an infectious disease, including COVID-19, may also result in a period of business disruption, and in reduced operations, including employee absenteeism and delays in payments from our customers, any of which could materially affect our business, financial condition and results of operations. Although, as of the date of this Annual Report on Form 10-K, we do not expect any material impact on our long-term activity, the extent to which COVID-19 impacts our business will depend on future developments, which are highly uncertain and cannot be predicted, including new information which may emerge concerning the severity of COVID-19 and the actions to contain COVID-19 or treat its impact, among others.

27

Our success depends on our ability to protect our intellectual property and our proprietary technologies.

Our commercial success depends in part on our ability to obtain and maintain patent protection and trade secret protection for our product candidates, proprietary technologies, and their uses as well as our ability to operate without infringing upon the proprietary rights of others. We can provide no assurance that our patent applications or those of our licensors will result in additional patents being issued or that issued patents will afford sufficient protection against competitors with similar technologies, nor can there be any assurance that the patents issued will not be infringed, designed around or invalidated by third parties. Even issued patents may later be found unenforceable or may be modified or revoked in proceedings instituted by third parties before various patent offices or in courts. The degree of future protection for our proprietary rights is uncertain. Only limited protection may be available and may not adequately protect our rights or permit us to gain or keep any competitive advantage. Composition-of-matter patents on the biological or chemical active pharmaceutical ingredients are generally considered to offer the strongest protection of intellectual property and provide the broadest scope of patent protection for pharmaceutical products, as such patents provide protection without regard to any method of use or any method of manufacturing. While we have an issued patentpatents in the United States with a claim for a composition directed to a vector comprising a promoter linked to a pancreatic and duodenal homeobox 1 (PDX-1) polypeptide, and a carrier, we cannot be certain that the claimclaims in our issued patent will not be found invalid or unenforceable if challenged.

We cannot be certain that the claims in our issued United States methods of use patents will not be found invalid or unenforceable if challenged.

We cannot be certain that the pending applications covering composition-of-matter of our transdifferentiatedamong others the bioconjugates comprising sulfated polysaccharides; ranpirnase and other ribonucleases for treating viral diseases; therapeutic compositions comprising exosomes, bioxomes, and redoxomes; automated devices for supporting cell populationstherapies; immune cells, ribonucleases, or antibodies for treating COVID-19; chimeric antigen receptors (CARs); or cell-conditioned medium will be considered patentable by the United States Patent and Trademark Office (USPTO), and courts in the United States or by the patent offices and courts in foreign countries, nor can we be certain that the claims in our issued patents will not be found invalid or unenforceable if challenged. Even if our patent applications covering populations of transdifferentiated cellsthese inventions issue as patents, the patents protect a specific transdifferentiated cell productproducts and may not be enforced against competitors making and marketing a product that has the same activity. Method-of-use patents protect the use of a product for the specified method or for treatment of a particular indication. ThisThese type of patents may not be enforced against competitors making and marketing a product that has cells that may provideprovides the same activity but is used for a method not included in the patent. Moreover, even if competitors do not actively promote their product for our targeted indications, physicians may prescribe these products “off-label.” Although off-label prescriptions may infringe or contribute to the infringement of method-of-use patents, the practice is common and such infringement is difficult to prevent or prosecute.

WeIn addition, we own or have exclusive rights to ten (10)twenty eight (28) United States (US) patents, onethirty six (36) foreign-issued patents, twenty five (25) pending patent applications in the United States, forty five (45) pending patent applications in foreign jurisdictions, including Australia, Brazil, Canada, China, Europe, Hong Kong, India, Israel, Japan, Mexico, New Zealand, North Korea, Russia, Singapore, South Africa, and South Korea, and two (2) international Patent Cooperation Treaty (“PCT”) patent applications. These patents and patent applications relate, among others, to (1) dendritic and macrophages based vaccines, and their use for treating cancer and viral diseases; (2) compositions comprising ranpirnase and other ribonucleases for treating viral diseases; (3) tumor infiltrating lymphocytes (TILs) and their use for treating cancer; (4) compositions comprising immune cells, ribonucleases, or antibodies for treating COVID-19; (5) whole-cell antiviral vaccines; (6) therapeutic compositions comprising exosomes, bioxomes, and redoxomes; (7)bioreactors for cell culture and automated systems and devices for supporting cell therapies; and(8) scaffolds, including alginate and sulfated alginate scaffolds, polysaccharides thereof, and scaffolds for use for cell propagation, transplantations, and in the treatment of which isautoimmune diseases.

28

We have pending U.S. patent applications directed, among others, to a composition comprising a vector comprising a promoter linked to PDX-1dendritic and having a term of 2021, three (3) having a term of 2023macrophages based vaccines, and directed, among others, to methods of inducing endogenous PDX-1 expressiontheir use for treating cancer and viral diseases. If issued, this application would expire in a human differentiated primary non-pancreatic cell, inducing or enhancing a pancreatic islet cell phenotype in non-pancreatic cells, and increasing PDX-1 induction in non-pancreatic primary cells; one (1) having a term of 2024 and directed, among others, to components of a bioreactor; two (2) having a term of 2026 and directed, among others, to a bioconjugate molecules comprising a sulfated polysaccharide; one (1) having a term of 2027 and directed, among others, to an epithelial organoid; one (1) having a term of 2033 and directed, among others, to methods for producing scaffolds; and one (1) having a term of 2034 and directed, among others, to methods for producing transdifferentiated cells. Further, we2038.

We have exclusive rights to nineteen (19) foreign issued patents (six (6) patents granted in Australia, France, Germany, Israel, Switzerland, and the United Kingdom, having a term between 2025 and 2027,pending U.S. patent applications directed, among others, to compositions comprising sulfated polysaccharide bioconjugate,ranpirnase and to epithelial organoids having liver phenotype; eight (8)other ribonucleases for treating viral diseases. If issued, these applications would expire between 2039 and 2040. Counterpart patents grantedapplications were filed in Australia, Canada, France, Germany, Israel, Italy,China, Europe, Hong Kong, Japan, Mexico, New Zealand, North Korea, Russian Federation, Singapore, South Africa, and were also filed as International (“PCT”) applications. If issued, these applications would expire between 2035 and 2037. These expiration dates do not include any patent term extensions that might be available following the United Kingdom, having a term between 2020 and 2024, andgrant of marketing authorizations.

We have pending U.S. patent applications directed, among others, to methodstherapeutic compositions comprising exosomes, bioxomes, and redoxomes. If issued, these applications would expire in 2040. Counterpart patents applications were filed in Australia, Brazil, Canada, China, Europe, India, Israel, India, Japan and South Korea. If issued, these applications would expire in 2039. These expiration dates do not include any patent term extensions that might be available following the grant of inducing pancreatic hormone expression, methods of inducing a beta cell phenotype, and methods of producing hydrogel; and five (5) patents granted in Austria, France, Germany, Israel, and the United Kingdom, having a term of 2024 andmarketing authorizations.

We have pending U.S. patent applications directed, among others, to components of a bioreactor. We also have nine (9) pendingautomated devices for supporting cell therapies. If issued, these applications in the United States, which if granted would expire between 2035 and 2038.

We have a pending U.S. provisional patent application directed, among others, to tumor infiltrating lymphocytes (TILs) and their use for treating cancer. If converted into a non-provisional application and issued, this application would expire in 2041, without including any patent term extensions that might be available following the grant of 2036-2038; thirty two (32)marketing authorizations.

We have pending applications in foreign jurisdictions, including Europe, Australia, Brazil, Canada, China , Eurasia, Israel, Japan, South Korea, Mexico, and Singapore, which if they were to be granted would have a term of 2026-2036; and four (4) International Patent Cooperation Treaty (“PCT”)U.S. provisional patent applications which if fileddirected, among others, to compositions comprising immune cells, ribonucleases, or antibodies for treating COVID-19. If converted into a non-provisional application and granted as national phase applicationsissued, this application would have aexpire in 2041, without including any patent term extensions that might be available following the grant of 2028. These pending applicationsmarketing authorizations.

Granted U.S. patents, which are directed among others to the trans-differentiation of cells (including hepatic cells) to cells having pancreatic β-cell phenotype and function, and their use in the treatment of degenerative pancreatic disorders including diabetes, pancreatic cancer, and pancreatitis; and to scaffolds, including alginate and sulfated alginate scaffolds, polysaccharides thereof, and scaffolds for use for cell propagation, transdifferentiation,transplantations, and transplantation in the treatment of autoimmune diseases, including diabetes.will expire between 2025 and 2036. Counterpart patents granted in Australia, France, Germany, Israel, Switzerland, and the United Kingdom, will expire between 2026 and 2035. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

 

We have pending U.S. patent applications directed, among others, to bioconjugates comprising sulfated polysaccharides and diverse bioactive peptides, and their use in the treatment of inflammatory conditions. If issued, these applications would expire in 2038. Counterpart patents applications were filed in China, Europe, Israel, Japan, and South Korea. If issued, these applications would expire between 2026 and 2038. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

Orgenesis Ltd, has exclusive rights to six (6) United States patents, fourteen (14) foreign-issued patents, five (5) pending patent applications in the United States, twenty six (26) pending patent applications in foreign jurisdictions, including Australia, Brazil, Canada, China, Europe, India, Israel, Japan, Mexico, Panama, Singapore, and South Korea. These patents and patent applications relate, among others, to the trans-differentiation of cells (including hepatic cells) to cells having pancreatic β-cell-like phenotype and function and to their use in the treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis. Granted U.S. patents, which are directed among others to trans-differentiation to pancreatic β-cell-like phenotype and function cells and to their use in the treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis, will expire between 2024 and 2035. Counterpart patents granted in Australia, France, Germany, Israel, Switzerland, and the United Kingdom, will expire between 2024 and 2035. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

29

Orgenesis Ltd, has pending U.S. patent applications directed, among others, to the trans-differentiation of cells, to cells having pancreatic β-cell-like phenotype and function and to their use in the treatment of degenerative pancreatic disorders, including diabetes, pancreatic cancer and pancreatitis. If issued, these applications would expire between 2038 and 2040. Counterpart patents applications were filed in Australia, Brazil, Canada, China, Europe, India, Israel, Mexico, Panama, Singapore, South Korea, and were also filed as International (“PCT”) applications. If issued, these applications would expire between 2034 and 2039. These expiration dates do not include any patent term extensions that might be available following the grant of marketing authorizations.

The patent application process is subject to numerous risks and uncertainties, and there can be no assurance that we or any of our future development partners will be successful in protecting our product candidates by obtaining and defending patents. These risks and uncertainties include the following:

-35-

·the USPTO and various foreign governmental patent agencies require compliance with a number of procedural, documentary, fee payment and other provisions during the patent process. There are situations in which noncompliance can result in abandonment or lapse of a patent or patent application, resulting in partial or complete loss of patent rights in the relevant jurisdiction. In such an event, competitors might be able to enter the market earlier than would otherwise have been the case;
·patent applications may not result in any patents being issued;
·patents that may be issued or in-licensed may be challenged, invalidated, modified, revoked, circumvented, found to be unenforceable or otherwise may not provide any competitive advantage;
·our competitors, many of whom have substantially greater resources and many of whom have made significant investments in competing technologies, may seek or may have already obtained patents that will limit, interfere with or eliminate our ability to make, use, and sell our potential product candidates;
·there may be significant pressure on the U.S. government and international governmental bodies to limit the scope of patent protection both inside and outside the United States for disease treatments that prove successful, as a matter of public policy regarding worldwide health concerns; and
·countries other than the United States may have patent laws less favorable to patentees than those upheld by U.S. courts, allowing foreign competitors a better opportunity to create, develop and market competing product candidates.

In addition, we rely on the protection of our trade secrets and proprietary know-how. Although we have taken steps to protect our trade secrets and unpatented know-how, including entering into confidentiality agreements with third parties, and confidential information and inventions agreements with employees, consultants and advisors, we cannot provide any assurances that all such agreements have been duly executed, and third parties may still obtain this information or may come upon this or similar information independently. Additionally, if the steps taken to maintain our trade secrets are deemed inadequate, we may have insufficient recourse against third parties for misappropriating its trade secrets. If any of these events occurs or if we otherwise lose protection for our trade secrets or proprietary know-how, our business may be harmed.

30

 

If product liability lawsuits are brought against us, we may incur substantial liabilities and may be required to limit commercialization of our product candidates.

We face an inherent risk of product liability as a result of the clinical testing of our product candidates and will face an even greater risk if we commercialize any products. For example, we may be sued if our product candidates cause or are perceived to cause injury or are found to be otherwise unsuitable during clinical testing, manufacturing, marketing or sale. Any such product liability claims may include allegations of defects in manufacturing, defects in design, a failure to warn of dangers inherent in the product, negligence, strict liability or a breach of warranties. Claims could also be asserted under state consumer protection acts. If we cannot successfully defend ourselves against product liability claims, we may incur substantial liabilities or be required to limit commercialization of our product candidates. Even a successful defense would require significant financial and management resources. Regardless of the merits or eventual outcome, liability claims may result in:

·decreased demand for our products;
·injury to our reputation;
·withdrawal of clinical trial participants and inability to continue clinical trials;
·initiation of investigations by regulators;
·costs to defend the related litigation;
·a diversion of management’s time and our resources;
·substantial monetary awards to trial participants or patients;
·product recalls, withdrawals or labeling, marketing or promotional restrictions;
·loss of revenue;
·exhaustion of any available insurance and our capital resources;
·the inability to commercialize any product candidate; and
·a decline in our share price.

Because most of our products have not reached clinical or commercial stage, we do not currently need to carry clinical trial or extensive product liability insurance. In the future, our inability to obtain additional sufficient product liability insurance at an acceptable cost to protect against potential product liability claims could prevent or inhibit the commercialization of products we develop, alone or with collaborators. Such insurance policies may also have various exclusions, and we may be subject to a product liability claim for which we have no coverage.

-36-

It may be difficult to enforce a U.S. judgment against us, our officers and directors and the foreign persons named in this Annual Report on Form 10-K in the United States or in foreign countries, , or to assert U.S. securities laws claims in foreign countries or serve process on our officers and directors and these experts.

While we are incorporated in the State of Nevada, currently a majority of our directors and executive officers are not residents of the United States, and the foreign persons named in this Annual Report on Form 10-K are located in Israel and Belgium.outside of the United States. The majority of our assets are located outside the United States. Therefore, it may be difficult for an investor, or any other person or entity, to enforce a U.S. court judgment based upon the civil liability provisions of the U.S. federal securities laws against us or any of these persons in a U.S. or foreign court, or to effect service of process upon these persons in the United States. Additionally, it may be difficult for an investor, or any other person or entity, to assert U.S. securities law claims in original actions instituted in foreign countries in which we operate. Foreign courts may refuse to hear a claim based on a violation of U.S. securities laws on the grounds that foreign countries are not necessary the most appropriate forum in which to bring such a claim. Even if a foreign court agrees to hear a claim, it may determine that foreign law and not U.S. law is applicable to the claim. If U.S. law is found to be applicable, the content of applicable U.S. law must be proved as a fact, which can be a time-consuming and costly process. Certain matters of procedure will also be governed by foreign countries law. There is little binding case law in foreign countries addressing the matters described above.

Risks RelatedWe may be subject to Our CDMO Businessnumerous and varying privacy and security laws, and our failure to comply could result in penalties and reputational damage.

WhileWe are subject to laws and regulations covering data privacy and the protection of personal information, including health information. The legislative and regulatory landscape for privacy and data protection continues to evolve, and there ishas been an increasing numberfocus on privacy and data protection issues which may affect our business. In the U.S., numerous federal and state laws and regulations, including state security breach notification laws, state health information privacy laws, and federal and state consumer protection laws, govern the collection, use, disclosure, and protection of product candidatespersonal information. Each of these laws is subject to varying interpretations by courts and government agencies, creating complex compliance issues for us. If we fail to comply with applicable laws and regulations we could be subject to penalties or sanctions, including criminal penalties if we knowingly obtain or disclose individually identifiable health information from a covered entity in clinical trials with a smaller numbermanner that is not authorized or permitted by the Health Insurance Portability and Accountability Act of 1996, as amended by the Health Information Technology for Economic and Clinical Health Act, or HIPAA.

31

Numerous other countries have, reached commercial production, cell therapy isor are developing, laws governing the collection, use and transmission of personal information as well. The EU and other jurisdictions have adopted data protection laws and regulations, which impose significant compliance obligations. In the EU, for example, effective May 25, 2018, the GDPR replaced the prior EU Data Protection Directive (95/46) that governed the processing of personal data in the European Union. The GDPR imposes significant obligations on controllers and processors of personal data, including, as compared to the prior directive, higher standards for obtaining consent from individuals to process their personal data, more robust notification requirements to individuals about the processing of their personal data, a developing industrystrengthened individual data rights regime, mandatory data breach notifications, limitations on the retention of personal data and a significant global market for manufacturing services may never emerge.

Cell therapy is in its early stagesincreased requirements pertaining to health data, and is still a developing areastrict rules and restrictions on the transfer of research, with few cell therapy products approved for clinical use. Manypersonal data outside of the existing cellular therapy candidates are basedEU, including to the U.S. The GDPR also imposes additional obligations on, novel cell technologiesand required contractual provisions to be included in, contracts between companies subject to the GDPR and their third-party processors that are inherently riskyrelate to the processing of personal data. The GDPR allows EU member states to make additional laws and regulations further limiting the processing of genetic, biometric or health data.

Adoption of the GDPR increased our responsibility and liability in relation to personal data that we process and may not be understood require us to put in place additional mechanisms to ensure compliance. Any failure to comply with the requirements of GDPR and applicable national data protection laws of EU member states, could lead to regulatory enforcement actions and significant administrative and/or accepted by the marketplace, making it difficult for their own fundingfinancial penalties against us (fines of up to enable themEuro 20,000,000 or up to continue their business. In addition to providing in-house process development and manufacturing expertise for our own product candidates in development, Masthercell Global provides development and manufacturing of cell and tissue-based therapeutic products in clinical and pre-clinical trials. The number of people who may use cell or tissue-based therapies, and the demand for cell processing services, is difficult to forecast. If cell therapies under development by us or by others to treat disease are not proven safe and effective, demonstrate unacceptable risks or side effects or, where required, fail to receive regulatory approval, our manufacturing business will be significantly impaired. While the therapeutic application of cells to treat serious diseases is currently being explored by a number of companies, to date there are only a handful of approved cell therapy products in the U.S. Ultimately, our success in deriving revenue from manufacturing depends on the development and growth of a broad and profitable global market for cell-, gene- and tissue-based therapies and services and our ability to capture a share of this market through our global CDMO network.

Masthercell Global's revenues may vary dramatically change from period to period making it difficult to forecast future results.

Masthercell Global (which includes our CDMO subsidiaries) (“Masthercell” or “Masthercell Global”) recorded revenues of approximately $22 million for the year ended November 30, 2018, representing an increase of 92% over the same period last year. The nature and duration of Masthercell’s contracts with customers often involve regular renegotiation of the scope, level and price of the services we are providing. If our customers reduce the level of their spending on research and development or are unsuccessful in attaining or retaining product sales due to market conditions, reimbursement issues or other factors, our results of operations may be materially impacted. In addition, other factors, including the rate of enrollment for clinical studies, will directly impact the level and timing of the products and services we deliver. As such, the levels of our revenues and profitability can fluctuate significantly from one period to another and it can be difficult to forecast the level of future revenues with any certainty. Furthermore, a dramatic change in our future revenue may result an impairment of our goodwill.

The loss of one or more of MaSTherCell’s major clients or a decline in demand from one or more of these clients could harm MaSTherCell’s business.

-37-

MaSTherCell has a limited number of major clients that together account for a large percentage4% of the total revenues earned. Overworldwide annual turnover of the pastpreceding financial year, MaSTherCell has increased its client portfoliowhichever is higher), and diversified sourcecould adversely affect our business, financial condition, cash flows and results of revenues, butoperations.

We are increasingly dependent on information technology and our systems and infrastructure face certain risks, including cybersecurity and data storage risks.

Significant disruptions to our information technology systems or breaches of information security could adversely affect our business. In the ordinary course of business, we collect, store and transmit confidential information, and it is critical that we do so in a secure manner in order to maintain the confidentiality and integrity of such confidential information. Our information technology systems are potentially vulnerable to service interruptions and security breaches from inadvertent or intentional actions by our employees, partners, vendors, or from attacks by malicious third parties. Maintaining the secrecy of this confidential, proprietary, and/or trade secret information is important to our competitive business position. While we have taken steps to protect such information and invested in information technology, there can be no assurance that such clientsour efforts will continue to use MaSTherCell’s services atprevent service interruptions or security breaches in our systems or the same levelunauthorized or at all. A reductioninadvertent wrongful access or delaydisclosure of confidential information that could adversely affect our business operations or result in the loss, dissemination, or misuse of critical or sensitive information. A breach of our security measures or the accidental loss, inadvertent disclosure, unapproved dissemination or misappropriation or misuse of trade secrets, proprietary information, or other confidential information, whether as a result of theft, hacking, or other forms of deception, or for any other cause, could enable others to produce competing products, use our proprietary technology and/or adversely affect our business position. Further, any such interruption, security breach, loss or disclosure of MaSTherCell’s services, including reductions or delays dueconfidential information could result in financial, legal, business, and reputational harm to market, economic or competitive conditions,us and could have a material adverse effect on MaSTherCell’s business, operating results and financial condition.

MaSTherCell’s business is subject to risks associated with a single manufacturing facility.

The majority of Masthercell Global’s contract manufacturing services are from MaSTherCell S.A., our Belgian subsidiary and are dependent upon its single fully operational facility located in Gosselies (Belgium). A catastrophic loss of the use of all or a portion of the manufacturing facility due to accident, fire, explosion, labor issues, weather conditions, other natural disaster or otherwise, whether short or long-term, could have a material adverse effect on MaSTherCell’s customer relationships and financial results. While its global network partners offer alternative manufacturing sites as part of a disaster recovery plan, this may require it to invest significant time and effort in tech transfer.

If MaSTherCell loses electrical power at its manufacturing facility, its business operations may be adversely affected.

If MaSTherCell loses electrical power at its manufacturing facility for more than a few hours, MaSTherCell would be unable to continue its manufacturing operations for an extended period of time. Additionally, MaSTherCell does not have an alternative manufacturing location located nearby. While MaSTherCell implemented remediation measures to address this risk by setting up a back-up generator allowing it to provide for its manufacturing power consumption needs for a few hours and by being granted a priority access to power in case of global power outage, in the industrial park in Belgium where its premises are located, these measures may not prevent a significant disruption in MaSTherCell’s manufacturing operations which could materially and adversely affect its business operations during an extended period of power outage.

The logistics associated with the distribution of materials produced by MaSTherCell for third parties and for us are significant, complex and expensive and may negatively impact our ability to generate and meet future demand for our products and improve profitability.

Current cell therapy products and product candidates have a limited shelf life, in certain instances limited to less than 12 hours. Thus, it is necessary to minimize the amount of time between when the cell product is extracted from a patient, arrives at our facility for processing, and is returned for infusion in the patient. To do so, we need our cell therapy facilities to be located in major population centers in which patients are likely to be located and within close proximity of major airports. In the future, it may be necessary to build new facilities or invest into new technologies enabling final formulation at point of care, which would require a significant commitment of capital and may not then be available to us. Even if we are able to establish such new facilities or technologies, we may experience challenges in ensuring that they are compliant with cGMP standards, EMEA requirements, and/or applicable state or local regulations. We cannot be certain that we would be able to recoup the costs of establishing a facility in a given market. Given these risks, we could choose not to expand our cell processing and manufacturing services into new geographic markets which will limit our future growth prospects.

Product liability and uninsured risks may adversely affect MaSTherCell’s continuing operations and damage its reputation.

MaSTherCell operates in an industry susceptible to significant product liability claims. MaSTherCell may be liable if it manufactures any product that causes injury, illness, or death for intentional or gross fault on its part. In addition, product liability claims may be brought against MaSTherCell’s clients, in which case MaSTherCell’s clients or others may seek contribution from MaSTherCell if they incur any loss or expenses related to such claims. These claims may be brought by individuals seeking relief or by groups seeking to represent a class. While MaSTherCell’s liability may be limited to instances where it was grossly negligent, nonetheless, the defense of such claims may be costly and time-consuming and could divert the attention of MaSTherCell’s management and technical personnel.

-38-

A breakdown or breach of MaSTherCell’s information technology systems could subject MaSTherCell to liability.

MaSTherCell relies upon its information technology systems and infrastructure for its business. The size and complexity of MaSTherCell’s computer systems make it potentially vulnerable to breakdown and unauthorized intrusion. MaSTherCell could also experience a business interruption, theft of confidential information, or reputational damage from industrial espionage attacks, malware or other cyber-attacks, which may compromise MaSTherCell’s system infrastructure or lead to data leakage, either internally or at MaSTherCell’s third-party providers.

Similarly, data privacy breaches by those who access MaSTherCell’s systems may pose a risk that sensitive data, including intellectual property, trade secrets or personal information belonging to MaSTherCell or its employees, clients or other business partners, may be exposed to unauthorized persons or to the public. Even if MaSTherCell runs regular IT security audits by third-parties, there can be no assurance that MaSTherCell’s efforts to protect its data and information technology systems will prevent breakdowns or breaches in MaSTherCell’s systems that could adversely affect its business and result in financial and reputational harm to MaSTherCell.

We face competition from other third party contact manufacturers, as well as more general competition from companies and academic and research institutions that may choose to self-manufacture rather than utilize a contract manufacturer.

We face competition from companies that are large, well-established manufacturers with financial, technical, research and development and sales and marketing resources that are significantly greater than those that we currently possess. In addition, certain of our leading competitors, such as Lonza Group, WuXi AppTec and PCT have international capabilities that we do not currently possess though we are pursuing.

More generally, we face competition inherent in any third-party manufacturer’s business - namely, that potential customers may instead elect to invest in their own facilities and infrastructure, affording them greater control over their products and the hope of long-term cost savings compared to a third party contract manufacturer. To be successful, we will need to convince potential customers that our current and expanding capabilities are more innovative, of higher-quality and more cost-effective than could be achieved through internal manufacturing and that our experience and quality manufacturing and process development expertise are unique in the industry. Our ability to achieve this and to successfully compete against other manufacturers will depend, in large part, on our success in developing technologies that improve both the quality and profitability associated with cell therapy manufacturing. If we are unable to successfully compete against other manufacturers, we may not be able to develop our CDMO business plans which may harm our business, financial condition andposition, results of operations.operations and/or cash flow.

32

 

Extensive industry regulation has had, and will continue to have, a significant impact on our CDMO business, and it may require us to substantially invest in our development, manufacturing and distribution capabilities and may negatively impact our ability to generate and meet future demand for our products and improve profitability.

Although we seek to conduct our business in compliance with applicable governmental healthcare laws and regulations, these laws and regulations are exceedingly complex and often subject to varying interpretations. The cell therapy industry is the topic of significant government interest, and thus the laws and regulations applicable to our business are subject to frequent change and/or reinterpretation. As such, thereThere can be no assurance that we will be able to develop in-house sales and commercial distribution capabilities or will haveestablish or maintain relationships with third-party collaborators to successfully commercialize any product in the resources, to maintain compliance with all such healthcare lawsUnited States or overseas, and regulations. Failure to comply with such healthcare laws and regulations couldas a result, in significant enforcement actions, civil or criminal penalties, which along with the costs associated with such compliance or with enforcement of such healthcare laws and regulations, may have a material adverse effect on our operations or may require restructuring of our operations or impair our ability to operate profitably.

-39-

Joint-venture partnerships integration into our global CDMO network would be subject to various risks and uncertainties and may involve significant time and attention, all of which could disrupt or adversely affect our business and harm our reputation.

We need our cell therapy facilities to be located in major population centers in which patients are likely to be located and within close proximity of major airports. To do so, we intend to build up a global CDMO network partnership offering alternative manufacturing sites for our third-party clients currently operating out of Belgium, Korea and Israel. The failure to provide harmonized manufacturing quality standards between the current and any future sites to our clients and compliance with local regulatory agencies requirements could have a material adverse effect on our reputation, business, operating results and financial condition.

We are highly dependent on our key personnel, and if we are not successful in attracting, motivating and retaining highly qualified personnel, we may not be able to successfully implementgenerate product revenue.

A variety of risks associated with operating our business strategy.internationally could materially adversely affect our business. We plan to seek regulatory approval of our product candidates outside of the United States and, accordingly, we expect that we, and any potential collaborators in those jurisdictions, will be subject to additional risks related to operating in foreign countries, including:

differing regulatory requirements in foreign countries, unexpected changes in tariffs, trade barriers, price and exchange controls, and other regulatory requirements;
economic weakness, including inflation, or political instability in particular foreign economies and markets;
compliance with tax, employment, immigration, and labor laws for employees living or traveling abroad;
foreign taxes, including withholding of payroll taxes;
foreign currency fluctuations, which could result in increased operating expenses and reduced revenue, and other obligations incident to doing business in another country;
difficulties staffing and managing foreign operations;
workforce uncertainty in countries where labor unrest is more common than in the United States;
potential liability under the Foreign Corrupt Practices Act of 1977 or comparable foreign laws;
challenges enforcing our contractual and intellectual property rights, especially in those foreign countries that do not respect and protect intellectual property rights to the same extent as the United States;
production shortages resulting from any events affecting raw material supply or manufacturing capabilities abroad; and
business interruptions resulting from geo-political actions, including war, and terrorism or disease outbreaks (such as the recent outbreak of COVID-19, or the novel coronavirus).

Our ability to compete in the highly competitive biotechnologyThese and pharmaceutical industries depends uponother risks associated with our planned international operations may materially adversely affect our ability to attract, motivate and retain highly qualified managerial, scientific and medical personnel. Weattain or maintain profitable operations.

If we are highly dependent onunable to integrate acquired businesses effectively, our management and on our trained staff turnover. If the staff turnover increases, it could result in additional hiring and training expenses, potentially delays in product development and manufacturing and harmoperating results may be adversely affected.

From time to time, we seek to expand our business and our growth. Competition for skilled personnel is intense and the turnover rate can be high, whichthrough acquisitions. We may limit our ability to hire and retain highly qualified personnel on acceptable terms or at all.

To induce valuable employees to remain at our company, in addition to salary and cash incentives, we have provided stock option grants that vest over time. The value to employees of these equity grants that vest over time may be significantly affected by movements in our stock price that are beyond our control and may at any time be insufficient to counteract more lucrative offers from other companies. Although we have employment agreements with our key employees, some of these employment agreements provide for at-will employment, which means that any of our employees could leave our employment at any time, with or without notice. We do not maintain “key man” insurance policies on the lives of all of these individuals or the lives of any of our other employees.

Masthercell Global may not receive the future payments pursuant to the Stock Purchase Agreement with GPP-II.

The purchase price for the Masthercell Global Preferred Stock was up to $25 million, subject to certain adjustments, of which $11.8 million was paid in cash at closing. The Stock Purchase Agreement also requires GPP-II to make up to two additional payments to Masthercell Global if certain specified EBITDA (as defined in the Stock Purchase Agreement) and revenue targets are satisfied by Masthercell Global during each of years 2018 and 2019. For each of those fiscal years in which such specified EBITDA and revenue targets are satisfied by Masthercell Global, GPP-II will be obligated to pay an additional $6.6 million, subject to adjustment, to Masthercell Global shortly after the end of that fiscal year.

To earn such contingent payment for the 2018 fiscal year, Masthercell Global must have (i) during any twelve month period ending on or prior to December 31, 2018, generated Net Revenue equal to or greater than €14,100,000 and EBITDA equal to or greater than €1,800,000, and (ii) by December 31, 2018, obtained stockholder approval of the Stockholders’ Agreement Terms in accordance with law the and in a manner that will ensure that GPP-II is able to exercise its rights under the Stockholders’ Agreement without any further action or approval by GPP-II, us, our stockholders, or any other person, which includes the stockholder approval sought in our proxy statement for our annual meeting of stockholders (“Proper Approval”).   In satisfaction of these conditions, Masthercell Global achieved the specified EBITDA and revenues targets in 2018 as described in the SPA and received $6,600,000 of the Future Payments on January 16, 2019 and received approval from the requisite shareholders on October 23, 2018.

To earn such contingent payment for the 2019 fiscal year, Masthercell Global must (i) during any twelve-month period ending on or prior to December 31, 2019, generate Net Revenue equal to or greater than €19,100,000 and EBITDA equal to or greater than €3,900,000, and (ii) by December 31, 2019, obtain Proper Approval, if not already obtained. Accordingly, if our stockholders do not approve the Stockholders’ Agreement Terms and do not meet the applicable Net Revenue and EBITDA targets, Masthercell Global will not be eligible to receive the future payments. In addition, in such event, GPP-II will obtain the right to put to us (or, at our discretion, to Masthercell Global if Masthercell Global shall then have the funds available to consummate the transaction) its shares in Masthercell Global.

-40-

GPP-II may force the sale of Masthercell Global which may result in GPP-II receiving a greater value than the Company and its shareholders.

At any time following the earlier to occur of (i) after June 28, 2020 or (ii) Masthercell Global’s failure to generate positive EBITDA for any twelve (12) month period as determined on a quarterly basis prior to June 28, 2020 or its failure to generate at least $1,000,000 of EBITDA during any such twelve (12) month period after June 28, 2020 (collectively, a “Material Underperformance Event”), GPP-II has the right, in its sole discretion, to approve and force the sale of Masthercell Global. While we have the right of first refusal with respect to acquiring Masthercell Global in its entirety, if GPP-II elects to exercise such a right and if we are not in the position to acquire Masthercell Global, GPP-II may cause the sale of Masthercell Global to any third party on terms GPP-II approves on an arm’s length basis and subject to the receipt of a fairness opinion. If this occurs, we are contractually obligated to approve such a sale and execute any documents as required by GPP-II. We must also share in any costs and expenses relating to such a sale on a pro rata basis. Based on this, there may be a situation where GPP-II approves a sale that is more valuable or beneficial to GPP-II than to our Company and our shareholders, and we will not be able to prevent such a transaction. A sale of Masthercell Globalsuccessfully integrate acquired businesses and, where desired, their product portfolios into ours, and therefore we may not be able to realize the intended benefits. If we fail to successfully integrate acquisitions or product portfolios, or if they fail to perform as we anticipate, our existing businesses and our revenue and operating results could have impacts tobe adversely affected. If the CDMO activities of Orgenesis as conducted through Masthercell Global and to Orgenesis’ overall value as a whole.

GPP-II may, under certain circumstances, assume controldue diligence of the Boardoperations of Directors ofacquired businesses performed by us and by third parties on our subsidiary, Masthercell Global, which wouldbehalf is inadequate or flawed, or if we later discover unforeseen financial or business liabilities, acquired businesses and their assets may not perform as expected. Additionally, acquisitions could result in our inability to controldifficulties assimilating acquired operations and, direct the activities of such subsidiary.

Currently, the Board of Directors of Masthercell Global is comprised of seven (7) directors, four (4) of whom are appointed by us (one of whom must be an industry expert (the “Industry Expert Director”)) and three (3) of whom are appointed by GPP-II. In the event the Industry Expert Director is removed or replaced without the prior written approval of GPP-II, orwhere deemed desirable, transitioning overlapping products into a Material Underperformance Event has occurred after June 28, 2020, GPP-II has the right to increase the size of the Board of Directors of Masthercell Global and appoint additional directors to fill such vacancies so that GPP-II appointments represent a majority of the directors. If this were to occur, GPP-II would control the Board of Directors of Masthercell Global and will be entitled to direct its activities and approve any transactions of Masthercell Global, even if such transactions provide greater value to GPP-II than they do to Orgenesis and its stockholders. This lack of control could diminish the value of Masthercell Global as it relates to Orgenesis’ overall activity and significantly impact CDMO activities of Orgenesis as conducted through Masthercell Global.

GPP-II has the right to buy our shares in Masthercell Global upon the occurrence of certain events resulting in Orgenesis not holding any shares in Masthercell Global.

GPP-II has the right to purchase all of the shares of stock we hold in Masthercell Global if any of the following occurs: (i) there is an Activist Shareholder (as defined in the Stockholders’ Agreement) of the Company; (ii) the Chief Executive Officer and/or Chairman of the Board of Directors of the Company is replaced prior to June 28, 2023; (iii) there is a Change of Control (as defined in the Stockholders’ Agreement) of the Company; or (iv) the Industry Expert Director is removed or replaced without the prior written consent of GPP-II. If any of these events occur, GPP-II, upon notice to the Company, can force the Company to sell all of the securities it holds in Masthercell Global to GPP-II based upon a valuation of Masthercell Global to be determined by one of the top ten (10) independent third-party accounting firms in the United States with experience in performing valuations as selected by GPP-II. This right of GPP-II expires if GPP-II fails to exercise this right within three (3) years from the first occurrence of any of the events listed above. In the event GPP-II does exercise its right following the occurrence of any such event, Orgenesis shall cease to be a stockholder of Masthercell Global and will no longer derive any benefits from this subsidiary or its activities. This would also affect the CDMO activities being conducted by Orgenesis through Masthercell Global.

-41-

GPP-II has the right to effectuate a spin-off of our subsidiary, Masthercell Global

In addition to the other rights GPP-II has obtained under the Stockholders’ Agreement, GPP-II also has the right to effectuate a spin-off of Masthercell Global upon the earlier to occur of: (i) any of the four events listed in the section above or (ii) ninety (90) days after GPP-II provides the Company of its intent to exercise this right provided that such notice cannot be delivered by GPP-II before June 28, 2020. Such a spin-off would be based on a valuation of Masthercell Global as determined in accordance with the terms of the Stockholders’ Agreement. If such a spin-off was completed, there is a chance that Masthercell Global would no longer be a subsidiary of Orgenesis, which would result in a significant loss of value to Orgenesis. In addition, without control or ownership of Masthercell Global, Orgenesis’ ability to conduct CDMO activities independently would also be greatly affected in the event such a spin-off is completed.

If GPP-II opts to exchange its Masthercell Global Preferred Stock for shares of our common stock, we could potentially issue a substantial number of shares of our common stock to GPP-II, which may result in significant dilution to our existing stockholders.

The Stockholders’ Agreement provides that GPP-II is entitled, at any time, to convert its share capital in Masthercell Global for our common stock (such exchange option being the “Stock Exchange Option”). Under the Stock Exchange Option, GPP-II is entitled to exchange the Masthercell Global Preferred Stock for our common stock based on an exchange price (as defined in the Stockholders’ Agreement) that is not currently known. If GPP-II opts to exchange its Masthercell Global Preferred Stock for shares of our common stock, we could potentially issue a substantial number of shares of our common stock to GPP-II. The common stock issuable to GPP-II upon exchange of the Masthercell Global Preferred Stock for our common stock could have a depressive effect on the market price of our common stock by increasing the number of shares of common stock outstanding. Such downward pressure could encourage short sales by certain investors, which could place further downward pressure on the price of the common stock. Accordingly, the number of shares of outstanding common stock may increase significantlysingle product line and the ownership interestsdiversion of capital and proportionate voting powermanagement’s attention away from other business issues and opportunities. The failure to integrate acquired businesses effectively may adversely impact our business, results of the existing stockholders may be significantly diluted.operations or financial condition.

Risks Related to Our Trans-Differentiation Technologies for Diabetes

THM is entitled to cancel the THM License Agreement.

Pursuant to the terms of the THM License Agreement with THM, Orgenesis Ltd, the Israeli Subsidiary, must develop, manufacture, sell and market the products pursuant to the milestones and time schedule specified in the development plan. In the event the Israeli Subsidiary fails to fulfill the terms of the development plan under the THM License Agreement, THM shall be entitled to terminate the THM License Agreement by providing the Israeli Subsidiary with written notice of such a breach and if the Israeli Subsidiary does not cure such breach within one year of receiving the notice. If THM cancels the THM License Agreement, our PT business may be materially adversely affected. THM may also terminate the THM License Agreement if the Israeli Subsidiary breaches an obligation contained in the THM License Agreement and does not cure it within 180 days of receiving notice of the breach. Any terminationWe also run the risk that THM may attempt cancel or, at the very least challenge, the License Agreement with Orgenesis Ltd. as we continue to expand our focus to other therapies and business activities. We believe that our expanded focus to such other therapies and business activities may continue to prompt THM to inquire of such activities as they may relate to our compliance with the terms or direction of resources toward the THM License Agreement. While we have not received any notice of cancellation of the THM License Agreement, is likelywe have received an allegation regarding the scope of the rights by THM that may present future challenges for our Israeli Subsidiary to materially adversely affect our business and prospects.

Our success will depend on strategic collaborations with third partiescontinue to develop, manufacture, sell and commercializemarket the products pursuant to the milestones and time schedule specified in the development plan of the THM License Agreement.

33

We have developed a technology that demonstrates the capacity to induce a shift in the developmental fate of cells from the liver and differentiating (converting) them into “pancreatic beta cell-like” insulin-producing cells for patients with diabetes. Our intention is to develop our technology to the clinical stage for regeneration of functional insulin-producing cells, thus enabling normal glucose regulated insulin secretion, via cell therapy. By using therapeutic agents that efficiently convert a sub-population of liver cells into pancreatic islets phenotype and function, this approach allows the diabetic patient to be the donor of his/her own therapeutic tissue and to start producing his/her own insulin in a glucose-responsive manner, thereby eliminating the need for insulin injections. Because this is a new approach to treating diabetes, developing and commercializing our product candidates and we may not have control oversubjects us to a number of key elements relating to the development and commercialization of any such product candidate.challenges, including:

A key aspect of our strategy is to seek collaboration with a partner, such as a large pharmaceutical organization, that is willing to further develop and commercialize a selected product candidate. To date, we have not entered into any such collaborative arrangement. By entering into any such strategic collaboration, we may rely on our partner for financial resources and for development, regulatory and commercialization expertise. Our partner may fail to develop or effectively commercialize our product candidate because they:

·doobtaining regulatory approval from the FDA, EMA and other regulatory authorities that have very limited experience with the commercial development of our technology for diabetes;
developing and deploying consistent and reliable processes for engineering a patient’s liver cells ex vivo and infusing the engineered cells back into the patient;
developing processes for the safe administration of these products, including long-term follow-up for all patients who receive our products;
sourcing clinical and, if approved, commercial supplies for the materials used to manufacture and process our products;
developing a manufacturing process and distribution network with a cost of goods that allows for an attractive return on investment;
establishing sales and marketing capabilities after obtaining any regulatory approval to gain market acceptance; and
maintaining a system of post marketing surveillance and risk assessment programs to identify adverse events that did not have sufficient resources or decide not to devoteappear during the necessary resources due to internal constraints such as limited cash or human resources;
·decide to pursue a competitive potential product developed outside of the collaboration;
·cannot obtain the necessary regulatory approvals;drug approval process.

-42-

·determine that the market opportunity is not attractive; or
·cannot manufacture or obtain the necessary materials in sufficient quantities from multiple sources or at a reasonable cost.

We may not be ableRisks Related to enter into a collaboration on acceptable terms, if at all. We face competition in our search for partners from other organizations worldwide, manyDevelopment and Regulatory Approval of whom are largerOur Therapies and are able to offer more attractive deals in terms of financial commitments, contribution of human resources, or development, manufacturing, regulatory or commercial expertiseProduct Candidates

Research and support. If we are not successful in attracting a partner and entering into a collaboration on acceptable terms, we may not be able to complete development of or commercialize any product candidate. In such event, our ability to generate revenues and achieve or sustain profitability would be significantly hindered and we may not be able to continue operations as proposed, requiring us to modify our business plan, curtail various aspects of our operations or cease operations.biopharmaceutical products is inherently risky.

Third parties to whom we may license or transfer development and commercialization rights for products covered by intellectual property rightsWe may not be successful in theirour efforts to use and asenhance our technology platform to create a result,pipeline of product candidates and develop commercially successful products. Furthermore, we may expend our limited resources on programs that do not receive future royaltyyield a successful product candidate and fail to capitalize on product candidates or other milestone payments relating to those productsdiseases that may be more profitable or rights.

for which there is a greater likelihood of success. If we are unablefail to successfully acquire, develop or commercialize new products,additional product candidates, our operating results will suffer. Our future results of operations will depend to a significant extent upon our ability to successfully develop and commercialize our technology and businesses in a timely manner. There are numerous difficulties in developing and commercializing new technologies and products, including:

·successfully achieving major developmental steps required to bring the product to a clinical testing stage and clinical testing may not be positive;
·developing, testing and manufacturing products in compliance with regulatory standards in a timely manner;
·the failure to receive requisite regulatory approvals for such products in a timely manner or at all;
·developing and commercializing a new product is time consuming, costly and subject to numerous factors, including legal actions brought by our competitors, that may delay or prevent the development and commercialization of our product;
·incomplete, unconvincing or equivocal clinical trials data;
·experiencing delays or unanticipated costs;
·significant and unpredictable changes in the payer landscape, coverage and reimbursement for our future product;
·experiencing delays as a result of limited resources at the U.S. Food and Drug Administration (“FDA”) or other regulatory agencies; and
·changing review and approval policies and standards at the FDA and other regulatory agencies.

As a result of these and other difficulties, products in development by us may not receive timely regulatory approvals, or approvals at all, necessary for marketing by us or other third-party partners. If any of our future products are not approved in a timely fashion or, when acquired or developed and approved, cannot be successfully manufactured, commercialized or reimbursed, our operating results could be adversely affected. We cannot guarantee that any investment we make in developing productcommercial opportunity will be recouped, evenlimited. Even if we are successful in continuing to build our pipeline, obtaining regulatory approvals and commercializing these products.

Our research and development programs are based on novel technologiesadditional product candidates will require substantial additional funding and are inherently risky.

We are subjectprone to the risks of failure inherent in medical product development. Investment in biopharmaceutical product development involves significant risk that any potential product candidate will fail to demonstrate adequate efficacy or an acceptable safety profile, gain regulatory approval, and become commercially viable. We cannot provide you any assurance that we will be able to successfully advance any of these additional product candidates through the development of products based on new technologies. The novel nature of our cell therapy technology creates significant challenges with respect to product development and optimization, manufacturing, government regulation and approval, third-party reimbursement and market acceptance. For example, the FDA and EMA have relatively limited experience with the development and regulation of cell therapy products and, therefore, the pathway to marketing approval for our cell therapyprocess. Our research programs may initially show promise in identifying potential product candidates, yet fail to yield product candidates for clinical development or commercialization for many reasons, including the following:

our platform may not be successful in identifying additional product candidates;
we may not be able or willing to assemble sufficient resources to acquire or discover additional product candidates;
our product candidates may not succeed in preclinical or clinical testing;
a product candidate may on further study be shown to have harmful side effects or other characteristics that indicate it is unlikely to be effective or otherwise does not meet applicable regulatory criteria;
competitors may develop alternatives that render our product candidates obsolete or less attractive;
product candidates we develop may nevertheless be covered by third parties’ patents or other exclusive rights;
the market for a product candidate may change during our program so that the continued development of that product candidate is no longer reasonable;
a product candidate may not be capable of being produced in commercial quantities at an acceptable cost, or at all; and
a product candidate may not be accepted as safe and effective by patients, the medical community or third- party payers, if applicable.

If any of these events occur, we may accordingly be more complex, lengthy and uncertain thanforced to abandon our development efforts for a more conventional product candidate. The indications of use for whichprogram or programs, or we choose to pursue development may have clinical effectiveness endpoints that have not previously been reviewed or validated by the FDA or EMA, which may complicate or delay our effort to ultimately obtain FDA or EMA approval. Our efforts to overcome these challenges may not prove successful, and any product candidate we seek to develop may not be successfully developedable to identify, discover, develop, or commercialized.commercialize additional product candidates, which would have a material adverse effect on our business and could potentially cause us to cease operations.

-43-
34

Extensive industry regulation has had, and will continue to have, a significant impact on our business, especially our product development, manufacturing and distribution capabilities.

All pharmaceutical companies are subject to extensive, complex, costly and evolving government regulation. For the U.S., this is principally administered by the FDA and to a lesser extent by the Drug Enforcement Administration (“DEA”) and state government agencies, as well as by varying regulatory agencies in foreign countries where products or product candidates are being manufactured and/or marketed. The Federal Food, Drug and Cosmetic Act, the Controlled Substances Act and other federal statutes and regulations, and similar foreign statutes and regulations, govern or influence the testing, manufacturing, packing, labeling, storing, record keeping, safety, approval, advertising, promotion, sale and distribution of our future products. Under these regulations, we may become subject to periodic inspection of our facilities, procedures and operations and/or the testing of our future products by the FDA, the DEA and other authorities, which conduct periodic inspections to confirm that we are in compliance with all applicable regulations. In addition, the FDA and foreign regulatory agencies conduct pre-approval and post-approval reviews and plant inspections to determine whether our systems and processes are in compliance with current good manufacturing practice (“cGMP”) and other regulations. Following such inspections, the FDA or other agency may issue observations, notices, citations and/or warning letters that could cause us to modify certain activities identified during the inspection. FDA guidelines specify that a warning letter is issued only for violations of “regulatory significance” for which the failure to adequately and promptly achieve correction may be expected to result in an enforcement action. We may also be required to report adverse events associated with our future products to FDA and other regulatory authorities. Unexpected or serious health or safety concerns would result in labeling changes, recalls, market withdrawals or other regulatory actions.

The range of possible sanctions includes, among others, FDA issuance of adverse publicity, product recalls or seizures, fines, total or partial suspension of production and/or distribution, suspension of the FDA’s review of product applications, enforcement actions, injunctions, and civil or criminal prosecution. Any such sanctions, if imposed, could have a material adverse effect on our business, operating results, financial condition and cash flows. Under certain circumstances, the FDA also has the authority to revoke previously granted drug approvals. Similar sanctions as detailed above may be available to the FDA under a consent decree, depending upon the actual terms of such decree. If internal compliance programs do not meet regulatory agency standards or if compliance is deemed deficient in any significant way, it could materially harm our business.

The European Medicines Agency (“EMA”) will regulate our future products in Europe. Regulatory approval by the EMA will be subject to the evaluation of data relating to the quality, efficacy and safety of our future products for its proposed use. The time taken to obtain regulatory approval varies between countries. Different regulators may impose their own requirements and may refuse to grant, or may require additional data before granting, an approval, notwithstanding that regulatory approval may have been granted by other regulators.

Regulatory approval may be delayed, limited or denied for a number of reasons, including insufficient clinical data, the product not meeting safety or efficacy requirements or any relevant manufacturing processes or facilities not meeting applicable requirements.

Further trials and other costly and time-consuming assessments of the product may be required to obtain or maintain regulatory approval. Medicinal products are generally subject to lengthy and rigorous pre-clinical and clinical trials and other extensive, costly and time-consuming procedures mandated by regulatory authorities. We may be required to conduct additional trials beyond those currently planned, which could require significant time and expense. In addition, even after the technology approval, both in the U.S. and Europe, we will be required to maintain post marketing surveillance of potential adverse and risk assessment programs to identify adverse events that did not appear during the clinical studies and drug approval process. All of the foregoing could require an investment of significant time and expense.

35

We have never generated anylimited revenue from therapeutic product sales, and our ability to generate any significant revenue from product sales and become profitable depends significantly on our success in a number of factors.

-44-

We have noa limited number of therapeutic products approved for commercial sale, and we have not generated anyonly limited revenue from product sales, and do not anticipate generating any revenue from product sales until sometime after we have received regulatory approval for the commercial sale of a product candidate.sales. Our ability to generate revenue of more significant scale and achieve profitability depends significantly on our success in many factors, including:

·completing research regarding, and nonclinical and clinical development of, our product candidates;
·obtaining regulatory approvals and marketing authorizations for product candidates for which we complete clinical studies;
·developing a sustainable and scalable manufacturing process for our product candidates, including establishing and maintaining commercially viable supply relationships with third parties and establishing our own manufacturing capabilities and infrastructure;
·launching and commercializing product candidates for which we obtain regulatory approvals and marketing authorizations, either directly or with a collaborator or distributor;
·obtaining market acceptance of our product candidates as viable treatment options;
·addressing any competing technological and market developments;
·identifying, assessing, acquiring and/or developing new product candidates;
·negotiating favorable terms in any collaboration, licensing, or other arrangements into which we may enter;
·maintaining, protecting, and expanding our portfolio of intellectual property rights, including patents, trade secrets, and know-how; and
·attracting, hiring, and retaining qualified personnel.

Even if one or more of the product candidates that we develop isare approved for commercial sale, we anticipate incurring significant costs associated with commercializing any approved product candidate. Our expenses could increase beyond expectations if we are required by the U.S. Food and Drug Administration, or the FDA, or other regulatory agencies, domestic or foreign, to change our manufacturing processes or assays, or to perform clinical, nonclinical, or other types of studies in addition to those that we currently anticipate. If we are successful in obtaining regulatory approvals to market one or more of our product candidates, our revenue will be dependent, in part, upon the size of the markets in the territories for which we gain regulatory approval, the accepted price for the product, the ability to get reimbursement at any price, and whether we own the commercial rights for that territory. If the number of our addressable disease patients is not as significant as we estimate, the indication approved by regulatory authorities is narrower than we expect, or the reasonably accepted population for treatment is narrowed by competition, physician choice or treatment guidelines, we may not generate significant revenue from sales of such products, even if approved. If we are not able to generate revenue from the sale of any approved products, we may never become profitable.

We have concentrated our research and development efforts on technology using cell-based therapy, and our future success is highly dependent on the successful development of that technology for diabetes.

We have developed a technology that demonstrates the capacity to induce a shift in the developmental fate of cells from the liver and differentiating (converting) them into “pancreatic beta cell-like” insulin-producing cells for patients with diabetes. Based on licensed know-how and patents, our intention is to develop our technology to the clinical stage for regeneration of functional insulin-producing cells, thus enabling normal glucose regulated insulin secretion, via cell therapy. By using therapeutic agents (i.e., PDX-1, and additional pancreatic transcription factors in an adenovirus-vector) that efficiently convert a sub-population of liver cells into pancreatic islets phenotype and function, this approach allows the diabetic patient to be the donor of his/her own therapeutic tissue and to start producing his/her own insulin in a glucose-responsive manner, thereby eliminating the need for insulin injections. Because this is a new approach to treating diabetes, developing and commercializing our product candidates subjects us to a number of challenges, including:

·obtaining regulatory approval from the FDA, EMA and other regulatory authorities that have very limited experience with the commercial development of our technology for diabetes;
·developing and deploying consistent and reliable processes for engineering a patient’s liver cells ex vivo and infusing the engineered cells back into the patient;
·developing processes for the safe administration of these products, including long-term follow-up for all patients who receive our products;

-45-

·sourcing clinical and, if approved, commercial supplies for the materials used to manufacture and process our products;
·developing a manufacturing process and distribution network with a cost of goods that allows for an attractive return on investment;
·establishing sales and marketing capabilities after obtaining any regulatory approval to gain market acceptance; and
·maintaining a system of post marketing surveillance and risk assessment programs to identify adverse events that did not appear during the drug approval process

When we commence ourany clinical trials, we may not be able to conduct our trials on the timelines we expect.

Clinical testing is expensive, time consuming, and subject to uncertainty. We cannot guarantee that any clinical studies will be conducted as planned or completed on schedule, if at all. We expect that our early clinical work will help support the filing with the FDA of an IND for our product in fiscal 2019.2020. However, we cannot be sure that we will be able to submit an IND in this time-frame, and we cannot be sure that submission of an IND will result in the FDA allowing clinical trials to begin. Moreover, even if these trials begin, issues may arise that could suspend or terminate such clinical trials. A failure of one or more clinical studies can occur at any stage of testing, and our future clinical studies may not be successful. Events that may prevent successful or timely completion of clinical development include:

·the inability to generate sufficient preclinical or other in vivo or in vitro data to support the initiation of clinical studies;
·delays in reaching a consensus with regulatory agencies on study design;
·delays in establishing CMC (Chemistry, Manufacturing, and Controls) which is a cornerstone in clinical study submission and later on, the regulatory approval;
·the FDA not allowing us to use the clinical trial data from a research institution to support an IND if we cannot demonstrate the comparability of our product candidates with the product candidate used by the relevant research institution in its clinical studies;
·delays in obtaining required Institutional Review Board, or IRB, approval at each clinical study site;
·imposition of a temporary or permanent clinical hold by regulatory agencies for a number of reasons, including after review of an IND application or amendment, or equivalent application or amendment;

·36

a result of a new safety finding that presents unreasonable risk to clinical trial participants;
·a negative finding from an inspection of our clinical study operations or study sites;
·developments on trials conducted by competitors for related technology that raises FDA concerns about risk to patients of the technology broadly;
·if the FDA finds that the investigational protocol or plan is clearly deficient to meet its stated objectives;
·delays in recruiting suitable patients to participate in our clinical studies;
·difficulty collaborating with patient groups and investigators;
·failure to perform in accordance with the FDA’s current good clinical practices, or cGCPs, requirements, or applicable regulatory guidelines in other countries;
·delays in having patients'patients’ complete participation in a study or return for post-treatment follow-up;
·patients dropping out of a study;
·occurrence of adverse events associated with the product candidate that are viewed to outweigh its potential benefits;
·changes in regulatory requirements and guidance that require amending or submitting new clinical protocols;
·changes in the standard of care on which a clinical development plan was based, which may require new or additional trials;
·the cost of clinical studies of our product candidates being greater than we anticipate;
·clinical studies of our product candidates producing negative or inconclusive results, which may result in our deciding, or regulators requiring us, to conduct additional clinical studies or abandon product development programs; and

-46-

·delays in manufacturing, testing, releasing, validating, or importing/exporting sufficient stable quantities of our product candidates for use in clinical studies or the inability to do any of the foregoing.

Any inability to successfully complete preclinical and clinical development could result in additional costs to us or impair our ability to generate revenue. In addition, if we make manufacturing or formulation changes to our product candidates, we may be required to, or we may elect to conduct additional studies to bridge our modified product candidates to earlier versions. Clinical study delays could also shorten any periods during which our products have patent protection and may allow our competitors to bring products to market before we do, which could impair our ability to successfully commercialize our product candidates and may harm our business and results of operations.

Our clinical trial results may also not support approval, whether accelerated approval, conditional marketing authorizations, or regular approval. The results of preclinical and clinical studies may not be predictive of the results of later-stage clinical trials, and product candidates in later stages of clinical trials may fail to show the desired safety and efficacy despite having progressed through preclinical studies and initial clinical trials. In addition, our product candidates could fail to receive regulatory approval for many reasons, including the following:

·the FDA or comparable foreign regulatory authorities may disagree with the design or implementation of our clinical trials;
·the population studied in the clinical program may not be sufficiently broad or representative to assure safety in the full population for which we seek approval;
·we may be unable to demonstrate that our product candidates’ risk-benefit ratios for their proposed indications are acceptable;
·the results of clinical trials may not meet the level of statistical significance required by the FDA or comparable foreign regulatory authorities for approval;
·we may be unable to demonstrate that the clinical and other benefits of our product candidates outweigh their safety risks;
·the FDA or comparable foreign regulatory authorities may disagree with our interpretation of data from preclinical studies or clinical trials;
·the data collected from clinical trials of our product candidates may not be sufficient to the satisfaction of the FDA or comparable foreign regulatory authorities to obtain regulatory approval in the United States or elsewhere;
·the FDA or comparable foreign regulatory authorities may fail to approve the manufacturing processes, our own manufacturing facilities, or our third-party manufacturers’ facilities with which we contract for clinical and commercial supplies; and
·the approval policies or regulations of the FDA or comparable foreign regulatory authorities may significantly change in a manner rendering our clinical data insufficient for approval.

37

Further, failure to obtain approval for any of the above reasons may be made more likely by the fact that the FDA and other regulatory authorities have very limited experience with commercial development of our cell therapy for the treatment of Type 1 Diabetes.

Our product candidates may cause undesirable side effects or have other properties that could halt their clinical development, prevent their regulatory approval, limit their commercial potential, or result in significant negative consequences.

As with most biological drug products, use of our product candidates could be associated with side effects or adverse events which can vary in severity from minor reactions to death and in frequency from infrequent to prevalent. Any of these occurrences may materially and adversely harm our business, financial condition and prospects.

-47-

Research and development of biopharmaceutical products is inherently risky.

We may not be successful in our efforts to use and enhance our technology platform to create a pipeline of product candidates and develop commercially successful products. Furthermore, we may expend our limited resources on programs that do not yield a successful product candidate and fail to capitalize on product candidates or diseases that may be more profitable or for which there is a greater likelihood of success. If we fail to develop additional product candidates, our commercial opportunity will be limited. Even if we are successful in continuing to build our pipeline, obtaining regulatory approvals and commercializing additional product candidates will require substantial additional funding and are prone to the risks of failure inherent in medical product development. Investment in biopharmaceutical product development involves significant risk that any potential product candidate will fail to demonstrate adequate efficacy or an acceptable safety profile, gain regulatory approval, and become commercially viable. We cannot provide you any assurance that we will be able to successfully advance any of these additional product candidates through the development process. Our research programs may initially show promise in identifying potential product candidates, yet fail to yield product candidates for clinical development or commercialization for many reasons, including the following:

·our platform may not be successful in identifying additional product candidates;
·we may not be able or willing to assemble sufficient resources to acquire or discover additional product candidates;
·our product candidates may not succeed in preclinical or clinical testing;
·a product candidate may on further study be shown to have harmful side effects or other characteristics that indicate it is unlikely to be effective or otherwise does not meet applicable regulatory criteria;
·competitors may develop alternatives that render our product candidates obsolete or less attractive;
·product candidates we develop may nevertheless be covered by third parties’ patents or other exclusive rights;
·the market for a product candidate may change during our program so that the continued development of that product candidate is no longer reasonable;
·a product candidate may not be capable of being produced in commercial quantities at an acceptable cost, or at all; and
·a product candidate may not be accepted as safe and effective by patients, the medical community or third- party payers, if applicable.

If any of these events occur, we may be forced to abandon our development efforts for a program or programs, or we may not be able to identify, discover, develop, or commercialize additional product candidates, which would have a material adverse effect on our business and could potentially cause us to cease operations.

Our product candidates are biologics and the manufacture of our product candidates is complex and we may encounter difficulties in production, particularly with respect to process development or scaling-out of our manufacturing capabilities.

If we encounter such difficulties, our ability to provide supply of our product candidates for clinical trials or our products for patients, if approved, could be delayed or stopped, or we may be unable to maintain a commercially viable cost structure. Our product candidates are biologics and the process of manufacturing our products is complex, highly regulated and subject to multiple risks. The manufacture of our product candidates involves complex processes, including the biopsy of tissue from a patient’s liver, propagation of the patient’s liver cells from that liver tissue to obtain the desired dose, trans-differentiating those cells into insulin-producing cells ex vivo and ultimately infusing the cells back into a patient’s body. As a result of the complexities, the cost to manufacture biologics is generally higher than traditional small molecule chemical compounds, and the manufacturing process is less reliable and is more difficult to reproduce.

-48-

Our manufacturing process will be susceptible to product loss or failure due to logistical issues associated with the collection of liver cells, or starting material, from the patient, shipping such material to the manufacturing site, shipping the final product back to the patient, and infusing the patient with the product, manufacturing issues associated with the differences in patient starting materials, interruptions in the manufacturing process, contamination, equipment or reagent failure, improper installation or operation of equipment, vendor or operator error, inconsistency in cell growth, failures in process testing and variability in product characteristics. Even minor deviations from normal manufacturing processes could result in reduced production yields, product defects, and other supply disruptions. If for any reason we lose a patient’s starting material or later-developed product at any point in the process, the manufacturing process for that patient will need to be restarted and the resulting delay may adversely affect that patient’s outcome. If microbial, viral, or other contaminations are discovered in our product candidates or in the manufacturing facilities in which our product candidates are made, such manufacturing facilities may need to be closed for an extended period of time to investigate and remedy the contamination. Because our product candidates are manufactured for each particular patient, we will be required to maintain a chain of identity and tractability of all reagents and viruses involved in the process with respect to materials as they move from the patient to the manufacturing facility, through the manufacturing process, and back to the patient. Maintaining such a chain of identity is difficult and complex, and failure to do so could result in adverse patient outcomes, loss of product, or regulatory action including withdrawal of our products from the market. Further, as product candidates are developed through preclinical to late stage clinical trials towards approval and commercialization, it is common that various aspects of the development program, such as manufacturing methods, are altered along the way in an effort to optimize processes and results. Such changes carry the risk that they will not achieve these intended objectives, and any of these changes could cause our product candidates to perform differently and affect the results of planned clinical trials or other future clinical trials.

Although we are working to develop commercially viable processes, doing so is a difficult and uncertain task, and there are risks associated with scaling to the level required for advanced clinical trials or commercialization, including, among others, cost overruns, potential problems with process scale-out, process reproducibility, stability issues, lot consistency, and timely availability of reagents or raw materials. We may ultimately be unable to reduce the cost of goods for our product candidates to levels that will allow for an attractive return on investment if and when those product candidates are commercialized.

38

We expect that continued development of our manufacturing facility via MaSTherCell and our global CDMO network will provide us with enhanced control of material supply for both clinical trials and the commercial market, enable the more rapid implementation of process changes, and allow for better long-term margins. We may establish multiple manufacturing facilities as we expand our commercial footprint to multiple geographies, which may lead to regulatory delays or prove costly. Even if we are successful, our manufacturing capabilities could be affected by cost-overruns, unexpected delays, equipment failures, labor shortages, natural disasters, power failures and numerous other factors that could prevent us from realizing the intended benefits of our manufacturing strategy and have a material adverse effect on our business.

In addition, the manufacturing process for any products that we may develop is subject to FDA and foreign regulatory authority approval process, and we will need to contract with manufacturers who can meet all applicable FDA and foreign regulatory authority requirements on an ongoing basis. If we are unable to reliably produce products to specifications acceptable to the FDA or other regulatory authorities, we may not obtain or maintain the approvals we need to commercialize such products. Even if we obtain regulatory approval for any of our product candidates, there is no assurance that either we or our CDMO subsidiaries and joint ventures will be able to manufacture the approved product to specifications acceptable to the FDA or other regulatory authorities, to produce it in sufficient quantities to meet the requirements for the potential launch of the product, or to meet potential future demand. Any of these challenges could delay completion of clinical trials, require bridging clinical trials or the repetition of one or more clinical trials, increase clinical trial costs, delay approval of our product candidate, impair commercialization efforts, increase our cost of goods, and have an adverse effect on our business, financial condition, results of operations and growth prospects.

-49-

The manufacture of biological drug products is complex and requires significant expertise and capital investment, including the development of advanced manufacturing techniques and process controls. Manufacturers of biologic products often encounter difficulties in production, particularly in scaling up or out, validating the production process, and assuring high reliability of the manufacturing process (including the absence of contamination). These problems include logistics and shipping, difficulties with production costs and yields, quality control, including stability of the product, product testing, operator error, availability of qualified personnel, as well as compliance with strictly enforced federal, state and foreign regulations. Furthermore, if contaminants are discovered in our supply of our product candidates or in the manufacturing facilities, such manufacturing facilities may need to be closed for an extended period of time to investigate and remedy the contamination. We cannot assure you that any stability failures or other issues relating to the manufacture of our product candidates will not occur in the future. Additionally, our manufacturers may experience manufacturing difficulties due to resource constraints or as a result of labor disputes or unstable political environments. If our manufacturers were to encounter any of these difficulties, or otherwise fail to comply with their contractual obligations, our ability to provide our product candidate to patients in clinical trials would be jeopardized. Any delay or interruption in the supply of clinical trial supplies could delay the completion of clinical trials, increase the costs associated with maintaining clinical trial programs and, depending upon the period of delay, require us to begin new clinical trials at additional expense or terminate clinical trials completely.

Cell-based therapies rely on the availability of reagents, specialized equipment, and other specialty materials, which may not be available to us on acceptable terms or at all. For some of these reagents, equipment, and materials, we rely or may rely on sole source vendors or a limited number of vendors, which could impair our ability to manufacture and supply our products.

Manufacturing our product candidates will require many reagents and viruses, which are substances used in our manufacturing processes to bring about chemical or biological reactions, and other specialty materials and equipment, some of which are manufactured or supplied by small companies with limited resources and experience to support commercial biologics production. We currently depend on a limited number of vendors for certain materials and equipment used in the manufacture of our product candidates. Some of these suppliers may not have the capacity to support commercial products manufactured under GMP by biopharmaceutical firms or may otherwise be ill-equipped to support our needs. We also do not have supply contracts with many of these suppliers and may not be able to obtain supply contracts with them on acceptable terms or at all. Accordingly, we may experience delays in receiving key materials and equipment to support clinical or commercial manufacturing.

For some of these reagents, viruses, equipment, and materials, we rely and may in the future rely on sole source vendors or a limited number of vendors. An inability to continue to source product from any of these suppliers, which could be due to regulatory actions or requirements affecting the supplier, adverse financial or other strategic developments experienced by a supplier, labor disputes or shortages, unexpected demands, or quality issues, could adversely affect our ability to satisfy demand for our product candidates, which could adversely and materially affect our product sales and operating results or our ability to conduct clinical trials, either of which could significantly harm our business.

As we continue to develop and scale our manufacturing process, we expect that we will need to obtain rights to and supplies of certain materials and equipment to be used as part of that process. We may not be able to obtain rights to such materials on commercially reasonable terms, or at all, and if we are unable to alter our process in a commercially viable manner to avoid the use of such materials or find a suitable substitute, it would have a material adverse effect on our business.

39

We currently have no marketing and sales organization and have no experience in marketing therapeutic products. If we are unable to establish marketing and sales capabilities or enter into agreements with third parties to market and sell our product candidates, we may not be able to generate product revenue.

-50-

We currently have no sales, marketing, or commercial therapeutic product distribution capabilities and have no experience in marketing products. We intend to develop an in-house marketing organization and sales force, which will require significant capital expenditures, management resources, and time. We will have to compete with other pharmaceutical and biotechnology companies to recruit, hire, train, and retain marketing and sales personnel. If we are unable to or decide not to establish internal sales, marketing and commercial distribution capabilities for any or all products we develop, we will likely pursue collaborative arrangements regarding the sales and marketing of our products. However, thereThere can be no assurance that we will be able to establish or maintain such collaborative arrangements, or if we are able to do so, that they will have effective sales forces. Any revenue we receive will depend upon the efforts of such third parties, which may not be successful. We may have little or no control over the marketing and sales efforts of such third parties, and our revenue from product sales may be lower than if we had commercialized our product candidates ourselves. We also face competition in our search for third parties to assist us with the sales and marketing efforts of our product candidates.

There can be no assurance that we will be able tofurther develop in-house sales and commercial distribution capabilities or establish or maintain relationships with third-party collaborators to successfully commercialize any product in the United States or overseas, and as a result, we may not be able to generate product revenue.

A variety of risks associated with operating our business internationally could materially adversely affect our business. We plan to seek regulatory approval of our product candidates outside of the United States and, accordingly, we expect that we, and any potential collaborators in those jurisdictions, will be subject to additional risks related to operating in foreign countries, including:

·differing regulatory requirements in foreign countries, unexpected changes in tariffs, trade barriers, price and exchange controls, and other regulatory requirements;
·economic weakness, including inflation, or political instability in particular foreign economies and markets;
·compliance with tax, employment, immigration, and labor laws for employees living or traveling abroad;
·foreign taxes, including withholding of payroll taxes;
·foreign currency fluctuations, which could result in increased operating expenses and reduced revenue, and other obligations incident to doing business in another country;
·difficulties staffing and managing foreign operations;
·workforce uncertainty in countries where labor unrest is more common than in the United States;
·potential liability under the Foreign Corrupt Practices Act of 1977 or comparable foreign laws;
·challenges enforcing our contractual and intellectual property rights, especially in those foreign countries that do not respect and protect intellectual property rights to the same extent as the United States;
·production shortages resulting from any events affecting raw material supply or manufacturing capabilities abroad; and
·business interruptions resulting from geo-political actions, including war and terrorism.

These and other risks associated with our planned international operations may materially adversely affect our ability to attain or maintain profitable operations.

We face significant competition from other biotechnology and pharmaceutical companies, and our operating results will suffer if we fail to compete effectively.

The biopharmaceutical industry, and the rapidly evolving market for developing cell-based therapies is characterized by intense competition and rapid innovation. Our competitors may be able to develop other compounds or drugs that are able to achieve similar or better results. Our potential competitors include major multinational pharmaceutical companies, established biotechnology companies, specialty pharmaceutical companies, universities, and other research institutions. Many of our competitors have substantially greater financial, technical and other resources, such as larger research and development staff and experienced marketing and manufacturing organizations as well as established sales forces. Smaller or early-stage companies may also prove to be significant competitors, particularly through collaborative arrangements with large, established companies. Mergers and acquisitions in the biotechnology and pharmaceutical industries may result in even more resources being concentrated in our competitors. Competition may increase further as a result of advances in the commercial applicability of technologies and greater availability of capital for investment in these industries. Our competitors, either alone or with collaborative partners, may succeed in developing, acquiring or licensing on an exclusive basis drug or biologic products that are more effective, safer, more easily commercialized, or less costly than our product candidates or may develop proprietary technologies or secure patent protection that we may need for the development of our technologies and products.

40

Specifically, we face significant competition from companies in the insulin therapy market. Insulin therapy is widely used for Insulin-Dependent Diabetes Mellitus (IDDM) patients who are not controlled with oral medications. The global diabetes market comprising the insulin, insulin analogues and other anti-diabetic drugs has been evolving rapidly. A look at the diabetes market reveals that it is dominated by a handful of participants such as Novo Nordisk A/S, Eli Lilly and Company, Sanofi-Aventis, Takeda Pharmaceutical Company Limited, Pfizer Inc., Merck KgaA, and Bayer AG. Even if we obtain regulatory approval of our product candidates, we may not be the first to market and that may affect the price or demand for our product candidates. Additionally, the availability and price of our competitors’ products could limit the demand and the price we are able to charge for our product candidates. We may not be able to implement our business plan if the acceptance of our product candidates is inhibited by price competition or the reluctance of physicians to switch from existing methods of treatment to our product candidates, or if physicians switch to other new drug or biologic products or choose to reserve our product candidates for use in limited circumstances. Additionally, a competitor could obtain orphan product exclusivity from the FDA with respect to such competitor’s product. If such competitor product is determined to be the same product as one of our product candidates, that may prevent us from obtaining approval from the FDA for such product candidate for the same indication for seven years, except in limited circumstances.

-51-

We are highly dependent on our key personnel, and if we are not successful in attracting, motivating and retaining highly qualified personnel, we may not be able to successfully implement our business strategy.

Our ability to compete in the highly competitive biotechnology and pharmaceutical industries depends upon our ability to attract, motivate and retain highly qualified managerial, scientific and medical personnel. We are highly dependent on our senior management, particularly our Chief Scientific Officer, Prof. Sarah Ferber, and our Chief Executive Officer, Vered Caplan. The loss of the services of any of our executive officers, other key employees, and other scientific and medical advisors, and our inability to find suitable replacements, could result in delays in product development and harm our business. Competition for skilled personnel is intense and the turnover rate can be high, which may limit our ability to hire and retain highly qualified personnel on acceptable terms or at all.

To induce valuable employees to remain at our company, in addition to salary and cash incentives, we have provided stock option grants that vest over time. The value to employees of these equity grants that vest over time may be significantly affected by movements in our stock price that are beyond our control and may at any time be insufficient to counteract more lucrative offers from other companies. Although we have employment agreements with our key employees, most these employment agreements provide for at-will employment, which means that any of our employees could leave our employment at any time, with or without notice. We do not maintain “key man” insurance policies on the lives of all of these individuals or the lives of any of our other employees.

Risks Related to our Common Stock

If we issue additional shares in the future, it will result in the dilution of our existing stockholders.

Our articles of incorporation authorizes the issuance of up to 145,833,334 shares of our common stock with a par value of $0.0001 per share. Our Board of Directors may choose to issue some or all of such shares to acquire one or more companies or products and to fund our overhead and general operating requirements. The issuance of any such shares will reduce the book value per share and may contribute to a reduction in the market price of the outstanding shares of our common stock. If we issue any such additional shares, such issuance will reduce the proportionate ownership and voting power of all current stockholders. Further, such issuance may result in a change of control of our company.

Our stock price and trading volume may be volatile, which could result in losses for our stockholders.

The equity trading markets have recently experienced high volatility resulting in highly variable and unpredictable pricing of equity securities. If the turmoil in the equity trading markets continues, the market for our common stock could change in ways that may not be related to our business, our industry or our operating performance and financial condition. In addition, the trading volume in our common stock may fluctuate and cause significant price variations to occur. Some of the factors that could negatively affect our share price or result in fluctuations in the price or trading volume of our common stock include:

·actual or anticipated quarterly variations in our operating results, including further impairment to unproved oil and gas properties;results;
·changes in expectations as to our future financial performance or changes in financial estimates, if any;
·announcements relating to our business;
·conditions generally affecting the oil and natural gasbiotechnology industry;
·the success of our operating strategy; and
·the operating and stock performance of other comparable companies.

-52-

Many of these factors are beyond our control, and we cannot predict their potential effects on the price of our common stock. In addition, the stock market is subject to extreme price and volume fluctuations. During the past 52 weeks ended November 30, 2018,December 31, 2020, our stock price has fluctuated from a low of $4.50$2.76 to a high of $14.68 (adjusted to account for the 1:12 reverse split implemented in November 2017).$7.84. This volatility has had a significant effect on the market price of securities issued by many companies for reasons unrelated to their operating performance and could have the same effect on our common stock.

No assurance can be provided that a purchaser of our common stock will be able to resell their shares of common stock at or above the price that they acquired those shares. We can provide no assurances that the market price of common stock will increase or that the market price of common stock will not fluctuate or decline significantly.

41

We do not intend to pay dividends on any investment in the shares of stock of our company.

We have never paid any cash dividends, and currently do not intend to pay any dividends for the foreseeable future. The Board of Directors has not directed the payment of any dividends and does not anticipate paying dividends on the shares for the foreseeable future and intends to retain any future earnings to the extent necessary to develop and expand our business. Payment of cash dividends, if any, will depend, among other factors, on our earnings, capital requirements, and the general operating and financial condition, and will be subject to legal limitations on the payment of dividends out of paid-in capital. Because we do not intend to declare dividends, any gain on an investment in our company will need to come through an increase in the stock’s price. This may never happen, and investors may lose all of their investment in our company.

ITEM 1B. UNRESOLVED STAFF COMMENTS

Not Applicable.applicable.

ITEM 2. PROPERTIES

We do not own any real property. A description of the leased premises we utilize in several of our facilities is as follows:

Entity

Property Description

Orgenesis Inc./Orgenesis Maryland Inc.

These are theour principal offices:

·

   Located at 20271 Goldenrod Lane, Germantown, MD 20876.

·   Occupy office space at the Germantown Innovation Center.

·   Cost is $200 per month on a month-to-month contract.

MaSTherCell SA, Cell Therapy Holding SA and
Orgenesis SPRLLtd.

All activities located in Gosselies, Belgium, in the I-Tech Incubator 2. Property consists of:

·     Operational production and Office area represent +/-2,400 m².

·     Monthly costs are approximately €25 thousand.

·     Lease agreement for the office and operational production area expires on March 31, 2030.

·     Additional offices are leased in a separate building to temporarily locate MaSTherCell corporate service and meeting rooms; it represents 480m² for a monthly cost of €7 thousand and termination lease agreement on February 29, 2020.

·     The new production area designed during 2016 was built in 2017-2018 and was operational at the end of 2018.

-53-

Orgenesis Ltd.

·   The development lab is located in 8 HaHaruv St.the Bar Lev Industrial Park M.P. MISGAV, Israel.

·     The Company’s offices●   Offices are in the science park of Ness Ziona. Monthly costs are approximately $5 thousand.

Masthercell

Orgenesis Korea

·   Operational production and Office area represent +/-2,234 m².approximately 2,234 square meters.

·   Monthly costs are approximately 21,232 thousand KRW.KRW, or approximately $19 thousand.

·   Lease agreement for the office and operational production area expires on July 14, 2020.January 1, 2023.

Atvio Biotech
Koligo Therapeutics Inc.

●   Production facility and development labs in New Albany, Indiana – approximately 4170 square feet (388 square meter) at monthly costs of about $5400

·●   Medical device maintenance and development labs in Leander, Texas – approximately 2000 square feet (186 square meter) at monthly costs of about $2500

Orgenesis Biotech Israel (previously Atvio Biotech)

   Located in 8 HaHaruv St.the Bar Lev Industrial Park M.P. MISGAV, Israel.

·   Operational production and Office area represent +/-1,264 m².

·   Monthly costs are approximately $10.5 thousand.

·   Lease agreement for the office and operational production area expires on July 31, 2023.

Orgenesis Belgium

●   Located near Namur, at Novalis Science Park, Orgenesis Belgium

42

We believe that our facilities are generally in good condition and suitable to carry on our business. We also believe that, if required, suitable alternative or additional space will be available to us on commercially reasonable terms.

ITEM 3. LEGAL PROCEEDINGS

We are not involved in any pending legal proceedings that we anticipate would result in a material adverse effect on our business or operations.

ITEM 4. MINE SAFETY DISCLOSURES

Not applicable.

PART II

ITEM 5. MARKET FOR REGISTRANT’S COMMON EQUITY, RELATED STOCKHOLDER MATTERS AND ISSUER PURCHASES OF EQUITY SECURITIES

Market Information

Until March 13, 2018, the Company’sour common shares were traded under OTC Market Group’s OTCQB. FromSince March 13, 2018, the Company'sour common stock beganhas been listed for trading on the Nasdaq Capital Market (Nasdaq CM)(“Nasdaq CM”) under the symbol “ORGS.”

As of February 11, 2019,March 9, 2021, there were 156205 holders of record of our common stock, and the last reported sale price of our common stock on the Nasdaq CMNasdaqCM on February 12, 2019March 8, 2021 was $4.88.$7.26. A significant number of shares of our common stock are held in either nominee name or street name brokerage accounts, and consequently, we are unable to determine the total number of beneficial owners of our stock.

-54-

Dividend Policy

To date, we have paid no dividends on our common stock and do not expect to pay cash dividends in the foreseeable future. We plan to retain all earnings to provide funds for the operations of our company. In the future, our Board of Directors will decide whether to declare and pay dividends based upon our earnings, financial condition, capital requirements, and other factors that our Board of Directors may consider relevant. We are not under any contractual restriction as to present or future ability to pay dividends.

Unregistered Sales of Equity Securities

During the fiscal year ended November 30, 2018,December 31, 2020, our financing activities consisted of the following:

(1) InOn January 2017, the Company20, 2020, we entered into definitive agreementsa Securities Purchase Agreement with an institutional investor for thecertain investors pursuant to which we issued and sold, in a private placement of 2,564,115 units of the Company’s securities for aggregate subscription proceeds to the Company of $16 million at $6.24 price per unit. Each unit is comprised of one share of the Company’s Common Stock and one warrant, exercisable over a three-years period from the date of issuance, to purchase one additional share(the “Offering”), 2,200,000 shares of Common Stock at a purchase price of $4.20 per share exercise price of $6.24 (“Unit”). The subscription proceeds have been paid to the Company on a periodic basis through October 2018.

In July 2018, the Company entered into definitive agreements with assignees of the aforementioned institutional investor whereby these assignees remitted $4.6 million in respect of the units available under the original subscription agreement that had been subscribed for, entitling such investors to 702,307 units, with each unit being comprised of (i) one share of the Company's common stock and (ii) one three-year warrant to purchase up to an additional one share of the Company’s common stock at a per share exercise price of $6.24.

During the year ended November 30, 2018 and 2017, the investor and the assignees remitted to the Company $11.5 and $4.5 million, respectively, and the Company issued 1,813,687 and 721,160 Units, respectively.

(2) During the fiscal year ended November 30, 2018, the Company entered into definitive agreements with accredited and other qualified investors relating to a private placement of 1,237,642 units. Each unit is comprised of (i) one share of the Company’s common stock and (ii) three-year warrant to purchase up to an additional one share of the Company’s Common Stock at a per share exercise price of $6.24, for aggregate proceeds to the Company of approximately $7.7 million.

(3) During the year ended November 30, 2018, investors exercised 136,646 warrants into 136,646 shares of the Company’s Common Stock, for aggregate proceeds of $853 thousand.

(4) In November 2018, we entered into unsecured convertible note agreements with accredited or offshore investors for an aggregate amount of $625 thousand. The loans bear an annual interest rate of 2% and mature in three years unless converted earlier. The holders, at their option, may convert the outstanding principal amount and accrued interest under those notes into 89,285 shares of our common stock and 89,285 three-year warrants to purchase up to an additional 89,2851,000,000 shares of our common stockCommon Stock at a per sharean exercise price of $7. In the initial two years, the holders have the right to convert the outstanding principal amount and accrued interest into shares of capital stock of Hemogenyx-Cell, a subsidiary of Hemogenyx Pharmaceuticals Plc, at a price$5.50 per share, based on a pre-money valuationwhich are exercisable between June 2021 and January 2023. We received gross proceeds of Hemogenyx-Cellapproximately $9.24 million before deducting related offering expenses in the amount of $12 million$0.8 million.

On April 7, 2020, we entered into an Asset Purchase Agreement (the “Hemogenyx Securities”“Tamir Purchase Agreement”) with Tamir Biotechnology, Inc. (“Tamir”), pursuant to which we agreed to acquire certain assets and liabilities of Tamir related to the collaboration agreement with Hemogenyx Pharmaceuticals Plc.discovery, development and testing of therapeutic products for the treatment of diseases and conditions in humans, including all rights to ranpirnase and use for antiviral therapy (collectively, the “Purchased Assets and Assumed Liabilities” and such acquisition, the “Tamir Transaction”). The Tamir Transaction closed on April 23, 2020. As aggregate consideration for the acquisition, we paid $2.5 million in cash and issued an aggregate of 3,400,000 shares of common stock to Tamir resulting in a total consideration of $20.2 million.

-55-
43

(5) In November 2018,On September 26, 2020, we entered into unsecured convertible note agreementsan Agreement and Plan of Merger and Reorganization (the “Merger Agreement”) by and among the Company, Orgenesis Merger Sub, Inc., a Delaware corporation and a wholly-owned subsidiary of the Company (“Merger Sub”), Koligo Therapeutics Inc., a Kentucky corporation (“Koligo”), the shareholders of Koligo (collectively, the “Shareholders”) and Long Hill Capital V, LLC, solely in its capacity as the representative, agent and attorney-in-fact of the Shareholders. The Merger Agreement provided for the acquisition of Koligo by the Company through the merger of Merger Sub with accredited or offshore investors forand into Koligo, with Koligo surviving as a wholly-owned subsidiary of the Company (the “Merger”). The Merger closed on October 15, 2020.

Pursuant to the terms of the Merger Agreement, an aggregate of 2,061,713 shares of Company common stock were issued to Koligo’s Shareholders who were accredited investors (with certain Shareholders who were not accredited investors being paid solely in cash in the amount of $625 thousand. The loan bearsapproximately $20 thousand) in accordance with the terms of the Merger Agreement. In connection with the Merger, the Company assumed an annual interest rateaggregate of 2% and maturesapproximately $1.9 million of Koligo’s liabilities, which were substantially all of Koligo’s liabilities at the closing of the Merger. In addition, we issued 66,910 shares to Maxim Group LLC for advisory services in three years unless converted earlier. The holders, at their option, may convertconnection with the outstanding principal amount and accrued interest under those notes into 89,285 shares of our common stock and 89,285 three-year warrants to purchase up to an additional 89,285 shares of our common stock at a per share exercise price of $7. In the initial two years, the holders have the right to convert the outstanding principal amount and accrued interest into shares of capital stock of Immugenyx, LLC, a subsidiary of Hemogenyx Pharmaceuticals Plc, at a price per share based on a pre-money valuation of Immugenyx of $8 million (the “Immugenyx Securities”), pursuant to the collaboration agreement with Immugenyx, LLC.Merger.

All of the securities issued in the transactions described above were issued without registration under the Securities Act in reliance upon the exemptions provided in Section 4(2) or Regulation S of the Securities Act. Except with respect to securities sold pursuant to Regulation S, the recipients of securities in each such transaction acquired the securities for investment only and not with a view to or for sale in connection with any distribution thereof. Appropriate legends were affixed to the share certificates issued in all of the above transactions. Each of the recipients also represented that they were “accredited investors” within the meaning of Rule 501(a) of Regulation D under the Securities Act or had such knowledge and experience in financial and business matters as to be able to evaluate the merits and risks of an investment in its common stock. All recipients had adequate access, through their relationships with the Company and its officers and directors, to information about the Company. None of the transactions described above involved general solicitation or advertising.

Issuer Purchases of Equity Securities

We do not have aOn May 14, 2020, our Board of Directors approved the stock repurchase program forplan (the “Stock Repurchase Plan”) pursuant to which we may, from time to time, purchase up to $10 million of our common stock and have not otherwise purchased anyoutstanding shares of our common stock. The shares may be repurchased from time to time in privately negotiated transactions or the open market, including pursuant to Rule 10b5-1 trading plans, and in accordance with applicable regulations of the SEC. The timing and exact amount of any repurchases will depend on various factors including, general and business market conditions, corporate and regulatory requirements, share price, alternative investment opportunities and other factors. The Repurchase Plan commenced on May 29, 2020 and does not obligate us to acquire any specific number of shares in any period, and may be expanded, extended, modified, suspended or discontinued by the Board of Directors at any time.

The following table summarizes the share repurchase activity from the inception of the Stock Repurchase Plan through December 31, 2020.

   Total Number of Shares
Purchased
  Average Price
Paid per Share
  Total Number of Shares Purchased as Part of Publicly Announced Plans or Programs  

 

Maximum Value that May Yet Be Purchased Under the Plans or Programs

 
            (in thousands) 
October 2020   8,807   4.47   8,807  $9,960 
November 2020   101   4.50   101   9,960 
December 2020   46,401   4.47   46,401   9,750 
    55,309   4.47   55,309  $9,750 

44

ITEM 6. SELECTED FINANCIAL DATA

Not applicable.

ITEM 7. MANAGEMENT’S DISCUSSION AND ANALYSIS OF FINANCIAL CONDITION AND RESULTS OF OPERATIONS

The following Management’s Discussion and Analysis of Financial Condition and Results of Operations is intended to provide information necessary to understand our audited consolidated financial statements for the two-year periodfiscal years ended November 30, 2018December 31, 2020 and December 31, 2019 and highlight certain other information which, in the opinion of management, will enhance a reader’s understanding of our financial condition, changes in financial condition and results of operations. In particular, the discussion is intended to provide an analysis of significant trends and material changes in our financial position and the operating results of our business during the fiscal year ended November 30, 2018,December 31, 2020, as compared to the fiscal year ended November 30, 2017.December 31, 2019. This discussion should be read in conjunction with our consolidated financial statements for the two-year periodfiscal years ended November 30, 2018December 31, 2020 and December 31, 2019 and related notes included elsewhere in this Annual Report on Form 10-K. These historical financial statements may not be indicative of our future performance. This Management’s Discussion and Analysis of Financial Condition and Results of Operations contains numerous forward-looking statements, all of which are based on our current expectations and could be affected by the uncertainties and risks described throughout this filing, particularly in “Item 1A. Risk Factors.”

The full extent to which the COVID-19 pandemic may directly or indirectly impact our business, results of operations and financial condition, will depend on future developments that are uncertain, including as a result of new information that may emerge concerning COVID-19 and the actions taken to contain it or treat COVID-19, as well as the economic impact on local, regional, national and international customers and markets. We have made estimates of the impact of COVID-19 within our financial statements, and although there is currently no major impact, there may be changes to those estimates in future periods. Actual results may differ from these estimates.

Corporate Overview

We areOrgenesis Inc., a biotechnologyNevada corporation, is a global biotech company specializing inworking to unlock the development, manufacturing and provisionpotential of technologies and services in the cell and gene therapies in an affordable and accessible format (“CGTs”).

CGTs can be centered on autologous (using the patient’s own cells) or allogenic (using master banked donor cells) and are part of a class of medicines referred to as advanced therapy industry.medicinal products (ATMPs). We operate through two platforms: (i)mostly focus on autologous therapies, with processes and systems that are developed for each therapy using a point-of-care (“POCare”) cell therapy platform (“PT”)closed and (ii)automated processing system approach that is validated for compliant production near the patient at their point of care for the treatment of patients. This approach has the potential to overcome the limitations of traditional commercial manufacturing methods that do not translate well to commercial production of advanced therapies due to their cost prohibitive nature and complex logistics to deliver the treatments to patients (ultimately limiting the number of patients that can have access to, or can afford, these therapies).

To achieve these goals, we have developed a Contract DevelopmentPoint of Care Platform comprised of three enabling components: a pipeline of licensed POCare Therapies that are designed to be processed and Manufacturing Organization (“CDMO”) platform conducted through our subsidiary, Masthercell Global. Through our PT business, our aim isproduced in closed, automated POCare Technology systems across a collaborative POCare Network. Via a combination of science, technology, engineering, and networking, we are working to further the development of Advanced Therapy Medicinal Products (“ATMPs”) through collaborationsprovide a more efficient and in-licensing with other pre-clinical and clinical-stage biopharmaceutical companies and research and healthcare institutes to bring such ATMPs to patients. We out-license these ATMPs through regional partners to whom we also provide regulatory, pre-clinical and training services to support their activity in orderscalable pathway for advanced therapies to reach patients in a point-of-care hospital setting. Through our CDMO platform, we are focusedmore rapidly at lowered costs. We also draw on providing contract manufacturing and development services for biopharmaceutical companies.

-56-

Our therapeutic development efforts in our PT business are focused on advancing breakthrough scientific achievements in ATMPs, and namelyextensive medical expertise to identify promising new autologous therapies which haveto leverage within the POCare Platform either via ownership or licensing.

The POCare Network brings together patients, doctors, industry partners, research institutes and hospitals worldwide with a curative potential. We base our development on therapeutic collaborations and in-licensing with other pre-clinical and clinical-stage biopharma companies as well as direct collaboration with research and healthcare institutes. We are engaging in therapeutic collaborations and in-licensing with other academic centers and research centers in order to pursue emerging technologiesgoal of other ATMPs in cell and gene therapy in such areas as cell-based immunotherapies, metabolic diseases, neurodegenerative diseases and tissue regeneration. Each of these customers and collaborations represents a growth opportunity and future revenue potential as we out-license these ATMPs through regional partners to whom we also provide regulatory, pre-clinical and training services to support their activity in order to reach patients in a point-of-care hospital setting.

We carry out our PT business through three wholly-owned and separate subsidiaries. This corporate structure allows us to simplify the accounting treatment, minimize taxation and optimize local grant support. The subsidiaries related to this business are Orgenesis Maryland Inc., in the U.S., Orgenesis SPRL, in the European Union and Orgenesis Ltd. in Israel.

Our subsidiary, Masthercell Global, is a CDMO specialized in cell therapy development for advanced therapeutically products. In the last decade, cell therapy medicinal products have gained significant importance, particularly in the fields of ex-vivo gene therapy and immunotherapy. While academic and industrial research has led scientific development in the sector, industrialization and manufacturing expertise remains insufficient. Masthercell Global plans to fill this gap by providing three types of services to its customers: (i) process and assay development services and (ii) current Good Manufacturing Practices (cGMP) contract manufacturing services and (iii) technology innovation and engineering services. These services offer a double advantage to Masthercell Global’s customers. First, customers can continue allocating their financial and human resources on their product/therapy, while relying on a long-term reliable and trusted partner for their process development/production. Second, through its subsidiaries, it allows customers to benefit from Masthercell Global’s expertise in cell therapy manufacturing and all related aspects.

Masthercell Global’s wholly-owned subsidiaries include MaSTherCell S.A., a Belgian-based subsidiary and a Contract Development and Manufacturing Organization (“CDMO”) specialized in cell therapyachieving harmonized, regulated clinical development and manufacturing for advanced medicinal products, Atvio Biotech Ltd. (“Atvio”), an Israeli-based CDMO,production of the therapies.

45

POCare Platform Operations via Subsidiaries

We currently conduct our core business operations ourselves and CureCell Co. Ltd. (“CureCell”through our subsidiaries which are all wholly-owned except as otherwise stated below (collectively, the “Subsidiaries”), a Korea-based CDMO.. The Subsidiaries are as follows:

We operate our CDMO and the PT businesses as two separate business segments.United States

Orgenesis Maryland Inc. (the “U.S. Subsidiary”) is the center of activity in North America and is currently focused on setting up the POCare Network.
Koligo Therapeutics Inc. (“Koligo”) is a Kentucky corporation that we acquired in 2020 and is currently focused on developing the POCare network and therapies. .

Europe

Orgenesis Belgium SRL (the “Belgian Subsidiary”) is the center of activity in Europe and is currently focused on process development and the preparation of European clinical trials.
Orgenesis Switzerland Sarl (the “Swiss Subsidiary”), was incorporated in October 2020, and is currently focused on providing management services to us.

Asia

Orgenesis Ltd. in Israel (the “Israeli Subsidiary”) is a provider of regulatory, clinical and pre-clinical services.
Orgenesis Biotech Israel Ltd. (“OBI”), is a provider of cell-processing services in Israel.
Korea: Orgenesis Korea Co. Ltd. (the “Korean Subsidiary”), is a provider of processing and pre-clinical services in Korea. We own 94.12% of the Korean Subsidiary.

Corporate History

We were incorporated in the state of Nevada on June 5, 2008 under the name Business Outsourcing Services, Inc. Effective August 31, 2011, we completed a merger with our subsidiary, Orgenesis Inc., a Nevada corporation, which was incorporated solely to effect a change in its name. As a result, the Companywe changed itsour name from “Business Outsourcing Services, Inc.” to “Orgenesis Inc.”

On October 11, 2011, we incorporated Orgenesis Ltd. as our wholly-owned subsidiary under the laws of Israel. On February 2, 2012, Orgenesis Ltd. signed and closed a definitive agreement to license from Tel Hashomer - Medical Research, Infrastructure and Services Ltd. (“THM”), a private company duly incorporated under the laws of Israel patents and know-how related tofor the development of AIP (Autologous Insulin Producing) cells.

On November 6, 2014, we entered into an agreement with the shareholders of MaSTherCell S.A. to acquire MaSTherCell S.A. On March 2, 2015, we closed on the acquisition of MaSTherCell whereby it became aan independent, and wholly-owned subsidiary of Orgenesis.Orgenesis INC. Through MaSTherCell, we became engaged in the CDMO business. Currently, most of the Company’s revenues are generated through MaSTherCell.

-57-

On June 28, 2018, the Company,we, Masthercell Global, Great Point Partners, LLC, a manager of private equity funds focused on growing small to medium sized heath care companies (“Great Point”), and certain of Great Point’s affiliates, entered into a series of definitive strategic agreements intended to finance, strengthen and expand Orgenesis’our CDMO business. In connection therewith, the Company,we, Masthercell Global and GPP-II Masthercell, LLC, a Delaware limited liability company (“GPP-II”) and an affiliate of Great Point, entered into a Stock Purchase Agreement (the “SPA”) pursuant to which GPP-II purchased 378,000 shares of newly designated Series A Preferred Stock of Masthercell Global (the “Masthercell Global Preferred Stock”), representing 37.8% of the issued and outstanding share capital of Masthercell Global, for cash consideration to be paid into Masthercell Global of up to $25 million, subject to certain adjustments (the “Consideration”). Orgenesis holdsAt such time, we held 622,000 shares of Masthercell Global’s Common Stock, representing 62.2% of the issued and outstanding equity share capital of Masthercell Global. An initial cash payment of $11.8 million of the Consideration was remitted at closing by GPP-II, with a follow up payment of $6,600,000 to be made in each of years 2018 and 2019, (the “Future Payments”), or an aggregate of $13.2 million (the “Future Payments”), if (a) Masthercell Global achievesachieved specified EBITDA and revenues targets during each of these years, and (b) the Orgenesis’ shareholders approveapproved certain provisions of the Stockholders’ Agreement referred to below on or before December 31, 2019. None2020. Both of the future Consideration amounts, if any, will result in an increase in GPP-II’s equity holdings in Masthercell Global beyond the 378,000 shares of Series A Preferred Stock issued to GPP-II at closing.these conditions were met and we received both milestone payments.

46

Contemporaneous with the execution of the SPA, Orgenesiswe and Masthercell Global entered into a Contribution, Assignment and Assumption Agreement pursuant to which Orgenesiswe contributed to Masthercell Global the Orgenesis’our assets relating to the CDMO Business (as defined below), including the CDMO subsidiaries (the “Corporate Reorganization”). In furtherance thereof, Masthercell Global, as Orgenesis’our assignee, acquired all of the issued and outstanding share capital of Atvio, the Company’sOBI, our Israel based CDMO partner since May 2016, and 94.12% of the share capital of CureCell, the Company’sKorean Subsidiary, our Korea based CDMO partner since March 2016. Orgenesis exercised the” call option”

On August 7, 2019, we, Masthercell Global and GPP (the “Parties”) entered into a Transfer Agreement (the “Transfer Agreement”). As a result of the Transfer Agreement, Masthercell Global transferred all of its equity interests of OBI and the Korean Subsidiary to which it was entitled under the joint ventureus in exchange for one dollar ($1.00). The Transfer Agreement also contains agreements made with each of these entitiesrespect to purchase from the former shareholders their equity holding. The considerationcertain intercompany loans. We accounted for the outstanding share equity in eachTransfer Agreement as a transaction with non-controlling interest.

Discontinued Operations

Until December 31, 2019, we operated the POCare Platform as one of Atviotwo business separate business segments.

Historically, the second separate business segment was operated as a Contract Development and CureCellManufacturing Organization (“CDMO”) platform, providing third party contract manufacturing and development services for biopharmaceutical companies (the “CDMO Business”). The CDMO platform was historically operated mainly through majority owned Masthercell Global (which consisted solely of Orgenesis common stock. In respect of the acquisition of Atvio, Orgenesis issued tofollowing two subsidiaries: MaSTherCell S.A. in Belgium (“MaSTherCell”), and Masthercell U.S., LLC in the former Atvio shareholders an aggregate of 83,965 shares of Orgenesis common stock. In respect of the acquisition of CureCell, Orgenesis Inc. issued to the former CureCell shareholders an aggregate of 202,846 shares of Orgenesis Common Stock subject to a third-party valuation. Together with MaSTherCell S.A., Atvio and CureCell are directly held subsidiaries under United States (“Masthercell GlobalU.S.”) (collectively, the “Masthercell Global Subsidiaries”)).

In February 2020, we and GPP-II Masthercell LLC (“GPP”) sold 100% of the outstanding equity interests of Masthercell (the “Masthercell Business”), which comprised the majority of our CDMO Business, to Catalent Pharma Solutions, Inc. for an aggregate nominal purchase price of $315 million, subject to customary adjustments (the “Masthercell Sale”). After accounting for GPP’s liquidation preference and equity stake in Masthercell as well as other investor interests in our Belgian subsidiary MaSTherCell, distributions to Masthercell option holders and transaction costs, we received approximately $126.7 million. We incurred an additional approximately $5.6 million in transaction costs.

We determined that the Masthercell Business (“Discontinued Operation”) meets the criteria to be classified as a discontinued operation as of the first quarter of 2020. The Discontinued Operation includes the vast majority of the previous CDMO Business, including majority-owned Masthercell, including MaSTherCell, Masthercell U.S. and all of the Masthercell Global Subsidiaries.

Since the Masthercell Sale, we entered into new joint venture agreements with new partners in various jurisdictions. This has allowed us to grow our infrastructure and expand our processing sites into new markets and jurisdictions. In addition, we have engaged some of these joint venture partners to perform research and development services to further develop and adapt our systems and devices for specific purposes. We have been investing manpower and financial resources to focus on developing, manufacturing and rolling out several types of OMPULs to be used and/or distributed through our POCare Network of partners, collaborators, and joint ventures.

The Chief Executive Officer (“CEO”) is our chief operating decision-maker who reviews financial information prepared on a consolidated basis. Effective from the first quarter of 2020, all of our continuing operations are in the point-of-care business via our POCare Platform. Therefore, no segment report has been presented.

47

Orgenesis Inc., a Nevada corporation, is a global biotech company working to unlock the potential of cell and gene therapies (“CGT”s) in an affordable and accessible format.

CGTs can be centered on autologous (using the patient’s own cells) or allogenic (using master banked donor cells) and are part of a class of medicines referred to as advanced therapy medicinal products (ATMP). We mostly focus on autologous therapies, with processes and systems that are developed for each therapy using a closed and automated processing system approach that is validated for compliant production near the patient at their point of care. This approach has the potential to overcome the limitations of traditional commercial manufacturing methods that do not translate well to commercial production of advanced therapies due to their cost prohibitive nature and complex logistics to deliver the treatments to patients (ultimately limiting the number of patients that can have access to, or can afford, these therapies).

To achieve these goals, we have developed a Point of Care Platform comprised of three enabling components: a pipeline of licensed POCare Therapies that are designed to be processed and produced in closed, automated POCare Technology systems across a collaborative POCare Network. Via a combination of science, technology, engineering, and networking, we are working to provide a more efficient and scalable pathway for advanced therapies to reach patients more rapidly at lowered costs. We also draw on extensive medical expertise to identify promising new autologous therapies to leverage within the POCare Platform either via ownership or licensing. The POCare Network brings together patients, doctors, industry partners, research institutes and hospitals worldwide with a goal of achieving harmonized, regulated clinical development and production of the therapies.

Material Developments During Fiscal 20182020

Funding from SFPIAcquisitions and Dispositions

On November 15, 2017, we, MaSTherCell and the Belgian Sovereign Funds Société Fédérale de Participations et d'Investissement (“SFPI”) entered into a Subscription and Shareholders Agreement (the “ SFPI Agreement”) pursuant to which SFPI completed an equity investment in MaSTherCell in the aggregate amount of €5 million (approximately $5.9 million), for approximately 16.7% of MaSTherCell. The equity investment commitment included the conversion of the outstanding loan and accrued interest of Euro 1.07 million (approximately $1.18 million), previously made by SFPI to MaSTherCell.

Following the SFPI investment in MaSTherCell, in November 2017, MaSTherCell announced the expansion by 600m² of its facility in Belgium with a dedicated, late-stage clinical and commercial cGMP unit, which was opened in the first quarter of 2019. This new expansion enables MaSTherCell to augment its commercial capabilities in Europe with five state-of-the-art advanced manufacturing units and extended GMP-accredited quality control (QC) laboratories. On June 13, 2018, SPFI has paid the balance of Euro 1.9 million (approximately $2.3 million) to MaSTherCell.

-58-

Consolidation of CDMO Entities and Strategic Funding

On June 28, 2018, the Company, Masthercell Global Inc., a Delaware company and a newly formed subsidiary of the Company that holds our business relating to the third party contract manufacturing for cell therapy companies (CDMO) (“Masthercell Global”), Great Point Partners, LLC, a manager of private equity funds focusedAs mentioned above, on growing small to medium sized heath care companies (“Great Point”), and certain of Great Point’s affiliates, entered into a series of definitive strategic agreements intended to finance, strengthen and expand Orgenesis’ CDMO business. In connection therewith, the Company, Masthercell Global and GPP-II Masthercell, LLC, a Delaware limited liability company (“GPP-II”) and an affiliate of Great Point entered into Stock Purchase agreement (the “SPA”) pursuant to which GPP-II purchased 378,000 shares of newly designated Series A Preferred Stock of Masthercell Global (the “Masthercell Global Preferred Stock”), representing 37.8% of the issued and outstanding share capital of Masthercell Global, for cash consideration to be paid into Masthercell Global of up to $25 million, subject to certain adjustments (the “Consideration”). Orgenesis holds 622,000 shares of Masthercell Global’s Common Stock, representing 62.2% of the issued and outstanding equity share capital of Masthercell Global. An initial cash payment of $11.8 million of the Consideration was remitted at closing, with a follow up payment of $6,600,000 to be made in each of years 2018 and 2019 (the “Future Payments”), or an aggregate of $13.2 million, if (a) Masthercell Global achieves specified EBITDA and revenues targets during each of these years, and (b) the Orgenesis’ shareholders approve certain provisions of the Stockholders’ Agreement referred to below on or before December 31, 2019. None of the future Consideration amounts, if any, will result in an increase in GPP-II’s equity holdings in Masthercell Global beyond the 378,000 shares of Series A Preferred Stock issued to GPP-II at closing. The proceeds of the investment will be used to fund the activities of Masthercell Global and its consolidated subsidiaries. Notwithstanding the foregoing, GPP-II may, in its sole discretion, elect to pay all or a portion of the future Consideration amounts even if the financial targets described above have not been achieved and the Orgenesis Stockholder Approval has not been obtained. In satisfaction of the first of the two conditions described above, Masthercell Global achieved the specified EBITDA and revenues targets in 2018 as described in the SPA and received $6,600,000 of the Future Payments on January 16, 2019.

In connection with the entry into the SPA described above, each of the Company, Masthercell Global and GPP-II entered into the Masthercell Global Inc. Stockholders’ Agreement (the “Stockholders’ Agreement”) providing for certain restrictions on the disposition of Masthercell Global securities, the provisions of certain options and rights with respect to the management and operations of Masthercell Global, certain favorable, preferential rights to GPP-II (including, without limitation, a tag right, drag right and certain protective provisions), a right to exchange the Masthercell Global Preferred Stock for shares of Orgenesis common stock and certain other rights and obligations. In addition, after the earlier of the second anniversary of the closing or certain enumerated circumstances, GPP-II is entitled to effectuate a spinoff of Masthercell Global and the Masthercell Global Subsidiaries (the “Spinoff”). The Spinoff is required to reflect a market value determined by one of the top ten independent accounting firms in the U.S. selected by GPP-II, provided that under certain conditions, such market valuation shall reflect a valuation of Masthercell Global and the Masthercell Global Subsidiaries of at least $50 million. In addition, upon certain enumerated events as described below, GPP-II is entitled, at its option, to put to the Company (or, at Company’s discretion, to Masthercell Global if Masthercell Global shall then have the funds available to consummate the transaction) its shares in Masthercell Global or, alternatively, purchase from the Company its share capital in Masthercell Global at a purchase price equal to the fair market value of such equity holdings as determined by one of the top ten independent accounting firms in the U.S. selected by GPP-II, provided that the purchase price shall not be greater than three times the price per share of Masthercell Global Preferred Stock paid by GPP-II and shall not be less than the price per share of Masthercell Global Preferred Stock paid by GPP-II . GPP-II may exercise its put or call option upon the occurrence of any of the following: (i) there is an Activist Shareholder of the Company; (ii) the Chief Executive Officer and/or Chairman of the board of directors of the Company resigns or is replaced, removed, or terminated for any reason prior to June 28, 2023; (iii) there is a Change of Control event of the Company; or (iv) the industry expert director appointed to the board of directors of Masthercell Global is removed or replaced (or a new such director is appointed) without the prior written consent of GPP-II. For the purposes of the foregoing, the following definitions shall apply: (A) “Activist Shareholder” shall mean any Person who acquires shares of capital stock of the Company who either: (x) acquires more than a majority of the voting power of the Company, (y) actively takes over and controls a majority of the board of directors of the Company, or (z) is required to file a Schedule 13D with respect to such Person’s ownership of the Company and has described a plan, proposal or intent to take action with respect to exerting significant pressure on the management of or directors of, the Company; and (B) “Change of Control” shall mean any of: (a) the acquisition, directly or indirectly (in a single transaction or a series of related transactions) by a Person or group of Persons of either (I) a majority of the common stock of the Company (whether by merger, consolidation, stock purchase, tender offer, reorganization, recapitalization or otherwise), or (II) all or substantially all of the assets of the Company and its Subsidiaries (but only if such transaction includes the transfer of Securities held by the Company), (b) if any four (4) of the directors of the Company as of June 28, 2018 are removed or replaced or for any other reason cease to serve as directors of the Company, (c) the filing of a petition in bankruptcy or the commencement of any proceedings under bankruptcy laws by or against the Company, provided that such filing or commencement shall be deemed a Change of Control immediately if filed or commenced by the Company or after sixty (60) days if such filing is initiated by a creditor of the Company and is not dismissed; (d) insolvency of the Company that is not cured by the Company within thirty (30) days; (e) the appointment of a receiver for the Company, provided that such appointment shall constitute an Change of Control immediately if the appointment was consented to by the Company or after sixty (60) days if not consented to by the Company and such appointment is not terminated; or (f) or dissolution of the Company .

-59-

The Stockholders’ Agreement further provides that GPP-II is entitled, at any time, to convert its share capital in Masthercell Global for the Company’s common stock in an amount equal to the lesser of (a)(i) the fair market value of GPP-II’s shares of Masthercell Global Preferred Stock to be exchanged, as determined by one of the top ten independent accounting firms in the U.S. selected by GPP-II and the Company, divided by (ii) the average closing price per share of Orgenesis Common Stock during the thirty (30) day period ending on the date that GPP-II provides the exchange notice (the “Exchange Price”) and (b)(i) the fair market value of GPP-II’s shares of Masthercell Global Preferred Stock to be exchanged assuming a value of Masthercell Global equal to three and a half (3.5) times the revenue of Masthercell Global during the last twelve (12) complete calendar months immediately prior to the exchange divided by (ii) the Exchange Price; provided, that in no event will (A) the Exchange Price be less than a price per share that would result in Orgenesis having an enterprise value of less than $250,000,000 and (B) the maximum number of shares of Orgenesis Common Stock to be issued shall not exceed 2,704,247 shares of outstanding Orgenesis Common Stock (representing approximately 19.99% of then outstanding Orgenesis Common Stock), unless Orgenesis obtains shareholder approval for the issuance of such greater amount of shares of Orgenesis Common Stock in accordance with the rules and regulations of the Nasdaq Stock Market. Such shareholder approval for a greater number was obtained on October 23, 2018.

Great Point and Masthercell Global entered into an advisory services agreement pursuant to which Great Point is to provide management services to Masthercell Global for which Great Point will be compensated at an annual base compensation equal to the greater of (i) $250,000 per each 12 month period or (ii) 5% of the EBITDA for such 12 month period, payable in arrears in quarterly installments; provided, that these payments will (A) begin to accrue immediately, but shall not be paid in cash to Great Point until such time as Masthercell Global generates EBITDA of at least $2,000,000 for any 12 month period or the sale of or change in control of Masthercell Global, and (B) shall not exceed an aggregate annual amount of $500,000. Such compensation accrues but is not owed to Great Point until the earlier of (i) Masthercell Global generating EBITDA of at least $2 million for any 12 months period following the date of the agreement or (ii) a Sale of the Company or Change of Control of the Company (as both terms are defined therein).

GPP Securities, LLC, a Delaware limited liability company and an affiliate of Great Point and Masthercell Global entered into a transaction services agreement pursuant to which GPP Securities, LLC is to provide certain brokerage services to Masthercell Global for which GPP Securities LLC will be entitled to a certain Exit Fee and Transaction Fee (as both terms are defined in the agreement), such fees not to be less thanFebruary 2, percent of the applicable transaction value.

Corporate Reorganization

Contemporaneous with the execution of the SPA and the Stockholders’ Agreement, Orgenesis and Masthercell Global entered into a Contribution, Assignment and Assumption Agreement pursuant to which Orgenesis contributed to Masthercell Global the Orgenesis’ assets relating to the CDMO Business (as defined below), including the CDMO subsidiaries (the “Corporate Reorganization”). In furtherance thereof, Masthercell Global, as Orgenesis’ assignee, acquired all of the issued and outstanding share capital of Atvio, the Company’s Israel based CDMO partner since May 2016, and 94.12% of the share capital of CureCell, the Company’s Korea based CDMO partner since March 2016. Orgenesis exercised the” call option” to which it was entitled under the joint venture agreements with each of these entities to purchase from the former shareholders their equity holding. The consideration for the outstanding share equity in each of Atvio and CureCell consisted solely of Orgenesis common stock. In respect of the acquisition of Atvio, Orgenesis issued to the former Atvio shareholders an aggregate of 83,965 shares of Orgenesis common stock. In respect of the acquisition of CureCell, Orgenesis Inc. issued to the former CureCell shareholders an aggregate of 202,846 shares of Orgenesis Common Stock subject to a third-party valuation. Together with MaSTherCell S.A., Atvio and CureCell are directly held subsidiaries under Masthercell Global (collectively, the “Masthercell Global Subsidiaries”).

-60-

Masthercell Global, through the Masthercell Global Subsidiaries, will be engaged in the business of providing manufacturing and development services to third parties related to cell therapy products, and the creation and development of technology, and optimizations in connection with such manufacturing and development services for third parties (the “CDMO Business”). Under the terms of the Stockholders’ Agreement, Orgenesis has agreed that so long as it owns equity in Masthercell Global and for two years thereafter it will not engage in the CDMO Business, except through Masthercell Global (but may continue to engage in its other areas of business). In addition, except for certain limited circumstances, each of Orgenesis and GPP-II agreed in the Stockholders’ Agreement to not recruit or solicit or hire any officer or employee of Masthercell Global that was or is involved in the CDMO Business.

We intend, through our direct subsidiaries, to continue to engage in the manufacturing, researching, marketing, developing, selling and commercializing (either alone or jointly with third parties) products that are not directly related to the CDMO business, including, joint ventures, collaboration, partnership or similar arrangement with a third party.

Change of Fiscal Year

On October 22, 2018, the Board of Directors of the Company approved a change in the Company’s fiscal year end from November 30 to December 31 of each year. This change to the calendar year reporting cycle began January 1, 2019. As a result of the change, the Company will have a December 2018 fiscal month transition period, the results of which will be separately reported in the Company’s Quarterly Report on Form 10-Q for the calendar quarters ending March 31, 2019, June 30, 2019 and September 30, 2019, and in the Company’s Annual Report on Form 10-K for the calendar year ending December 31, 2019.

Collaboration Agreements with Immugenyx and Hemogenyx

On October 16, 2018,2020, we entered into a collaboration agreementPurchase Agreement with Immugenyx, LLC (“Immugenyx”), a wholly owned subsidiary of Hemogenyx Pharmaceuticals Plc (“Hemogenyx”). Immugenyx will collaborate withGPP, Masthercell and Catalent Pharma Solutions, Inc. pursuant to which the Company to further the development and commercialization of its advanced hematopoietic chimeras (“AHC”). AHC, a new type of humanized mouse with a functional human immune system, is being developed by Immugenyx as an in vivo platform for disease modelling, drug and cell therapy development. Pursuant to the termsSellers sold 100% of the agreement, we shall receive the worldwide rights to market the products and shall serve as a global distributor of Immugenyx’s products. Immugenyx will retain exclusive rights to manufacture, make and supply to the Company or our affiliates all the Immugenyx technology and/or licensed products that are marketed, sold or otherwise commercialized by the Company. In consideration for the license, we and/or our affiliates will advance to Immugenyx a convertible loan in an amount of no less than $1.0 million for advancing the development of humanized mice models and related antibody development. We also agreed to pay a royalty of 12%outstanding equity interests of our net revenues resulting from the sale or licensingMasthercell Business for an aggregate nominal purchase price of products involving the use of Immugenyx’s AHC technology. As of November 30, 2018,$315 million, subject to customary adjustments. After accounting for GPP’s liquidation preference and equity stake in Masthercell as well as other investor interests in its Belgian subsidiary MaSTherCell, S.A. (“MaSTherCell”), distributions to Masthercell option holders and transaction costs, we have funded $0.5received approximately $126.7 million. We incurred an additional approximately $5.6 million in transaction costs.

On October 18, 2018,April 7, 2020, we entered into a collaboration agreement with Hemogenyx Pharmaceuticals Plc to collaborate on the development and commercialization of Hemogenyx’s Human Postnatal Hemogenic Endothelial (“Hu-PHEC”) technology. Hu-PHEC is a cell replacement product candidate that is being designed to generate cancer-free, patient-matched blood stem cells after transplantation into the patient. Pursuant to the terms of the agreement, we shall manufacture and supply all Hu-PHEC related products both during and following completion of clinical trials. We shall also receive the worldwide rights to market the products and shall serve as a global distributor of Hemogenyx’s Hu-PHEC related products. In consideration for the license, we and/or our affiliates will advance to Hemogenyx a convertible loan in an amount of no less than $1.0 million for advancing the development of the Hu-PHEC technology. We also agreed to pay a royalty of 12% of our net revenues resulting from the sale or licensing of products involving the use of Hemogenyx’s Hu-PHEC technology. As of November 30, 2018, we have funded $0.5 million.

-61-

Convertible Note Agreements

During November 2018, we entered into private placement subscription agreementsAsset Purchase Agreement (the “Hemogenyx-Cell Subscription“Tamir Purchase Agreement”) with certain accredited investors (the “Investors”Tamir Biotechnology, Inc. (“Tamir” or “Seller”), pursuant to which we agreed to sellacquire certain assets and liabilities of Tamir related to the discovery, development and testing of therapeutic products for the treatment of diseases and conditions in humans, including all rights to ranpirnase and use for antiviral therapy (collectively, the “Purchased Assets and Assumed Liabilities” and such acquisition, the “Tamir Transaction”). The Tamir Transaction closed on April 23, 2020. As aggregate consideration for the acquisition, we paid $2.5 million in cash and issued an aggregate principal amount of $625,0003,400,000 shares (the “Shares”) of Common Stock to Tamir resulting in a 2% Unsecured Convertible Notetotal consideration of $20.2 million.

On September 26, 2020, we entered into an Agreement and Plan of Merger and Reorganization (the “Hemogenyx Convertible Note”“Merger Agreement”) by and among ourselves, Orgenesis Merger Sub, Inc., which is convertible, at the discretion of the Investor, into either (i) units, each unit consisting of one share of common stocka Delaware corporation and a wholly-owned subsidiary of the Company par value $0.0001(“Merger Sub”), Koligo Therapeutics Inc., a Kentucky corporation (“Koligo”), the shareholders of Koligo (collectively, the “Shareholders”), and Long Hill Capital V, LLC (“Long Hill”), solely in its capacity as the representative, agent and attorney-in-fact of the Shareholders. The Merger Agreement provides for the acquisition of Koligo by us through the merger of Merger Sub with and into Koligo, with Koligo surviving as our wholly-owned subsidiary (the “Merger”). The Merger was announced in a Current Report on Form 8-K filed with the Securities and Exchange Commission on October 1, 2020, to which a copy of the Merger Agreement, along with copies of certain other ancillary agreements, were annexed as exhibits. The Merger closed on October 15, 2020.

Koligo was a privately-held US regenerative medicine company. Koligo’s first commercial product is KYSLECEL® (autologous pancreatic islets) for chronic and acute recurrent pancreatitis. Koligo’s 3D-V technology platform incorporates the use of advanced 3D bioprinting techniques and vascular endothelial cells to support development of transformational cell and tissue products for serious diseases.

48

In addition, according to the agreement between the parties, we also funded an additional cash consideration of $500 thousand (with $100 thousand of such reducing the ultimate consideration payable to Koligo) for the acquisition of the assets of Tissue Genesis, LLC (“Tissue Genesis”) by Koligo that was consummated on October 14, 2020. The Tissue Genesis assets include the entire inventory of Tissue Genesis Icellator® devices, related kits and reagents, a broad patent portfolio to protect the technology, registered trademarks, clinical data, and existing business relationships for commercial and development stage use of the Icellator technology.

Private Placement

On January 20, 2020, we entered into a Securities Purchase Agreement with certain investors pursuant to which we issued and sold, in a private placement, 2,200,000 shares of Common Stock at a purchase price of $4.20 per share (“Common Stock”) and one three-year warrantwarrants to purchase one shareup to 1,000,000 shares of Common Stock at an exercise price of $7.00$5.50 per share at a conversion pricewhich are exercisable between June 2021 and January 2023. We received gross proceeds of $7.00 per unit (the “Units”approximately $9.24 million before deducting related offering expenses in the amount of $0.8 million.

Other Developments and Agreements During Fiscal 2020

Joint Ventures, Collaborations and License Agreements During Fiscal 2020

During 2020, we entered into joint venture agreements (“JVA”) or amended existing JVAs (“AJVA”) (which superseded previous JVAs), master service agreements for POC development revenue (“MSA DEV”) and master service agreements for procured services (“MSA PS”), with third parties as per the following table:

Name of Party (and country of origin)

Nature of

Agreement

TerritoryNotes
Theracell Advanced BiotechnologyAJVAGreece, Turkey, Cyprus, Israel, and Balkans(1)
Broaden Bioscience and Technology CorpAJVACertain projects in China and the Middle East(1)

Mircod LLC

(US)

JVARussia(2)

Image Securities FZC (UAE)

(a related party)

AJVA &

MSA PS

India

(1)

Cure TherapeuticsJVAKorea and Japan
Kidney Cure LtdJVAN/A(5)
Sescom LtdJVAN/A(6)

Educell D.O.O

(Slovenia)

AJVA &

MSA PS &

MSA DEV

Croatia, Serbia and Slovenia(1)

Med Centre for Gene and

Cell Therapy FZ-LLC (UAE)

AJVA &

MSA PS &

MSA DEV

UAE(1)
Mida Biotech B.V. (Netherlands)

AJVA &

MSA PS &

MSA DEV

Netherlands, Lithuania, Spain, Switzerland,

Germany, Belgium and other countries

within West Europe

(7)
Butterfly Biosciences SarlJVAN/A(8)

Notes:

(1) The parties will collaborate in POC processing, regulatory and therapy development including the setting up one or more point of care processing facilities in institutions or hospitals in the territory, the supply of our products and services within the Territory, and the clinical development and commercialization of the relevant third-party products worldwide.

(2) The parties will collaborate in POC processing, regulatory and therapy development including the setting up one or more point of care processing facilities in institutions or hospitals in the territory, the supply of our products and services within the Territory and clinical, regulatory, development and commercialization of cell and gene therapies in the Territory.

49

(5) The parties will collaborate in the (i) implementation of a point-of-care strategy; (ii) sharesassessment of capital stockthe options for development and manufacture of Hemogenyx-Cell,various cell-based types (including kidney derived cells, MSC cells, exosomes, gene therapies) development; and (iii) development of protocols and tests for kidney therapies.

(6) The parties will collaborate in (i) the assessment of relevant tools and technologies to be used in our information security system (the “ISS”); (ii) the implementation of the ISS within the Company and in our point-of-care network; and (iii) the operation and maintenance of the ISS.

(7) The parties will collaborate in POC processing, regulatory and therapy development including the setting up one or more point of care processing facilities in institutions or hospitals in the territory and the establishment of an induced pluripotent stem cells R&D and automation platforms and other early-stage development activities.

(8) We and Kidney Cure Ltd own 49% and 51%, respectively, of Butterfly Biosciences Sarl (“BB”). BB is the entity through which the Kidney Cure JVA activities will be completed.

Other License Agreements

We are now working on the completion of all the IND enabling requirements in order to get into Phase I studies under the Sponsored Research Agreement (the “SRA”) and Exclusive License Agreement between ourselves and the Trustees of Columbia University in the City of New York, a subsidiaryNew York corporation (“Columbia University”). In 2019, we entered into an SRA with Columbia University whereby we will provide financial support for studying the utility of Hemogenyx, atserological tumor marker for tumor dynamics monitoring. Also in 2019, we and Columbia University entered into an Exclusive License Agreement (the “Columbia License Agreement”) whereby Columbia University granted to us an exclusive license to discover, develop, manufacture and sell product in the field of cancer therapy. In consideration of the licenses granted under the Columbia License Agreement, we shall pay to Columbia University (i) a price per share based on a pre-money valuationroyalty of Hemogenyx-Cell5% of $12,000,000 (the “Hemogenyx Securities”) pursuant to the previously disclosed collaboration agreement with Hemogenyx.net sales of any patented product sold and (ii) 2.5% of net sales of other products.

In addition, on such same month,On May 15, 2019, we entered into a private placement subscriptionJoint Venture Agreement with SBH Sciences, Inc., a Massachusetts corporation (“SBH”), for the establishment of a joint venture with SBH for the purpose of collaborating in the field of gene and cell therapy development, process and services of bio-exosome therapy products and services in the areas of diabetes, liver cells and skin applications, including wound healing.

In October 2019, we concluded a license agreement (the “Immugenyx Subscription Agreement”) with the Investors,Caerus Therapeutics Corporation (a related party), a Virginia company (“Caerus”), pursuant to which Caerus granted us, among others, an exclusive license to all Caerus IP relating to Advance Chemeric Antigen Vectors for Targeting Tumors for the development and/or commercialization of certain licensed products. In consideration for the license granted to us under this agreement, we agreedshall pay Caerus feasibility fees, annual maintenance fees and royalties of sales of up to sell an aggregate principal amount5% and up to 18% of $625,000 insub-license fees. Through this joint venture, the parties co-develop a 2% Unsecured Convertible Note (the “Immugenyx Convertible Note ”).novel CART and CAR-NK platform for the treatment of solid tumors. The Investors may convert all or any portiondevelopment is at a pre-clinical stage.

On December 20, 2019, we and the Regents of the outstanding principal amountUniversity of California (“University”) entered into a joint research agreement in the Immugenyx Convertible Note, plus accrued interest thereon, intofield of therapies and processing technologies according to an agreed upon work plan. According to the atagreement, we will pay the discretionUniversity royalties of up to 5% (or up to 20% of sub-licensing sales) in the Investors, either (i) units, each unit consistingevent of one sharesales that includes certain types of common stockUniversity owned IP.

During the third quarter of 2020, we purchased the Company, par value $0.0001 per share (“Common Stock”)IP and one three-year warrant to purchase one share of Common Stock at an exercise price of $7.00 per share, atrelated EV technology from a conversion price of $7.00 per unitservice provider (the “Units”) or (ii) shares of capital stock of Immugenyx , at a price per share based on a pre-money valuation of Immugenyx of $8,000,000 (the “Immugenyx Securities”“Service Provider”) pursuant to an EV agreement (the “EV agreement”). According to the previously disclosed collaborationEV agreement, with Immugenyx.the Service Provider sold to us all of its rights in the EV technology that it had produced, in the amount of $500 thousand, to be paid in installments over the next 12 months from September 2020. In addition, the Service Provider granted us an exclusive worldwide license to use the EV IP technology for any purpose.

50

The entire principal amount, plus accrued interest thereon, shall automatically convert into Units if at any time from and afterIncluded in the date hereof, the closing pricepurchased assets of the Company’s Common Stock onTamir Biotechnology Inc. acquisition was the Nasdaq Capital Market (or other national stock exchangeassumption by us of a worldwide license to a private company of certain Tamir technologies in the field of treatment, amelioration, mitigation or market on whichprevention of diseases or conditions of the Common Stock is then listed or quoted) equals or exceeds $20.00 per share (which amount may be adjustedeye and its adnexa in return for certain capital events, such as stock splits) for thirty (30) consecutive trading days.

The Convertible Notes contain standarddevelopment and customary events of default including, but not limitedsales milestone payments to failurebe paid to makeTamir. This license fee and the right to receive future milestone payments when due, failure(of up to observe or perform covenants or agreements contained$11 million assuming that certain milestones are reached) and royalties (of up to $35 million based on net sales milestones), were assumed by us in connection with the Convertible Notes, the breach of any material representation or warranty contain therein or the bankruptcy or insolvency of the Company. If any event of default occurs, subject to any cure period, the full principal amount,Tamir Purchase Agreement together with interest (including default interest of 12% per annum) and other amounts owing in respect thereof to thea less than 10% share interest. To date, of acceleration shall become, at the Investor’s election, immediately due and payable in cash.no milestones have been reached.

The WarrantsAs mentioned above, included in the Units expire three years fromKoligo acquisition were the dateassets of issuanceTissue Genesis, LLC (“Tissue Genesis”). We are committed to paying the previous owners of Tissue Genesis up to $500 thousand upon the achievement of certain performance milestones and have an exercise price of $7.00 per share. If at any time from and afterearn-out payments on future sales provided that in no event will the date of issuance, the closing price of our Common Stock on the Nasdaq Capital Market (or other national stock exchange or market on which the Common Stock is then listed or quoted) equals or exceeds $20.00 per share (which amount may be adjusted for certain capital events, such as stock splits, as described herein) for thirty (30) consecutive trading days, then the Company shall have the right to require the holder to exercise all or any portionaggregate of the Warrant still unexercised for a cash exercise into shares of Common Stock in accordance with the terms of the Warrant.earn-out payments exceed $4 million.

 

-62-

Results of Operations

Comparison of the year ended November 30, 2018Year Ended December 31, 2020 to the year ended November 30, 2017Year Ended December 31, 2019.

Our financial results for the year ended November 30, 2018December 31, 2020 are summarized as follows in comparison to the year ended November 30, 2017:December 31, 2019:

 Year Ended November 30,  Year Ended December 31, 
 2018 2017  2020  2019 
  (in thousands)  (in thousands) 
Revenues $18,655 $10,089               $6,177  $2,629 
Cost of sales 10,824  6,807 
Research and development expenses, net 6,464  2,478 
Revenues from related party  1,475   1,270 
Research and development expenses and Research and development service expenses, net  83,986   14,014 
Amortization of intangible assets 1,913 1,631   478   430 
Selling, general and administrative expenses 16,303  9,189   18,973   11,451 
Other income  (2,930) -   (4)  (21)
Share in losses of associated company  731 1,214 
Share in income of associated company  (106)  - 
Financial expense, net  3,117 2,447   1,061   843 
Loss before income taxes $17,767 $13,677 
     
Loss from continuing operation before income taxes $96,736  $22,818 

 

Revenues

  Year Ended November 30,
  2018 2017
   (in thousands) 
Services $14,065 $8,024 
Goods  4,590  2,065 
Total $18,655 $10,089 
          

AllThe following table shows our revenues were derived from the CDMO segment, most of which were generated from our Belgian Subsidiary, MaSTherCell S.A. We believe thatby major revenue diversification by source in the CDMO segment, together with a leading position in immunotherapy and, in particular, CAR T-cell therapy development and manufacturing, strengthened MaSTherCell’s resilience in the industry.streams:

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Revenue stream:        
POC and hospital services $6,068  $3,109 
Cell process development services  1,584   790 
Total $7,652  $3,899 

Our revenues for the year ended November 30, 2018December 31, 2020 were $18,655$7,652 thousand, as compared to $10,089$3,899 thousand for the corresponding period in 2017,year ended December 31, 2019, representing an increase of 85%96%. The increase in revenues for the year ended November 30, 2018December 31, 2020 compared to the corresponding periodyear ended December 31, 2019 is mainly attributable to increased POC services revenue.

51

POC services are mainly the result of agreements between us and our joint venture partners (See note 11). Pursuant to the agreements, we provide certain services in 2017support of partners’ activity. We have signed master services agreements partners in the aggregate amount of over $38 million for services to be provided from 2021 to 2022.

A breakdown of the revenues per customer that constituted at least 10% of revenues is as follows:

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Revenue earned:        
Customer A $2,857  $1,420 
Customer B  1,577   - 
Customer C – related party  1,475   1,270 
Customer D  1,412   857 

Research and Development and Research and Development Services, net:

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Salaries and related expenses $5,175  $3,064 
Stock-based compensation  481   776 
Professional fees and consulting services  3,463   3,419 
Lab expenses  2,348   3,229 
First Choice JVA (See Note 11)  -   2,741 
Tamir Purchase Agreement (See Note 4)  19,225   - 
Depreciation expenses, net  603   521 
Other research and development expenses  52,887   1,076 
Less – grant  (196)  (812)
Total $83,986  $14,014 

Research and development expenses for the year ended December 31, 2020 were $83,986 thousand, as compared to $14,014 thousand for the year ended December 31, 2019, representing an increase of 499%.

The increase is mainly attributable to the following:

expansion of our pipeline of licensed CGTs with a harmonized pathway for regulatory approval;
expansion of our POC capacity globally;
investment in automated processing units & processes;
developing owned and licensed advanced therapies to enable commercial production;
works with partners to enable efficient closed processing system technologies addressing POCare needs;
an increase in salaries and related expenses and other research and development expenses. Additional R&D staff were hired as we expanded our research and development to the evaluation and development of new cell therapies and related technologies in the field of immune-oncology (our novel CD19 CAR-T and CD19.22 CAR-T programs, cellular vaccination for solid cancers, advanced tumor infiltrating lymphocyte, NK-based therapies, etc.), liver pathologies, stem cell-based therapies and other cell-based technologies such as the novel delivery system, Bioxomes. We invested in converting biological processes to GMP-compliant processes as these therapies progress to clinical stage;
In 2020 we made significant investments in the development of several types of Orgenesis Mobile Processing Units and Labs (OMPULs) with the expectation of use and/or distribution through our POCare Network of partners, collaborators, and joint ventures. OMPULs are designed for the purpose of validation, development, performance of clinical trials, manufacturing and/or processing of potential or approved cell and gene therapy products in a safe, reliable, and cost-effective manner at the point of care, as well as the manufacturing of such CGTs in a consistent and standardized manner in all locations. The design delivers a potential industrial solution for us to deliver CGTs to practically any clinical institution at the point of care; and
The Tamir purchase agreement (See Note 4).

52

Selling, General and Administrative Expenses

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Salaries and related expenses $3,379  $2,332 
Stock-based compensation  1,915   1,855 
Accounting and legal fees  6,946   2,388 
Professional fees  1,571   1,553 
Rent and related expenses  407   214 
Business development  3,477   1,148 
Expenses related to collaboration with Theracell  -   689 
Depreciation expenses, net  101   113 
Other general and administrative expenses  1,177   1,159 
Total $18,973  $11,451 

Selling, general and administrative expenses for the year ended December 31, 2020 were $18,973 thousand, as compared to $11,451 thousand for the year ended December 31, 2019, representing an increase of 66%. The increase for the year ended December 31, 2020 is primarily attributable to:

(i)An increase in salaries and related expenses of $1,047 thousand, as a result of additional managerial appointments and increased salaries;
(ii)An increase in accounting and legal fees of $4,558 thousand, which is mainly attributable to additional legal fees incurred for recent business and collaboration agreements; and
(iii)An increase in business development of $2,329 thousand, as a result of increased activities to establish our presence in new markets.

Financial Expenses, net

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Decrease in fair value financial liabilities and assets measured at fair value $-  $63 
Interest expense on convertible loans and loans  1,254   498 
Foreign exchange loss, net  160   395 
Other income  (353)  (113)
Total $1,061  $843 

Financial expenses, net for the year ended December 31, 2020 were $1,061 thousand, as compared to $843 thousand for the year ended December 31, 2019, representing an increase of 26%. The increase for the year ended December 31, 2020 is primarily attributable to an increase in interest expense on convertible loans and loans of $756 thousand.

53

Tax income

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Tax income $1,609  $229 
Total $1,609  $229 

Tax income, net for the projects provided by MaSTherCell S.A, resultingyear ended December 31, 2020 were $1,609 thousand, as compared to $229 thousand for the year ended December 31, 2019, representing an increase of 603%.  The increase for the year ended December 31, 2020 is primarily fromattributable due to the release of a tax asset up to the amount of Koligo’s net tax liability. See Note 4.

Discontinued Operations

Discontinued operations relate to the Masthercell Business. The following table presents the financial results associated with the Masthercell Business operation as reflected in our Consolidated Comprehensive loss:

OPERATIONS Year Ended December 31, 
  2020  2019 
  (in thousands): 
Revenues $2,556  $31,053 
Cost of revenues  1,482   18,318 
Cost of research and development and research and development services, net  7   54 
Amortization of intangible assets  137   1,631 
Selling, general and administrative expenses  1,896   13,886 
Other (income) expenses, net  305   (207)
Operating loss  1,271   2,629 
Financial expenses, net  (29)  31 
Loss before income taxes  1,242   2,660 
Tax expenses (income)  (30)  792 
Net loss from discontinuing operation, net of tax $1,212  $3,452 

Revenues are attributable to the extension of existing customer service contracts with biotechnology clients as well asand from revenues generated from existing manufacturing agreements.

In addition, we acquired all the issued and outstanding share capital of Atvio, our Israel-based CDMO partner since August 2016, and 94.12% of the share capital of CureCell, our Korea-based CDMO partner since March 2016, which are both reflected in the increase in our revenues from services provided of $1,174 thousand during the year ended November 30, 2018.

Backlog

We define our backlog as products that we are obligated to deliver or services to be rendered based on firm commitments relating to purchase orders received from customers. As of November 30, 2018, MaSTherCell S.A. had a backlog of approximately $12.6 million, consisting of services that we expect to deliver into fiscal year 2019.However, no assurance can be provided that such contracts will not be cancelled, in which case we will not be authorized to deliver and record the anticipated revenues.

-63-

Expenses

Cost of Revenues

  Year Ended November 30,
  2018 2017
   (in thousands) 
Salaries and related expenses $4,915 $2,642 
Stock-based compensation  126  - 
Professional fees and consulting services  145  - 
Raw materials  4,614  2,692 
Depreciation and amortization expenses, net  391  986 
Other expenses  633  487 
  $10,824 $6,807 
          

Cost of revenues forwere in line with the year ended November 30, 2018 were $10,824 thousand, as compared to $6,807 thousand during the same periodgrowth in 2017, representing an increaserevenues and employment of 59%. The increase for the year ended November 30, 2018 as compared to the corresponding period in 2017 is primarily attributed to the following:

(i)An increase in salaries and related expenses of $2,273 thousand, primarily attributable to an increase of activities andadditional operational staff. This is in line with the increase in revenue of MaSTherCell S.A., as well as the inclusion of salaries and related expenses of Atvio and CureCell for the five months ended in November 30, 2018 (not included in the prior year).

(ii)An increase of stock-based compensation for the year ended November 30, 2018 generated from options granted to employees.

(iii)An increase of $1,922 thousand in raw materials, mainly attributed to the growth in the volume of services provided by MaSTherCell S.A., both from existing and new manufacturing agreements.

(iv)A decrease of $595 thousand in depreciation and amortization expenses. This was primarily attributable to the increase in the production facility and laboratory equipment useful life from 10 to 20 years and from 5 to 3 years, respectively. This change occurred in the last quarter of 2017.

Research and Development Expenses

  Year Ended November 30, 
  2018 2017 
  (in thousands) 
Salaries and related expenses $2,077 $1,181 
Stock-based compensation  659  711 
Professional fees and consulting services  605  854 
Lab expenses  3,370  287 
Depreciation expenses, net  320  110 
Other research and development expenses  355  183 
Less – grant  (922)  (848) 
Total $6,464 $2,478 

The increase in research and development expenses reflects management’s determination to move transdifferentiating technology to the next the stage towards clinical studies. In the fiscal year ended 2018, we focused primarily on combining the in vitro research to increase insulin production and secretion with pre-clinical studies aiming to evaluate the efficacy and safety of the product in rodents' model. In addition, we evaluated new transplantation methods during this period. Sourcing of the starting material (liver sampling and cell collection) and upscaling of virus production and cell propagation using advanced technologies complement this effort with the target to establish start to end production capabilities.

The scope of research and development expenses was also expanded to the evaluation and development of new cell therapies related technologies in the field of immunoncology, liver pathologies and others. In furtherance of these developments, salaries and related expenses increased for the year ended November 30, 2018 compared to 2017, primarily due to the expansion of our development team in Israel and Belgium.

-64-

Research and development expenses (net) for the fiscal year ended November 30, 2018 were $6,464 thousand, as compared to $2,478 thousand for the same period in 2017, representing an increase of 160%. The increase in research and development, net expenses in the year ended November 30, 2018 is primarily attributable to the following:

(i)An increase of $896 thousand in salaries and related expenses primarily attributable to an increase of activities and operational staff.

(ii)A decrease of $249 thousand in the expenses of professional fees and consulting services, mostly related to the conclusion of the BIRD and KORIL projects in Orgenesis Ltd. and decrease in consultant service in Orgenesis Maryland Inc

(iii)An increase of $3,083 thousand of lab expenses, mostly attributed to new therapeutics projects and to the DGO6 project.

Selling, General and Administrative Expenses

  Year Ended November 30,
  2018 2017
   (in thousands) 
Salaries and related expenses $4,581 $2,862 
Stock-based compensation  3,399  1,155 
Accounting and legal fees  2,528  1,773 
Professional fees  2,000  2,017 
Rent and related expenses  1,281  859 
Business development  1,557  599 
Other general and administrative expenses  957  (76) 
Total $16,303 $9,189 
          

Selling, general and administrative expenses forincluded additional managerial appointments, increased professional fees, additional rental space including in the fiscal year ended November 30, 2018 were $16,303 thousand, as compared to $9,189 thousand for the same period in 2017, representingU.S., and an increase of 77%. The increase is primarily attributable to:business development expenses.

(i)An increase of $1,719 thousand in salaries and related expenses as a result of additional managerial appointments, and salaries and related expenses of CureCell, Atvio and Masthercell Global not previously consolidated.

(ii)An increase of $2,244 thousand in stock-based compensation as a result of options granted to employees and consultants.

(iii)An increase of $755 thousand in accounting and legal fees mainly attributed to expenses related to the Company’s Nasdaq listing, and legal and accounting services related to the strategic agreements with GPP-II and the CureCell and Atvio consolidation.

(iv)An increase of $422 thousand in rent and related expenses mainly related to the occupation of additional space rented by MaSTherCell S.A. and to rent and related expenses in 2018 of CureCell and Atvio (not previously consolidated).

(v)An increase of $958 thousand in business development expenses related to the increase in the related activities during the year.
(vi)(vi) An increase of $1,033 thousand in other general and administrative expenses related to the increase in filing and other fees.

-65-

Financial Expenses, net

  Year Ended November 30,
  2018 2017
   (in thousands) 
Changes in fair value financial liabilities and assets measured at fair value $50 $(902) 
Stock-based compensation related to warrants granted debt holders  180  1,497 
Interest expense on convertible loans and loans  2,753  1,233 
Foreign exchange loss, net  129  562 
Other expenses  7  57 
Total $3,117 $2,447 
          

Financial expenses, net for the fiscal year ended November 30, 2018, increased by $670 thousand, compared to the same period in 2017. The increase in financial expenses is primarily attributable to:

(i)An increase of $952 thousand in fair value of the put options of Atvio.

(ii)A decrease of $1,317 thousand in stock-based compensation related to warrants granted to bondholders.

(iii)An increase of $1,520 thousand in interest expenses on convertible loans and loans.

Tax expenses (income)

  Year Ended November 30,
  2018 2017
   (in thousands) 
Tax expenses (income) $1,337 $(1,310) 
Total $1,337 $(1,310) 
          

Tax expenses for the fiscal year ended November 30, 2018, increased by $2,647 thousand, compared to the same period in 2017. The increase in tax expenses is mainly due to a decrease in deferred taxes related to carryforward losses in MaSTherCell S.A.

Working Capital

 November 30,  December 31, 
 2018  2017  2020  2019 
 (in thousands)  (in thousands) 
Current assets$30,297 $7,295  $50,077  $78,348 
Current liabilities 17,145  16,914  $16,285  $42,434 
Working capital (deficiency)$13,152 $(9,619) 
Working capital $33,792  $35,914 

Current assets increaseddecreased by $23,002$28,271 thousand between December 31, 2019 and December 31, 2020, which was primarily attributable to the following: (i) an increase in cash and cash equivalents due to proceeds from private placements of debtthe Masthercell sale; and equity securities and a cash payment and receivable of $16.9 million from GPP-II to our subsidiary, Masthercell Global; (ii) an increase in inventory and accounts receivable as a result of POC revenues.

Current liabilities decreased by $26,149 thousand between December 31, 2019 and December 31, 2020, which was primarily attributable to the following: (i) an increase in accounts payable and accrued expenses due to the acquisitionexpanded operations, (ii) an increase in current maturities of CureCell and (iii) higher sales of MaSTherCell.convertible loans.

-66-
54

Liquidity and Capital Resources

 Year Ended November 31, Year Ended December 31, 
 2018 2017 2020  2019 
  (in thousands)  (in thousands) 
Net loss $(19,104) $(12,367)  $579  $(26,041)
              
Net cash used in operating activities  (15,682) (3,833)   (78,046)  (13,220)
Net cash used in investing activities  (6,268) (3,404) 
Net cash provided by (used in) investing activities  105,610   (13,778)
Net cash provided by financing activities  35,060 8,365   5,881   24,098 
Increase in cash and cash equivalents $13,110 $1,128 
     
Net change in cash and cash equivalents and restricted cash $33,445  $(2,900)

Since inception,On February 2, 2020, we have funded our operations primarily through private placementsentered into a Stock Purchase Agreement (the “Purchase Agreement”) with GPP-II Masthercell LLC (“GPP” and debt instrumentstogether with us, the “Sellers”), Masthercell Global Inc. (“Masthercell”) and through revenues generated fromCatalent Pharma Solutions, Inc. (the “Buyer”). Pursuant to the activitiesterms and conditions of the Purchase Agreement, on February 10, 2020 the Sellers sold 100% of the outstanding equity interests of Masthercell to Buyer (the “Masthercell Sale”) for an aggregate nominal purchase price of $315 million, subject to customary adjustments. After accounting for GPP’s liquidation preference and equity stake in Masthercell as well as SFPI – FPIM’s interest in MaSTherCell S.A., our Belgian Subsidiary. Asdistributions to Masthercell option holders and transaction costs, we received approximately $126.7 million, of November 30, 2018, we had positive working capitalwhich $7.2 million was used for the repayment of $13.2 million, including cashintercompany loans and cash equivalents and restricted cash of $16.5 million.payables.

Net cash used in operating activities for the year ended December 31, 2020 was approximately $15.7$78 million, for the fiscal year ended November 30, 2018, as compared withto net cash used in operating activities of approximately $3.8$13 million for the same periodyear ended December 31, 2019. Since the Masthercell Sale, we entered into new joint venture agreements with new partners in 2017.various jurisdictions. This has allowed us to grow our infrastructure and expand our processing sites into new markets and jurisdictions. In addition, we engaged some of these joint venture partners to perform research and development services to further develop and adapt our systems and devices for specific purposes. We expanded our pre-clinical studies in the U.S., Israel, Belgiuminvested manpower and South Korea. The increase reflects management’sfinancial resources to focus on movingdeveloping, manufacturing and rolling out several types of OMPULs to be used and/or distributed through our trans-differentiation technology with first indication in Type 1 DiabetesPOCare Network of partners, collaborators, and joint ventures.

Net cash provided by investing activities for the year ended December 31, 2020 was approximately $106 million, as compared to the next stage towards clinical trials. We also expanded our global activity of the CDMO business with Masthercell Global, while maintaining the same level of cash used in operating activities as a result of the increased revenues at our subsidiaries MaSTherCell, Cure Cell and Atvio, thereby increasing gross profit and generating cash to pay our ongoing operating expenses. Additionally, we improved payment terms to our service providers.

Netnet cash used in investing activities for the fiscal year ended November 30, 2018 wasof approximately $6.3 million, as compared with approximately $3.4$14 million for the same period in 2017. Net cash used in investing activities was primarily for additions to fixed assets at our subsidiaries, MaSTherCell, CureCelland Atvio.

During the year ended November 30, 2018, our financing activities consisted of proceeds from private placements of our equity securities, warrants exercise and equity-linked instruments inDecember 31, 2019. This was mainly attributable to the net amount of approximately $21.5 million and $12.6 million from SFPI and GPP.Masthercell sale.

Liquidity and Capital Resources Outlook

WeBased on our current cash resources and commitments, we believe that we will be able to maintain our business plan will provide sufficient liquidity to fund ourcurrent planned activities and expected level of expenditures for at least 12 months from the date of the issuance of the financial statements. If increases are incurred in operating needscosts in general and administrative expenses for the next 12 months. However, there are factors that can impact our ability continue to fund our operating needs, including:

·our ability to expand sales volume, which is highly dependent on implementing our growth strategy in Masthercell Global;
·restrictions on our ability to continue receiving government funding for our PT business;
·additional CDMO expansion into other regions that we may decide to undertake; and
·the need for us to continue to invest in operating activities to remain competitive or acquire other businesses and technologies and to complement our products, expand the breadth of our business, enhance our technical capabilities or otherwise offer growth opportunities.

Iffacilities expansion, research and development, commercial and clinical activity or if we cannot effectively manage these factors,experience decreases in revenues from customers, we may need to raiseseek additional capital before such datefinancing. In addition, additional funds may be necessary to fund our operating needs.

From December 1, 2017 to the date of this Annual Report on Form 10-K, we funded our operations primarily from the proceeds from private placementsfinance some of our equity securitiescollaborations and convertible debt and from revenues generated by Masthercell Global, mainly revenues generated from MaSTherCell. From December 1, 2017 through November 30, 2018, we received, through MaSTherCell, proceeds of approximately $17.3 million in revenues and accounts receivable from customers, $12.6 million from SFPI and GPP-II and $21.5 million from private placements to accredited investors of our equity and convertible debt, net of finders’ fees, and exercise of warrants. In addition, from December 1, 2018 through February 13, 2019, we raised $0.25 million from the private placement of our equity-linked securities, $6.6 million from GPP and proceeds of approximately $4.7 million in accounts receivable from customers of MaSTherCell.joint ventures.

-67-

We believe that the investment consummated in June 2018 by an affiliate of GPP in our newly formed subsidiary, Masthercell Global, which has included to the date of this report a total net amount of $16.9 million and, subject to meeting certain specified financial targets and other conditions over the course of 2019, an additional $6.6 million, should cover the costs associated with the current business plan of Masthercell Global.

In December 2018, we entered into a Controlled Equity Offering Sales Agreement, or Sales Agreement, with Cantor Fitzgerald & Co., or Cantor, pursuant to which we may offer and sell, from time to time through Cantor, shares of our common stock having an aggregate offering price of up to $25.0 million. We will pay Cantor a commission rate equal to 3.0% of the aggregate gross proceeds from each sale. Shares sold under the Sales Agreement will be offered and sold pursuant to our Shelf Registration Statement on Form S-3 (Registration No. 333-223777) that was declared effective by the Securities and Exchange Commission on March 28, 2018, or the Shelf Registration Statement, and a prospectus supplement and accompanying base prospectus that we filed with the Securities and Exchange Commission on December 20, 2018. We have not yet sold any shares of our common stock pursuant to the Sales Agreement.

55

Critical Accounting Policies and Estimates

 

Our significant accounting policies are more fully described in the notes to our financial statements included in this Annual Report on Form 10-K for the fiscal year ended November 30, 2018.December 31, 2020. We believe that the accounting policies below are critical for one to fully understand and evaluate our financial condition and results of operations.

Fair Value Measurement

The fair value measurement guidance clarifies that fair value is an exit price, representing the amount that would be received to sell an asset or paid to transfer a liability in an orderly transaction between market participants. As such, fair value is a market-based measurement that should be determined based on assumptions that market participants would use in the valuation of an asset or liability. It establishes a fair value hierarchy that prioritizes the inputs to valuation techniques used to measure fair value. The hierarchy gives the highest priority to unadjusted quoted prices in active markets for identical assets or liabilities (Level 1 measurements) and the lowest priority to unobservable inputs (Level 3 measurements). The three levels of the fair value hierarchy under the fair value measurement guidance are described below:

Level 1 - Unadjusted quoted prices in active markets that are accessible at the measurement date for identical assets or liabilities;

Level 2 - Quoted prices in markets that are not active, or inputs that are observable, either directly or indirectly, for substantially the full term of the asset or liability; or

Level 3 - Prices or valuation techniques that require inputs that are both significant to the fair value measurement and unobservable (supported by little or no market activity).

We did not have any Level 1 or Level 2 assets and liabilities as of November 30, 2018 and 2017.

The derivative liabilities are Level 3 fair value measurements; we did not have any Level 3 assets and liabilities as of November 30, 2018.

Business Combination

The Company allocatesWe allocate the purchase price of an acquired business to the tangible and intangible assets acquired and liabilities assumed based upon theirour estimated fair values on the acquisition date. Any excess of the purchase price over the fair value of the net assets acquired is recorded as goodwill. Acquired in-process backlog, customer relations, brand name know, technology and knowIPR&D how are recognized at fair value. The purchase price allocation process requires management to make significant estimates and assumptions, especially at the acquisition date with respect to intangible assets. Direct transaction costs associated with the business combination are expensed as incurred. The allocation of the consideration transferred in certain cases may be subject to revision based on the final determination of fair values during the measurement period, which may be up to one year from the acquisition date. The Company includesWe include the results of operations of the business that it haswe have acquired in itsour consolidated results prospectively from the date of acquisition.

-68-

If the business combination is achieved in stages, the acquisition date carrying value of the acquirer’s previously held equity interest in the acquiree is re-measured to fair value at the acquisition date; any gains or losses arising from such re-measurement are recognized in profit or loss.

Redeemable Non-controlling InterestGoodwill

Non-controlling interests with embedded redemption features, whose settlement is not at the Company’s discretion, are considered redeemable non-controlling interest. Redeemable non-controlling interests are considered to be temporary equity and are therefore presented as a mezzanine section between liabilities and equity on the Company's consolidated balance sheets. Subsequent adjustment of the amount presented in temporary equity is required only if the Company's management estimates that it is probable that the instrument will become redeemable. Adjustments of redeemable non-controlling interest to its redemption value are recorded through additional paid-in capital.

Revenue Recognition

The Company recognizes revenue for services linked to cell process development and cell manufacturing services based on individual contracts in accordance with Accounting Standards Codification (“ASC”) 605,Revenue Recognition,when the following criteria have been met: persuasive evidence of an arrangement exists; delivery of the processed cells has occurred or the services that are milestones based have been provided; the price is fixed or determinable and collectability is reasonably assured. The Company determines that persuasive evidence of an arrangement exists based on written contracts that define the terms of the arrangements. In addition, the Company determines that services have been delivered in accordance with the arrangement. The Company assesses whether the fee is fixed or determinable based on the payment terms associated with the transaction and whether the sales price is subject to refund or adjustment. Service revenues are recognized as the services are provided. In addition, as part of the services, the Company recognizes revenue based on use of consumables, which it received as reimbursement on a cost-plus basis on certain expenses.

Goodwill

Goodwill represents the excess of the purchase price of acquired businessconsideration transferred over the estimated fair value assigned to the net tangible and identifiable intangible assets of businesses acquired. Goodwill is allocated to reporting units expected to benefit from the identifiable net assets acquired.business combination. Goodwill is not amortized but israther tested for impairment at least annually (at November 30), atin the reporting unit levelfourth quarter, or more frequently if events or changes in circumstances indicate that the asset mightgoodwill may be impaired. Following the sale of Masthercell, we manage the business as one operating segment and one reporting unit. Goodwill impairment is recognized when the quantitative assessment results in the carrying value exceeding the fair value, in which case an impairment charge is recorded to the extent the carrying value exceeds the fair value.

There were no impairment charges to goodwill during the periods presented.

Impairment of Long-lived Assets

We will periodically evaluate the carrying value of long-lived assets to be held and used when events and circumstances warrant such a review. The goodwill impairment testcarrying value of a long-lived asset is appliedconsidered impaired when the anticipated undiscounted cash flow from such asset is separately identifiable and is less than its carrying value. In that event, a loss is recognized based on the amount by performing a qualitative assessment before calculatingwhich the carrying value exceeds the fair value of the reporting unit. If, on the basis of qualitative factors, it is considered not more likely than not that the fairlong-lived asset. Fair value of the reporting unit is less than the carrying amount, further testing of goodwill for impairment would not be required. Otherwise, goodwill impairment is tested using a two-step approach.

The first step involves comparing the fair value of the reporting unit to its carrying amount. If the fair value of the reporting unit is determined primarily using the anticipated cash flows discounted at a rate commensurate with the risk involved. Losses on long-lived assets to be greater than its carrying amount, there is no impairment. If the reporting unit’s carrying amount is determined to be greater than the fair value, the second step must be completed to measure the amountdisposed of impairment, if any. The second step involves calculating the implied fair value of goodwill by deducting the fair value of all tangible and intangible assets, excluding goodwill, of the reporting unit from the fair value of the reporting unit asare determined in step one. The implieda similar manner, except that fair value ofvalues are reduced for the goodwill in this step is comparedcost to the carrying value of goodwill. If the implied fair value of the goodwill is less than the carrying value of the goodwill, an impairment loss equivalent to the difference is recorded.dispose.

56

Income Taxes

Deferred income tax assets and liabilities are computed for differences between the financial statement and tax basis of assets and liabilities that will result in taxable or deductible amounts in the future based on enacted tax laws and rates applicable to the periods in which the differences are expected to affect taxable income. Valuation allowances are established when necessary to reduce deferred tax assets to the amount expected to be realized. Income tax expense is the tax payable or refundable for the period plus or minus the change during the period in deferred tax assets and liabilities.

-69-

In addition, our management performs an evaluation of all uncertain income tax positions taken or expected to be taken in the course of preparing our income tax returns to determine whether the income tax positions meet a “more likely than not” standard of being sustained under examination by the applicable taxing authorities. This evaluation is required to be performed for all open tax years, as defined by the various statutes of limitations, for federal and state purposes.

On December 22, 2017, the President of the United States signed and enacted into law H.R. 1 (the “Tax Reform Law”). The Tax Reform Law, effective for tax years beginning on or after January 1, 2018, except for certain provisions, resulted in significant changes to existing United States tax law, including various provisions that are expected to impact us. The Tax Reform Law reduces the federal corporate tax rate from 35% to 21% effective January 1, 2018. We have analyzed the provisions of the Tax Reform Law to assess the impact on our consolidated financial statements.

Impairment of LongLived Assets

We will periodically evaluate the carrying value of longlived assets to be held and used when events and circumstances warrant such a review and at least annually. The carrying value of a longlived asset is considered impaired when the anticipated undiscounted cash flow from such asset is separately identifiable and is less than its carrying value. In that event, a loss is recognized based on the amount by which the carrying value exceeds the fair value of the longlived asset. Fair value is determined primarily using the anticipated cash flows discounted at a rate commensurate with the risk involved. Losses on longlived assets to be disposed of are determined in a similar manner, except that fair values are reduced for the cost to dispose.

Recently Issued Accounting Standards

In January 2016, the Financial Accounting Standards Board (“FASB”) issued guidance on recognition and measurement of financial assets and financial liabilities (ASU No. 2016-01) that will supersede most current guidance. Changes to the U.S. GAAP model primarily affect the accounting for equity investments, financial liabilities under the fair value option and the presentation and disclosure requirements for financial instruments. In addition, the FASB clarified guidance related to the valuation allowance assessment when recognizing deferred tax assets resulting from unrealized losses on available-for-sale debt securities. The accounting for other financial instruments, such as loans, investments in debt securities, and financial liabilities, is largely unchanged. The classification and measurement guidance became effective as of December 1, 2018. We do not expect the implementation of this new pronouncement to have a material impact on our consolidated financial statements.

In May 2014, the FASB issued Accounting Standards Update (“ASU”) 2014-09 “RevenueASC 606 - Revenue from Contracts with Customers (Topic 606)” (“Topic 606”)

Our agreements are primarily service contracts that will supersede most currentrange in duration. We recognize revenue recognition guidance, including industry specific guidance. Under the new standard, a good or servicewhen control of these services is transferred to the customer when (or as) the customer obtains control of the good or service, which differs from the risk and rewards approach under current guidance. The guidance provides a five-step analysis of transactionsfor an amount, referred to determine when and how revenue is recognized. Other major provisions include capitalization of certain contract costs, consideration of the time value of money inas the transaction price, and allowing estimates of variablewhich reflects the consideration to which we are expected to be recognized before contingenciesentitled in exchange for those goods or services.

A contract with a customer exists only when:

the parties to the contract have approved it and are committed to perform their respective obligations;
we can identify each party’s rights regarding the distinct goods or services to be transferred (“performance obligations”);
we can determine the transaction price for the goods or services to be transferred; and
the contract has commercial substance and it is probable that we will collect the consideration to which it will be entitled in exchange for the goods or services that will be transferred to the customer.

For the majority of our contracts, we receive non-refundable upfront payments. We do not adjust the promised amount of consideration for the effects of a significant financing component since we expect, at contract inception, that the period between the time of transfer of the promised goods or services to the customer and the time the customer pays for these goods or services to be generally one year or less. Our credit terms to customers are resolved in certain circumstances. The guidance also requires enhanced disclosures regardingaverage between thirty and ninety days.

We do not disclose the nature, amount, timing and uncertaintyvalue of revenue and cash flows arising from an entity’sunsatisfied performance obligations for contracts with customers. original expected duration of one year or less.

Disaggregation of Revenue

The guidance is effective in annual reporting periods beginning after December 15, 2017, including interim reporting periods within that reporting period.  The Company will implement the guidance forfollowing table disaggregates our annual period ending on December 31, 2019 and interim periods within such annual periods, using the modified retrospective method and will adjust the accumulated deficit and deferredrevenues by major revenue asstreams:

  Year Ended December 31, 
Revenue stream: 2020  2019 
  (in thousands) 
POC and hospital services $6,068  $3,109 
Cell process development services  1,584   790 
Total $7,652  $3,899 

Nature of the adoption date. Revenue Streams

Under current GAAP, the Company recognizesWe have two main revenue for services linked to cell process development based on both the input and output methods of measurement. The Company has evaluated the application of the requirements of ASC 606 to ‘recognize revenue when or as the entity satisfies a performance obligation to its business. The Company has several types of revenue contracts:

-70-

a)Cell process development services

The Company has concluded that under the revised standard, contracts forstreams being cell process development services areand POC development services which includes and hospital supplies.

57

POC Development Services

Revenue recognized under contracts for POC development services may, in some cases a single performance obligation (where promises offered to customers are not distinct within the context of the contract), and in other cases havecontracts, represent multiple performance obligations (where promises to the customers are distinct) in circumstances in which the work packages are not interrelated or the customer is able to complete the services performed.

For arrangements that include multiple performance obligations, the transaction price is allocated to the identified performance obligations based on their relative standalone selling prices.

We recognize revenue when, or as, it satisfies a performance obligation. At contract inception, we determine whether the services are transferred over time or at a point in time. Performance obligations that have no alternative use and that we have the right to payment for performance completed to date, at all times during the contract term, are recognized over time. All other Performance obligations are recognized as revenues by the company at point of time (upon completion).

Included in POC development services is hospital supplies revenue which is derived principally from the sale or lease of products and the performance of services to hospitals or other medical providers. Revenue is earned and recognized when product and services are received by the customer.

Significant Judgement and Estimates

Significant judgment is required to identifying the distinct performance obligations and estimating the standalone selling price of each distinct performance obligation, and identifying which performance obligations create assets with alternative use to us, which results in revenue recognized upon completion, and which performance obligations are transferred to the customer over time.

Cell Process Development Services (mainly discontinued operations)

Revenue recognized under contracts for cell process development services may, in some contracts, represent multiple performance obligations (where promises to the customers are distinct) in circumstances in which the work packages and milestones are not interrelated or the customer is able to complete the services performed independently or by using our competitors. In all casesother contracts when the above circumstances are not met, the promises are not considered distinct and the contract represents one performance obligation. All performance obligations are satisfied over time. Undertime, as there is no alternative use to the new standard,services it performs, since, in nature, those services are unique to the Company will recognizecustomer, which retain the ownership of the intellectual property created through the process. Additionally, due to the non-refundable upfront payment the customer pays, together with the payment term and cancellation fine, it has a right to payment (which include a reasonable margin), at all times, for work completed to date, which is enforceable by law.

For arrangements that include multiple performance obligations, the transaction price is allocated to the identified performance obligations based on their relative standalone selling prices. For these contracts, the standalone selling prices are based on our normal pricing practices when sold separately with consideration of market conditions and other factors, including customer demographics and geographic location.

We measure the revenue to be recognized over time using on a contract by contract basis, determining the use of either a cost-based input method or output method, depending on whichever fairlybest depicts the transfer of control over the life of the performance obligation.

Tech Transfer Services (discontinued operations)

Revenue recognized under contracts for tech transfer services are considered a single performance obligation, as appropriate.all work packages (including data collection, GMP documentation, validation runs) and milestones are interrelated. Additionally, the customer is unable to complete services of work performed independently or by using our competitors. Revenue is recognized over time using a cost-based input method where progress on the performance obligation is measured by the proportion of actual costs incurred to the total costs expected to complete the contract.

b)Cell manufacturing services

Regarding revenuesCell Manufacturing Services (discontinued operations)

Revenues from cell manufacturing services the Company concluded that these comprised representa single performance obligation.obligation which is recognized over time. The progress towards completion will continue to be measured on an output measure based on direct measurement of the value transferred to the customer (units produced).

c)58Tech transfer

The Company has concluded that underReimbursed Expenses

We include reimbursed expenses in revenues and costs of revenue as we are primarily responsible for fulfilling the revised standard, contracts for Tech Transferpromise to provide the specified service, including the integration of the related services are consideredinto a single performance obligation and will be measured over time using a cost based input method where progress on the performance obligation is measured by the proportion of actual costs incurredcombined output to the totalcustomer, which are inseparable from the integrated service. These costs expected to complete the contract.include such items as consumable, reagents, transportation and travel expenses, over which we have discretion in establishing prices.

The cost-based and output methods of revenue recognition require the Company to make estimates of costs to complete its projects and the percentage of completeness on an ongoing basis. Significant judgment is required to evaluate assumptions related to these estimates. The effect of revisions to estimates related to the transaction price (including variable consideration relating to reimbursement on a cost-plus basis on certain expenses) or costs to complete a project are recordedChange Orders

Changes in the period in which the estimate is revised. The adoptionscope of the new standard is not expected towork are common and can result in a materialchange in transaction price, equipment used and payment terms. Change orders are evaluated on a contract-by-contract basis to determine if they should be accounted for as a new contract or as part of the existing contract. Generally, services from change orders are not distinct from the original performance obligation. As a result, the effect that the contract modification has on the total stockholders’ equitycontract revenue, and measure of progress, is recognized as of December 1, 2018.

In November 2016, the FASB issued ASU 2016-18, Statement of Cash Flows (Topic 230): Restricted Cash (a Consensus of the FASB Emerging Issues Task Force) (“ASU 2016-18”), which requires entities to include amounts generally described as restricted cash and restricted cash equivalents in cash and cash equivalents when reconciling beginning-of-period and end-of-period total amounts shown on the statement of cash flows. ASU 2016-18 is effective for annual reporting periods (including interim periods within those annual reporting periods) beginning after December 15, 2017. The Company adopted this standard during the year ended November 30, 2018.

In February 2016, the FASB issued ASU 2016-02, “Leases (Topic 842)” (“ASU 2016-02”), which supersedes the existing guidance for lease accounting, “Leases (Topic 840)”. ASU 2016-02 requires lessees to recognize leases on their balance sheets, and leaves lessor accounting largely unchanged. The amendments in ASU 2016-02 are effective for fiscal years beginning after December 15, 2018 and interim periods within those fiscal years. Early application is permitted for all entities. ASU 2016-02 requires a modified retrospective approach for all leases existing at, or entered into after, the date of initial application, with an option to elect to use certain transition relief. We expect to apply the ASU without adjusting the comparative periods and, if applicable, recognizing a cumulative effect adjustment to revenue when they occur.

Costs of Revenue (Discontinued Operations)

Costs of revenue include (i) compensation and benefits for billable employees and personnel involved in production, data management and delivery, and the opening balancecosts of retained earnings inacquiring and processing data for our information offerings; (ii) costs of staff directly involved with delivering services offerings and engagements; (iii) consumables used for the periodservices; and (iv) other expenses directly related to service contracts such as courier fees, laboratory supplies, professional services and travel expenses.

Contract Assets and Liabilities

Contract assets are mainly comprised of adoption. We are currently evaluating the impacttrade receivables net of this new standard on our consolidated financial statements.

In June 2016, the FASB issued ASU 2016-13, “Financial Instruments-Credit Losses (Topic 326)” (“ASU 2016-13”). ASU 2016-13 requires that financial assets measured at amortized cost be presented at the net amount expected to be collected. The allowance for credit lossesdoubtful debts, which includes amounts billed and currently due from customers.

The activity for trade receivables is a valuation account thatcomprised of:

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Balance as of beginning of period $1,831  $129 
Acquisition of Koligo  228   - 
Additions  6,997   2,079 
Collections  (5,982)  (364)
Exchange rate differences  11   (13)
Balance as of end of period $3,085  $1,831 

The activity for contract liabilities is deducted from the amortized cost basis. The income statement reflects the measurement of credit losses for newly recognized financial assets, as well as the expected credit losses during the period. The measurement of expected credit losses is based upon historical experience, current conditions, and reasonable and supportable forecasts that affect the collectability of the reported amount. Credit losses relating to available-for-sale debt securities will be recorded through an allowance for credit losses rather than as a direct write-down to the security. ASU 2016-13 is effective for fiscal years beginning after December 15, 2019, including interim periods within those fiscal years. Early adoption is permitted as of the fiscal years beginning after December 15, 2018, including interim periods within those fiscal years. We do not expect the implementation of this new pronouncement to have a material impact on our consolidated financial statements.comprised of:

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Balance as of beginning of period $325  $56 
Additions  597   1,126 
Realizations  (862)  (854)
Exchange rate differences  (1)  (3)
Balance as of end of period $59  $325 

-71-
59

In May 2017, the FASB issued ASU 2017-09, “Compensation - Stock Compensation (Topic 718): Scope of Modification Accounting” (“ASU 2017-09”), which gives direction on which changes to the terms or conditions of share-based payment awards require an entity to apply modification accounting in Accounting Standard Codification (“ASC”) Topic 718. In general, entities will apply the modification accounting guidance if the value, vesting conditions or classification of the award changes. ASU 2017-09 is effective for fiscal years beginning after December 15, 2017, including interim periods within those fiscal years. Early adoption is permitted, including adoption in an interim period. We do not expect the implementation of this new pronouncement to have a material impact on our consolidated financial statements.See note 2(z).

In June 2018, the FASB issued ASU 2018-07, “Compensation – Stock Compensation (Topic 718): Improvements to Nonemployee Share-Based Payment Accounting” that expands the scope of ASC Topic 718 to include share-based payment transactions for acquiring goods and services from nonemployees. An entity should apply the requirements of ASC Topic 718 to nonemployee awards except for certain exemptions specified in the amendment. The guidance is effective for fiscal years beginning after December 15, 2018, including interim reporting periods within that fiscal year. Early adoption is permitted, but no earlier than an entity’s adoption date of Topic 606. We do not expect the implementation of this new pronouncement to have a material impact on our consolidated financial statements.See note (x).

In January 2017, FASB issued Accounting Standards Update (ASU) 2017-04, Intangibles—Goodwill and Other (Topic 350): Simplifying the Test for Goodwill Impairment, which eliminated the calculation of implied goodwill fair value. Instead, companies will record an impairment charge based on the excess of a reporting unit’s carrying amount of goodwill over its fair value. This guidance simplifies the accounting as compared to prior GAAP. The guidance is effective for fiscal years beginning after December 15, 2019. The Company does not expect the implementation of this new pronouncement to have a material impact on its consolidated financial statements.See note 2(y).

60

Off-Balance Sheet Arrangements

We have no off-balance sheet arrangements that have or are reasonably likely to have a current or future effect on our financial condition, changes in financial condition, revenues or expenses, results of operations, liquidity, capital expenditures or capital resources that is material to stockholders.

ITEM 7A. QUANTITATIVE AND QUALITATIVE DISCLOSURES ABOUT MARKET RISK

Not applicable.

ITEM 8. FINANCIAL STATEMENTS AND SUPPLEMENTARY DATA

The information called for by Item 8 is included following the “Index to Financial Statements” on page F-1 contained in this Annual Report on Form 10-K.

ITEM 9. CHANGES IN AND DISAGREEMENTS WITH ACCOUNTANTS ON ACCOUNTING AND FINANCIAL DISCLOSURE

None.

ITEM 9A. CONTROLS AND PROCEDURES

Evaluation of Disclosure Controls and Procedures

Our management, with the participation of our Chief Executive Officer and Chief Financial Officer, has evaluated the effectiveness of our disclosure controls and procedures (as such term is defined in Rules 13a-15(e) and 15d-15(e) under the Exchange Act and regulations promulgated thereunder) as of November 30, 2018,December 31, 2020, or the Evaluation Date. Based on such evaluation, those officersour Chief Executive Officer and Chief Financial Officer have concluded that, as of the Evaluation Date, our disclosure controls and procedures are effective.

 

Management’s Report on Internal Control over Financial Reporting

Our management, under the supervision of theChief Executive Officer and Chief Financial Officer, is responsible for establishing and maintaining adequate internal control over financial reporting for our company. Internal control over financial reporting is defined in Rule 13a-15(f) or 15d-15(f) promulgated under the Exchange Act as a process designed by, or under the supervision of, the company’s principal executive and principal financial officers and effected by the company’s board of directors, management and other personnel, to provide reasonable assurance regarding the reliability of financial reporting and the preparation of financial statements for external purposes in accordance with GAAP and includes those policies and procedures that: (i) pertain to the maintenance of records that, in reasonable detail, accurately and fairly reflect the transactions and dispositions of the assets of the company; (ii) provide reasonable assurance that transactions are recorded as necessary to permit preparation of financial statements in accordance with generally accepted accounting principles, and that receipts and expenditures of our company are being made only in accordance with authorizations of management and directors of the company; and (iii) provide reasonable assurance regarding prevention or timely detection of unauthorized acquisition, use, or disposition of our company’s assets that could have a material effect on the financial statements.

-72-

The CureCell and Atvio acquisitionsKoligo acquisition which werewas completed in the second half of 2018 wereOctober 2020 was excluded from management’s evaluation of internal control over financial reporting as of November 30, 2018. Curecell and Atvio, collectively, represent 5%December 31, 2020 because the business was acquired in a transaction accounted for as a business combination during 2020. Koligo, represents approximately 2% of our total consolidated assets and 6%approximately 3% of our total consolidated revenues as of and for the year ended November 30, 2018.December 31, 2020.

61

Our management, with the participation of ourChief Executive Officer and Chief Financial Officer, evaluated the effectiveness of our internal control over financial reporting as of November 30, 2018.December 31, 2020. In making this evaluation, our management used the criteria set forth in the Internal Control — Integrated Framework (2013) issued by the Committee of Sponsoring Organizations of the Treadway Commission.

Based on this evaluation, management concluded that our internal control over financial reporting was effective as of November 30, 2018December 31, 2020 based on those criteria.

Attestation Report of Independent Registered Public Accounting Firm

The effectivenessThis annual report does not include an attestation report of our registered public accounting firm on internal control over financial reporting as of November 30, 2018 has been audited by Kesselman & Kesselman,because we are is a member firm of PricewaterhouseCoopers International Limited (“PwC”), an independent registered public accounting firm, as stated in their report which is included under "Item 8- Financial Statements".smaller reporting company and non-accelerated filer.

Changes in Internal Control Over Financial Reporting

Prior to listing our common stock on the Nasdaq Capital Market, we identified a material weaknessThere were no changes in our internal control over financial reporting as of November 30, 2017. As defined in Regulation 12b-2 underthat occurred during the Securities Exchange Act of 1934, a “material weakness” is a deficiency,quarter ended December 31, 2020 that have materially affected, or combination of deficiencies, in internal control over financial reporting, such that there is a reasonable possibility that a material misstatement of our company’s annual or interim financial statements will not be prevented or detected on a timely basis. The deficiency inare reasonably likely to materially affect, our internal control over financial reporting was due to the applied risk-based approach which is indicative of many small companies with limited number of staff in corporate functions which have insufficient segregation of duties and insufficient controls over period end financial disclosure and reporting processes. Subsequently, the following had occurred in order to remediate the material weakness: (a) during the fiscal year ended November 30, 2018, we hired additional qualified personnel to the corporate finance team, as well as to our subsidiaries; (b) in the beginning of the third quarter of 2018, we engaged an internal control Sarbanes and Oxley (SOX) expert to assist us in improving our internal processes and in reviewing the design and implementation of our internal control over financial reporting; (c) during the year ended November 30, 2018, our procedures of internal control over financial reporting and made changes to our processes to improve controls and increase efficiency, by implementing new, more efficient consolidating activities, and migrating processes. Management believes that due to the foregoing, as of November 30, 2018, it has remediated the material weakness previously identified.reporting.

ITEM 9B. OTHER INFORMATION

None.

-73-

PART III

ITEM 10. DIRECTORS, EXECUTIVE OFFICERS AND CORPORATE GOVERNANCE

The following table sets forth certain information regarding our each of our current Directors and Executive Officers. The age of each Director and Executive Officer listed below is givenOfficers as of November 30, 2018.March 9, 2021.

NameAgePosition
Vered Caplan5052Chief Executive Officer and Chairperson of the Board of Directors
Neil Reithinger4851Chief Financial Officer, Secretary and Treasurer
Sarah FerberDavid Sidransky (1) (2) (4)6460Chief Scientific OfficerDirector
Denis BedoretGuy Yachin (1) (2) (3) (4)3853Managing Director of MaSTherCell S.A
David SidranskyYaron Adler (1)(2) (3)5850Director
Guy YachinAshish Nanda (1)(3)5155Director
Yaron AdlerMario Philips (1)4851Director
Ashish Nanda(1)53Director

(1)A member on each of the audit committee.
(2)A member on the compensation andcommittee.
(3)A member on the nominating and corporate governance committees.committee.
(4)A member of the research and development committee.

Our Executive Officers

Vered Caplan – Chief Executive Officer and Chairperson of the Board of Directors

Ms. Caplan has served as our CEO and Chairperson of the Board of Directors since August 14, 2014, prior to which she served as Interim President and CEO commencing on December 23, 2013. She joined our Board of Directors in February 2012. She has 2526 years of industry experience, previously holding positions asCEO of Kamedis Ltd.from 2009 to 2014, CEO of GammaCan International Inc. from 2004 to 2007,2007. She also served as a director of the following companies: Opticul Ltd., Inmotion Ltd., Nehora Photonics Ltd., Ocure Ltd., Eve Medical Ltd., Menopause and PMS and Biotech Investment Corp. Ms. Caplan holds a M.Sc. in biomedical engineering from Tel Aviv University specializing in signal processing; management for engineers from Tel Aviv University specializing in business development; and a B.Sc. in mechanical engineering from the Technion– Israel Institute of Technology specialized in software and cad systems.

62

Neil Reithinger – Chief Financial Officer, Secretary and Treasurer

Mr. Reithinger was appointed Chief Financial Officer, Secretary and Treasurer on August 1, 2014. Mr. Reithinger is the Founder and President of Eventus Advisory Group, LLC, a private, CFO-services firm incorporated in Arizona, which specializes in capital advisory and SEC compliance for publicly-traded and emerging growth companies. He is also the President of Eventus Consulting, P.C., a registered CPA firm in Arizona. Prior to forming Eventus, Mr. Reithinger was Chief Operating Officer & CFO from March 2009 to December 2009 of New Leaf Brands, Inc., a branded beverage company, CEO of Nutritional Specialties, Inc. from April 2007 to October 2009, a nationally distributed nutritional supplement company that was acquired by Nutraceutical International, Inc., Chairman, CEO, President and director of Baywood International, Inc. from January 1998 to March 2009, a publicly-traded nutraceutical company and Controller of Baywood International, Inc. from December 1994 to January 1998. Mr. Reithinger earned a B.S. in Accounting from the University of Arizona and is a Certified Public Accountant. He is a Member of the American Institute of Certified Public Accountants and the Arizona Society of Certified Public Accountants.

Prof. Sarah Ferber – Chief Scientific Officer

Prof. Ferber has served as the Company’s Chief Scientific Officer since her appointment on February 2, 2012. Since 2017, Prof. Ferber has been the Principal Investigator of cell therapy for TMU DiaCure. Prof. Ferber studied biochemistry at the Technion under the supervision of Professor Avram Hershko and Professor Aharon Ciechanover, winners of the Nobel Prize in Chemistry in 2004. Most of the research was conducted in Prof. Ferber’s Endocrine Research Lab. Prof. Ferber received Teva, Lindner, Rubin and Wolfson awards for this research. Prof. Ferber’s research work has been funded over the past 15 years by the JDRF, the Israel Academy of Science foundation (ISF), BIODISC and DCure. Prof. Ferber earned her B.Sc. from Technion-Haifa, a M.Sc. in Biochemistry from Technion-Haifa and a Ph.D. in Medical Sciences from Technion-Haifa. She also holds a Post Doctorate degree in Molecular Biology from Harvard Medical School and a degree in Cell Therapy Sciences from UTSW, Dallas.

-74-

Dr. Denis Bedoret – General Manager of MaSTherCell, S.A.

Dr. Bedoret has served as the General Manager of MaSTherCell since his appointment on July 6, 2017. Dr. Bedoret joined MaSTherCell in October 2016 as Chief Business and Administration Officer. Prior to joining MaSTherCell, from January 2014 to September 2016, he held the position of Chief Operations Officer at Quality Assistance, a leading European analytical CRO where he was also member of the board of directors. Between September 2011 and January 2014, Dr. Bedoret served as Engagement Manager at McKinsey & Company, focusing on bio-pharmaceutical projects. Through those experiences, he gained a strong expertise in biologicals, FDA and EMA regulations, as well as team management. He holds a degree in Veterinary Medicine, a Ph.D. in Life Sciences from ULg and a post-doctorate degree in Immunology from Harvard Medical School.

On September 5, 2018, Dr. Bedoret was promoted to Managing Director of MaSTherCell. On January 22, 2019, Dr. Bedoret was appointed as President of Masthercell Global.

Our Directors

Dr. David Sidransky – Director

Dr. Sidransky has served as a director since his appointment on July 18, 2013. Dr. Sidransky is a renowned oncologist and research scientist named and profiled by TIME magazine in 2001 as one of the top physicians and scientists in America, recognized for his work with early detection of cancer. Since 1994, Dr. Sidransky has been the Director of the Head and Neck Cancer Research Division at Johns Hopkins University School of Medicine’s Department of Otolaryngology and Professor of Oncology, Cellular & Molecular Medicine, Urology, Genetics, and Pathology at the John Hopkins University School of Medicine. Dr. Sidransky is one of the most highly cited researchers in clinical and medical journals in the world in the field of oncology during the past decade, with over 460560 peer reviewed publications. Dr. Sidransky is a founder of a number of biotechnology companies and holds numerous biotechnology patents. Dr. Sidransky has served as Vice Chairman of the board of directors, and was, until the merger with Eli Lilly, a director of ImClone Systems, Inc., a global biopharmaceutical company committed to advancing oncology care. He is serving, or has served on, the scientific advisory boards of MedImmune, LLC, Roche, Amgen Inc. and Veridex, LLC (a Johnson & Johnson diagnostic company), among others and is currently on the board of Directors of Galmed and Rosetta Genomics Ltd.Champions Oncology. and chairs the board of directors of Advaxis and Champions Oncology, Inc.Ayala . Dr. Sidransky served as Director from 2005 until 2008 of the American Association for Cancer Research (AACR). He was the chairperson of AACR International Conferences during the years 2006 and 2007 on Molecular Diagnostics in Cancer Therapeutic Development: Maximizing Opportunities for Personalized Treatment. Dr. Sidransky is the recipient of a number of awards and honors, including the 1997 Sarstedt International Prize from the German Society of Clinical Chemistry, the 1998 Alton Ochsner Award Relating Smoking and Health by the American College of Chest Physicians, and the 2004 Richard and Hinda Rosenthal Award from the American Association of Cancer Research. Dr. Sidransky received his BS in Chemistry from Brandies University and his medical degree from Baylor College of medicine where he also completed his residency in internal medicine. His specialty in Medical Oncology was completed at Johns Hopkins University and Hospital.

We believe Dr. Sidransky is qualified to serve on our Board of Directors because of his education, medical background, experience within the life science industry and his business acumen in the public markets.

Guy Yachin – Director

Mr. Yachin has served as a director since his appointment on April 2, 2012. Mr. Yachin has served as the President and CEO of Serpin Pharma, a clinical stage Virginia-based company focused on the development of anti-inflammatory drugs, since April 2013. Mr. Yachin is the CEO of Oasis Management, a Maryland-based consulting company, since 2010. Mr. Yachin is the CEO of NasVax Ltd., a company focused on the development of improved immunotherapeutics and vaccines. Prior to joining NasVax, Mr. Yachin served as CEO of MultiGene Vascular Systems Ltd., a cell therapy company focused on blood vessels disorders, leading the company through clinical studies in the U.S. and Israel, financial rounds, and a keystone strategic agreement with Teva Pharmaceuticals Industries Ltd. He was CEO and founder of Chiasma Inc., a biotechnology company focused on the oral delivery of macromolecule drugs, where he built the company’s presence in Israel and the U.S., concluded numerous financial rounds, and guided the company’s strategy and operation for over six years. Earlier, he was CEO of Naiot Technological Center Ltd., and provided seed funding and guidance to more than a dozen biomedical startups such as Remon Medical Technologies Ltd., Enzymotec Ltd. and NanoPass Technologies Ltd. He holds a BSc. in Industrial Engineering and Management and an MBA from the Technion – Israel Institute of Technology. Mr. Yachin served on the board of Peak Pharmaceuticals, Inc. from March 2014 to April 2016.

-75-
63

We believe Mr. Yachin is qualified to serve on our Board of Directors because of his education, experience within the life science industry and his business acumen in the public markets.

Yaron Adler – Director

Mr. Adler has served as a director since his appointment on April 17, 2012. Mr. Adler is the chairman of ExitValley Ltd., an equity-based crowdfunding platform, since April 2014 and the co-founder of a startup incubator, We Group Ltd. In 1999, Mr. Adler co-founded IncrediMail Ltd. and served as its CEO until 2008 and President until 2009. In 1999, prior to founding IncrediMail, Mr. Adler consulted Israeli startup companies regarding Internet products, services and technologies. Mr. Adler served as a product manager from 1997 to 1999, and as a software engineer from 1994 to 1997, at Tecnomatix Technologies Ltd., a software company that develops and markets production engineering solutions to complex automated manufacturing lines that fill the gap between product design and production, and which was acquired by UGS Corp. in April 2005. In 1993, Mr. Adler held a software engineer position at Intel Israel Ltd. He has a B.A. in computer sciences and economics from Tel Aviv University.

We believe Mr. Adler is qualified to serve on our Board of Directors because of his education, success with early-stage enterprises and his business acumen in the public markets.

Ashish Nanda – Director

Mr. Nanda has served as a director since his appointment on February 22, 2017. Since 1998, Mr. Nanda has been the Managing Director of Innovations Group, one of the largest outsourcing companies in the financial sector that employs close to 14,000 people working across various financial sectors. Since 1992, Mr. Nanda has served as the Managing Partner of Capstone Insurance Brokers LLC and, since 2009, has served as Managing Partner of Dive Tech Marine Engineering Services L.L.C. From 1991 to 1994, Mr. Nanda held the position of Asst. Manager Corporate Banking at Emirates Banking Group where he was involved in establishing relationships with business houses owned by UAE nationals and expatriates in order to set up banking limits and also where he managed portfolios of USD $26 billion. Mr. Nanda holds a Chartered Accountancy from the Institute of Chartered Accountants from India.

We believe that Mr. Nanda is qualified to serve on our Board of Directors because of his business experience and strategic understanding of advancing the valuation of companies in emerging industries.

There are no family relationships between any of the above executive officers or directors or any other person nominated or chosen to become an executive officer or a director. Pursuant to an agreement entered into between us and Image Securities fzc. (“Image”), forso long as Image’s ownership of our company is 10% or greater, itwas granted the right to nominate a director to our Board of Directors. Mr. Nanda was nominated for a directorship at the 2017 annual meeting in compliance with our contractual undertakings.

Mario Philips – Director

Mr. Philips has served as a director since his appointment on January 9, 2020. Since November 2020, Mr. Philips has been Chief Executive Officer of Polyplus, a leading Biotech supplier of transfection reagents for cell & gene therapy as well as the research life sciences market. Mario is also chairmen of the Board of PLL Therapeutics, a drug company based in France that has developed a diagnostic platform technology for neurodegenerative diseases in combination with a therapy to cure neurodegenerative diseases such as ALS and Parkinson’s.

Prior to that Mario acted as VP/GM for Danaher Pall Biotech business with full P&L responsibility for a $1,3B business unit. Mario joined Pall in February 2014, as part of the Pall acquisition of ATMI Life Sciences, and was appointed to Vice President and General Manager to lead the Single-Use Technologies BU. In this role he was responsible for leading and executing an aggressive investment and growth strategy.

64

Mario joined ATMI in 1999 with ATMI’s acquisition of MST Analytics, Inc., serving as European Sales Manager for ATMI Analytical Systems. In 2004, Mario was appointed to General Manager of ATMI Packaging, a role he held through 2010 when he was promoted to the position of Senior Vice President and General Manager, ATMI Life Sciences. In that role, he was responsible for developing and executing all business strategies, including the introduction of new products and service solutions for the Life Sciences industry. A strong leading innovative IP portfolio was created, Pall acquired the business in 2014.

Mario also held in the past several board member positions in the life sciences industry with Clean Biologics, Austar Life Sciences (China), Disposable Lab (France) and Artelis (Belgium).

We believe that Mr. Philips is qualified to serve on our Board of Directors because of his business experience and strategic understanding of advancing the valuation of companies in emerging industries.

Board of Directors

Our Board of Directors currently consists of fivesix (6) members. All directors hold office until the next annual meeting of stockholders. At each annual meeting of stockholders, the successors to directors whose terms then expire are elected to serve from the time of election and qualification until the next annual meeting following election.

Management has been delegated the responsibility for meeting defined corporate objectives, implementing approved strategic and operating plans, carrying on our business in the ordinary course, managing cash flow, evaluating new business opportunities, recruiting staff and complying with applicable regulatory requirements. The Board of Directors exercises its supervision over management by reviewing and approving long-term strategic, business and capital plans, material contracts and business transactions, and all debt and equity financing transactions and stock issuances.

-76-

Director Independence

Our Board of Directors is comprised of a majority of independent directors. In determining director independence, the Company uses the definition of independence in Rule 5605(a)(2) of the listing standards of The Nasdaq Stock Market.

The Board has concluded that each of Dr. Sidransky, and Messrs. Yachin, Adler, Philips and Nanda is “independent” based on the listing standards of the Nasdaq Stock Market, having concluded that any relationship between such director and our company, in its opinion, does not interfere with the exercise of independent judgment in carrying out the responsibilities of a director.

Board Committees

Our Board of Directors has established an Audit Committee, a Compensation Committee and a Nominating and Corporate Governance Committee, with each comprised of independent directors in accordance with the rules of The Nasdaq Stock Market and applicable federal securities laws and regulations. The members of each committeethe Audit Committee are Dr. Sidransky and Messrs. Yachin and Philips. The members of the Compensation Committee are Dr. Sidransky and Messrs. Adler and Yachin. The members of the Nominating and Corporate Governance Committee are Messrs. Nanda, Adler and Yachin. The members of the Research and Development Committee are Mr. Yachin and Dr. Sidransky.

Each committee operates under a written charter that has been approved by our Board of Directors. Copies of our committee charters are available on the investor relations section of our website, which is located athttp://www.orgenesis.com.www.orgenesis.com.

65

Audit Committee

The Audit Committee (a) assists the Board of Directors in fulfilling its oversight of: (i) the quality and integrity of our financial statements; (ii) our compliance with legal and regulatory requirements relating to our financial statements and related disclosures; (iii) the qualifications and independence of our independent auditors; and (iv) the performance of our independent auditors; and (b) prepares any reports that the rules of the SEC require be included in our proxy statement for our annual meeting.

The Audit Committee held nine9 meetings in fiscal 2018.2020. In addition, the Audit Committee reviewed and approved various corporate items by way of written consent during the fiscal year 2018.2020. The Board has determined that each member of the Audit Committee is an independent director in accordance with the rules of The Nasdaq Stock Market and applicable federal securities laws and regulations. In addition, the Board has determined that Dr. Sidransky is an “audit committee financial expert” within the meaning of Item 407(d)(5) of Regulation S-K and has designated him to fill that role. See “Directors, Executive Officers and Corporate Governance – Directors” above for descriptions of the relevant education and experience of each member of the Audit Committee.

At no time since the commencement of the Company’sour most recently completed fiscal year was a recommendation of the Audit Committee to nominate or compensate an external auditor not adopted by the Board of Directors.

The Audit Committee is responsible for the oversight of our financial reporting process on behalf of the Board of Directors and such other matters as specified in the Audit Committee’s charter or as directed by the Board of Directors. Our Audit Committee is directly responsible for the appointment, compensation, retention and oversight of the work of any registered public accounting firm engaged by us for the purpose of preparing or issuing an audit report or performing other audit, review or attest services for us (or to nominate the independent registered public accounting firm for stockholder approval), and each such registered public accounting firm must report directly to the Audit Committee. Our Audit Committee must approve in advance all audit, review and attest services and all non-audit services (including, in each case, the engagement and terms thereof) to be performed by our independent auditors, in accordance with applicable laws, rules and regulations.

-77-

Compensation Committee

The Compensation Committee (i) assists the Board of Directors in discharging its responsibilities with respect to compensation of our executive officers and directors, (ii) evaluates the performance of our executive officers, and (iii) administers our stock and incentive compensation plans and recommends changes in such plans to the Board as needed.

The Compensation Committee acted by unanimous written consent or held one meeting5 meetings in fiscal 2018.2020. In addition, the Compensation Committee reviewed and approved various corporate items by way of written consent during the fiscal year 2018.2019. The Board of Directors has determined that each member of the Compensation Committee is an independent director in accordance with the rules of The Nasdaq Stock Market and applicable federal securities laws and regulations.

Nominating and Corporate Governance Committee

The Nominating and Corporate Governance Committee assists the Board in (i) identifying qualified individuals to become directors, (ii) determining the composition of the Board and its committees, (iii) developing succession plans for executive officers, (iv) monitoring a process to assess Board effectiveness, and (v) developing and implementing our corporate governance procedures and policies.

The Nominating and Corporate Governance Committee acted by unanimous written consent or held one4 meeting in fiscal 2018.2020. The Board has determined that each member of the Nominating and Corporate Governance Committee is an independent director in accordance with the rules of The Nasdaq Stock Market and applicable federal securities laws and regulations.

Research and Development Committee

The Research and Development Committee assists the Board in fulfilling the Board’s responsibilities to oversee the Company’s research and development programs, and strategies.

The Research and Development Committee was established in January 2021. The Board has determined that each member of the Research and Development Committee is an independent director in accordance with the rules of The Nasdaq Stock Market and applicable federal securities laws and regulations.

66

DELINQUENT SECTION 16(a) BENEFICIAL OWNERSHIP REPORTING COMPLIANCEREPORTS

Section 16(a) of the Securities Exchange Act of 1934, as amended (the “Exchange Act”), requires officers and directors of the Company and persons who beneficially own more than ten percent (10%) of the Common Stock outstanding to file initial statements of beneficial ownership of Common Stock (Form 3) and statements of changes in beneficial ownership of Common Stock (Forms 4 or 5) with the SEC. Officers, directors and greater than 10% stockholders are required by SEC regulation to furnish us with copies of all such forms they file.

Our records reflect that all reports which were required to be filed pursuant to Section 16(a) of the Securities Exchange Act of 1934, as amended, were filed on a timely basis, except that two reports, covering an aggregate of twofive transactions, were filed late by Vered Caplan, two reports, covering an aggregate of two transaction, were filed late by David Sidransky, one report, covering an aggregate of one transaction, was filed late by Guy Yachin, one report, covering an aggregate of one transaction, was filed late by Yaron Adler, one report, covering an aggregate of one transaction, was filed late by Sarah Ferber, one report, covering an aggregate of eight transactions, was filed late by Hugues Bultot, an initial report of ownership was filed late by David SidranskyGuy Yachin, and each of Ashish Nanda and Denis Bedoret did not timely file one report, covering an aggregate of one transaction.transaction, was filed late by Ashish Nanda.

Corporate Code of Conduct and Ethics

Our Board of Directors has adopted a written code of business conduct and ethics that applies to our directors, officers and employees, including our principal executive officer, principal financial officer, principal accounting officer or controller, or persons performing similar functions. Copies of our corporate code of conduct and ethics are available, without charge, upon request in writing to Orgenesis Inc., 20271 Goldenrod Lane, Germantown, MD, 20876, Attn: Secretary and are posted on the investor relations section of our website, which is located at www.orgenesis.com. The inclusion of our website address in this Annual Report on Form 10-K does not include or incorporate by reference the information on our website into this Annual Report on Form 10-K. We also intend to disclose any amendments to the Corporate Code of Conduct and Ethics, or any waivers of its requirements, on our website.

-78-

ITEM 11. EXECUTIVE COMPENSATION

The following table shows the total compensation paid or accrued during the last two fiscal yearyears ended November 30, 2018,December 31, 2020 to our Chief Executive Officer ourand Chief Financial Officer and our two next most highly compensatedOfficer. As of December 31, 2020, there were no other executive officers who earned more than $100,000 during the fiscal year ended November 30, 2018December 31, 2020 and were serving as executive officers as of such date (the “named executive officers”).

67

Summary Compensation Table

Name and

Principal

Position

 Year  

Salary

($)

  

Bonus

($)

  

Stock

Awards

($)

  

Option

Awards

($) (1)

  

Non-Equity

Incentive

Plan

Compensa-

tion

($)

  

Non-qualified

Deferred

Compensation

Earnings

($)

  

All Other

Compensa-

tion

($) (2)

  Total
($)
 
Vered Caplan
  2020   250,000   400,000         -   163,239            -            -   215,640   1,028,879 
CEO(3)  2019   250,000   200,000   -   871,036   -   -   77,020   1,398,056 
                                     
Neil Reithinger CFO, Treasurer &  2020   255,231   200,000   -   57,331   -   -   -   512,562 
Secretary  2019   213,653   -   -   22,970   -   -   -   236,623 

Name and

Principal

Position

Year

Salary

($)

Bonus

($)

Stock

Awards

($)

Option

Awards

($)(1)

Non-Equity

Incentive

Plan

Compensa-

tion

($)

Non qualified

Deferred

Compensation

Earnings

($)

All Other

Compensation

($)(2)

Total($)

Vered Caplan

CEO

2018

2017

226,122(3)

156,232(3)

350,000

150,000

-

-

1,318,771

685,318

-

-

-

-

80,697

63,262

1,975,590

1,054,812

Neil Reithinger

CFO, Treasurer & Secretary

2018

2017

266,452(4)

112,652(4)

-

-

-

-

139,590

136,148

-

-

-

-

-

-

406,042

248,800

Sarah Ferber

Chief Scientific Officer

2018

2017

225,523(5)

128,907(5)

-

-

-

-

464

-

-

-

-

-

42,796(5)

43,328(5)

268,783

172,235

Denis Bedoret,Managing Director of MaSTherCell

2018

2017

211,847(6)

208,542(6)

56,539

31,281

-

-

20,214

-

-

-

-

-

-

-

288,600

239,823

 (1)In accordance with SEC rules, the amounts in this column reflect the fair value on the grant date of the option awards granted to the named executive, calculated in accordance with ASC Topic 718. Stock options were valued using the Black-Scholes model. The grant-date fair value does not necessarily reflect the value of shares which may be received in the future with respect to these awards. The grant-date fair value of the stock options in this column is a non-cash expense for the Company that reflects the fair value of the stock options on the grant date and therefore does not affect our cash balance. The fair value of the stock options will likely vary from the actual value the holder receives because the actual value depends on the number of options exercised and the market price of our Common Stock on the date of exercise. For a discussion of the assumptions made in the valuation of the stock options, see Note 15 to this Annual Report on Form 10-K for the year ended  November 30, 2018.December 31, 2020.
(2)For 20182020 and 2017,2019, represents the compensation as described under the caption “All Other Compensation” below.
(3)Due to cash flow considerations, part of the amounts earned have been deferred periodically and, as of November 30, 2018, an aggregate of $195,501 has been deferred by agreement and accrued by the Company.  See below under “Employment/Consulting Agreements – Vered Caplan.”
(4)As of November 30, 2018, an aggregate of $18,276 has been deferred and accrued by agreement and accrued the Company.  See below under “Employment/Consulting Agreements – Neil Reithinger.”
(5)Due to cash flow considerations, Prof. Ferber has been deferring part of her salary and social benefits due thereon until such time as our cash position permits payment of salary and benefits in full without interfering with our ability to pursue our plan.  As of November 30, 2018, such deferred amount totaled an aggregate of $404,791 for the years 2013 to 2018.
(6)On July 6, 2017, MaSTherCell’s Board of Directors appointed Denis Bedoret as General Manager and day-to-day manager of MaSTherCell, effective as of July 11, 2017.  On September 5, 2018, Mr. Bedoret was promoted to Managing Director of MaSTherCell.  On January 22, 2019, Mr. Bedoret was appointed as President of Masthercell Global. Out of the 2018 amounts earned, $241,835 was paid and $26,552 was deferred by agreement with MaSTherCell.

-79-

All Other Compensation

The following table provides information regarding each component of compensation for 2018fiscal years 2020 and 20172019 included in the All Other Compensation column in the Summary Compensation Table above. Represents amounts paid in New Israeli Shekels (NIS) or Swiss Franks and converted at average exchange rates for the year.

Name Year  

Automobile and Communication

Related

Expenses

$ (1)

  

Social

Benefits

$ (2)

  

Total

$

 
Vered Caplan  2020   13,172   202,468   215,640 
   2019   18,876   58,144   77,020 

NameYear

Automobile and Communication

Related

Expenses

$(1)

Israel-

related

Social

Benefits

$(2)

Total

$(3)

Vered Caplan

2018

2017

31,027

21,921

49,670

41,371

80,697

63,262

Prof. Sarah Ferber

2018

2017

5,379

5,144

37,418

38,183

42,797

43,328

(1)Represents for Ms. Caplan, a leased automobile and communication expenses.
(2)These are comprised of contributioncontributions by the Company to savings, severance, pension, disability and insurance plans generally provided in Israel and Switzerland, including education funds and managerial insurance funds.  For Ms. Caplan, thisfunds, and redeemed vacation pay. This amount represents Israeli severance fund payments, managerial insurance funds, disability insurance, supplemental education fund contribution, and social securities.  For Prof Ferber, this amount represents IsraeliSwiss severance fund payments, managerial insurance funds, disability insurance, supplemental education fund contribution and social securities. See discussion below under “Employment/Consulting Agreements“Narrative Disclosure to Summary Compensation Table – Vered Caplan and Sarah Ferber.Caplan.

68

Outstanding Equity Awards at November 30, 2018December 31, 2020

The following table summarizes the outstanding equity awards held by each named executive officer of our company as of November 30, 2018.December 31, 2020.

Name Grant Date Number of Shares Underlying Unexercised Options (#) Exercisable Number of Shares Underlying Unexercised Options (#) Unexercisable Option Exercise Price ($) Option Expiration Date Grant Date Number of Shares
Underlying
Unexercised
Options (#)
Exercisable
 Number of Shares
Underlying
Unexercised
Options (#)
Unexercisable
 Option
Exercise
Price ($)
 Option
Expiration
Date
                
Vered Caplan 02-Feb-12(1) 278,191 - 0.0012 02-Feb-22 02-Feb-12(1)  278,191   -   0.012  02-Feb-22
 22-Aug-14(1) 230,189 - 0.0012 22-Aug-24 22-Aug-14(1)  230,189   -   0.0012  22-Aug-24
 09-Dec-16(2) 145,834 20,833 4.8 09-Dec-26 09-Dec-16(1)  166,667   -   4.80  09-Dec-26
 06-Jun-17(3) 83,334 - 7.2 06-Jun-27 06-Jun-17(1)  83,334   -   7.20  06-Jun-27
 28-Jun-18(4) - 250,001 8.36 28-Jun-28 28-Jun-18(1)  250,000   -   8.36  28-Jun-28
 22-Oct-18(5) - 85,000 5.99 22-Oct-28 22-Oct-18(3)  42,500   42,500   5.99  22-Oct-28
 19-Mar-20(2)  31,875   53,125   2.99  18-Mar-30
                
Neil Reithinger 01-Aug-14(6) 16,667   6 01-Aug-19 09-Dec-16(1)  83,334   -   4.80  09-Dec-26
 09-Dec-16(2) 72,918 10,416 4.8 09-Dec-26 08-Mar-19(2)  6,250   18,750   5.07  08-Mar-29

Dr. Denis Bedoret

 14-May-18(7) 1,875 13,125 8.43 14-May-28
Prof. Sarah Ferber 02-Feb-12(1) 231,826 - 0.0012 02-Feb-22
 22-Oct-18(5) - 3,750 5.99 22-Oct-28 19-Mar-20(2)  5,625   9,375   2.99  18-Mar-30

-80-

(1)The options were fully vested as of November 30, 2018.   December 31, 2020.
(2)The options vestedvest on a quarterly basis over a period of two years from the date of grant.
(3)The options vested intwoequal installments on December 6, 2018 and on June 6, 2018.
(4)The option vested in two annual installments of 125,001 and 125,000 on each of the 6th and 12th month anniversaries from the date of grant.
(5)The options vest on a quarterly basis over a period of four years from the date of grant.
(6)One quarter of the options vest at the end of each three months from the date of grant.
(7)The options vested on a quarterly basis over a period of two years from the date of grant and were fully vested as of June 30, 2018.

Option Exercises in 2018

There were no option exercises by our named executive officers during our fiscal year ended November 30, 2018.December 31, 2019 and 2020.

Narrative Disclosure to Summary Compensation Table

Vered Caplan

On August 14, 2014, our Board of Directors confirmed that Ms. Vered Caplan, who hashad served as our President and Chief Executive Officer on an interim basis since December 23, 2013, was appointed as our President and Chief Executive Officer. In connection with her appointment as our President and Chief Executive Officer, on August 22, 2014, our wholly-owned Israeli Subsidiary, Orgenesis Ltd., entered into a Personal Employment Agreement with Ms. Caplan (the “Caplan Employment Agreement”). The Caplan Employment Agreement replaced a previous employment agreement with Ms. Caplan dated April 1, 2012 pursuant to which she had served as Vice President.

On March 30, 2017, we and Ms. Caplan entered into an employment agreement replacing the Caplan Employment Agreementa previous employment agreement dated August 22, 2014 (the “Amended Caplan Employment Agreement”). Under the Amended Caplan Employment Agreement, which took effect April 1, 2017, Ms. Caplan'sCaplan’s annual salary continued at $160,000 per annum, subject to adjustment to $250,000 per annum upon the listing of the Company’s securities on an Exchange. Ms. Caplan is also entitled to an annual cash bonus with a target of 25% of base salary, provided that the actual amount of such bonus may be greater or less than the target amount. Ms. Caplan was entitled to a signing bonus of $150,000 upon execution of the Amended Caplan Employment Agreement. Under the Amended Caplan Employment Agreement, Ms. Caplan is entitled to the following social benefits typically provided to Israeli employees, computed on the basis of her base salary: (i) Manager's Insurance under Israeli law pursuant to which the Company contributes between 6.5% and 7.5% (and Ms. Caplan contributes an additional 6%), (ii) severance pay under Israeli law pursuant to which the Company contributes 8 1/3% and (iii) Education fund pursuant to which the Company continues to contribute $3,677 a year. In addition, Ms. Caplan is also entitled to paid annual vacation days, annual recreation allowance, sick leave and expenses reimbursement. In addition, we provide Ms. Caplan with a leased company car and a mobile phone.

Either we or Ms. Caplan may terminate the employment under the Amended Caplan Employment Agreement upon six months prior written notice. Upon termination by us of Ms. Caplan’s employment without cause (as defined therein) or by Ms. Caplan for any reason whatsoever, in addition to any accrued but unpaid base salary and expense reimbursement, she shall be entitled to receive an amount equal to 12 months of base salary at the highest annualized rate in effect at any time before the employment terminates payable in substantially equal installments. Upon termination by us of Ms. Caplan’s employment without cause (as defined therein) or by Ms. Caplan for any reason following a Change of Control (as defined therein), in addition to any accrued but unpaid base salary and expense reimbursement, she shall be entitled to receive an amount equal to 18 months of one and a half times annual base salary at the highest annualized rate in effect at any time before the employment terminates payable in substantially equal installments.

-81-

On May 10, 2017, we and Ms. Caplan further amended the Amended Caplan Employment Agreement pursuant to which Ms. Caplan isbecame entitled to a grant under the 2017 of options (the “Initial Option”) to purchase 83,334 shares of the Company’s common stock at a per share exercise price equal to the Fair Market Value (as defined in our 2017 Equity Incentive Plan (the “2017 Plan”)) of the Company’s common stock on the date of grantgrant. The amendment further providesprovided that beginning in fiscal 2018, subject to approval by the compensation committee, Ms. Caplan isbecame entitled to an additional option (the “Additional Option”; together with the Initial Option, the “Options”) under the 2017 Plan for up to 250,000 shares of common stock of the Company to be awarded in such amounts per fiscal year as shall be consistent with the Plan, in each case at a per share exercise price equal to the Fair Market Value (as defined in the Plan) of the Company’s common stock on the date of grant.

In 2018, following the listing of the company’sCompany’s securities on the NASDAQ market,Nasdaq, Ms. Caplan’s annual salary was raised to $250,000. On June 6, 2017 and June 28, 2018 the compensation committee approved a grant of 83,334 and 250,001

For additional information regarding Ms. Caplan’s stock options respectively. In October 2018, Ms. Caplan was awarded a further bonus of $200,000 and 85,000 stock options. For additional information,awards, see the Outstanding Equity Awards table above.

TheOn November 19, 2020, we and Ms. Caplan entered into an executive directorship agreement, effective as of October 1, (the “Executive Directorship Agreement”), that supersedes and replaces the Amended Caplan Employment Agreement (the “Prior Agreement”). Pursuant to the Executive Directorship Agreement, Ms. Caplan will continue to serve the Company as its Chairperson of the Board of Directors (the “Board”) and shall receive in consideration for her serving as Chairperson of the Board an annual regular Board fee in the amount of $75,000 payable by the Company in equal quarterly installments in advance. In addition, Ms. Caplan may be eligible for non-recurring special Board fees as reviewed and approved by the Compensation Committee of the Board (the “Compensation Committee”) and then reviewed and ratified by the Board. In addition, Ms. Caplan may be granted option awards from time to time at the discretion of the Compensation Committee.

69

Ms. Caplan’s position as Chairperson of the Board under the Executive Directorship Agreement may be terminated for any reason by either Ms. Caplan or the Company upon 90 days prior written notice (the “Notice Period”), provided that the Company may terminate such appointment as Chairperson at any time during the Notice Period subject to certain conditions. Such termination as Chairperson of the Board will be deemed a termination even if Ms. Caplan remains as a regular director of the Board. Upon termination by the Company of Ms. Caplan’s employment other than for cause or by Ms. Caplan for any reason whatsoever, in addition to any Accrued Obligations (as defined therein) she shall be entitled to receive a lump sum payment equal to the sum of (i) the annual regular Board fee (the “Board Fee”) and (ii) the greater of actual or target annual performance bonus to which she may have been entitled to as of the termination date (in each case, less all customary and required taxes and related deductions).

Ms. Caplan’s position under the Executive Directorship Agreement may be terminated in the event of a Change of Control (as defined therein) by the Company other than for cause or by Ms. Caplan for any reason whatsoever. In the event of a Change of Control and if, within one year following such Change of Control, employment under the Executive Directorship Agreement is terminated by the Company other than for cause or by Ms. Caplan for any reason whatsoever, in addition to any Accrued Obligations, she shall be entitled to receive a lump sum payment equal to one and a half times the sum of (i) the Board Fee and (ii) the target annual performance remuneration to which she may have been entitled as of the termination date (in each case, less all customary and required taxes and related deductions).

In addition, on November 19, 2020, Orgenesis Services Sàrl, a Swiss corporation and wholly-owned, direct subsidiary of the Company (“Orgenesis Services”), and Ms. Caplan entered into a personal employment agreement (the “Swiss Employment Agreement” and together with the Executive Directorship Agreement, the “Agreements”), pursuant to which Ms. Caplan will serve as Chief Executive Officer, President and Chairperson of the Board of Directors of Orgenesis Services and will be a material provider of services to the Company pursuant to a services agreement between the Company and Orgenesis Services. The Swiss Employment Agreement provides that Ms. Caplan is entitled to a monthly base salary of CHF 13,345.05 (equivalent to $14,583 based on the current exchange rate at signing), and an annual representation fee of CHF 24,000 (equivalent to $26,226 based on the current exchange rate at signing), payable in monthly installments of CHF 2,000. Ms. Caplan is eligible to receive a bonus at the absolute discretion of Orgenesis Services and its compensation committee. Ms. Caplan may also contains restrictive covenantsbe granted option awards from time to time, as per the recommendation of the compensation committee of Orgenesis Services as reviewed and approved by the Compensation Committee. Under the Swiss Employment Agreement, Ms. Caplan is entitled to paid annual vacation days, monthly travel allowance, sick leave, expenses reimbursement and a mobile phone. The Swiss Employment Agreement has an effective date as of October 1, 2020.

Employment under the Swiss Employment Agreement may be terminated for any reason by Ms. Caplan or by Orgenesis Services other than for just cause (as defined therein) upon six months prior written notice or by Orgenesis Services other than for just cause in the event of a Change of Control (as defined therein) of the Company upon at least 12 months prior written notice. Upon termination by Orgenesis Services of Ms. Caplan’s employment without just cause or by Ms. Caplan for any reason whatsoever, in addition to any Accrued Obligations (as defined therein), she shall be entitled to receive a lump sum payment equal to the sum of (i) her Base Salary (as defined therein) at the rate in effect as of the termination date and (ii) the greater of actual or target annual performance bonus to which she may have been entitled to for the year in which employment terminates (in each case, less all customary and required taxes and employment-related deductions). In the event of a Change of Control and if, within one year following such Change of Control, employment is terminated by Orgenesis Services other than for cause or by Ms. Caplan for any reason whatsoever, in addition to any Accrued Obligations she shall be entitled to receive a lump sum payment equal to one and a half times the sum of (i) her Base Salary and (ii) the target annual performance bonus to which she may have been entitled to for the year in which employment terminates (in each case, less all customary and required taxes and employment-related deductions).

The Swiss Employment Agreement provides for customary protections of the Company'sOrgenesis Services’ confidential information and intellectual property.

On November 19, 2020, the Compensation Committee approved a special remuneration of $400,000 to Ms. Caplan for her outstanding service in the business development of the Company and its affiliates. The payment of such remuneration was made at the time of entry into the Agreements.

70

Neil Reithinger

Mr. Reithinger was appointed Chief Financial Officer, Treasurer and Secretary on August 1, 2014. Mr. Reithinger’s employment agreement stipulates a monthly salary of $1,500; payment of an annual bonus as determined by the Company in its sole discretion, participation in the Company’s pension plan; grant of stock options as determined by the Company; and reimbursement of expenses. In addition, on August 1, 2014, the Company entered into a financial consulting agreement with Eventus Consulting, P.C., an Arizona professional corporation, of which Mr. Reithinger is the sole shareholder (“Eventus”), pursuant to which Eventus has agreed to provide financial consulting services to the Company. In consideration for Eventus’ services, the Company agreed to pay Eventus according to its standard hourly rate structure. The term of the consulting agreement was for a period of one year from August 1, 2014 and automatically renews for additional one-year periods upon the expiration of the term unless otherwise terminated. Eventus is owned and controlled by NeilMr. Reithinger. On December 16, 2020, the Compensation Committee of the Board of Directors of the Company, approved a special one-time bonus of $200,000 was paid prior to December 31, 2020. As of November 30, 2018,December 31, 2020, Eventus was owed $18,276$28 thousand for accrued and unpaid services under the financial consulting agreement.

Prof. Sarah Ferber

Our wholly-owned Israeli Subsidiary, Orgenesis Ltd., entered into a Personal Employment Agreement with Prof. Ferber on February 2, 2012 to serve as Chief Scientific Officer (the “Ferber Employment Agreement”) on a part time basis. Under the Ferber Employment Agreement, Prof. Ferber earned an annual salary of the current New Israeli Shekel equivalent of $232,000 since September 2013. However, in order to reduce operating expenses and conserve cash, Prof. Ferber has been deferring a part of her salary and social benefits due thereon until such time as our cash position permits payment of salary in full without interfering with our ability to pursue our plan of operations, and, as of November 30, 2018, such deferred amount totaled an aggregate of $404,791. Under the Ferber Employment Agreement, Prof. Ferber is entitled to the following social benefits out of her base salary typically provided to Israeli employees: (i) Manager’s Insurance under Israeli law pursuant to which the Company contributes 2.5% (and Prof. Ferber contributes an additional 3.5%) and, in addition, the Company contributes 1.25% towards loss of working capacity disability insurance, (ii) pension plan to which the Company contributes 3.75% (and Prof. Ferber contributes an additional 3.5%), (iii) severance pay under Israeli law pursuant to which the Company contributes 8 1/3% and (iv) Education fund pursuant to which the Company contributes 7.5% (and Prof. Farber contributes an additional 2.5%). In addition, Prof. Ferber is also entitled to paid annual vacation days, annual recreation allowance, sick leave and expenses reimbursement. In addition, we provide Prof. Ferber with a mobile phone.

-82-

The Ferber Employment Agreement does not specify a stated term and either we or Ms. Ferber are entitled to terminate Prof. Ferber’s employment upon four months’ notice other than in the case of a termination for cause. The Ferber Employment Agreement contains customary provisions regarding confidentiality of information, non-competition and assignment of inventions.

In October 2018, Prof. Ferber was awarded 3,750 options. The options shall vest in equal quarterly installments over four years.

Denis Bedoret

Effective October 24, 2017, our subsidiary, MaSTherCell, entered into a management agreement with BM&C SPRL/BVBA, a Belgian company owned by Denis Bedoret, for certain services to be performed by Dr. Bedoret on an exclusive and full-time basis (the “Bedoret Agreement”). The agreement appoints Dr. Bedoret as General Manager of MaSTherCell, requires him to work 220 days annually and stipulates compensation based on revenue with (i) a daily rate of Euro 800 until such time that MaSTherCell’s annual revenue reaches Euro 10 million, (ii) a daily rate of Euro 850 until such time that MaSTherCell’s annual revenue reaches Euro 15 million and (iii) a daily rate of Euro 900 until such time that MaSTherCell’s annual revenue exceeds Euro 15 million. Dr. Bedoret is also entitled to expense reimbursement and a bonus equivalent to up 15% of the annual fees approved by MaSTherCell’s Board of Directors, subject to goals and achievements to be agreed upon by the parties. Dr. Bedoret is also entitled to participation in Orgenesis’ equity incentive plan after six months after the effective date. The Bedoret Agreement also contains customary termination clauses.

In May 2018, Dr. Bedoret was awarded 15,000 options. The options shall vest in equal quarterly installments over two years.

On September 5, 2018, Dr. Bedoret was promoted to Managing Director of MaSTherCell. On January 22, 2019, Dr. Bedoret was appointed to President of Masthercell Global.

Potential Payments upon Change of Control or Termination following a Change of Control

Our employment agreements with our named executive officers provide incremental compensation in the event of termination, as described herein. Generally, we currently do not provide any severance specifically upon a change in control nor do we provide for accelerated vesting upon change in control. Termination of employment also impacts outstanding stock options.

Due to the factors that may affect the amount of any benefits provided upon the events described below, any actual amounts paid or payable may be different than those shown in this table. Factors that could affect these amounts include the basis for the termination, the date the termination event occurs, the base salary of an executive on the date of termination of employment and the price of our common stock when the termination event occurs.

The following table sets forth the compensation that would have been received by each of the Company’s executive officers had they been terminated as of November 30, 2018.December 31, 2020.

        Accrued    

 

Name

 

Salary

Continuation

  

 

Bonus

  Vacation Pay  Total Value 
Vered Caplan $*  $62,500  $111,731   $174,231 
Prof. Sarah Ferber $                       -  $    -  $183,534  $183,534 
Name

Salary

Continuation

Vered Caplan$*

 

(*)

Termination by Company without cause: $250,000

Termination without cause following a change in control: $375,000

 

-83-

Director Compensation

The following table sets forth for each non-employee director that served as a director during the year ended December 31, 2020 certain information concerning his or her compensation for the year ended November 30, 2018:December 31, 2020 and the December 2018 transition period:

Name

Fees

Earned

or

Paid in

Cash

($)

Stock

Awards

($)

Option

Awards

($)(1)

Non-equity

Incentive Plan

Compensation

($)

Nonqualified

Deferred

Compensation

Earnings

($)

All Other

Compensation

($)

Total

($)

Guy Yachin147,290-83,480---230,770
Yaron Adler6,680-83,480---90,160
Dr. David Sidransky15,195-104,537---119,732
Hugues Bultot1,480-----1,480
Ashish Nanda4,375-12,900---17,275
71

Year Ended December 31, 2020

Name 

Fees

Earned

or

Paid in

Cash

($)

  

Stock

Awards

($)

  

Option

Awards

($) (1)

 

Non-equity

Incentive Plan

Compensation

($)

  

Nonqualified

Deferred

Compensation

Earnings

($)

  

All Other

Compensation

($)

  

Total

($)

 
Guy Yachin  52,500       -  45,462 (2)          -           -           -   97,962 
Yaron Adler  46,250   -  45,306 (3)  -   -   -   91,556 
Dr. David Sidransky  75,000   -  45,518 (4)  -   -   -   120,518 
Ashish Nanda  52,500   -  45,257 (5)  -   -   -   97,757 
Mario Philips  37,500      27,514 (6)              65,014 

(1)In accordance with SEC rules, the amounts in this column reflect the fair value on the grant date of the option awards granted to the named executive, calculated in accordance with ASC Topic 718.  Stock options were valued using the Black-Scholes model.  The grant-date fair value does not necessarily reflect the value of shares which may be received in the future with respect to these awards.  The grant-date fair value of the stock options in this column is a non-cash expense for the Company that reflects the fair value of the stock options on the grant date and therefore does not affect our cash balance.  The fair value of the stock options will likely vary from the actual value the holder receives because the actual value depends on the number of options exercised and the market price of our common stock on the date of exercise.  For a discussion of the assumptions made in the valuation of the stock options, see Note 15 (Stock Based Compensation) to our financial statements, which are included in thethis Annual Report on Form 10-K.
(2)

Aggregate number of option awards outstanding as of December 31, 2020 was 150,934 of which (i) 122,184 options are exercisable as of December 31, 2020, (ii) 12,500 options are exercisable on April 1, 2021 and (iii) 16,250 options are exercisable on December 17, 2021. Does not include $192 thousand related to options held by Caerus Therapeutics LLC over which Mr. Yachin does not have beneficial control.

(3)

Aggregate number of option awards outstanding as of December 31, 2020 was 169,325 of which (i) 141,825 options are exercisable as of December 31, 2020, (ii) 12,500 options are exercisable as of April 1, 2021 and (iii) 15,000 options are exercisable on December 17, 2021.

(4)

Aggregate number of option awards outstanding as of December 31, 2020 was 133,401 of which (i) 104,201 options are exercisable as of December 31, 2020, (ii) 12,500 options are exercisable on April 1, 2021 and (iii) 16,700 options are exercisable on December 17, 2021.

(5)Aggregate number of option awards outstanding as of December 31, 2020 was 66,700 of which (i) 39,600 options are exercisable as of December 31, 2020, (ii) 12,500 options are exercisable on April 1, 2021 and (ii) 14,600 options are exercisable on December 17, 2021.
(6)Aggregate number of option awards outstanding as of December 31, 2020 was 32,500 of which (i) 2,083 options are exercisable on January 9, 2021 (ii) 12,500 options are exercisable on April 1, 2021 (iii) 13,750 options are exercisable on December 17, 2021 (iv) 2,084 options are exercisable on January 9, 2022 and (v) 2,084 options are exercisable on January 9, 2023.

All directors receive reimbursement for reasonable out of pocket expenses in attending Board of Directors meetings and for participating in our business.

 

On April 2, 2012, we entered into an agreement with Guy Yachin to serve as a member of our Board of Directors for a consideration of $2,500 per month and an additional payment for every Board of Directors’ meeting at the rate of $300 for the first hour of attendance and $200 for each additional hour or portion of an hour.

On April 17, 2012, we entered into an agreement with Yaron Adler to serve as a member of our Board of Directors. In consideration for Dr. Sidransky’s services, we pay for his attendance at Board of Directors’ meetings at the rate of $300 for the first hour of attendance and $200 for each additional hour or portion of an hour.

On July 17, 2013 we entered into an agreement with Dr. David Sidransky to serve as a member of our Board of Directors. In consideration for Dr. Sidransky’s services, we pay for his attendance at Board of Directors’ meetings at the rate of $300 for the first hour of attendance and $200 for each additional hour or portion of an hour.

Compensation Policy for Non-Employee Directors.

In October 2018, the Board of Directors adopted a compensation policy for non-employee directors which replaced the non-employee director compensation terms discussed above. By its terms, the policy became effective November 2018. Under the adopted policy, each director is to receive an annual cash compensation of $30,000 and the Chairman and Vice Chairman is paid an additional $15,000 per annum. Each committee member will be paid an additional $7,500 per annum and each committee chairman is to receive $15,000 per annum. Cash compensation will be made on a quarterly basis.

All newly appointed directors also receive options to purchase up to 6,250 shares of the Company’s common stock. All directors are entitled on an annual bonus of options for 12,500 shares and each committee member is an entitled to a further option to purchase up to 1,250 shares of common stock and each committee chairperson to options for an additional 2,100 shares of common stock. In addition, the Chairman and Vice Chairman shall be granted an option to purchase 4,200 shares of the Company’s ordinary shares. In all cases, the options are granted at a per share exercise price equal to the closing price of the Company’s publicly traded stock on the date of grant and the vesting schedule is determined by the compensation committee at the time of grant.

-84-
72

Compensation Committee Interlocks and Insider Participation

None of our executive officers has served as a member of the Board of Directors, or as a member of the compensation or similar committee, of any entity that has one or more executive officers who served on our Board of Directors or Compensation Committee during the fiscal year ended November 30, 2018.December 31, 2020.

ITEM 12. SECURITY OWNERSHIP OF CERTAIN BENEFICIAL OWNERS AND MANAGEMENT AND RELATED STOCKHOLDER MATTERS

The following table sets forth certain information with respect to the beneficial ownership of our common stock as of February 13, 2019March 9, 2021 for (a) the named executive officers, (b) each of our directors, (c) all of our current directors and executive officers as a group and (d) each stockholder known by us to own beneficially more than 5% of our common stock. Beneficial ownership is determined in accordance with the rules of the SEC and includes voting or investment power with respect to the securities. We deem shares of common stock that may be acquired by an individual or group within 60 days of February 13, 2019March 9, 2021 pursuant to the exercise of options or warrants to be outstanding for the purpose of computing the percentage ownership of such individual or group but are not deemed to be outstanding for the purpose of computing the percentage ownership of any other person shown in the table. Except as indicated in footnotes to this table, we believe that the stockholders named in this table have sole voting and investment power with respect to all shares of common stock shown to be beneficially owned by them based on information provided to us by these stockholders. Percentage of ownership is based on 15,620,97124,199,674 shares of common stock outstanding on February 13, 2019March 9, 2021.

Security Ownership of Greater than 5% or Greater Beneficial Owners

Name and Address of

Beneficial Owner

Amount and Nature of

Beneficial Ownership(1)

Percent(1) 

Amount and Nature of

Beneficial Ownership (1)

  Percent(1) 

Oded Shvartz

130 Biruintei Blvd.

Pantelmon

Ilfov, Romania

1,830,65811.72%

Image Securities fzc.

2310, 23rd floor, Tiffany

Towers, JLT

Dubai, UAE

2,336,390(2)13.39%  3,126,434(2)  12.92%

SFPI - FPIM (Societe Federale de Participations et d'lnvestissement) SA

Avenue Louise 32, bte 4 at 1050 Bruxelles, Belgium, BCE n° 253.445.063

947,055(3)5.72%
Yehuda Nir
c/o Orgenesis Inc.
20271 Goldenrod Lane
Germantown, MD 20876
  2,175,152(3)  8.99%
Gakasa Holding, LLC
c/o Knoll Capital Management
5 East 44th Street
New York, NY 10017
  1,316,364(4)  5.44%

73

Security Ownership of Directors and Executive Officers

Name and Address of

Beneficial Owner

 

Amount and Nature of

Beneficial Ownership (1)

  Percent(1) 
Vered Caplan
c/o Orgenesis Inc.
20271 Goldenrod Lane
Germantown, MD 20876
  1,104,006(5)  4.56%
Neil Reithinger
14201 N. Hayden Road, Suite A-1
Scottsdale, AZ 85260
  112,709(6)  <1%
Guy Yachin
c/o Orgenesis Inc.
20271 Goldenrod Lane
Germantown, MD 20876
  134,684(7)  <1%
Dr. David Sidransky
c/o Orgenesis Inc.
20271 Goldenrod Lane
Germantown, MD 20876
  116,701(8)  <1%
Yaron Adler
c/o Orgenesis Inc.
20271 Goldenrod Lane
Germantown, MD 20876
  217,629(9)  <1%
Ashish Nanda
c/o Orgenesis Inc.
20271 Goldenrod Lane
Germantown, MD 20876
  52,100(10)  <1%
Mario Philips
c/o Orgenesis Inc.
20271 Goldenrod Lane
Germantown, MD 20876
  14,583(11)  <1%
Directors & Executive Officers as a Group (7 persons)  1,752,412   7.24%

Notes:

Name and Address of

Beneficial Owner

Amount and Nature of

Beneficial Ownership(1)

Percent(1)

Vered Caplan

c/o Orgenesis Inc.

20271 Goldenrod Lane

Germantown, MD 20876

894,006(4)5.41%

Neil Reithinger

14201 N. Hayden Road, Suite A-1

Scottsdale, AZ 85260

100,001(5)<1%

-85-

Prof. Sarah Ferber

c/o Orgenesis Inc.

20271 Goldenrod Lane

Germantown, MD 20876

232,295(6)1.47%

Dr. Denis Bedoret

c/o Orgenesis Inc.

20271 Goldenrod Lane

Germantown, MD 20876

5,625(7)<1%

Guy Yachin

c/o Orgenesis Inc.

20271 Goldenrod Lane

Germantown, MD 20876

80,934(8)<1%

Dr. David Sidransky

c/o Orgenesis Inc.

20271 Goldenrod Lane

Germantown, MD 20876

62,501(9)<1%

Yaron Adler

c/o Orgenesis Inc.

20271 Goldenrod Lane

Germantown, MD 20876

163,879(10)1.04%

Ashish Nanda

c/o Orgenesis Inc.

20271 Goldenrod Lane

Germantown, MD 20876

-(11)-
Directors & Executive Officers as a Group (8 persons)1,539,2419%

Notes:

(1)Percentage of ownership is based on 15,620,97124,167,784 shares of our common stock outstanding as of February 13, 2019.March 9, 2021.  Except as otherwise indicated, we believe that the beneficial owners of the common stock listed above, based on information furnished by such owners, have sole investment and voting power with respect to such shares, subject to community property laws where applicable.  Beneficial ownership is determined in accordance with the rules of the SEC and generally includes voting or investment power with respect to securities.  Shares of common stock subject to options or warrants currently exercisable or exercisable within 60 days, are deemed outstanding for purposes of computing the percentage ownership of the person holding such option or warrants but are not deemed outstanding for purposes of computing the percentage ownership of any other person.
(2)IncludingConsists of (i) 1,494,217 ordinary shares and (ii) 1,832,538ordinary shares issuable upon exercise of outstanding warrants at a price of$6.24 per share. The warrants are exercisable over a three yearthree-year period from the date of issuance.
(3)Under the terms of the SFPI Agreement since the Company uplisted to NASDAQ, SFPI is entitled to convert its MaSTherCell’s equity interest into shares of our Common Stock based upon a conversion price of $6.24 (using an exchange rate of approximately $0.85), the exercise period of the option is 3 years from the closing date of the SFPI Agreement. The $6.24 conversion price represents the price after the previous stock split of the Company.
(4)Consists of (i) 508,380309,464 ordinary shares issuable upon exercise of outstanding warrants at a price of $6.24 per share, exercisable until June 30, 2021, (ii) 153,846 ordinary shares issuable upon exercise of outstanding warrants at a price of $6.24 per share, exercisable until June 9, 2021, (iii) 50,000 ordinary shares issuable upon exercise of outstanding warrants at a price of $7.00 per share, exercisable until October 3, 2022, and (iv) 1,661,842 ordinary shares issuable upon exercise of convertible debt at a price of $7.00 per share.
(4)Consists of 1,316,364 ordinary shares.

74

(5)

Consists of (i) 278,191 ordinary shares issuable upon exercise of outstanding options at a price of$0.00120.012 per share, (ii)166,667(ii) 230,189 ordinary shares issuable upon exercise of outstanding options at a price of$4.800.0012 per share,(iii) 83,334166,667 ordinary shares issuable upon exercise of outstanding options at a price of$7.20 4.80 per share, (iv) 125,00083,334 ordinary shares issuable upon exercise of outstanding options at a price of$8.367.20 per share,and (v) 10,625 (vi) 250,000 ordinary shares issuable upon exercise of outstanding options at a price of$5.998.36 per share. and (v) 53,125 ordinary shares issuable upon exercise of outstanding options at a price of $5.99 per share and(vii) 42,500 ordinary shares issuable upon exercise of outstanding options at a price of $2.99 per share. Does not include (i) options for (i) 125,001 shares of common stock with an exercise price of $8.36 per share that are exercisable on June 28, 2019 and (ii) 74,37531,875 shares of common stock with an exercise price of $5.99 per sharethat are exercisable quarterly after DecemberApril 22, 2018.

2021 and (ii) option for 42,500 shares of common stock with an exercise price of $2.99 per share that are exercisable quarterly after March 31, 2021.

-86-

(5)
(6)

Consists of (i) 16,66783,334 ordinary shares issuable upon exercise of outstanding options at a price of$6.004.80 per share and (ii) 83,33421,875 ordinary shares issuable upon exercise of outstanding options at a price of$4.805.07 per share.

(6)Consists of (i) 231,826share (iii) 7,500 ordinary shares issuable upon exercise of outstanding options at a price of $2.99 per share. Does not include (i) options for 3,125 shares of common stock with an exercise price of $5.07 per share that are exercisable quarterly after April 1, 2021 and (ii) option for 7,500 shares of common stock with an exercise price of $2.99 per share that are exercisable quarterly after March 31, 2021.

(7)

Consists of (i) 39,267 ordinary shares issuable upon exercise of outstanding options at a price of $0.001210.2 per share and (ii) 46941,667 ordinary shares issuable upon exercise of outstanding options at a price of $4.80 per share and (iii) 28,750 ordinary shares issuable upon exercise of outstanding options at a price of $5.99 per share and (iv) 25,000 ordinary shares issuable upon exercise of outstanding options at a price of $2.99 per share. Does not include options exercisable at a price per share of $7.00 into 70,000 ordinary shares held by Caerus Therapeutics LLC for which Mr. Yachin does not have beneficial control.

(8)

Consists of (i) 20,834 ordinary shares issuable upon exercise of outstanding options at a price of $9 per share and (ii) 41,667 ordinary shares issuable upon exercise of outstanding options at a price of $4.80 per share and (iii) 29,200 ordinary shares issuable upon exercise of outstanding options at a price of $5.99 per share and (iv) 25,000 ordinary shares issuable upon exercise of outstanding options at a price of $2.99 per share.

(9)

Consists of (i) 63,304 ordinary shares, (ii) 58,908 ordinary shares issuable upon exercise of outstanding options at a price of $9.48 per share and (iii) 41,667 ordinary shares issuable upon exercise of outstanding options at a price of $4.80 per share and (iv) 28,750 ordinary shares issuable upon exercise of outstanding options at a price of $5.99 per share and (iiv) 25,000 ordinary shares issuable upon exercise of outstanding options at a price of $2.99 per share.

(10)

Consists of (i) 27,100 ordinary shares issuable upon exercise of outstanding options at a price of $5.99 per share.  Does not include options for 3,281 shares of common stock with an exercise price of $5.99 per sharethat are exercisable quarterly after December 22, 2018.

(7)Consists of 5,625 and (ii) 25,000 ordinary shares issuable upon exercise of outstanding options at a price of$8.43 $2.99 per share. Does not include options for 9,375 shares of common stock with an exercise price of $8.43 per sharethat are exercisable quarterly after December 30, 2018.

(8)(11)Consists of (i) 39,2672,083 ordinary shares issuable upon exercise of outstanding options at a price of$10.2 $4.7 per share and (ii) 41,66712,500 ordinary shares issuable upon exercise of outstanding options at a price of$4.80 $2.99 per share. Does not include options for 28,7504,167 shares of common stock with an exercise price of $5.99 per share that are exercisable on October 22, 2019.
(9)Consists of (i) 20,834ordinary shares issuable upon exercise of outstanding options at a price of$9 per share and (ii) 41,667ordinary shares issuable upon exercise of outstanding options at a price of$4.80 per share.  Does not include options for 29,200 shares of common stock with an exercise price of $5.99$4.70 per share that are exercisable in October 22, 2019.three equal instalments over three anniversaries starting on January 9, 2022.

 
(10)Includes (i) 58,908ordinary shares issuable upon exercise of outstanding options at a price of$9.48 per share and (ii) 41,667ordinary shares issuable upon exercise of outstanding options at a price of$4.80 per share.  Does not include options for 28,750 shares of common stock with an exercise price of $5.99 per share that are exercisable on October 22, 2019.
75 
(11)Does not include options for 27,100 shares of common stock with an exercise price of $5.99 per share that are exercisable on October 22, 2019.

Securities Authorized for Issuance Under Existing Equity Compensation Plans

The following table summarizes certain information regarding our equity compensation plans as of November 30, 2018:December 31, 2020:

-87-

Plan Category

Number of Securities

to be Issued Upon

Exercise of

Outstanding Options

  

Weighted-Average

Exercise Price of

Outstanding Options

  

Number of Securities

Remaining Available for

Future Issuance Under

Equity Compensation

Plans (Excluding

Securities Reflected in

Column (a))

 
  (a)  (b)  (c) 
Equity compensation plans approved by security holders (1)  2,503,002  $4.64   1,496,998 
Equity compensation plans not approved by security holders  963,806  $3.55   141,668 
Total  3,466,808  $4.34   1,638,666 

 

 

 

 

 

 

Plan Category

 

 

Number of Securities

to be Issued Upon

Exercise of

Outstanding Options

 

 

 

Weighted-Average

Exercise Price of

Outstanding Options

Number of Securities

Remaining Available for

Future Issuance Under

Equity Compensation

Plans (Excluding

Securities Reflected in

Column (a))

 (a)(b)(c)
Equity compensation plans approved by security holders(1)1,040,942$7.05709,058
Equity compensation plans not approved by security holders(2)1,805,465$3.37300,009
Total2,846,407$4.721,009,067

(1)Consists of the 2017 Equity Incentive Plan and the Global Share Incentive Plan (2012). For a short description of those plans, see Note 15 to our 20182020 Consolidated Financial Statements included in this Annual Report on Form 10-K for the year ended November 30, 2018.December 31, 2020.

ITEM 13. CERTAIN RELATIONSHIPSAND RELATED TRANSACTIONS, AND DIRECTORSDIRECTOR INDEPENDENCE

Transactions with Related Persons

Except as set out below, as of November 30, 2018,December 31, 2020, there have been no transactions, or currently proposed transactions, in which we were or are to be a participant and the amount involved exceeds the lesser of $120,000 or one percent of the average of our total assets at year-end for the last two completed fiscal years, and in which any of the following persons had or will have a direct or indirect material interest:

any director or executive officer of our company;
any person who beneficially owns, directly or indirectly, shares carrying more than 5% of the voting rights attached to our outstanding shares of common stock;
any promoters and control persons; and
any member of the immediate family (including spouse, parents, children, siblings and in laws) of any of the foregoing persons.

On September 15, 2014, the Company received a loan in the principal amount of $100,000 from Yaron Adler Investments (1999) Ltd., an entity of which Mr. Yaron Adler, one of the Company’s non-employee director, is the sole shareholder. The loan, with an original interest rate of 6% per annum, was repayable on or before March 15, 2015. The Loan currently bears a default interest rate of 24% per annum and, as of November 30, 2017, the outstanding balance on the note was $166,581. The loan was converted into our common stock in 2018.

In January 2017, the Company entered into definitive agreements with Image Securities fzc. (“Image”) for the private placement of 2,564,115 units of the Company’s securities for aggregate subscription proceeds to the Company of $16 million at $6.24 price per unit. Each unit is comprised of one share of the Company’s Common Stock and a warrant, exercisable over a three-years period from the date of issuance, to purchase one additional share of Common Stock at a per share exercise price of $6.24. The subscription proceeds were payable on a periodic basis. Each periodic payment of subscription proceeds was evidenced by the Company’s standard securities subscription agreement. During the year ended November 30, 2017, Image remitted $4.5 million to the Company, in consideration of which, the investor received 721,160 shares of the Company’s Common Stock and three-year warrants to purchase up to an additional 721,160 shares of the Company’s Common Stock at a per share exercise price of $6.24.

In July 2018, the Company entered into definitive agreements with assignees of Image whereby these assignees remitted $4.6 million in respect of the units available under the original subscription agreement that have not been subscribed for, entitling such investors to 702,307 units, with each unit being comprised of (i) one share of the Company'sCompany’s common stock and (ii) one three-year warrant to purchase up to an additional one share of the Company’s common stock at a per share exercise price of $6.24.

-88-
76

In July 2018, the Company entered into definitive agreements with assignees of Image whereby these assignees remitted $4.6 million in respect of the units available under the original subscription agreement that have not been subscribed for, entitling such investors to 702,307 units, with each unit being comprised of (i) one share of the Company’s common stock and (ii) one three-year warrant to purchase up to an additional one share of the Company’s common stock at a per share exercise price of $6.24.

During 2018, the Company raised $6.9 million from Image entitling it to 1,111,380 shares of Common Stock and three-year warrants for an additional 1,111,380 shares of the Company’s Common Stock at a per share exercise price of $6.24. Following this remittance and those referred to in the previous paragraph, the Company received a total of $16 million out of the committed $16 million subscription proceeds under such agreement

Pursuant to an agreement entered into between the Company and Image,so long as Image’s ownership of the company is 10% or greater, itis entitled to nominate a director to the Company’s Board of Directors. Mr. Nanda was nominated for a directorship at the 2018 annual meeting in compliance with our contractual undertakings.

Pursuant to our Audit Committee charter adopted in March 2017, the Audit Committee is responsible for reviewing and approving, prior to our entry into any such transaction, all transactions in which we are a participant and in which any parties related to us have or will have a direct or indirect material interest.

Pursuant to agreements with Image, the Company procured services from Image in the amount of $4.8 million during the year ended December 31, 2020, and earned revenues from Image in the amount of $1.5 million and $1.3 million for the years ended December 31, 2020 and December 31, 2019, respectively. In addition, the company earned interest income in the amount of $169 thousand and $112 thousand for the years ended December 31, 2020 and December 31, 2019, respectively.

Named Executive Officers and Current Directors

For information regarding compensation for our named executive officers and current directors, see “Executive Compensation.”

Director Independence

See “Directors, Executive Officers and Corporate Governance – Director Independence” and “Directors, Executive Officers and Corporate Governance – Board Committees” in Item 10 above.

ITEM 14. PRINCIPAL ACCOUNTANT FEES AND SERVICES

The Boardof Directors of the Companyhas appointed Kesselman & Kesselman, a member firm of PricewaterhouseCoopers International Limited (“PwC”) as our independent registered public accounting firm (the “Independent Auditor”) for the fiscal year ending November 30, 2018.ended December 31, 2020. The following table sets forth the fees billed to the Company for professional services rendered by PwC for the years ended November 30, 2018December 31, 2020 and 2017:December 31, 2019:

Services 2018  2017
Audit Fees (1)$365,300 $211,000
Audit-Related fees (2) 16,475  22,000
Tax fees (3) 31,822  -
Total fees$413,597 $233,000
  Year Ended December 31, 
Services: 2020  2019 
Audit Fees (1) $267,231  $426,040 
Audit-Related Fees (2)  67,405   26,900 
Tax Fees (3)  12,500   18,300 
All Other Fees  10,000   49,500 
Total fees $357,136  $520,740 

(1)Audit fees consisted of audit work performed in the preparation of financial statements, as well as work generally only the independent registered public accounting firm can reasonably be expected to provide, such as statutory audits.

77

(2)Audit related fees consisted principally of audits of employee benefit plans and special procedures related to regulatory filings in 2018.2020.

(3)The tax fees were paid for reviewing various tax related matters.

Policy on Audit Committee Pre-Approval of Audit and Permissible Non-audit Services of Independent Public Accountant

Consistent with SEC policies regarding auditor independence, the Audit Committee has responsibility for appointing, setting compensation and overseeing the work of our independent registered public accounting firm. In recognition of this responsibility, the Audit Committee has established a policy to pre-approve all audit and permissible non-audit services provided by our independent registered public accounting firm.

-89-

Prior to engagement of an independent registered public accounting firm for the next year’s audit, management will submit an aggregate of services expected to be rendered during that year for each of four categories of services to the Audit Committee for approval.

1. Auditservices include audit work performed in the preparation of financial statements, as well as work that generally only an independent registered public accounting firm can reasonably be expected to provide, including comfort letters, statutory audits, and attest services and consultation regarding financial accounting and/or reporting standards.

2. Audit-Relatedservices are for assurance and related services that are traditionally performed by an independent registered public accounting firm, including due diligence related to mergers and acquisitions, employee benefit plan audits, and special procedures required to meet certain regulatory requirements.

 

3. Taxservices include all services performed by an independent registered public accounting firm’s tax personnel except those services specifically related to the audit of the financial statements, and includes fees in the areas of tax compliance, tax planning, and tax advice.

 

4. Other Fees are those associated with services not captured in the other categories. The Company generally does not request such services from our independent registered public accounting firm.

Prior to engagement, the Audit Committee pre-approves these services by category of service. The fees are budgeted and the Audit Committee requires our independent registered public accounting firm and management to report actual fees versus the budget periodically throughout the year by category of service. During the year, circumstances may arise when it may become necessary to engage our independent registered public accounting firm for additional services not contemplated in the original pre-approval. In those instances, the Audit Committee requires specific pre-approval before engaging our independent registered public accounting firm.

The Audit Committee may delegate pre-approval authority to one or more of its members. The member to whom such authority is delegated must report, for informational purposes only, any pre-approval decisions to the Audit Committee at its next scheduled meeting.

PART IV

ITEM 15. EXHIBITSEXHIBIT AND FINANCIAL STATEMENT SCHEDULES

(a)
(1)c.Financial Statements

Our consolidated financial statements are set forth in Part II, Item 8 of this Annual Report on Form 10-K and are incorporated herein by reference.

(2)d.Financial Statement Schedules

No financial statement schedules have been filed as part of this Annual Report on Form 10-K because they are not applicable or are not required or because the information is otherwise included herein.

78

(3)e.Exhibits required by Regulation S-K

No.Description
3.12.1Articles of Incorporation (incorporatedStock Purchase Agreement, dated February 2, 2020, by and among Orgenesis, Inc., GPP-II Masthercell LLC, Masthercell Global Inc. and Catalent Pharma Solutions, Inc.(incorporated by reference to an exhibitExhibit 2.1 to our registration statementthe Registrant’s Current Report on Form S-1,8-K filed with the SEC on April 2, 2009)February 3, 2020).
3.22.2CertificateAgreement and Plan of Change Pursuant to Nevada Revised Statutes Section 78.209Merger and Reorganization, dated as of September 26, 2020 by and among Orgenesis Inc., Orgenesis Merger Sub, Inc., Koligo Therapeutics Inc., the Shareholders of Koligo and Long Hill Capital V, LLC, solely in its capacity as representative of the Shareholders (incorporated by reference to an exhibit to our current report on Form 8-K, filed on SeptemberOctober 2, 2011)2020)
3.33.1Articles of MergerIncorporation, as amended (incorporated by reference to an exhibit to our current reportregistration statement on Form 8-K,S-8, filed on September 2, 2011)August 7, 2020)

-90-

No.Description
3.43.2Certificate of Amendment to Articles of Incorporation (incorporated by reference to an exhibit to our current report on Form 8-K, filed on September 21, 2011)
3.5Amended and Restated Bylaws (incorporated by reference to an exhibit to our current report on Form 8-K, filed on September 21, 2011)
3.64.1CertificateDescription of Correction, dated February 27, 2012Securities (incorporated by reference to an exhibit to our currentannual report on Form 8-K/A,10-K filed on March 16, 2012)9, 2020)
3.74.2CertificateForm of Change Pursuant to Nevada Revised Statutes Section 78.209, as filed by Orgenesis Inc. on November 13, 2017Warrant (incorporated by reference to an exhibit to our current report on Form 8-K, filed on November 16, 2017)January 22, 2020)
4.310.2Form of Stock Option Agreement (incorporated by reference to an exhibit to our current report on Form S-8, filed on August 7, 2020)
10.1Convertible Loan Agreement, dated December 6, 2013, with Mediapark A.G. (incorporated by reference to an exhibit to our current report on Form 8-K, filed on December 16, 2013)
10.310.2Investment Agreement, dated December 13, 2013, with Kodiak Capital Group, LLC (incorporated by reference to an exhibit to our current report on Form 8-K, filed on December 16, 2013)
10.410.3Registration Rights Agreement, dated December 13, 2013, with Kodiak Capital Group, LLC (incorporated by reference to an exhibit to our current report on Form 8-K, filed on December 16, 2013)
10.510.4Form of subscription agreement (incorporated by reference to an exhibit to our current report on Form 8-K, filed on March 4, 2014)
10.610.5Form of warrant (incorporated by reference to an exhibit to our current report on Form 8-K, filed on March 4, 2014)
10.710.6Consulting Agreement, dated April 3, 2014, with Aspen Agency Limited (incorporated by reference to an exhibit to our current report on Form 8-K, filed on April 7, 2014)
10.810.7Stock Option Agreement, dated April 3, 2014, with Aspen Agency Limited (incorporated by reference to an exhibit to our current report on Form 8-K, filed on April 7, 2014)
10.910.8Form of subscription agreement with form of warrant (incorporated by reference to an exhibit to our current report on Form 8-K, filed on April 28, 2014)
10.1010.9Convertible Loan Agreement, dated May 29, 2014, with Nine Investments Limited (incorporated by reference to an exhibit to our current report on Form 8-K, filed on May 30, 2014)
10.1110.10Service Agreement between Orgenesis SPRL and MaSTherCell S.A., dated July 3, 2014 (incorporated by reference to an exhibit to our current report on Form 8-K, filed on July 7, 2014)
10.1210.11Financial Consulting Agreement, dated August 1, 2014, with Eventus Consulting, P.C. (incorporated by reference to an exhibit to our current report on Form 8-K, filed on August 5, 2014)
10.1310.12Personal Employment Agreement, dated August 1, 2014, by and between Orgenesis Inc. and Neil Reithinger (incorporated by reference to an exhibit to our current report on Form 8-K, filed on August 5, 2014)
10.1410.13Personal Employment Agreement, dated July 23, 2014, by and between Orgenesis Maryland Inc. and Scott Carmer (incorporated by reference to an exhibit to our current report on Form 8-K, filed on August 7, 2014)
10.15Release Agreement, dated November 18, 2016, by and between Orgenesis Maryland Inc. and Scott Carmer (incorporated by reference to an exhibit to our current report on Form 8-K, filed on November 23, 2016)
10.16Strategic Advisory Agreement, dated November 18, 2016, by and between Orgenesis Inc. and Scott Carmer (incorporated by reference to an exhibit to our current report on Form 8-K, filed on November 23, 2016)
10.17Executive Employment Agreement, dated March 30, 2017, between Orgenesis Inc. and Vered Caplan (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on July 24, 2017)
10.1810.14Amendment No. 1, dated May 10, 2017, to Executive Employment Agreement, dated as of March 30, 2017, between Orgenesis Inc. and Vered Caplan (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on July 24, 2017)

79

No.Description
10.1910.15Share Exchange Agreement, dated November 3, 2014, by and between Orgenesis Inc. and MaSTherCell S.A. and Cell Therapy Holding SA (collectively “MaSTherCell”) and each of the shareholders of MaSTherCell (incorporated by reference to an exhibit to our current report on Form 8-K, filed on November 10, 2014)

-91-

No.Description
10.2010.16Addendum No. 1, dated March 2, 2015, to Share Exchange Agreement, dated November 3, 2014, by and between Orgenesis Inc., MaSTherCell, and each of the shareholders of MaSTherCell (incorporated by reference to an exhibit to our current report on Form 8-K, filed on March 5, 2015)
10.2110.17Escrow Agreement, dated February 27, 2015, by and between Orgenesis Inc., the shareholders of MaSTherCell S.A. and Cell Therapy Holding SA, the bondholders of MaSTherCell S.A. and Securities Transfer Corporation (incorporated by reference to an exhibit to our current report on Form 8-K, filed on March 5, 2015)
10.2210.18Orgenesis Inc. Board of Advisors Consulting Agreement, dated March 16, 2015 (incorporated by reference to an exhibit to our current report on Form 8-K, filed on March 17, 2015)
10.2310.19Addendum No. 2, dated November 12, 2015, to Share Exchange Agreement, dated November 3, 2014, by and between Orgenesis Inc., MaSTherCell, and each of the shareholders of MaSTherCell (incorporated by reference to an exhibit our current report on Form 8-K, filed on November 13, 2015)
10.2410.20Joint Venture Agreement, dated March 14, 2016, by and between Orgenesis Inc. and CureCell Co., Ltd. (incorporated by reference to an exhibit to our annual report on Form 10-K, filed on February 28, 2017)
10.2510.21Joint Venture Agreement, dated May 10, 2016, by and between Orgenesis Inc. and Atvio Biotech Ltd. (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on April 19, 2017)
10.2610.22Private Placement Subscription Agreement, dated January 26, 2017, between Orgenesis Inc. and Image Securities FZC (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on April 19, 2017)
10.2710.23Amendment No. 1, dated February 9, 2017, to the Private Placement Subscription Agreement, dated January 26, 2017, between Orgenesis Inc. and Image Securities FZC (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on April 19, 2017)
10.2810.242017 Equity Incentive Plan (incorporated by reference to an exhibit to our definitive proxy statement on Schedule 14A, filed on March 30, 2017)
10.2910.25Collaboration and License Agreement, dated as of June 8, 2018, between Orgenesis Inc. and Mircod Limited (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on October 12, 2018)
10.3010.26Private Placement Subscription Agreement, dated November 13, 2018, between Orgenesis Inc. and Avner Sonnino (incorporated by reference to an exhibit to our current report on Form 8-K, filed on November 20, 2018)
10.3110.27Private Placement Subscription Agreement, dated November 21, 2018, between Orgenesis Inc. and an accredited investor (incorporated by reference to an exhibit to our current report on Form 8-K, filed on November 28, 2018)
10.3210.28Private Placement Subscription Agreement, dated November 30, 2018, between Orgenesis Inc. and an accredited investor (incorporated by reference to an exhibit to our current report on Form 8-K, filed on December 6, 2018)
10.3310.29Private Placement Subscription Agreement, dated December 10, 2018, between Orgenesis Inc. and an accredited investor (incorporated by reference to an exhibit to our current report on Form 8-K, filed on December 14, 2018)
10.3410.30Controlled Equity Offering Sales Agreement, dated December 20, 2018, between Orgenesis Inc. and Cantor Fitzgerald & Co. (incorporated by reference to an exhibit to our current report on Form 8-K, filed on December 20, 2018)
10.3121.1*Joint Venture Agreement between the Company and First Choice International Company, Inc. dated March 12, 2019 (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on May 8, 2019)

80

No.Description
10.32Convertible Loan Agreement between Orgenesis Maryland Inc. and Yosef Ram dated April 12, 2019 (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on May 8, 2019)
10.33Joint Venture Agreement between the Company and KinerjaPay Corp. dated May 6, 2019  (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on May 8, 2019)
10.34Convertible Loan Agreement, dated April 10, 2019, by and between the Company and Investor (incorporated by reference to an exhibit to our quarterly report on form 10-Q, filed on November 7, 2019)
10.35Form of Subscription Agreement, dated May 17, 2019, by and between the Company and Investor (incorporated by reference to an exhibit to our quarterly report on form 10-Q, filed on November 7, 2019)
10.36Form of Subscription Agreement, dated May 30, 2019, by and between the Company and Investor (incorporated by reference to an exhibit to our quarterly report on form 10-Q, filed on November 7, 2019)
10.37Form of Subscription Agreement, dated June 6, 2019, by and between the Company and Investor (incorporated by reference to an exhibit to our quarterly report on Form 10-Q, filed on November 7, 2019)
10.38Transfer Agreement, dated as of August 7, 2019 by and among Masthercell Global, Orgenesis Inc. and GPP-II Masthercell, LLC (incorporated by reference to our current report on Form 8-K, filed on August 13, 2019)
10.392017 Equity Incentive Plan (incorporated by reference to an exhibit to our definitive proxy statement on Schedule 14A, filed on March 30, 2017)
10.40Securities Purchase Agreement, dated January 20, 2020, by and among the Company and the Investors (incorporated by reference to an exhibit to our current report on Form 8-K, filed on January 22, 2020)
10.41Registration Rights Agreement, dated January 20, 2020, by and among the Company and the Investors (incorporated by reference to an exhibit to our current report on Form 8-K, filed on January 22, 2020)
10.42Asset Purchase Agreement by and between Orgenesis Inc. and Tamir Biotechnology, Inc, dated April 12, 2020 (incorporated by reference to an exhibit to our current report on Form 8-K, filed on April 13, 2020)
10.43Form of Registration Rights and Lock-Up Agreement between the Company, Long Hill Capital V, LLC and Maxim Group, LLC (incorporated by reference to an exhibit to our current report on Form 8-K, filed on October 1, 2020)
10.44Form of Shareholders Lock-Up Agreement between the Company and Shareholders other than Long Hill Capital V, LLC (incorporated by reference to an exhibit to our current report on Form 8-K, filed on October 1, 2020)
10.45*Executive Directorship Agreement between the Company and Vered Caplan dated November 19, 2020
10.46*Swiss Employment Agreement between the Company and Vered Caplan dated November 19, 2020
21.1*List of Subsidiaries of Orgenesis Inc.
23.1*Consent of independent registered public accounting firm
31.1*Certification Statement of the Chief Executive Officer pursuant to Section 302 of the Sarbanes Oxley Act of 2002
31.2*Certification Statement of the Chief Financial Officer pursuant to Section 302 of the Sarbanes Oxley Act of 2002

-92-

No.Description
32.1**Certification Statement of the Chief Executive Officer pursuant to Section 906 of the Sarbanes Oxley Act of 2002
32.2**Certification Statement of the Chief Financial Officer pursuant to Section 906 of the Sarbanes Oxley Act of 2002
99.1Global Share Incentive Plan (2012) (incorporated by reference to an exhibit to our current report on Form 8-K, filed on May 31, 2012)
99.2Appendix – Israeli Taxpayers Global Share Incentive Plan (2012) (incorporated by reference to an exhibit to our current report on Form 8-K, filed on May 31, 2012)
101.INS*XBRL Instance Document
101.SCH*XBRL Taxonomy Extension Schema Document
101.CAL*XBRL Taxonomy Extension Calculation Linkbase Document
101.DEF*XBRL Taxonomy Extension Definition Linkbase Document
101.LAB*XBRL Taxonomy Extension Label Linkbase Document
101.PRE*XBRL Taxonomy Extension Presentation Linkbase Document

 

*Filed herewith

**Furnished herewith

ITEM 16. FORM 10-K SUMMARY

Not applicable.

-93-
81

SIGNATURES

Pursuant to the requirements of Section 13 or 15(d) of the Securities Exchange Act of 1934, the registrant has duly caused this report to be signed on its behalf by the undersigned, thereunto duly authorized.

ORGENESIS INC.

By:/s/ Vered Caplan
Vered Caplan
Chief Executive Officer and Chairperson of the Board of Directors (Principal Executive Officer)
Date:  February 13, 2019March 9, 2021

By:/s/ Neil Reithinger
Neil Reithinger

Chief Financial Officer, Treasurer and Secretary

(Principal Financial and Accounting Officer)

Date:  February 13, 2019March 9, 2021

Pursuant to the requirements of the Securities Exchange Act of 1934, this report has been signed below by the following persons on behalf of the registrant and in the capacities and on the dates indicated.

By:/s/ Vered Caplan
Vered Caplan
Chief Executive Officer and Chairperson of the Board of Directors (Principal Executive Officer)
Date:  February 13, 2019March 9, 2021

By:/s/ Neil Reithinger
Neil Reithinger
Chief Financial Officer, Treasurer and Secretary (Principal Financial Officer and Principal Accounting Officer)
Date:  February 13, 2019March 9, 2021

By:/s/ Guy Yachin
Guy Yachin
Director
Date:  February 13, 2019March 9, 2021

By:/s/ David Sidransky
David Sidransky
Director
Date:  February 13, 2019March 9, 2021

By:/s/ Yaron Adler
Yaron Adler
Director
Date:  February 13, 2019March 9, 2021

By:/s/ Ashish Nanda
Ashish Nanda
Director
Date:  March 9, 2021

By:/s/ Mario Philips 
Ashish NandaMario Philips 
Director 
Date:  February 13, 2019March 9, 2021 

-94-


82

ITEM 8. FINANCIAL STATEMENTS AND SUPPLEMENTARY DATA

ORGENESIS INC.

CONSOLIDATED FINANCIAL STATEMENTS AS OF DECEMBER 31, 2020

TABLE OF CONTENTS

ORGENESIS INC.Page
CONSOLIDATED FINANCIAL STATEMENTS AS OF NOVEMBER 30, 2018
TABLE OF CONTENTS

Page
REPORT OF INDEPENDENT REGISTERED PUBLIC ACCOUNTING FIRMF-2
CONSOLIDATED FINANCIAL STATEMENTS:
Consolidated Balance SheetsF-4F-3
Consolidated Statements of Comprehensive Loss (Income)F-6F-5
Consolidated Statements of Changes in EquityF-7F-6
Consolidated Statements of Cash FlowsF-9F-8
Notes to Consolidated Financial StatementsF-10F-9 to F-46F-52

F-1


F-1

REPORT OF INDEPENDENT REGISTERED PUBLIC ACCOUNTING FIRM

To the Board of Directors and Shareholdersshareholders of Orgenesis Inc.:

Opinions Opinion on the Financial Statements and Internal Control over Financial Reporting

We have audited the accompanying consolidated balance sheets of Orgenesis Inc. and its subsidiaries (the “Company”) as of November 30, 2018December 31, 2020 and 2017,2019, and the related consolidated statements of comprehensive loss (income), changes in equity and cash flows for each of the two years in the periodthen ended, November 30, 2018, including the related notes (collectively referred to as the “consolidated financial statements”). We also have audited the Company's internal control over financial reporting as of November 30, 2018, based on criteria established inInternal Control - Integrated Framework (2013) issued by the Committee of Sponsoring Organizations of the Treadway Commission (COSO).

In our opinion, the consolidated financial statements referred to above present fairly, in all material respects, the financial position of the Company as of November 30, 2018December 31, 2020 and 2017,2019, and the results of its operations and its cash flows for each of the two years in the periodthen ended November 30, 2018 in conformity with accounting principles generally accepted in the United States of America. Also

Changes in our opinion, the Company maintained, in all material respects, effective internal control over financial reporting as of November 30, 2018, based on criteria established inInternal Control - Integrated Framework (2013) issued by the COSO.

Basis for OpinionsAccounting Principle

The Company's management is responsible for these

As discussed in Note 2(x) to the consolidated financial statements, the Company changed the manner in which it accounts for maintaining effective internal control overleases in 2019.

Basis for Opinion

These consolidated financial reporting, and for its assessmentstatements are the responsibility of the effectiveness of internal control over financial reporting, included in the accompanying Management’s Report on Internal Control over Financial Reporting.Company’s management. Our responsibility is to express opinionsan opinion on the Company’s consolidated financial statements and on the Company's internal control over financial reporting based on our audits. We are a public accounting firm registered with the Public Company Accounting Oversight Board (United States) (PCAOB) and are required to be independent with respect to the Company in accordance with the U.S. federal securities laws and the applicable rules and regulations of the Securities and Exchange Commission and the PCAOB.

We conducted our audits of these consolidated financial statements in accordance with the standards of the PCAOB. Those standards require that we plan and perform the auditsaudit to obtain reasonable assurance about whether the consolidated financial statements are free of material misstatement, whether due to error or fraud, and whether effectivefraud. The Company is not required to have, nor were we engaged to perform, an audit of its internal control over financial reporting. As part of our audits we are required to obtain an understanding of internal control over financial reporting was maintained in all material respects.but not for the purpose of expressing an opinion on the effectiveness of the Company’s internal control over financial reporting. Accordingly, we express no such opinion.

Our audits of the consolidated financial statements included performing procedures to assess the risks of material misstatement of the consolidated financial statements, whether due to error or fraud, and performing procedures that respond to those risks. Such procedures included examining, on a test basis, evidence regarding the amounts and disclosures in the consolidated financial statements. Our audits also included evaluating the accounting principles used and significant estimates made by management, as well as evaluating the overall presentation of the consolidated financial statements. Our audit of internal control over financial reporting included obtaining an understanding of internal control over financial reporting, assessing the risk that a material weakness exists, and testing and evaluating the design and operating effectiveness of internal control based on the assessed risk. Our audits also included performing such other procedures as we considered necessary in the circumstances. We believe that our audits provide a reasonable basis for our opinions.opinion.

Critical Audit Matters

The critical audit matter communicated below is a matter arising from the current period audit of the consolidated financial statements that was communicated or required to be communicated to the audit committee and that (i) relates to accounts or disclosures that are material to the consolidated financial statements and (ii) involved our especially challenging, subjective, or complex judgments. The communication of critical audit matters does not alter in any way our opinion on the consolidated financial statements, taken as a whole, and we are not, by communicating the critical audit matter below, providing a separate opinion on the critical audit matter or on the accounts or disclosures to which it relates.

Revenue Recognition - Point-of-Care (“POC”) Cell Therapy Platform

As described in Management’s Report on Internal Control over Financial Reporting, management has excluded CureCellNotes 1 and Atvio from its assessment of internal control over2(w) to the consolidated financial reporting as of November 30, 2018 because they were acquired bystatements, the Company generated approximately $5.9 million in purchase business combinations during 2018.  We have also excluded CureCell and Atviorevenue from our audit of internal control over financial reporting. CureCell and Atvio are wholly-owned subsidiaries whose total assets and total revenues excluded from management’s assessment and our audit of internal control over financial reporting represent 5% and 6% respectively, of the related consolidated financial statement amounts as of andPOC services for the year ended November 30, 2018.December 31, 2020. The transaction price from those POC services is allocated by management to each distinct performance obligation based on its relative standalone selling price. The Company recognizes revenue when, or as, it satisfies a performance obligation. At contract inception, the Company determines whether the services are transferred over time or at a point in time. Revenue related to performance obligations that have no alternative use and that the Company has the right to payment for performance completed to date, at all times during the contract term, are recognized over time. Revenue from all other performance obligations are recognized as revenues by the Company at point of time (upon completion).

F-2


The principal considerations for our determination that performing procedures relating to revenue recognition - POC cell therapy platform is a critical audit matter are that there was significant judgment by management in (1) identifying the distinct performance obligations and estimating the standalone selling price of each distinct performance obligation, and (2) identifying which performance obligations create assets with alternative use to the Company, which results in revenue recognized upon completion, and which performance obligations are transferred to the customer over time. This in turn led to significant auditor judgment and effort in performing procedures to evaluate management’s significant judgment in identifying distinct performance obligations and determining whether those performance obligations create assets with alternative use to the Company.

Addressing the matter involved performing procedures and evaluating audit evidence in connection with forming our overall opinion on the consolidated financial statements. These procedures included testing the effectiveness of controls relating to the revenue recognition process. These procedures also included, among others, on a test basis, testing the completeness and accuracy of management’s identification of the distinct performance obligations by evaluating customer arrangements; and testing management’s process for determining the appropriate amount of revenue recognition based on the performance obligations identified in relevant contracts.

Definition and Limitations of Internal Control over Financial Reporting/s/ Kesselman & Kesselman

Certified Public Accountants (Isr.)

A company’s internal control over financial reporting is a process designed to provide reasonable assurance regarding the reliabilitymember firm of financial reporting and the preparation of financial statements for external purposes in accordance with generally accepted accounting principles. A company’s internal control over financial reporting includes those policies and procedures that (i) pertain to the maintenance of records that, in reasonable detail, accurately and fairly reflect the transactions and dispositions of the assets of the company; (ii) provide reasonable assurance that transactions are recorded as necessary to permit preparation of financial statements in accordance with generally accepted accounting principles, and that receipts and expenditures of the company are being made only in accordance with authorizations of management and directors of the company; and (iii) provide reasonable assurance regarding prevention or timely detection of unauthorized acquisition, use, or disposition of the company’s assets that could have a material effect on the financial statements.PricewaterhouseCoopers International Limited

 Because of its inherent limitations, internal control over financial reporting may not prevent or detect misstatements. Also, projections of any evaluation of effectiveness to future periods are subject to the risk that controls may become inadequate because of changes in conditions, or that the degree of compliance with the policies or procedures may deteriorate.

/s/Kesselman & Kesselman
Certified Public Accountants (Isr.)
A member firm of PricewaterhouseCoopers International Limited
Tel-Aviv, Israel
February 13, 2019

Tel-Aviv, Israel

March 9, 2021

We have served as the Company’s auditor since 2012.

F-3



ORGENESIS INC.
CONSOLIDATED BALANCE SHEETS
(U.S. Dollars, in thousands)F-2

  November 30, 
  2018  2017 
                                                                                 Assets      
CURRENT ASSETS:      
     Cash and cash equivalents$16,064 $ 3,519 
     Restricted Cash 392  - 
     Accounts receivable, net 4,151  1,336 
     Prepaid expenses and other receivables 913  841 
     GPP receivable, see Note 3 6,600  - 
     Receivables from related party -  691 
     Grants receivable 441  183 
     Inventory 1,736  725 
Total current assets 30,297  7,295 
NON CURRENT ASSETS:      
   Bank deposits 85  - 
   Loan to related party, see Note 11(e) 1,007  - 
   Call option derivative -  339 
   Investments in associates, net -  1,321 
   Property and equipment, net 11,901  5,104 
   Intangible assets, net 16,700  15,051 
   Goodwill 15,165  10,684 
   Other assets 292  78 
Total non-current assets 45,150  32,577 
TOTAL ASSETS$ 75,447 $ 39,872 

F-4



ORGENESIS INC.

CONSOLIDATED BALANCE SHEETS

(U.S. Dollars, in thousands)

  2020  2019 
  December 31, 
  2020  2019 
Assets      
CURRENT ASSETS:        
Cash and cash equivalents $44,923  $107 
Restricted cash  645   467 
Accounts receivable, net  3,085   1,831 
Prepaid expenses and other receivables  1,070   382 
Grants receivable  169   204 
Inventory  185   136 
Current assets of discontinued operations (See Note 3)  -   75,221 
Total current assets  50,077   78,348 
NON CURRENT ASSETS:        
Deposits $296  $299 
Loan to related party  -   2,623 
Investments in associates, net  175   - 
Property, plants and equipment, net  3,073   2,305 
Intangible assets, net  13,023   3,348 
Operating lease right-of-use assets  1,474   725 
Goodwill  8,745   4,812 
Other assets  821   35 
Total non-current assets  27,607   14,147 
TOTAL ASSETS $77,684  $92,495 

ORGENESIS INC.
CONSOLIDATED BALANCE SHEETS
(U.S. Dollars, in thousands)F-3

  November 30, 
  2018  2017 
                                                            Liabilities and equity      
CURRENT LIABILITIES:      
       Accounts payable$ 3,804 $ 3,914 
       Accrued expenses and other payables 2,269  1,435 
       Employees and related payables 3,006  2,961 
       Related parties -  116 
       Advance payments on account of grant 1,724  1,719 
       Short-term loans and current maturities of long term loans 647  378 
       Deferred income 5,317  3,611 
       Current maturities of convertible loans 378  2,780 
TOTAL CURRENT LIABILITIES 17,145  16,914 
       
LONG-TERM LIABILITIES:      
     Loans payable$ 1,662 $ 2,118 
     Convertible loans 1,038  2,415 
     Retirement benefits obligation 265  6 
     Deferred taxes 1,702  690 
       Other long term liabilities 833  - 
TOTAL LONG-TERM LIABILITIES 5,500  5,229 
TOTAL LIABILITIES 22,645  22,143 
COMMITMENTS      
REDEEMABLE NON CONTROLLING INTERESTEQUITY: 24,153  3,606 
Common stock of $0.0001 par value, 145,833,334 shares authorized,
14,951,783 and 9,872,659 shares issued as of November 30, 2018 and
November 30, 2017, respectively
 

1
  

1
 
       Additional paid-in capital 88,082  55,334 
       Receipts on account of shares to be allotted 2,253  1,483 
       Accumulated other comprehensive income 425  1,425 
       Accumulated deficit (62,411) (44,120)
Equity attributable to Orgenesis Inc. 28,350  14,123 
Non-controlling interests 299  - 
TOTAL EQUITY 28,649  14,123 
       
TOTAL LIABILITIES AND EQUITY$ 75,447 $ 39,872 

ORGENESIS INC.

CONSOLIDATED BALANCE SHEETS

(U.S. Dollars, in thousands)

  December 31, 
  2020  2019 
Liabilities and equity        
CURRENT LIABILITIES:        
Accounts payable $8,649  $5,549 
Accrued expenses and other payables  792   1,615 
Income tax payable  7   - 
Employees and related payables  1,463   1,672 
Advance payments on account of grant  692   523 
Short-term loans and current maturities of long-term loans  145   391 
Contract liabilities  59   325 
Current maturities of finance leases  19   - 
Current maturities of operating leases  485   357 
Current maturities of convertible loans  3,974   416 
Current liabilities of discontinued operations (See Note 3)  -   31,586 
TOTAL CURRENT LIABILITIES  16,285   42,434 
         
LONG-TERM LIABILITIES:        
Non-current operating leases $1,020  $455 
Convertible loans  7,200   12,143 
Retirement benefits obligation  74   41 
Deferred taxes  -   58 
Long-term debt and finance leases  64   - 
Other long-term liabilities  313   331 
TOTAL LONG-TERM LIABILITIES  8,671   13,028 
TOTAL LIABILITIES  24,956   55,462 
COMMITMENTS        
REDEEMABLE NON CONTROLLING INTEREST OF DISCONTINUED OPERATIONS (See Note 3)  -   30,955 
EQUITY:        
Common stock of $0.0001 par value, 145,833,334 shares authorized, 24,223,093 and 16,140,962 shares issued as of December 31, 2020 and December 31, 2019, respectively  3   2 
Additional paid-in capital  140,397   94,691 
Accumulated other comprehensive income  748   213 
Treasury stock at December 31, 2020 55,309 shares  (250)  - 
Accumulated deficit  (88,319)  (89,429)
Equity attributable to Orgenesis Inc.  52,579   5,477 
Non-controlling interests  149   601 
TOTAL EQUITY  52,728   6,078 
TOTAL LIABILITIES AND EQUITY $77,684  $92,495 

The accompanying notes are an integral part of these consolidated financial statements.

F-5



ORGENESIS INC.
CONSOLIDATED STATEMENTS OF COMPREHENSIVE LOSS
(U.S. Dollars, in thousands, except share and per share amounts)F-4

  Year ended 
  November 30, 
  2018  2017 
REVENUES$ 18,655 $ 10,089 
COST OF REVENUES 10,824  6,807 
GROSS PROFIT 7,831  3,282 
       
RESEARCH AND DEVELOPMENT EXPENSES,net 6,464  2,478 
AMORTIZATION OF INTANGIBLE ASSETS 1,913  1,631 
SELLING, GENERAL AND ADMINISTRATIVEEXPENSES 16,303  9,189 
OTHER INCOME,net (2,930) - 
SHARE IN LOSSES OF ASSOCIATED COMPANY 731  1,214 
OPERATING LOSS 14,650  11,230 
FINANCIAL EXPENSES,net 3,117  2,447 
LOSS BEFORE INCOME TAXES 17,767  13,677 
TAX EXPENSES (INCOME) 1,337  (1,310)
NET LOSS$ 19,104 $ 12,367 
NET INCOME ATTRIBUTABLE TO NON-CONTROLLING INTERESTS (INCLUDINGREDEEMABLE) (813) - 
       
NET LOSS ATTRIBUTABLE TO THE COMPANYLOSS PER SHARE:$18,291 $ 12,367 
       Basic$ 1.43 $ 1.28 
       Diluted$ 1.43 $ 1.31 
WEIGHTED AVERAGE NUMBER OF SHARES USEDIN COMPUTATION OF BASIC AND DILUTEDLOSS PER SHARE:    
       Basic 13,374,103  9,679,964 
       Diluted 13,374,103  9,714,252 
       
COMPREHENSIVE LOSS -      
       Net loss$ 19,104 $ 12,367 
       Other Comprehensive (income) loss – Translation adjustment 1,000  (2,630)
Comprehensive loss$ 20,104 $ 9,737 
Comprehensive income attributed to non-controlling interests (including redeemable) (813) - 
COMPREHENSIVE LOSS ATTRIBUTED TOORGENESIS INC.$ 19,291 $ 9,737 

ORGENESIS INC.

CONSOLIDATED STATEMENTS OF COMPREHENSIVE LOSS (INCOME)

(U.S. Dollars, in thousands, except share and per share amounts)

  2020  2019 
  Year ended December 31, 
  2020  2019 
Revenues $6,177  $2,629 
Revenues from related party  1,475   1,270 
Total revenues  7,652   3,899 
Cost of research and development and research and development services, net  83,986   14,014 
Amortization of intangible assets  478   430 
Selling, general and administrative expenses  18,973   11,451 
Other income, net  (4)  (21)
Operating loss  95,781   21,975 
Financial expenses, net  1,061   843 
Share in net income of associated companies  (106)  - 
Loss from continuing operation before income taxes  96,736   22,818 
Tax income  (1,609)  (229)
Net loss from continuing operation  95,127   22,589 
Net loss (income) from discontinued operations, net of tax  (95,706)  3,452 
Net loss (income) $(579) $26,041 
Net loss attributable to non-controlling interests (including redeemable) from continuing operation  (39)  (99)
Net loss attributable to non-controlling interests (including redeemable) from discontinued operations  (492)  (1,821)
Net loss (income) attributable to Orgenesis Inc. $(1,110) $24,121 
Loss (income) per share:        
Basic and diluted from continuing operations $4.46  $1.41 
Basic and diluted from discontinued operations $(4.75) $0.36 
Basic and diluted $(0.29) $1.77 
         
Weighted average number of shares used in computation of Basic and Diluted loss per share:        
Basic and diluted  21,320,314   15,907,995 
         
Comprehensive loss (income):        
Net loss from Continuing Operation $95,127  $22,589 
Net loss (income) from Discontinued Operations, Net of Tax  (95,706)  3,452 
Other Comprehensive (income) loss – Translation adjustment  (341)  456 
Release of translation adjustment due to sale of subsidiary  (194)  - 
Comprehensive loss (income) $(1,114) $26,497 
Comprehensive loss attributed to non-controlling interests (including redeemable)  (39)  (99)
Comprehensive loss attributed to non-controlling interests (including redeemable) from discontinued operations  (492)  (1,821)
Comprehensive loss (income) attributed to Orgenesis Inc. $(1,645) $24,577 

The accompanying notes are an integral part of these consolidated financial statements.

F-6



F-5

ORGENESIS INC.

CONSOLIDATED STATEMENTS OF CHANGES IN EQUITY

(U.S. Dollars, in thousands, except share amounts)

  1  2  3  4  5  

6

  

7

  Total 
  Common Stock  Accumulated
Other
     Equity
Attributable
       
  Number  Par Value  Additional Paid-in Capital  Comprehensive Income
(loss)
  Accumulated Deficit  

to
Orgenesis Inc.

  

Non- Controlling

Interest

  Total 
BALANCE AT JANUARY 1, 2019  15,540,333  $2  $90,597  $669  $(65,163) $26,105  $645  $26,750 
Changes during the Year ended December 31, 2019:                                
Stock-based compensation to employees and directors  -   -   2,106   -       2,106   58   2,164 
Stock-based compensation to service providers  75,629   *-   893   -   -   893   -   893 
Stock-based compensation for Tamir purchase agreement (See Note 3)  -                             
Exercise of options  -                             
Beneficial conversion
feature of convertible loans
                                
Issuance of shares and warrants  -                             
Issuance of shares related to acquisition of Koligo  -                             
Sale of subsidiaries                                
Repurchase of treasury stock  -                             
Stock-based compensation to strategic collaborations  525,000   *-   2,641           2,641   -   2,641 
Issuance and modification of warrants and Beneficial conversion feature of convertible loans  -   -   515   -   (145)  370   -   370 
Transaction with non-controlling interest GPP (See Note 1)  -   -   2,034   -       2,034   -   2,034 
Adjustment to redemption value of redeemable non-controlling interest  -   -   (4,095)  -   -   (4,095)  -   (4,095)
Comprehensive loss for the year      -   -   (456)  (24,121)  (24,577)  (102)  (24,679)
BALANCE AT DECEMBER 31, 2019  16,140,962  $2  $94,691  $213  $(89,429) $5,477  $601  $6,078 

*Represents an amount lower than $1 thousand

The accompanying notes are an integral part of these consolidated financial statement

F-6

ORGENESIS INC.

CONSOLIDATED STATEMENTS OF CHANGES IN EQUITY

(U.S. Dollars, in thousands, except share amounts)

  1  2  3  4  5  6  

7

  

8

  Total 
  Common Stock  Accumulated Other        Equity Attributable       
  Number  Par Value  Additional Paid-in Capital  Comprehensive Income
(loss)
  Treasury Shares  Accumulated Deficit  

to

Orgenesis
Inc.

  

Non- Controlling

Interest

  Par Value 
BALANCE AT JANUARY 1, 2020  16,140,962  $2  $94,691  $213  $-  $(89,429) $5,477  $601  $6,078 
Changes during the Year ended  December 31, 2020:                                    
Stock-based compensation to employees and directors  -   -   1,470   -   -   -   1,470   -   1,470 
Stock-based compensation to
service providers
  **270,174   1   1,376   -   -   -   1,377   -   1,377 
Stock-based compensation for Tamir purchase agreement (See Note 4)  3,400,000   *-   17,748   -   -   -   17,748   -   17,748 
Exercise of options  83,334   *-   300   -   -   -   300   -   300 
Beneficial conversion
feature of convertible loans
  -   -   42   -   -   -   42   -   42 
Issuance of shares and warrants  2,200,000   -   8,438   -   -   -   8,438   -   8,438 
Issuance of shares related to acquisition of Koligo  2,128,623   *-   11,172   -   -   -   11,172   -   11,172 
Sale of subsidiaries  -   -   -   -   -   -   -   (413)  (413)
Adjustment to redemption value of redeemable non-controlling interest  -   -   5,160   -   -   -   5,160   -   5,160 
Repurchase of treasury stock  (55,309)  -   -   -   (250)  -   (250)  -   (250)
Comprehensive income (loss) for the period  -   -   -   535   -   1,110   1,645   (39)  1,606 
BALANCE AT DECEMBER 31, 2020  24,167,784  $3  $140,397  $748  $(250) $(88,319) $52,579  $149  $52,728 

*Represents an amount lower than $1 thousand
CONSOLIDATED STATEMENTS OF CHANGES IN EQUITY
(U.S. Dollars, in thousands, except share amounts)**out of which 30,000 shares have additional restrictions on transfer until services have been provided.

  Common Stock                      
           Receipts on  Accumulated        Non-    
        Additional  Account of  Other     Equity  Controlling     
     Par  Paid-in  Share to be  Comprehensive  Accumulated  Attributable to       
  Number  Value  Capital  Allotted  Income (loss)  Deficit  Orgenesis Inc.  Interest  Total 
BALANCE AT DECEMBER1, 2016 9,508,068 $ 1 $ 45,454 $ - $ (1,205)$ (31,753)$ 12,497 $ - $ 12,497 
Changes during the Yearended November 30, 2017:                  
Stock-based compensation to employees and directors     1,536        1,536    1,536 
Stock-based compensation to service providers 79,167  *  1,828        1,828    1,828 
Beneficial conversion feature of convertible loans and warrants issued     2,814        2,814    2,814 
Issuances of shares and warrants from equity investments and Issuance, cancellation of contingent shares, and receipts on account of shares and warrants to be allotted 285,424  *  3,702  1,483      5,185    5,185 
Comprehensive income (loss) for the year         2,630  (12,367) (9,737)   (9,737)
                            
BALANCE AT NOVEMBER30, 2017 9,872,659 $ 1 $ 55,334 $ 1,483 $ 1,425 $ (44,120)$ 14,123 $ - $ 14,123 

F-7



ORGENESIS INC.
CONSOLIDATED STATEMENTS OF CHANGES IN EQUITY
(U.S. Dollars, in thousands, except share amounts)

           Receipts on  Accumulated     Equity       
        Additional  Account of  Other     Attributable  Non-    
     Par  Paid-in  Share to be  Comprehensive  Accumulated  to Orgenesis  Controlling    
  Number  Value  Capital  Allotted  Income (loss)  Deficit  Inc.  Interest  Total 
BALANCE ATDECEMBER 1, 2017 9,872,659 $ 1 $ 55,334 $ 1,483 $ 1,425 $ (44,120)$ 14,123 $ - $ 14,123 
Changes during the Yearended November 30, 2018:                  
Stock-based compensation to employees and directors     2,426        2,426    2,426 
Stock-based compensation to service providers 315,198  *  1,938        1,938    1,938 
Issuance of shares and warrants due to conversion of convertible loans and shares in escrow account 1,486,722  *  7,511        7,511    7,511 
Issuance of shares related to acquisition of Atvio and CureCell 286,811  *  2,452        2,452  299  2,751 
Issuance of warrants and Beneficial conversion feature of convertible loans     438        438    438 
Issuance of shares and warrants and receipts on account of shares to be allotted 2,853,747  *  18,021  770      18,791    18,791 
Issuance of shares due to exercise of warrants 136,646    846        846    846 
Adjustment to redemption value of redeemable non-controlling interest     (884)         (884)   (884)
                       
Comprehensive loss for the year         (1,000) (18,291) (19,291)   (19,291)
                            
BALANCE ATNOVEMBER 30, 2018 14,951,783 $ 1 $ 88,082 $ 2,253 $ 425 $ (62,411)$ 28,350 $ 299 $ 28,649 

*Represents an amount lower than $ 1 thousand

The accompanying notes are an integral part of these consolidated financial statements.

F-8



ORGENESIS INC.
CONSOLIDATED STATEMENTS OF CASH FLOWS
(U.S. Dollars, in thousands)F-7

  Year ended November 30, 
  2018  2017 
CASH FLOWS FROM OPERATING ACTIVITIES:      
     Net loss$ (19,104)$ (12,367)
     Adjustments required to reconcile net loss to net cash used in operating activities:      
         Stock-based compensation 4,364  3,364 
         Share in losses of associated company 731  1,214 
         Depreciation and amortization expenses 2,624  2,598 
Net gain on remeasurement of previously equity interest in Atvio and CureCell to acquisition date at fair value (4,509) - 
         Change in fair value of warrants and embedded derivatives 26  (826)
         Change in fair value of convertible bonds    (192)
         Interest expense accrued on loans and convertible loans (including amortization of beneficial conversion feature) 2,564  1,110 
         Increase (decrease) in deferred taxes 1,982  (1,310)
     Changes in operating assets and liabilities:      
         Decrease (increase) in accounts receivable, net (2,901) 33 
         Increase in inventory (931) (265)
         Increase in other assets (19) (3)
         Decrease (increase) in prepaid expenses, other accounts receivable and GPP receivable 380  (107)
         Decrease in related parties, net (532) (583)
         Decrease in accounts payable (796) (933)
         Increase in accrued expenses 428  92 
         Increase (decrease) in employee and related payables (105) 1,142 
         Increase in deferred income 1,309  1,044 
         Increase (decrease) in advance payments and receivables on account of grant (193) 2,156 
               Net cash used in operating activities (15,682) (3,833)
CASH FLOWS FROM INVESTING ACTIVITIES:      
   Purchase of property and equipment (5,556) (975)
   Investments in associates -  (2,429)
   Long-term bank deposits (15) - 
   Increase in loan to JV with a related party (1,000) - 
   Acquisition of CureCell, net of cash acquired (see Note 4) 58  - 
   Acquisition of Atvio, net of cash acquired (see Note 4) 245  - 
           Net cash used in investing activities (6,268) (3,404)
CASH FLOWS FROM FINANCING ACTIVITIES:      
     Short-term line of credit -  (21)
     Proceeds from issuance of shares and warrants (net of transaction costs) 17,392  5,297 
     Redeemable non-controlling interest 14,058  2,349 
     Proceeds from receipts on account of shares to be allotted 2,252  - 
     Repayment of short and long-term debt (377) (1,108)
     Repayment of convertible loans and convertible bonds (177) (4,051)
     Proceeds from issuance of convertible loans (net of transaction costs) 1,912  5,899 
             Net cash provided by financing activities 35,060  8,365 
NET CHANGE IN CASH AND CASH EQUIVALENTS 13,110  1,128 
EFFECT OF EXCHANGE RATE CHANGES ON CASH ANDCASH EQUIVALENTS (173) 1,497 
CASH AND CASH EQUIVALENTS AT BEGINNING OF YEAR 3,519  891 
CASH, CASH EQUIVALENTS AND RESTRICTED CASH AT END OF YEAR$ 16,456 $ 3,519 
SUPPLEMENTAL NON-CASH FINANCING ACTIVITY      
Conversion of principal amount and accrued interest of convertible loans and bonds to common stock and warrants$ 7,511 $ 1,277 
Classification of loan receivable into services to be received from CureCell$836  - 
Leases of Fixed assets$ 955  - 
Receivable from GPP$6,600  - 
SUPPLEMENTAL INFORMATION ON INTEREST PAID IN CASH - $ 903 

ORGENESIS INC.

CONSOLIDATED STATEMENTS OF CASH FLOWS(*)

(U.S. Dollars, in thousands)

  2020  2019 
  Year ended December 31, 
  2020  2019 
CASH FLOWS FROM OPERATING ACTIVITIES:        
Net income (loss) $579  $(26,041)
Adjustments required to reconcile net income (loss) to net cash used in operating activities:        
Stock-based compensation  2,847   3,057 
Stock-based compensation for strategic collaborations  -   2,641 
Stock-based compensation for Tamir Purchase Agreement (See Notes 4)  17,048   - 
Capital loss (gain), net  22   (29)
Gain on disposal of subsidiaries  (96,918)  - 
Share in income of associated company  (106)  - 
Depreciation and amortization expenses  1,435   3,806 
Effect of exchange differences on inter-company balances  (618)  214 
Net changes in operating leases  14   (339)
Interest expense accrued on loans and convertible loans (including amortization of beneficial conversion feature)  927   387 
     Changes in operating assets and liabilities:        
Increase in accounts receivable  (1,350)  (5,308)
Increase in inventory  (84)  (414)
Increase in other assets  (24)  (46)
Increase in prepaid expenses, other accounts receivable  (1,073)  (112)
Increase in accounts payable  1,985   4,626 
Increase (decrease) in accrued expenses and other payable  (1,156)  271 
Increase (decrease) in employee and related payables  (170)  474 
Increase (decrease) in contract liabilities  (166)  3,536 
Change in advance payments and receivables on account of grant, net  140   (247)
Increase (decrease) in deferred taxes  (1,378)  304 
Net cash used in operating activities $(78,046) $(13,220)
CASH FLOWS FROM INVESTING ACTIVITIES:        
Increase in loan to JV partner, a related party  (500)  (1,500)
Repayment in loan to JV partner, a related party  3,000   - 
Sale of property, plants and equipment  7   79 
Purchase of property, plants and equipment  (1,525)  (12,129)
Acquisition of Koligo, net of cash acquired (See Note 4)  (955)  - 
Proceed from sale of subsidiaries, net  105,634   - 
Investment in associated company  (69)  - 
Repayment (investment) in short term deposits  18   (228)
Net cash provided by (used) in investing activities $105,610  $(13,778)
CASH FLOWS FROM FINANCING ACTIVITIES:        
Repurchase of treasury stock  (250)  - 
Increase in redeemable non-controlling interests received from GPP  -   13,200 
Proceeds from issuance of shares, warrants and exercise of options (net of transaction costs)  8,738   - 
Proceeds from issuance of convertible loans (net of transaction costs)  250   11,400 
Repayment of convertible loans and convertible bonds  (2,400)  - 
Repayment of short and long-term debt  (457)  (772)
Proceeds from issuance of loans payable  -   270 
Net cash provided by financing activities $5,881  $24,098 
NET CHANGE IN CASH AND CASH EQUIVALENTS AND RESTRICTED CASH  33,445   (2,900)
EFFECT OF EXCHANGE RATE CHANGES ON CASH AND CASH EQUIVALENTS $82  $(58)
CASH AND CASH EQUIVALENTS AND RESTRICTED CASH AT BEGINNING OF YEAR $12,041  $14,999 
CASH, CASH EQUIVALENTS AND RESTRICTED CASH AT END OF YEAR $45,568  $12,041 
         
SUPPLEMENTAL DISCLOSURE OF CASH FLOW TRANSACTIONS:        
Interest paid in cash during the year $-  $157 
Income taxes, net of refunds paid in cash during the year $-  $156 
         
SUPPLEMENTAL NON-CASH FINANCING AND INVESTING ACTIVITIES        
Finance Leases of property, plant and equipment $366  $355 
Right-of-use assets acquired in exchange for right-of-use liabilities $967  $8,229 
Purchase of property, plant and equipment included in accounts payable $241  $1,584 
Transaction costs of issuance of convertible loans $-  $546 
Acquisition of other asset in exchange for common stocks $700  $- 
Issuance of common stocks in connection with the acquisition of Koligo $11,172  $- 

(*)See Note 3 for information regarding the discontinued operations.

The accompanying notes are an integral part of these consolidated financial statements.

F-9



ORGENESIS INC.
NOTES TO CONSOLIDATED FINANCIAL STATEMENTS
FOR THE FISCAL YEARS ENDED NOVEMBER 30, 2018 AND 2017F-8

ORGENESIS INC.

NOTES TO CONSOLIDATED FINANCIAL STATEMENTS

NOTE 1 – DESCRIPTION OF BUSINESS

a.a. General

Orgenesis Inc., a Nevada corporation, (the “Company”), is a biotechnologyglobal biotech company specializingworking to unlock the potential of cell and gene therapies in an affordable and accessible format (“CGTs”).

CGTs can be centered on autologous (using the patient’s own cells) or allogenic (using master banked donor cells) and are part of a class of medicines referred to as advanced therapy medicinal products (ATMP). The Company mostly focusses on autologous therapies, with processes and systems that are developed for each therapy using a closed and automated processing system approach that is validated for compliant production near the patient at their point of care for treatment of the patient. This approach has the potential to overcome the limitations of traditional commercial manufacturing methods that do not translate well to commercial production of advanced therapies due to their cost prohibitive nature and complex logistics to deliver the treatments to patients (ultimately limiting the number of patients that can have access to, or can afford, these therapies).

To achieve these goals, the Company has developed a Point of Care Platform comprised of three enabling components: a pipeline of licensed POCare Therapies that are designed to be processed and produced in closed, automated POCare Technology systems across a collaborative POCare Network. Via a combination of science, technology, engineering, and networking, the Company is working to provide a more efficient and scalable pathway for advanced therapies to reach patients more rapidly at lowered costs. The Company also draws on extensive medical expertise to identify promising new autologous therapies to leverage within the POCare Platform either via ownership or licensing.

The POCare Network brings together patients, doctors, industry partners, research institutes and hospitals worldwide with a goal of achieving harmonized, regulated clinical development and production of the therapies.

Over time, the Company has worked to develop and validate POCare Technologies that can be combined within mobile production units for advanced therapies. In 2020, the Company made significant investments in the development manufacturingof several types of Orgenesis Mobile Processing Units and provisionLabs (OMPULs) with the expectation of servicesuse and/or distribution through our POCare Network of partners, collaborators, and joint ventures. As of the date of this report, the OMPULs are still in the development stage.

OMPULs are designed for the purpose of validation, development, performance of clinical trials, manufacturing and/or processing of potential or approved cell and gene therapy industry.products in a safe, reliable, and cost-effective manner at the point of care, as well as the manufacturing of such CGTs in a consistent and standardized manner in all locations. The design delivers a potential industrial solution for the Company operates throughto deliver CGTs to practically any clinical institution at the point of care.

Until December 31, 2019, the Company operated the POCare Platform as one of two platforms:(i) a point-of-care (“POCare”) cell therapy (“PT”) platform and (ii)business separate business segments.

Historically, the second separate business segment was operated as a Contract Development and Manufacturing Organization (“CDMO”) platform, conducted through its subsidiary, Masthercell Global Inc., a Delaware corporation. Through the PT business, the Company’s aim is to further the development of Advanced Therapy Medicinal Products (“ATMPs”) through collaborations and in-licensing with other pre-clinical and clinical-stage biopharmaceutical companies and research and healthcare institutes to bring such ATMPs to patients. The Company out-license these ATMPs through regional partners to whom the Company also provide regulatory, pre-clinical and training services to support their activity in order to reach patients in a point-of-care hospital setting. Through the CDMO platform, the Company is focused on providing contract manufacturing and development services for biopharmaceutical companies.

     Activities in the PT business include a multitude of cell therapies, including autoimmune, oncologic, neurologic and metabolic diseases and other indications. The Company provides services for its joint venture (“JV”) partners, pharmaceutical and biotech companies as well as research institutions and hospitals that have cell therapies in clinical development. Each of these customers and collaborations represents a revenue and growth opportunity upon regulatory approval. Furthermore, the Company’s trans-differentiation technology demonstrates the capacity to induce a shift in the developmental fate of cells from the liver or other tissues and transdifferentiating them into “pancreatic beta cell-like” Autologous Insulin Producing (“AIP”) cells for patients with Type 1 Diabetes, acute pancreatitis and other insulin deficient diseases. This technology, which has yet to be proven in human clinical trials, has shown in pre-clinical animal models that the human derived AIP cells produce insulin in a glucose-sensitive manner. This trans-differentiation technology is licensed by the Orgenesis Ltd (the “Israeli Subsidiary) and is based on the work of Prof. Sarah Ferber, the Company’s Chief Science Officer and a researcher at Tel Hashomer Medical Research Infrastructure and Services Ltd. (“THM”“CDMO Business”) in Israel. The development plan calls for conducting additional pre-clinical safety and efficacy studies with respect to diabetes and other potential indications prior to initiating human clinical trials.

. The CDMO platform operateswas historically operated mainly through majority owned Masthercell Global Inc. (“ Masthercell Global”), which currently consists(which consisted of the following two subsidiaries: MaSTherCell S.AS.A. in Belgium (“MaSTherCell”) in Belgium, Atvio Biotech Ltd. (“Atvio”) in Israel, CureCell Co., Ltd. (“CureCell”) in South Korea and Masthercell U.S., LLC in the United States (“Masthercell U.S.”) (collectively, the “ Masthercell“Masthercell Global Subsidiaries”)).

In February 2020, the Company and GPP-II Masthercell LLC (“GPP”) sold 100% of the outstanding equity interests of Masthercell (the “Masthercell Business”), having unique know-howwhich comprised the majority of the Company’s CDMO Business, to Catalent Pharma Solutions, Inc. for an aggregate nominal purchase price of $315 million, (the “Masthercell Sale”). After accounting for GPP’s liquidation preference and expertise for manufacturingequity stake in Masthercell as well as other investor interests in our Belgian subsidiary MaSTherCell, distributions to Masthercell option holders and transaction costs, the company received approximately $126.7 million. The Company incurred an additional approximately $5.6 million in transaction costs.

F-9

The Company determined that the Masthercell Business (“Discontinued Operation”) meets the criteria to be classified as a multitude of cell types. Masthercell Global strives to provide services that are all compliant with GMP requirements, ensuring identity, purity, stability, potency and robustness of cell therapy products for clinical phase I, II, III and through commercialization. (Masthercell U.S. LLC was incorporated in June 2018 and had no activitydiscontinued operation as of November 30, 2018).

the first quarter of 2020. The Discontinued Operation includes the vast majority of the previous CDMO Business, including majority-owned Masthercell, including MaSTherCell, Global, throughMasthercell U.S. and all of the Masthercell Global Subsidiaries, isSubsidiaries.

Since the Masthercell Sale, the Company has entered into new joint venture agreements with new partners in various jurisdictions. This has allowed the Company to grow its infrastructure and expand its processing sites into new markets and jurisdictions. In addition, the Company has engaged in the businesssome of providing manufacturingthese joint venture partners to perform research and development services to third parties relatedfurther develop and adapt its systems and devices for specific purposes. The Company has been investing manpower and financial resources to cell therapy products, and the creation and development of technology, and optimizations in connection with suchfocus on developing, manufacturing and development services for third parties (the “CDMO Business”).rolling out several types of OMPULs to be used and/or distributed through our POCare Network of partners, collaborators, and joint ventures.

The Chief Executive Officer (“CEO”) is the Company’s chief operating decision-maker who reviews financial information prepared on a consolidated basis. Effective from the first quarter of 2020, all of our continuing operations are in one segment, being the point-of-care business via our POCare Platform. Therefore, no segment report has been presented.

The Company operatescurrently conducts its CDMOcore CGT business operations through itself and PT businessits subsidiaries which are all wholly-owned except as two separate business segments.otherwise stated (collectively, the “Subsidiaries”). The Subsidiaries are as follows:

United States: Orgenesis Maryland Inc. (the “U.S. Subsidiary”) is the center of activity in North America currently focused on setting up of the POCare Network.
Koligo Therapeutics Inc. (“Koligo”) is a Kentucky corporation that was acquired in 2020 and is currently focused on developing the POCare network and therapies (See Note 4 for the acquisition of Koligo).
European Union: Orgenesis Belgium SRL (the “Belgian Subsidiary”) is the center of activity in Europe currently focused on process development and preparation of European clinical trials.
Orgenesis Switzerland Sarl (the “Swiss subsidiary) incorporated in October 2020 is currently focused on providing management services to the Company.
Israel: Orgenesis Ltd. (the “Israeli Subsidiary”) is a provider of regulatory, clinical and pre-clinical services, and Orgenesis Biotech Israel Ltd. (“OBI”) previously known as Atvio Biotech Ltd. (“Atvio”) is a provider of cell-processing services in Israel.
Korea: Orgenesis Korea Co. Ltd. (the “Korean Subsidiary”), previously known as CureCell Co. Ltd., is a provider of processing and pre-clinical services in Korea. The Company owns 94.12% of the Korean Subsidiary.

These consolidated financial statements include the accounts of Orgenesis Inc. and its subsidiaries including thosethe Discontinued Operation.

On April 7, 2020, the Company entered into an Asset Purchase Agreement (the “Tamir Purchase Agreement”) with Tamir Biotechnology, Inc. (“Tamir” or “Seller”), pursuant to which the Company agreed to acquire certain assets and liabilities of Tamir related to the Masthercell Global Subsidiaries; Orgenesis SPRL (the “Belgian Subsidiary”), a Belgian-based subsidiary which is engaged indiscovery, development and manufacturing activities together with clinical development studiestesting of therapeutic products for the treatment of diseases and conditions in Europe; Orgenesis Maryland Inc.humans, including all rights to Ranpirnase and use for antiviral therapy (collectively, the “Purchased Assets and Assumed Liabilities” and such acquisition, the “Tamir Transaction”). The Tamir Transaction closed on April 23, 2020. As aggregate consideration for the acquisition, the Company paid $2.5 million in cash and issued an aggregate of 3,400,000 shares (the “U.S. Subsidiary”“Shares”), of Common Stock to Tamir resulting in a Maryland corporation;total consideration of $20.2 million (See Note 4).

F-10

The Company’s common stock, par value $0.0001 per share (the “Common Stock”) is listed and Orgenesis Ltd., an Israeli corporation, (the “Israeli Subsidiary”).traded on the Nasdaq Capital Market under the symbol “ORGS.”

F-10


As used in this report and unless otherwise indicated, the term “Company” refers to Orgenesis Inc. and its subsidiaries (“Subsidiaries”).Subsidiaries. Unless otherwise specified, all amounts are expressed in United States Dollars.

     Until March 13, 2018, the Company’s common shares were traded on OTC Market Group’s OTCQB, at which point the Company's common stock began to be listed and traded on the Nasdaq Capital Market under the symbol “ORGS”.

b.Liquidity

b. Change in Fiscal Year End

     On October 22, 2018, the BoardAs of Directors of the Company approved a change in the Company’s fiscal year end from November 30 to December 31, of each year. This change to the calendar year reporting cycle began January 1, 2019. As a result of the change, the Company will have a December 2018 fiscal month transition period, the results of which will be separately reported in the Company’s Quarterly Report on Form 10-Q for the calendar quarter ending March 31, 2019, June 30, 2019, September 30, 2019 and in the Company’s Annual Report on Form 10-K for the calendar year ending December 31, 2019.

c. Liquidity

     As of November 30, 2018, the2020 ,the Company has accumulated losses of approximately $62.4 million. Although the Company is showing positive revenues and gross profit trends in the CDMO platform, the Company expects to incur further losses in the PT business.$88 Million.

     To date, the Company has been funding operations primarily from the proceeds from private placements of the Company’s equity securities and convertible debt and from revenues generated by Masthercell Global, mainly revenues generated from MaSTherCell S.A. in Belgium. From December 1, 2017 through November 30, 2018,

On February 10, 2020, the Company received through MaSTherCell,approximately $126.7 million, of which $7.2 million was used for the repayment of intercompany loans and payables, from the Masthercell Sale (See Note 3). In addition, on January 20, 2020, the Company entered into a Securities Purchase Agreement with certain investors pursuant to which the Company received gross proceeds of approximately $17.3 $9.24 million before deducting related offering expenses.

The Company invested significant resources in research and development and research and development services in 2020. The Company believes that these investments will enable it to substantially increase revenues and accounts receivable from customers, $12.6 million from SFPI and GPP (See also Note 3) and $21.5 million fromin the private placement to accredited investors of the Company's equity and convertible loans net of finders’ fees, convertible loans and exercise of warrants. In addition, from December 1, 2018 through February 13, 2019, the Company raised $0.25 million from the private placement referred to above of unsubscribed units under such investor’s subscription agreement, $6.6 million from GPP (See Note 3(b)) and proceeds of approximately $4.7 million in accounts receivable from customers of MaSTherCell.

next 12 months. Based on its current cash resources and commitments, the Company believes it will be able to maintain its current planned development activityactivities and correspondingexpected level of expenditures for at least 12 months from the date of the issuance of thethese financial statements, although no assurance can be given that it will not need additional funds prior to such time.statements. If there are unexpectedfurther increases in general and administrative expenses oroperating costs for facilities expansion, research and development, expenses,commercial and clinical activity or decreases in MaSTherCell's income,revenues from customers, the Company will needmay decide to seek additional financing.

NOTE 2 - SIGNIFICANT ACCOUNTING POLICIES

The consolidated financial statements are prepared in accordance with accounting policies adopted areprinciples generally consistent with those ofaccepted in the previous financial year.United States (“U.S. GAAP”).

a.Use of Estimates in the Preparation of Financial Statements

The preparation of theour consolidated financial statements in conformity with U.S. GAAP requires managementus to make estimates, judgments and assumptions that may affect the reported amounts of assets, liabilities, equity, revenues and liabilitiesexpenses and related disclosure of contingent assets and liabilities. On an ongoing basis, we evaluate our estimates, judgments and methodologies. We base our estimates on historical experience and on various other assumptions that we believe are reasonable, the results of which form the basis for making judgments about the carrying values of assets, liabilities atand equity, the financial statement dateamount of revenues and the reported expenses during the reporting periods.and determining whether an acquisition is a business combination or a purchase of asset. Actual results could differ from those estimates.

The full extent to which the COVID-19 pandemic may directly or indirectly impact our business, results of operations and financial condition, will depend on future developments that are uncertain, including as a result of new information that may emerge concerning COVID-19 and the actions taken to contain it or treat COVID-19, as well as the economic impact on local, regional, national and international customers and markets. We examined the impact of COVID-19 on our financial statements, and although there is currently no major impact, there may be changes to those estimates in future periods. Actual results may differ from these estimates.

b.Business Combination

The Company allocates the purchase price of an acquired business to the tangible and intangible assets acquired and liabilities assumed based upon their estimated fair values on the acquisition date. Any excess of the purchase price over the fair value of the net assets acquired is recorded as goodwill. Acquired in-process backlog, customer relations, technology, IPR&D, brand name and know how are recognized at fair value. The purchase price allocation process requires management to make significant estimates and assumptions, especially at the acquisition date with respect to intangible assets. Direct transaction costs associated with the business combination are expensed as incurred. The allocation of the consideration transferred in certain cases may be subject to revision based on the final determination of fair values during the measurement period, which may be up to one year from the acquisition date. The Company includes the results of operations of the business that it has acquired in its consolidated results prospectively from the date of acquisition.

F-11


F-11

 

If the business combination is achieved in stages, the acquisition date carrying value of the acquirer’s previously held equity interest in the acquire is re-measured to fair value at the acquisition date; any gains or losses arising from such re-measurement are recognized in profit or loss.

c.Other Investments

For other investments, the Company applies the measurement alternative upon the adoption of ASU 2016-01, and elected to record equity investments without readily determinable fair values at cost, less impairment, adjusted for subsequent observable price changes. In this measurement alternative method, changes in the carrying value of the equity investments are reflected in current earnings. Changes in the carrying value of the equity investment are required to be made whenever there are observable price changes in orderly transactions for the identical or similar investment of the same issuer.

d.Discontinued operations

Upon divestiture of a business, the Company classifies such business as a discontinued operation, if the divested business represents a strategic shift that has (or will have) a major effect on an entity’s operations and financial results. For disposals other than by sale such as abandonment, the results of operations of a business would not be recorded as a discontinued operation until the period in which the business is actually abandoned.

The Masthercell Business divestiture qualifies as a discontinued operation and therefore has been presented as such.

The results of businesses that have qualified as a discontinued operation have been presented as such for all reporting periods. Results of discontinued operations include all revenues and expenses directly derived from such businesses; general corporate overhead is not allocated to discontinued operations. Any loss or gain that arose from the divestiture of a business that qualifies as discontinued operations is included within the results of the discontinued operations. The Company included information regarding cash flows from discontinued operations (See Note 3).

e.Cash Equivalents

The Company considers cash equivalents to be all short term,short-term, highly liquid investments, which include short termmoney market instruments, that are not restricted as to withdrawal or use, and short-term bank deposits with original maturities of three months or less from the date of purchase that are not restricted as to withdrawal or use and are readily convertible to known amounts of cash, to be cash equivalents.cash.

d. Bank Deposits

     Bank deposits with maturityf.Cost of more than one year are considered long-term. The fair value of bank deposits approximates the carrying value since they bear interest at rates close to the prevailing market rates.

e. Research and Development, net

     Researchresearch and development expensesand research and development services, net

Cost of research and development and research and development services include costs directly attributable to the conduct of research and development programs,activities, including the cost of salaries, stock-based compensation expenses, payroll taxes and other employees'employees’ benefits, lab expenses, consumable equipment, courier fees, travel expenses, professional fees and consulting fees. All costs associated with research and developments are expensed as incurred. Participation from government departments and from research foundations for development of approved projects is recognized as a reduction of expense as the related costs are incurred. Research and development in-process acquired as part of an asset purchase, which has not reached technological feasibility and has no alternative future use, is expensed as incurred.

f. g.Principles of Consolidation

The consolidated financial statements include the accounts of the Company and its Subsidiaries. All intercompany transactions and balances have been eliminated in consolidation.

F-12

g. h.Non-Marketable Equity Investments

The Company’s investments in certain non-marketable equity securities in which it has the ability to exercise significant influence, but it does not control through variable interests or voting interest.interests. These are accounted for under the equity method of accounting and presented as Investment in associates, net, in the Company’s consolidated balance sheets. Under the equity method, the Company recognizes its proportionate share of the comprehensive income or loss of the investee. The Company’s share of income and losses from equity method investments is included in share in losses of associated company.

The Company reviews its investments accounted for under the equity method for possible impairment, which generally involves an analysis of the facts and changes in circumstances influencing the investments.

h.

i.Functional Currency

The currency of the primary economic environment in which the operations of the Company and part of its Subsidiaries are conducted is the U.S. dollar (“$” or “dollar”). The functional currency of the Belgian Subsidiaries is the Euro (“€” or “Euro”). The functional currency of CureCellOrgenesis Korea is the Won (“KRW”). Most of the Company’s expenses are incurred in dollars, and the source of the Company’s financing has been provided in dollars. Thus, the functional currency of the Company and its other subsidiaries is the dollar. Transactions and balances originally denominated in dollars are presented at their original amounts. Balances in foreign currencies are translated into dollars using historical and current exchange rates for nonmonetary and monetary balances, respectively. For foreign transactions and other items reflected in the statements of operations, the following exchange rates are used: (1) for transactions – exchange rates at transaction dates or average rates and (2) for other items (derived from nonmonetary balance sheet items such as depreciation) – historical exchange rates. The resulting transaction gains or losses are recorded as financial income or expenses. The financial statements of the Belgian Subsidiaries and CureCellOrgenesis Korea are included in the consolidated financial statements, translated into U.S. dollars. Assets and liabilities are translated at year-end exchange rates, while revenues and expenses are translated at yearly average exchange rates during the year. Differences resulting from translation of assets and liabilities are presented as other comprehensive income.

F-12


i. Inventory

j.Inventory

The Company’s inventory consists of raw material for use for the services provided. The Company periodically evaluates the quantities on year end.hand. Cost of the raw materials is determined using the weighted average cost method. The inventory is recorded at the lower of cost or net realizable value.

j.

k.Property, plant and Equipment

Property, plant and equipment are recorded at cost and depreciated by the straight-line method over the estimated useful lives of the related assets.

Annual rates of depreciation are presented in the table below:

SCHEDULE OF ANNUAL DEPRECIATION RATES, PROPERTY AND EQUIPMENT

 

Weighted Average

Useful Life (Years)

Production facility5-205 - 10
Laboratory equipment52 - 7
Office equipment and computers3-53 - 17

l.Intangible assets

Intangible assets and their useful lives are as follows:

SCHEDULE OF INTANGIBLE ASSETS AND THEIR USEFUL LIVE

 Weighted AverageAmortization Recorded at
 Useful Life (Years)

Amortization Recorded at Comprehensive

Loss Line Item

Customer Relationships2.5-10Amortization of intangible assets
Brand109.75Amortization of intangible assets
Know-How11.75-1212Amortization of intangible assets
BacklogTechnology1.515Amortization of intangible assets

F-13

Intangible assets are recorded at acquisition cost less accumulated amortization and impairment. Definite lived intangible assets are amortized over their estimated useful life using the straight-line method, which is determined by identifying the period over which the cash flows from the asset are expected to be generated.

k. Goodwill

m.Goodwill

Goodwill represents the excess of the purchase price of acquired businessconsideration transferred over the estimated fair value assigned to the net tangible and identifiable intangible assets of businesses acquired. Goodwill is allocated to reporting units expected to benefit from the identifiable net assets acquired.business combination. Goodwill is not amortized but israther tested for impairment at least annually (at November 30), atin the reporting unit levelfourth quarter, or more frequently if events or changes in circumstances indicate that the asset mightgoodwill may be impaired. The goodwillFollowing the sale of Masthercell the Company manages the business as one operating segment and one reporting unit. Goodwill impairment test is applied by performing a qualitativerecognized when the quantitative assessment before calculatingresults in the carrying value exceeding the fair value, ofin which case an impairment charge is recorded to the reporting unit. If, on the basis of qualitative factors, it is considered not more likely than not that the fair value of the reporting unit is less than the carrying amount, further testing of goodwill for impairment would not be required. Otherwise, goodwill impairment is tested using a two-step approach.

F-13


  The first step involves comparing the fair value of the reporting unit to its carrying amount. If the fair value of the reporting unit is determined to be greater than its carrying amount, there is no impairment. If the reporting unit's carrying amount is determined to be greater than the fair value, the second step must be completed to measure the amount of impairment, if any. The second step involves calculating the implied fair value of goodwill by deducting the fair value of all tangible and intangible assets, excluding goodwill, of the reporting unit from the fair value of the reporting unit as determined in step one. The implied fair value of the goodwill in this step is compared toextent the carrying value of goodwill. Ifexceeds the implied fair value of the goodwill is less than the carrying value of the goodwill, an impairment loss equivalent to the difference is recorded. value.

There were no 0impairment charges in 2018 and 2017.to goodwill during the periods presented.

l. n.Impairment of Long-lived Assets

The Company reviews its property, plants and equipment, intangible assets subject to amortization and other long-lived assets for impairment whenever events or changes in circumstances indicate that the carrying amount of an asset class may not be recoverable. Indicators of potential impairment include: an adverse change in legal factors or in the business climate that could affect the value of the asset; an adverse change in the extent or manner in which the asset is used or is expected to be used, or in its physical condition; and current or forecasted operating or cash flow losses that demonstrate continuing losses associated with the use of the asset. If indicators of impairment are present, the asset is tested for recoverability by comparing the carrying value of the asset to the related estimated undiscounted future cash flows expected to be derived from the asset. If the expected cash flows are less than the carrying value of the asset, then the asset is considered to be impaired and its carrying value is written down to fair value, based on the related estimated discounted cash flows. There were no0 impairment charges in 2018the year ended December 31, 2020 and 2017.2019.

m. Revenue Recognition

     The Company recognizes revenue for services linked to cell process development and cell manufacturing services based on individual contracts in accordance with Accounting Standards Codification (“ASC”) 605,Revenue Recognition,when the following criteria have been met: persuasive evidence of an arrangement exists; delivery of the processed cells has occurred or the services that are milestones based have been provided; the price is fixed or determinable and collectability is reasonably assured. The Company determines that persuasive evidence of an arrangement exists based on written contracts that define the terms of the arrangements. In addition, the Company determines that services have been delivered in accordance with the arrangement. The Company assesses whether the fee is fixed or determinable based on the payment terms associated with the transaction and whether the sales price is subject to refund or adjustment. Service revenues are recognized as the services are provided. In addition, as part of the services, the Company recognizes revenue based on use of consumables, which it received as reimbursement on a cost-plus basis on certain expenses.

n. Financial Liabilities Measured at Fair Value

1)Fair Value Option

     Topic 815 provides entities with an option to report certain financial assets and liabilities at fair value with subsequent changes in fair value reported in earnings. The election can be applied on an instrument by instrument basis. The Company elected the fair value option to its convertible bonds. The liability is measured both initially and in subsequent periods at fair value, with changes in fair value charged to finance expenses, net (See Note 17).

2)Derivatives

     Embedded derivatives are separated from the host contract and carried at fair value when (1) the embedded derivative possesses economic characteristics that are not clearly and closely related to the economic characteristics of the host contract and (2) a separate, standalone instrument with the same terms would qualify as a derivative instrument. The derivative is measured both initially and in subsequent periods at fair value, with changes in fair value charged to finance expenses, net. As to embedded derivatives arising from the issuance of convertible debentures (See Note 17).

o.Income Taxes

1) With respect to deferred taxes, income taxes are computed using the asset and liability method. Under the asset and liability method, deferred income tax assets and liabilities are determined based on the differences between the financial reporting and tax bases of assets and liabilities and are measured using the currently enacted tax rates and laws. A valuation allowance is recognized to the extent that it is more likely than not that the deferred taxes will not be realized in the foreseeable future.

F-14


2) The Company follows a two-step approach to recognizing and measuring uncertain tax positions. The first step is to evaluate the tax position for recognition by determining if the available evidence indicates that it is more likely than not that the position will be sustained on examination. If this threshold is met, the second step is to measure the tax position as the largest amount that is greater than 50%50% likely of being realized upon ultimate settlement.

3) Taxes that would apply in the event of disposal of investment in Subsidiaries have not been taken into account in computing the deferred income taxes, as it is the Company’s intention to hold these investments and not realize them.

F-14

p.Stock-based Compensation

The Company accounts for employeerecognizes stock-based compensation in accordance withfor the guidance of ASC Topic 718,Compensation - Stock Compensation, which requires all share-based payments to employees, including grants of employee stock options, to be recognized in the financial statements based on their grant date fair values. Theestimated fair value of the equity instrument is charged toshare-based awards. The Company measures compensation expense for share-based awards based on estimated fair values on the date of grant using the Black-Scholes option-pricing model. This option pricing model requires estimates as to the option’s expected term and creditedthe price volatility of the underlying stock. The Company amortizes the value of share-based awards to additional paid in capitalexpense over the vesting period during which services are rendered. The Company recorded stock based compensation expenses using the straight line method.

     The Company follows ASC Topic 505-50,Equity-Based Payments to Non-Employees, for stock options issued to consultants and other non-employees. In accordance with ASC Topic 505-50, these stock options issued as compensation for services provided to the Company are accounted for based upon the fair value of the options. The fair value of the options granted is measured on a final basis at the end of the related service period and is recognized over the related service period using the straight-line method.basis.

q.Redeemable Non-controlling Interest

Non-controlling interests with embedded redemptionfeatures,redemption features, whose settlement is not at the Company’s discretion, are considered redeemable non-controlling interest. Redeemable non-controlling interests are considered to be temporary equity and are therefore presented as a mezzanine section between liabilities and equity on the Company'sCompany’s consolidated balance sheets. Subsequent adjustment of the amount presented in temporary equity is required only if the Company'sCompany’s management estimates that it is probable that the instrument will become redeemable. Adjustments of redeemable non-controlling interest to its redemption value are recorded through additional paid-in capital.

r. Loss (income) per Share of Common Stock

Basic net loss (income) per share is computed by dividing the net loss (income) for the period by the weighted average number of shares of common stock outstanding for each period. Diluted net loss (income) per share is based upon the weighted average number of common shares and of common shares equivalents outstanding when dilutive. Common share equivalents include: (i) outstanding stock options and warrants which are included under the treasury share method when dilutive, and (ii) common shares to be issued under the assumed conversion of the Company’s outstanding convertible loans and debt, which are included under the if-converted method when dilutive (See Note 14).

s.Concentration of Credit Risk

Financial instruments that potentially subject the Company to concentration of credit risk consist of principally cash and cash equivalents, bank deposits and certain receivables. The Company held these instruments with highly rated financial institutions and the Company has not experienced any significant credit losses in these accounts and does not believe the Company is exposed to any significant credit risk on these instruments apart of accounts receivable. The Company performs ongoing credit evaluations of its customers for the purpose of determining the appropriate allowance for doubtful accounts. An appropriate allowance for doubtful accounts is included in the accounts and netted against accounts receivable. In the year ended November 30, 2018December 31, 2020 the Company has not experienced any material credit losses in these accounts and does not believe it is exposed to significant credit risk on these instruments. In the year ended November 30, 2017, the Company has recorded an allowance of $897 thousand.

F-15


Bad debt allowance is created when objective evidence exists of inability to collect all sums owed it under the original terms of the debit balances. Material customer difficulties, the probability of their going bankrupt or undergoing economic reorganization and insolvency or material delays in payments are all considered indicative of reduced debtor balance value.

t.Treasury shares

The Company repurchases its ordinary shares from time to time on the open market and holds such shares as treasury stock. The Company presents the cost to repurchase treasury stock as a reduction of shareholders’ equity. During the years ended December 31, 2020, the Company repurchased 55,309shares. The Company did not reissue nor cancel treasury shares during the year ended December 31, 2020.

u.Beneficial Conversion Feature (“BCF”)

When the Company issues convertible debt, if the stock price is greater than the effective conversion price (after allocation of the total proceeds) on the measurement date, the conversion feature is considered "beneficial"“beneficial” to the holder. If there is no contingency, this difference is treated as issued equity and reduces the carrying value of the host debt; the discount is accreted as deemed interest on the debt (See Note 8)7).

F-15

u. v.Other Comprehensive Loss

Other comprehensive loss represents adjustments of foreign currency translation.

v. Newly issued and recently adopted Accounting Pronouncements

1) In January 2016, the FASB issued guidance on recognition and measurement of financial assets and financial liabilities (ASU No. 2016-01) that will supersede most current guidance. Changes to the U.S. GAAP model primarily affect the accounting for equity investments, financial liabilities under the fair value option and the presentation and disclosure requirements for financial instruments. In addition, the FASB clarified guidance related to the valuation allowance assessment when recognizing deferred tax assets resulting from unrealized losses on available-for-sale debt securities. The accounting for other financial instruments, such as loans, investments in debt securities, and financial liabilities, is largely unchanged. The classification and measurement guidance became effective as of December 1, 2018. The Company does not expect to have a material impact on its consolidated financial statements.

2) In May 2014, the FASB issued Accounting Standards Update (“ASU”) 2014-09 “Revenuew.Revenue from Contracts with Customers (Topic 606)”

The Company recognizes revenue from contracts with customers according to ASC 606, Revenue from Contracts with Customers and the related amendments (“Topic 606”New Revenue Standard”) to all contracts.

The Company’s agreements are primarily service contracts that will supersede most currentrange in duration from a few months to one year. The Company recognizes revenue recognition guidance, including industry specific guidance. Under the new standard, a good or servicewhen control of these services is transferred to the customer when (or as) the customer obtains control of the good or service, which differs from the risk and rewards approach under current guidance. The guidance provides a five-step analysis of transactionsfor an amount, referred to determine when and how revenue is recognized. Other major provisions include capitalization of certain contract costs, consideration of the time value of money inas the transaction price, and allowing estimates of variablewhich reflects the consideration to which the Company is expected to be recognized before contingencies are resolvedentitled in certain circumstances. The guidance also requires enhanced disclosures regarding the nature, amount, timing and uncertainty of revenue and cash flows arising from an entity’s contractsexchange for those goods or services.

A contract with customers. The guidance is effective in annual reporting periods beginning after December 15, 2017, including interim reporting periods within that reporting period. a customer exists only when:

the parties to the contract have approved it and are committed to perform their respective obligations;
the Company can identify each party’s rights regarding the distinct goods or services to be transferred (“performance obligations”);
the Company can determine the transaction price for the goods or services to be transferred; and
the contract has commercial substance and it is probable that the Company will collect the consideration to which it will be entitled in exchange for the goods or services that will be transferred to the customer.

The Company will implement the guidance for our annual period ending on December 31, 2019 and interim periods within such annual periods, using the modified retrospective method and willdoes not adjust the accumulated deficit and deferred revenue aspromised amount of consideration for the effects of a significant financing component since the Company expects, at contract inception, that the period between the time of transfer of the adoption date.

     Under current GAAP,promised goods or services to the Company recognizes revenuecustomer and the time the customer pays for these goods or services linked to cell process development based on both the input and output methods of measurement.be generally one year or less. The Company has evaluated the application of the requirements of ASC 606 to recognize revenue when or as the entity satisfies a performance obligation to its business. The Company has several types of revenue contracts:

a) Cell process development services

     The Company has concluded that under the revised standard, contracts for cell process development services are in some cases a single performance obligation (where promises offeredCompany’s credit terms to customers are not distinct within the contextin average between thirty and one hundred and fifty days.

Nature of the contract),Revenue Streams

The Company’s main revenue streams from continuing operation are POC development services and Cell Process Development Services.

POC Development Services

Revenue recognized under contracts for POC development services may, in other cases havesome contracts, represent multiple performance obligations (where promises to the customers are distinct) in circumstances in which the work packages are not interrelated or the customer is able to complete the services performed.

For arrangements that include multiple performance obligations, the transaction price is allocated to the identified performance obligations based on their relative standalone selling prices.

The Company recognizes revenue when, or as, it satisfies a performance obligation. At contract inception, the Company determines whether the services are transferred over time or at a point in time. Performance obligations that have no alternative use and that the Company has the right to payment for performance completed to date, at all times during the contract term, are recognized over time. All other Performance obligations are recognized as revenues by the company at point of time (upon completion).

Included in POC development services is Hospital supplies revenue which is derived principally from the sale or lease of products and the performance of services to hospitals or other medical providers. Revenue is earned and recognized when product and services are received by the customer.

 Significant Judgement and Estimates

Significant judgment is required to identifying the distinct performance obligations and estimating the standalone selling price of each distinct performance obligation, and identifying which performance obligations create assets with alternative use to the Company, which results in revenue recognized upon completion, and which performance obligations are transferred to the customer over time.

Practical Expedients

As part of ASC 606, the Company has adopted several practical expedients including the Company’s determination that it need not adjust the promised amount of consideration for the effects of a significant financing component since the Company expects, at contract inception, that the period between when the Company transfers a promised service to the customer and when the customer pays for that service will be one year or less.

F-16

Cell Process Development Services (mainly discontinued operations)

Revenue recognized under contracts for cell process development services may, in some contracts, represent multiple performance obligations (where promises to the customers are distinct) in circumstances in which the work packages and milestones are not interrelated or the customer is able to complete the services performed independently or by using competitors of the Company. In all casesother contracts when the above circumstances are not met, the promises are not considered distinct and the contract represents one performance obligation. All performance obligations are satisfied over time. Undertime, as there is no alternative use to the new standard,services it performs, since, in nature, those services are unique to the customer, which retain the ownership of the intellectual property created through the process. Additionally, due to the non-refundable upfront payment the customer pays, together with the payment term and cancellation fine, it has a right to payment (which include a reasonable margin), at all times, for work completed to date, which is enforceable by law.

For arrangements that include multiple performance obligations, the transaction price is allocated to the identified performance obligations based on their relative standalone selling prices. For these contracts, the standalone selling prices are based on the Company’s normal pricing practices when sold separately with consideration of market conditions and other factors, including customer demographics and geographic location.

The Company will recognizemeasures the revenue to be recognized over time usingon a contract by contract basis, determining the use of either a cost-based input method or output method, depending on whichever fairlybest depicts the transfer of control over the life of the performance obligation.

Tech Transfer Services (discontinued operations)

Revenue recognized under contracts for tech transfer services are considered a single performance obligation, as appropriate.all work packages (including data collection, GMP documentation, validation runs) and milestones are interrelated. Additionally, the customer is unable to complete services of work performed independently or by using competitors of the Company. Revenue is recognized over time using a cost-based based input method where progress on the performance obligation is measured by the proportion of actual costs incurred to the total costs expected to complete the contract.

F-16


b) Cell manufacturing servicesManufacturing Services (discontinued operations)

     Regarding revenues

Revenues from cell manufacturing services the Company concluded that these comprisedrepresent a single performance obligation.obligation which is recognized over time. The progress towards completion will continue to be measured on an output measure based on direct measurement of the value transferred to the customer (units produced).

c) Tech transfer

Reimbursed Expenses (discontinued operations)

The Company has concluded that underincludes reimbursed expenses in revenues and costs of revenue as the revised standard contractsCompany is primarily responsible for Tech Transferfulfilling the promise to provide the specified service, including the integration of the related services are consideredinto a single performance obligation and will be measured over time using a cost-based input method where progress on the performance obligation is measured by the proportion of actual costs incurredcombined output to the totalcustomer, which are inseparable from the integrated service. These costs expected to complete the contract.

     The cost-basedinclude such items as consumable, reagents, transportation and output methods of revenue recognition requiretravel expenses, over which the Company to make estimates of costs to complete its projects and the percentage of completion on an ongoing basis. Significant judgment is required to evaluate assumptions related to these estimates. The effect of revisions to estimates related to the transaction price (including variable consideration relating to reimbursement on a cost-plus basis on certain expenses) or costs to complete a project are recordedhas discretion in establishing prices.

F-17

Change Orders

Changes in the period in which the estimate is revised.  The adoptionscope of the new standard is not expected towork are common and can result in a materialchange in transaction price, equipment used and payment terms. Change orders are evaluated on a contract-by-contract basis to determine if they should be accounted for as a new contract or as part of the existing contract. Generally, services from change orders are not distinct from the original performance obligation. As a result, the effect that the contract modification has on the total stockholders’ equitycontract revenue, and measure of progress, is recognized as an adjustment to revenue when they occur.

Costs of December 1, 2018.Revenue (discontinued operations)

3) In November 2016,

Costs of revenue include (i) compensation and benefits for billable employees and personnel involved in production, data management and delivery, and the FASB issued ASU 2016-18, Statementcosts of Cash Flows (Topic 230): Restricted Cash (a Consensusacquiring and processing data for the Company’s information offerings; (ii) costs of staff directly involved with delivering services offerings and engagements; (iii) consumables used for the FASB Emerging Issues Task Force) (“ASU 2016-18”), which requires entitiesservices; and (iv) other expenses directly related to include amounts generally describedservice contracts such as restricted cashcourier fees, laboratory supplies, professional services and restricted cash equivalents in cash and cash equivalents when reconciling beginning-of-period and end-of-period total amounts shown on the statement of cash flows. ASU 2016-18 is effective for annual reporting periods (including interim periods within those annual reporting periods) beginning after December 15, 2017. travel expenses.

x.Leases

The Company adopted thisthe new lease standard duringASC 842 and all the year ended November 30, 2018.related amendments on January 1, 2019.

4) In February 2016,

The Company determines if an arrangement is a lease at inception. Lease classification is governed by five criteria in ASC 842-10-25-2. If any of these five criteria is met, The Company classifies the FASB issued ASU 2016-02, “Leases (Topic 842)”lease as a finance lease; otherwise, the Company classifies the lease as an operating lease. When determining lease classification, the Company’s approach in assessing two of the mentioned criteria is: (i) generally 75% or more of the remaining economic life of the underlying asset is a major part of the remaining economic life of that underlying asset; and (ii) generally 90% or more of the fair value of the underlying asset comprises substantially all of the fair value of the underlying asset.

Operating leases are included in operating lease right-of-use (“ASU 2016-02”ROU”), which supersedes assets and operating lease liabilities in the existing guidanceconsolidated balance sheet.

ROU assets represent Orgenesis’s right to use an underlying asset for the lease accounting, “Leases (Topic 840)”. ASU 2016-02 requires lesseesterm and lease liabilities represent its obligation to recognize leasesmake lease payments arising from the lease. Operating lease ROU assets and liabilities are recognized at the commencement date based on their balance sheets, and leaves lessor accounting largely unchanged.the present value of lease payments over the lease term. The amendments in ASU 2016-02 are effectiveCompany uses its incremental borrowing rate based on the information available at the commencement date to determine the present value of the lease payments.

The standard also provides practical expedients for fiscal years beginning after December 15, 2018 and interim periods within those fiscal years. Early application is permitted for all entities. ASU 2016-02 requires a modified retrospective approachan entity’s ongoing accounting. The Company elected the short-term lease recognition exemption for all leases with a term shorter than 12 months. This means that for those leases, the Company does not recognize ROU assets or lease liabilities, including not recognizing ROU assets or lease liabilities for existing at,short-term leases of those assets in transition, but recognizes lease expenses over the lease term on a straight-line basis.

Lease terms will include options to extend or entered after,terminate the date of initial application, with anlease when it is reasonably certain that Orgenesis will exercise or not exercise the option to elect to use certain transition relief. The Company expects to applyrenew or terminate the ASU without adjusting the comparative periods and, if applicable, recognizing a cumulative effect adjustment to the opening balance of retained earnings in the period of adoption. The Company is currently evaluating the impact of this new standard on its consolidated financial statements.lease.

5)

y.Recently issued accounting pronouncements, not yet adopted

In June 2016, the FASB issued ASU 2016-13 “Financial Instruments-CreditInstruments—Credit Losses—Measurement of Credit Losses (Topic 326)on Financial Instruments.(“ASU 2016-13”). ASU 2016-13 requiresThis guidance replaces the current incurred loss impairment methodology with a methodology that financial assets measured at amortized cost be presented at the net amount expected to be collected. The allowance for credit losses is a valuation account that is deducted from the amortized cost basis. The income statement reflects the measurement of credit losses for newly recognized financial assets, as well as the expected credit losses during the period. The measurementand requires consideration of expected credit losses is based upon historical experience, current conditions, anda broader range of reasonable and supportable forecasts that affect the collectability of the reported amount. Credit losses relatinginformation to available-for-sale debt securitiesinform credit loss estimates. The guidance will be recorded througheffective for Smaller Reporting Companies (SRCs, as defined by the SEC) for the fiscal year beginning on January 1, 2023, including interim periods within that year. The Company is currently evaluating this guidance to determine the impact it may have on its consolidated financial statements.

F-18

In August 2020, the FASB issued Accounting Standards Update (“ASU”) 2020-06, Debt with Conversion and Other Options (Subtopic 470-20) and Derivatives and Hedging-Contracts in Entity’s Own Equity (Subtopic 815-40)-Accounting For Convertible Instruments and Contracts in an allowanceEntity’s Own Equity. The ASU simplifies accounting for credit losses rather thanconvertible instruments by removing major separation models required under current GAAP. Consequently, more convertible debt instruments will be reported as a direct write-downsingle liability instrument with no separate accounting for embedded conversion features. The ASU removes certain settlement conditions that are required for equity contracts to qualify for the security.derivative scope exception, which will permit more equity contracts to qualify for it. The ASU 2016-13also simplifies the diluted net income per share calculation in certain areas. The new guidance is effective for annual and interim periods beginning after December 15, 2021, and early adoption is permitted for fiscal years beginning after December 15, 2019, including interim periods within those fiscal years. Early adoption is permitted as of the fiscal years beginning after December 15, 2018, including2020, and interim periods within those fiscal years. The Company doesis currently evaluating the impact that this new guidance will have on its consolidated financial statements.

z.Newly issued and recently adopted accounting pronouncements

The Company early adopted ASU 2019-12 on January 1, 2020, which did not expect to have a material impact on itsthe Consolidated Financial Statements except for the removal of the exception related to intra-period tax allocations. Commencing from January 1, 2020, the Company followed the general intra-period allocation of tax expenses. The Company had incurred a loss from continuing operations and subsequent to the adoption of ASU 2019-12, the Company determined the amount attributable to continuing operations without regard to the tax effect of other items. The ASU 2019-12 amendment related to the intra-period tax allocation was applied prospectively.

Had the Company not adopted ASU 2019-12, an approximately $20 million tax benefit would have been recognized along with corresponding decreases to net loss from continuing operations with a corresponding increase in tax expenses and decrease in net income resulting from discontinued operations. The Company had no intra-period tax allocation items in prior years.

aa.Reclassifications

Certain reclassifications have been made to the prior years’ financial statements to conform to the current year presentation. These reclassifications had no net effect on previously reported results of operations.

NOTE 3 – DISCONTINUED OPERATION

On February 2, 2020, the Company entered into a Purchase Agreement with GPP, Masthercell and the Buyer. Pursuant to the terms and conditions of the Purchase Agreement, Sellers agreed to sell 100% of the outstanding equity interests of Masthercell to Buyer for an aggregate nominal purchase price of $315 million. The Company has determined that the Masthercell Business meets the criteria to be classified as discontinued operations.

On February 10, 2020, the Masthercell Sale was consummated in accordance with the terms of the Purchase Agreement. After accounting for GPP’s liquidation preference and equity stake in Masthercell, as well as SFPI – FPIM’s interest in MaSTherCell, distributions to Masthercell option holders and transaction costs, the Company received approximately $126.7 million at the closing of the Masthercell Sale, of which $7.2 million was used for the repayment of intercompany loans and payables, including $4.6 million of payables to MaSTherCell.

Due to the sale of the controlling interest in Masthercell, the Company retrospectively reclassified the assets and liabilities of these entities as assets and liabilities of discontinued operations and included the financial results of these entities as discontinued operations in the Company’s consolidated financial statements.

F-17


6) In May 2017, the FASB issued ASU 2017-09, “Compensation - Stock Compensation (Topic 718): Scope of Modification Accounting” (“ASU 2017-09”), which gives direction on which changesDiscontinued operations relate to the terms or conditions of share-based payment awards require an entity to apply modification accounting in Accounting Standard Codification (“ASC”) Topic 718. In general, entities will apply the modification accounting guidance if the value, vesting conditions or classificationMasthercell Business. The comprehensive loss and balance sheet for this operation are separately reported as discontinued operations for all periods presented.

F-19

The financial results of the award changes. ASU 2017-09 is effective for fiscal years beginning after December 15, 2017, including interim periods within those fiscal years. Early adoption is permitted, including adoption in an interim period.Masthercell Business are presented as income (loss) from discontinued operations, net of income taxes on the Company’s consolidated statement of comprehensive loss. The Company does not expectfollowing table presents the implementation of this new pronouncement to have a material impact on its consolidated financial statements.

7) In June 2018,results associated with the FASB issued ASU 2018-07, “Compensation – Stock Compensation (Topic 718): Improvements to Nonemployee Share-based Payment Accounting” that expands the scope of ASC Topic 718 to include share-based payment transactions for acquiring goods and services from nonemployees. An entity should apply the requirements of ASC Topic 718 to nonemployee awards except for certain exemptions specifiedMasthercell Business operation as reflected in the amendment. Company’s Consolidated Comprehensive loss (in thousands):

SCHEDULE OF DISCONTINUED OPERATION AND BALANCE SHEETS

  Year Ended December 31, 
 2020  2019 
OPERATIONS      
Revenues $2,556  $31,053 
Cost of revenues  1,482   18,318 
Cost of research and development and research and development services, net  7   54 
Amortization of intangible assets  137   1,631 
Selling, general and administrative expenses  1,896   13,886 
Other (income) expenses, net  305   (207)
Operating loss  1,271   2,629 
Financial expenses (income), net  (29)  31 
Loss before income taxes  1,242   2,660 
Tax expenses (income)  (30)  792 
Net loss from discontinuing operation, net of tax $1,212  $3,452 
         
DISPOSAL        
Gain on disposal before income taxes $96,918  $- 
Provision for income taxes  -  - 
Gain on disposal $96,918  $- 
         
Net profit (loss) from discontinuing operation, net of tax $95,706  $(3,452)

The guidancefollowing table is effective for fiscal years beginning after December 15, 2018, including interim reporting periods within that fiscal year. Early adoption is permitted, but no earlier than an entity’s adoption datea summary of Topic 606. the assets and liabilities of discontinued operations (in thousands):

  

December 31,

2019

 
Assets   
    
CURRENT ASSETS:    
Cash and cash equivalents $11,281 
Restricted cash  186 
Accounts receivable, net  6,654 
Prepaid expenses and other receivables  845 
Grants receivable  1,979 
Inventory  1,907 
Deposits  326 
Property and equipment, net  22,149 
Intangible assets, net (mainly Know How)  10,858 
Operating lease right-of-use assets  8,860 
Goodwill  10,129 
Other assets  47 
TOTAL CURRENT ASSETS OF DISCONTINUED OPERATIONS $75,221 

  

December 31,

2019

 
CURRENT LIABILITIES:    
Accounts payable $5,756 
Accrued expenses and other payables  372 
Employees and related payables  2,047 
Advance payments on account of grant  2,227 
Short-term loans and current maturities of long- term loans  372 
Contract liabilities  8,301 
Current maturities of long-term finance leases  291 
Current maturities of operating leases  1,365 
Non-current operating leases  7,069 
Loans payable  1,230 
Deferred taxes  1,868 
Long-term finance leases  688 
TOTAL CURRENT LIABILITIES OF DISCONTINUED OPERATIONS $31,586 

F-20

Property, plants and equipment, net and right-of-use assets by geographical location were as follows:

  

December 31,

2019

 
    
United States $16,707 
Belgium  14,302 
Total $31,009 

The Company don’t expectfollowing table represents the components of the cash flows from discontinued operations (in thousands):

  Year Ended December 31, 
  2020  2019 
       
Net cash flows used in operating activities $(2,409) $(1,248)
Net cash flows used in investing activities $(579) $(11,621)
Net cash flows (used in) provided by financing activities $(51) $12,570 

Disaggregation of Revenue

The following table disaggregates the Company’s revenues by major revenue streams related to have a material impact on its consolidated financial statements.discontinued operations (in thousands):

8) In January 2017, FASB issued Accounting Standards Update (ASU) 2017-04, Intangibles—Goodwill and Other (Topic 350): Simplifying the Test for Goodwill Impairment, which eliminated the calculationSCHEDULE OF DISAGGREGATION OF REVENUE RELATED TO DISCONTINUED OPERATIONS

  Year Ended December 31, 
  2020  2019 
Revenue stream:        
         
Cell process development services $2,556  $20,834 
Tech transfer services  -   5,396 
Cell manufacturing services  -   4,823 
Total $2,556  $31,053 

Redeemable Non-Controlling Interest of implied goodwill fair value. Instead, companies will record an impairment charge based on the excess of a reporting unit’s carrying amount of goodwill over its fair value. This guidance simplifies the accounting as compared to prior GAAP. The guidance is effective for fiscal years beginning after December 15, 2019. The Company does not expect the implementation of this new pronouncement to have a material impact on its consolidated financial statements.Discontinued Operations

NOTE 3 – REDEEMABLE NON CONTROLLING INTEREST

a.Subscription and Shareholders Agreement with Belgian Sovereign Funds Société Fédérale de Participations et d’Investissement (“SFPI”).

a.Subscription and Shareholders Agreement with Belgian Sovereign Funds Société Fédérale de Participationset d'Investissement ("SFPI").

On November 15, 2017, the Company, MaSTherCell and SFPI entered into a Subscription and Shareholders Agreement (“SFPI Agreement”) pursuant to which SFPI made an equity investment in MaSTherCell in the aggregate amount of Euro 5 million (approximately $5.9 million), for approximately 16.7% of MaSTherCell (SFPI received B-shares of MaSTherCell which have the same voting, dividend and other rights as the existing shares of MaSTherCell). The equity investment commitment included the conversion of the outstanding loan and accrued interest of Euro 1.07 million (approximately $1.18 million), previously made by SFPI to MaSTherCell. In November 2017, the initial subscription amount of Euro 2 million ($2.3 million) was paid by SFPI to MaSTherCell. The proceeds from the investment are to be used in accordance with the long-term business plan that was appended to the SFPI Agreement which includes, without limitation, expanding MaSTherCell’s facilities in Belgium with the addition of five new cGMP manufacturing cleanrooms. The agreement contains customary representations, warranties and covenants by MaSTherCell and the Company, in respect of which the Company has undertaken to indemnify SFPI for the consequences of any breach thereof by MaSTherCell or the Company.

     Under the Agreement, SFPI has the right to appoint one member to the board of directors of MaSTherCell’s five person board. In addition, the holders of the B-Shares have a right to, along with the Company, appoint an independent director who will serve as the chairman of the board of MaSTherCell for a renewable three year term. The agreement provides that, under certain specified circumstances, SFPI is entitled to transfer its equity interest in MaSTherCell to the Company at a price equal to the total investment amount, plus a specified annual premium ranging from 10% to 25%, depending on the year following the subscription in which the put is exercised.

     Under the terms of the agreement since the Company listed to Nasdaq, SFPI is entitled to convert its MaSTherCell’ s equity interest (using an exchange rate of approximately $0.85), into shares of Common Stock of the Company based upon a conversion price of $6.24, the exercise period of the option is 3 years from the closing date of the SFPI Agreement. The $6.24 conversion price represents the price after the previous stock split of the Company.

F-18


     Furthermore, under the agreement, the Company had the right to spin-off the CDMO business into a Subsidiary provided that the Subsidiary adhered to the terms of the agreement. In June 2018, the Company effectuated such a spin-off and consolidated the CDMO business into Masthercell Global and Masthercell Global adhered to the terms of this agreement. Also, the Company possesses a drag along right under the Agreement whereby if the Company transfers all or the majority of its shares in MaSTherCell, it can force SFPI to do the same. (See also Note 3(b)).

     On June 13, 2018, SFPI has paid into MaSTherCell the remaining amount of Euro 1.9 million (approximately $2.3 million) to complete its subscription obligations under the agreement.

Due to the embedded redemption feature of the SPFI agreement whose settlement iswas not at the Company discretion, the Company had accounted for the investment made by SFPIGPP as a redeemable non-controlling interest. As of November 30, 2018, and 2017 SFPI investment was presented as redeemable non-controlling interest in the balance sheet, in the amount of $5.8 million and $3.6, respectively.

F-21

b.Stock Purchase Agreement and Stockholders’ Agreement with Great Point Partners, LLC (“GPP”)Stock Purchase Agreement and Stockholders’ Agreement with Great Point Partners, LLC (“GPP”)

On June 28, 2018, the Company, Masthercell Global GPP, and certain of GPP’s affiliates, entered into a series of definitive strategic agreements intended to finance, strengthen and expand Orgenesis’ CDMO business. In connection therewith, the Company, Masthercell Global and GPP-II Masthercell, LLC, a Delaware limited liability company (“GPP-II”) and an affiliate of GPP entered into Stock Purchase Agreement (the “SPA”) pursuant to which GPP-II purchased 378,000 shares of newly designated Series A Preferred Stock of Masthercell Global (the “Masthercell Global Preferred Stock”), representing 37.8% of the issued and outstanding share capital of Masthercell Global, for a cash consideration to be paid into Masthercell Global of up to $25 million, of which $13.2 million was subject to certain contingencies described below (the “Consideration”). Orgenesis holds 622,000 shares of Masthercell Global’s Common Stock, representing 62.2% of the issued and outstanding equity share capital of Masthercell Global. An initial cash payment of $11.8 million of the Consideration was remitted at closing by GPP-II. $1.5 Million of the initial capital contributed to Masthercell Global was used to reimburse the investors for their fees and expenses incurred in conjunction with this transaction (net payment of $10.3 million). Under the terms of the SPA the follow up payments will be in the amount of $6.6 million to be made in each of years 2018 and 2019 (the “Future Payments”), if (a) Masthercell Global achieves specified EBITDA and revenues targets during each of these years, and (b) the Orgenesis’ shareholders approve certain provisions of the SPA entered into by these parties. Such shareholder approval was obtained on October 23, 2018. None of the future Consideration amounts, if any, will result in an increase in GPP-II’s equity holdings in MaSTherCell.

     In connection with the entry into the SPA as described above, each of the Company, Masthercell Global and GPP-II entered into the Masthercell Global Inc. Stockholders’ Agreement (the “Stockholders’ Agreement”) providing for certain restrictions on the disposition of Masthercell Global securities, the provisions of certain options and rights with respect to the management and operations of Masthercell Global, certain rights to GPP-II (including, without limitation, a tag along right, drag along right and certain protective provisions). After the earlier of the second anniversary of the closing or certain enumerated circumstances, GPP-II is entitled to effectuate a spinoff of Masthercell Global and the Masthercell Global Subsidiaries (the “Spinoff”).

     The Spinoff is required to reflect a market value, provided that under certain conditions, such market valuation shall reflect a valuation of Masthercell Global and its Subsidiaries of at least $50 million. In addition, upon certain enumerated events described below, GPP-II is entitled, at its option, to put to the Company (or, at Company’s discretion, to Masthercell Global if Masthercell Global shall then have the funds available to consummate the transaction) its shares in Masthercell Global or, alternatively, purchase from the Company its share capital in Masthercell Global at a purchase price equal to the fair market value provided that the purchase price shall not be greater than three times the price per share of Masthercell Global Preferred Stock paid by GPP-II and shall not be less than the price per share of Masthercell Global Preferred Stock paid by GPP-II. GPP-II may exercise its put or call option upon the occurrence of any of the following: (i) there is an Activist Shareholder of the Company; (ii) the Chief Executive Officer and/or Chairman of the board of directors of the Company resigns or is replaced, removed, or terminated for any reason prior to June 28, 2023; (iii) there is a change of control event of the Company as defined in the Stockholders’ Agreement; or (iv) the industry expert director appointed to the board of directors of Masthercell Global is removed or replaced (or a new such director is appointed) without the prior written consent of GPP-II.

F-19


Activist Shareholder shall mean any Person who acquires shares of capital stock of the Company who either: (x) acquires more than a majority of the voting power of the Company, (y) actively takes over and controls a majority of the board of directors of the Company, or (z) is required to file a Schedule 13D with respect to such Person’s ownership of the Company and has described a plan, proposal or intent to take action with respect to exerting significant pressure on the management of or directors of, the Company.

     The Stockholders’ Agreement further provides that GPP-II is entitled, at any time, to convert its share capital in Masthercell Global for the Company’s common stock in an amount equal to the lesser of (a)(i) the fair market value of GPP-II’s shares of Masthercell Global Preferred Stock to be exchanged, divided by (ii) the average closing price per share of the Company’s Common Stock during the thirty day period ending on the date that GPP-II provides the exchange notice (the “Exchange Price”) and (b)(i) the fair market value of GPP-II’s shares of Masthercell Global Preferred Stock to be exchanged assuming a value of Masthercell Global equal to three and a half (3.5) times the revenue of Masthercell Global during the last twelve (12) complete calendar months immediately prior to the exchange divided by (ii) the Exchange Price; provided, that in no event will (A) the Exchange Price be less than a price per share that would result in Orgenesis Inc. having an enterprise value of less than $250 million and (B) the maximum number of shares of the Company’s Common Stock to be issued shall not exceed 2,704,247 shares, unless the Company obtains shareholder approval for the issuance of such greater amount of shares of the Company in accordance with the rules and regulations of the Nasdaq Stock Market.

     Great Point and Masthercell Global entered into an advisory services agreement pursuant to which Great Point is to provide management services to Masthercell Global for which Great Point will be compensated at an annual base compensation equal to the greater of (i) $250 thousand per each 12 month period or (ii) 5% of the EBITDA for such 12 month period, payable in arrears in quarterly installments; provided, that these payments will (A) begin to accrue immediately, but shall not be paid in cash to Great Point until such time as Masthercell Global generates EBITDA of at least $2 million for any 12 month period or the sale of or change in control of Masthercell Global, and (B) shall not exceed an aggregate annual amount of $0.5 million. Such compensation accrues but is not owed to Great Point until the earlier of (i) Masthercell Global generating EBITDA of at least $2 million for any 12-month period following the date of the agreement or (ii) a Sale of the Company or Change of Control of the Company (as both terms are defined therein).

     GPP and Masthercell Global entered into a transaction services agreement pursuant to which GPP is to provide certain brokerage services to Masthercell Global for which GPP will be entitled to a certain exit fee and transaction fee (as both terms are defined in the agreement), such fees not to be less than 2 percent of the applicable transaction value.

Due to the embedded redemption feature of the GPP agreement whose settlement iswas not at the Company discretion, the Company had accounted for the investment made by GPP as a redeemable non-controlling interestinterest.

NOTE 4 – ACQUISITION AND REORGANIZATION

Tamir Biotechnology, Inc.

On April 7, 2020, the Company entered into the Tamir Purchase Agreement with Tamir, pursuant to which the Company agreed to acquire certain assets and liabilities of Tamir related to the discovery, development and testing of therapeutic products for the treatment of diseases and conditions in humans, including all rights to Ranpirnase and use for antiviral therapy. The Tamir Transaction closed on April 23, 2020.

As aggregate consideration for the acquisition, the Company paid $2.5 million in cash and issued an aggregate of 3,400,000 shares (the “Shares”) of Common Stock to Tamir resulting in a total consideration of $20.2 million based on the Company’s share price at the closing date. $59 thousand and 340,000 Shares are being held in an escrow account for a period of 18 months from closing to secure indemnification obligations of Tamir pursuant to the terms of the Tamir Purchase Agreement. $4.5 million of the consideration was attributable to research and development related inventory and most of the remaining amount reflected the cost of intangible assets. The Shares were registered for resale by the Company in November 2020.

The Company’s acquired right to Tamir’s intellectual property represents a single identifiable asset sourced from the agreement. Because substantially all (more than 90%) of the fair value of the gross assets acquired are concentrated in a single asset being the right to Tamir’s intellectual property and related assets (“IPR&D”), the Company determined that the acquisition is not considered a business in accordance with ASC 805-10-55-5A. Therefore, the Company accounted the transaction as an asset acquisition. The fair value associated with Tamir’s IPR&D in the amount of $18.4.

     On November 30, 2018 Masthercell Global achieved$19.5 million was charged to research and development expenses under ASC 730. The remaining amount was attributed to the specified EBITDA and revenues targets for 2018 as describedabove-mentioned share in the SPA. The Company reflected the $6.6 milliona private company, which is presented in the balance sheet as GPP receivable. On January 16, 2019, GPP-II remitted to Masthercell Global the $6.6 million.long term “other assets.

NOTE 4- CORPORATE REORGANIZATION AND EXERCISE OF CALL OPTIONS OF CURECELL AND ATVIO

Description of Koligo Acquisition during 2020

On September 26, 2020, the TransactionCompany entered into an Agreement and Plan of Merger and Reorganization (the “Merger Agreement”) by and among the Company, Orgenesis Merger Sub, Inc., a Delaware corporation and a wholly-owned subsidiary of the Company (“Merger Sub”), Koligo Therapeutics Inc., a Kentucky corporation (“Koligo”), the shareholders of Koligo (collectively, the “Shareholders”), and Long Hill Capital V, LLC (“Long Hill”), solely in its capacity as the representative, agent and attorney-in-fact of the Shareholders. The Merger Agreement provides for the acquisition of Koligo by the Company through the merger of Merger Sub with and into Koligo, with Koligo surviving as a wholly-owned subsidiary of the Company (the “Merger”). The acquisition was completed on October 15, 2020 (the “Effective Time”).

     Contemporaneous

Koligo is a privately-held US regenerative medicine company. Koligo’s first commercial product is KYSLECEL® (autologous pancreatic islets) for chronic and acute recurrent pancreatitis. Koligo’s 3D-V technology platform incorporates the use of advanced 3D bioprinting techniques and vascular endothelial cells to support development of transformational cell and tissue products for serious diseases.

Pursuant to the terms of the Merger Agreement, at the Effective Time, the shares of capital stock of Koligo that were issued and outstanding immediately prior to the Effective Time were automatically cancelled and converted into the right to receive, subject to customary adjustments, an aggregate of 2,061,713 shares of Company common stock which have been issued to Koligo’s accredited investors (with certain non-accredited investors being paid solely in cash in the amount of approximately $20 thousand). In addition, we issued 66,910 shares to Maxim Group LLC for advisory services in connection with the executionMerger. The share price was $5.26 at the day of the SPA andclosing.

F-22

The Merger Agreement contains customary indemnification provisions whereby the Stockholders’ Agreement (see Note 3),Shareholders of Koligo will indemnify the Company and Masthercell Globalcertain affiliated parties for any losses arising out of breaches of the representations, warranties and covenants of Koligo and the Shareholders under the Merger Agreement. As partial security for the indemnification and purchase price adjustment obligations of Koligo shareholders under the Merger Agreement, $7 thousand in cash and 328,587 shares of Company common stock of the merger consideration otherwise payable in the Merger to the Shareholders were placed in a third party escrow account. The aggregate indemnification obligations of the Koligo shareholders under the Merger Agreement is capped at the amounts in escrow, subject to certain limited exceptions.

In addition, according to the agreement between the parties, the Company has also funded an additional cash consideration of $500 thousand (with $100 thousand of such reducing the ultimate consideration payable to Koligo) for the acquisition of the assets of Tissue Genesis, LLC (“Tissue Genesis”) by Koligo that was consummated on October 14, 2020. The Tissue Genesis assets include the entire inventory of Tissue Genesis Icellator® devices, related kits and reagents, a broad patent portfolio to protect the technology, registered trademarks, clinical data, and existing business relationships for commercial and development stage use of the Icellator technology.

In connection with the Merger Agreement, the Company, Long Hill and Maxim Group LLC (“Maxim”) entered into a Contribution, AssignmentRegistration Rights and AssumptionLock-Up Agreement pursuant to which Long Hill will have one demand registration right to require the registration of the shares of Company common stock received by Long Hill in the Merger and Long Hill and Maxim will have certain piggyback registration rights. In addition, Long Hill agreed with the Company contributedthat, during the applicable Restriction Period (as defined below), it shall not sell or transfer, subject to Masthercell Global assets relatingcertain limited exceptions, the portion of the shares received in the Merger during the applicable Restriction Period, subject to a limitation on the CDMO platform including: (i)number of shares sold per any trading day not to exceed 10% of the average daily trading volume of the Common Stock, as reported by Bloomberg Financial L.P. “Restriction Period” means (a) in relation to 70% of all of the Company’s holdings in Masthercell Global Subsidiaries (ii) the debtshares received in the total amountMerger that Long Hill is entitled to receive under or in connection with the Merger Agreement, the period beginning on the date of $2.3 million owedthe closing and ending on the date that is the four month anniversary thereof, and (b) in relation to the Company by Atvio and CureCell (iii) the license agreement between the Company and MaSTherCell dated December 30, 2016; (v) the Joint Venture Agreement with Atvio dated May 10, 2016 (as amended on May 30, 2016); (vi) the SFPI Agreement (vii) the Joint Venture Agreement between Orgenesis and CureCell dated March 14, 2016 (the “Corporate Reorganization”). See Note 12(b).

F-20


      In furtherance thereof, Masthercell Global, as the Company assignee, acquiredremaining 30% of all of the issued and outstanding share capital of Atvio and 94.12%shares received in the Merger that Long Hill is entitled to receive under or in connection with the Merger Agreement, the period beginning on the date of the share capitalclosing and ending on the date that is the twelve month anniversary thereof. All of CureCell.the shares required to be registered by the Company pursuant to the Registration Rights and Lock-Up Agreement were registered by the Company in November 2020.

In addition, pursuant to separate Lock-Up Agreements entered into by the Shareholders other than Long Hill with the Company (the “Shareholders Lock-Up Agreement”), such Shareholders agreed that they will not transfer any of their shares received in the Merger except in accordance with the following lock-up release schedule whereby one fifth of such holder’s respective shares will be released from such restriction every six months, starting six months from the closing of the Merger. Each holder’s sales of such shares are subject to a resale limit of its pro rata portion of 10% of the average daily trading volume, allocated to the Shareholders other than Long Hill pro-rata.

The acquisition was accounted in accordance with Accounting Standards Codification Topic 805, “Business Combinations”. The allocation of the consideration transferred in certain cases may be subject to revision based on the final determination of fair values during the measurement period, which may be up to one year from the acquisition date. The Company exercisedincludes the "call option" to which it was entitled under the joint venture agreements with each of these entities to purchase from the former shareholders their equity holding. The consideration for the outstanding share equity in each of Atvio and CureCell consisted solely of the Company Common Stock.

     In respect of the acquisition of Atvio, the Company issued to the former Atvio shareholders an aggregate of 83,965 shares of Company’s Common Stock. In respect of the acquisition of CureCell, the Company issued the former CureCell shareholders an aggregate of 202,846 shares of the Company Common Stock. The exercise of the call options of CureCell and Atvio, pursuant to which the Company obtained effective control over such entities, was accounted for as a business combination. The results of operations of CureCell and Atvio have been includedthe business that it has acquired in the Company’s condensedits consolidated statements of operations startingresults prospectively from June 28, 2018, the date on which the Company obtained effective control of CureCell and Atvio. Before the closing date Atvio and CureCell were associated companies, see Note 12. The net gain on remeasurementacquisition.

Fair Value of the previously held equity interest in Atvio and CureCell to acquisition date fair value was $4.5 million.Consideration Transferred

CureCell

The following table summarizes the allocation of purchase price to the fair values of the assets acquired and liabilities assumed as of the transaction date:

SUMMARY OF ASSETS ACQUIRED AND LIABILITIES ASSUMED

  (in thousands) 
  (in thousands) 
Fair value of 8.8% of shared issued * *11,172 
Cash payment  1,115 
Total consideration transferred $12,287 

Total assets acquired:*
           Cash and cash equivalents$ 58
           Property and equipment, net1,104
           Inventory148
           Other assets300
           Other Intangible assets (a)3,933
           Goodwill (b)3,950
           Total assets9,493
Total liabilities assumed:
           Deferred income from the Company and others1,945
           Deferred taxes80
           Fair value of convertible loan from the Company892
           Non-controlling interests*299
           Other liabilities1,487
Total liabilities4,703
Total consideration transferred$ 4,790
Fair value of 36.4%the consideration is based on the company’s market share price.

F-23

Total assets acquired:    
Cash and cash equivalents $8 
Restricted Cash  152 
Accounts Receivable  228 
Inventory  34 
Other assets  25 
Property, plants and equipment, net  482 
Kyslecel Technology (a)(a) 9,340 
IPR&D (a)  641 
Operating lease right-of-use assets  238 
Goodwill (b)  3,704 
Total assets  14,852 
     
Total liabilities assumed:    
Operating leases  238 
Accounts Payable  216 
Accrued Expenses  4 
Orgenesis Inc loan  651 
Deferred taxes  1,293 
Notes Payable  162 
Other liabilities  1 
Total liabilities  2,565 
Total consideration transferred $12,287 

a.The allocation of shared issued * 1,853
Acquisition datethe purchase price to the net assets acquired and liabilities assumed resulted in the recognition of other intangible assets which comprised of: Kyslecel Technology of $9,340 and IPR&D of 641. Kyslecel Technology has a useful life of 15 years. The useful life of these intangible assets for amortization purposes was determined considering the period of expected cash flows generated by the assets used to measure the fair value of previously held equity interest2,937
Total consideration transferred$ 4,790the intangible assets adjusted as appropriate for the entity-specific factors, including legal, regulatory, contractual, competitive, economic or other factors that may limit the useful life of intangible assets.

* Fair value of the consideration is based on the company’s market share price.

a. The allocation of the purchase price to the net assets acquired and liabilities assumed resulted in the recognition of otherThese intangible assets which comprised of: Customer Relationships of $859 and “Know How” of $3,074. These other intangible assets have a useful life of 10 and 12 years, respectively. The useful life of the other intangible assets for amortization purposes was determined considering the period of expected cash flows generated by the assets used to measure the fair value of the intangible assets adjusted as appropriate for the entity-specific factors, including legal, regulatory, contractual, competitive, economic or other factors that may limit the useful life of intangible assets.

F-21


     The fair value of the Know How was estimated using a relief of royalties’ approach. Under this method, the fair value of the Know How is equal to the royalty fee that the owner of the Know How could profit from if he was to license the Know How out.

     Customer Relationships were estimated using a discounted cash flow method with the application of the multi-period excess earnings method. Under this method, an intangible asset’s fair value is equal to the present value of the incremental after-tax cash flows attributable only to the subject intangible asset after deducting contributory asset charges. An income and expenses forecast were built based upon revenue and expense estimates.

b.

b.The primary items that generate goodwill include the value of the synergies between the acquired company and the Company and the acquired assembled workforce, neither of which qualifies for recognition as an intangible asset. The Goodwill is not deductible for tax purposes.

Atvio

     The total consideration of Atvio of $890 thousand was attributed mainly to goodwill.

Pro forma Impact of Business Combination

The unaudited pro forma financial results have been prepared using the acquisition method of accounting and are based on the historical financial information of the Company Atvio and CureCell.Koligo. The unaudited pro forma condensed financial results have been prepared for illustrative purposes only and do not purport to be indicative of the results of operations that actually would have resulted had the acquisition of Atvio and CureCellKoligo occurred at the beginning of the fiscal year, or of future results of the combined entities. The unaudited pro forma condensed financial information does not reflect any operating efficiencies and expected realization of cost savings or synergies associated with the acquisition.

     The acquired business contributed revenues of $1.1 million for the period of June 28, 2018 to November 30, 2018. The following unaudited

Unaudited supplemental pro forma summary presents consolidation informationcombined results of the Company as if the business combination had occurred on December 1, 2017:

  Year Ended November 30, 
  2018  2017 
  (in thousands) 
Revenue$ 18,794 $ 10,216 
Net loss$ 20,919 $ 14,520 

NOTE 5 - SEGMENT INFORMATIONoperations (in thousands):

     The Chief Executive Officer ("CEO") is the Company’s chief operating decision-maker ("CODM"). Management has determined that there are two operating segments, based on the Company's organizational structure, its business activities and information reviewed by the CODM for the purposes of allocating resources and assessing performance.SCHEDULE OF UNAUDITED SUPPLEMENTAL PRO FORMA

  2020  2019 
  Year ended December 31, 
  2020  2019 
       
Revenues $8,239  $4,398 
Net loss $318  $27,263 
Loss per share:        
Basic $0.05  $1.91 

CDMO

     The CDMO platform is comprised of a specialization in cell therapy development and comprised of two types of services to its customers: (i) manufacturing and development services and (ii) cGMP contract manufacturing services. The CDMO platform operates through Masthercell Global, which currently consists of MaSTherCell in Belgium, Atvio in Israel and CureCell in South Korea, having unique know-how and expertise for manufacturing in a multitude of cell types. The CDMO activities include the operations of Masthercell Global, Atvio and CureCellKoligo’s related actual results from the date of acquisition to December 31, 2020 resulted in a loss of $513 thousand.

F-24

Koligo’s Acquisition-related Costs

Acquisition-related expenses consist of transaction costs which represent external costs directly related to the Corporate Reorganization.

PT Business

F-22


     Throughacquisition of Koligo and primarily include expenditures for professional fees such as legal, accounting and other directly related incremental costs incurred to close the PT business,acquisition by both the Company is focused on the development of proprietary cell and gene therapies, including the autologous trans-differentiation technology, and therapeutic collaborations and licensing with other pre-clinical and clinical-stage biopharmaceutical companies and research and healthcare institutes.Koligo.

     The CODM does not review assets by segment, therefore the measure of assets has not been disclosed for each segment.

     Segment dataAcquisition-related expenses for the year ended November 30, 2018 is as follows:

        Corporate    
     PT  and    
  CDMO  Business  Eliminations  Consolidated 
  (in thousands) 
Revenues from external customers$ 22,582 $ - $ (3,927)$ 18,655 
Cost of revenues (11,541)    1,235  (10,306)
Segment gross profit (loss) 11,041  -  (2,692  8,349 
Research and development expenses, net (39) (7,931) 2,485  (5,485)
Operating expenses (6,889) (6,224) 207  (12,906)
Other expenses (77) -     (77)
Segment operating profit (loss) 4,036  (14,155) -  (10,119)
Adjustments to presentation of segment Adjusted EBIT          
     Depreciation and amortization (2,613) (11)    (2,624)
Segment performance 1,423  (14,166)    (12,743)

ReconciliationDecember 31, 2020 were $682 thousand. These expenses were recorded to selling and general administrative expense in the consolidated statements of segment performancecomprehensive loss.

Cooperate reorganization, description of the Transactions Korea and OBI during 2019

On August 7, 2019, the Company, Masthercell Global and GPP-II Masthercell, LLC, a Delaware limited liability company (“GPP-II”), (the “Parties”) entered into a Transfer Agreement (the “Transfer Agreement”). As a result of the Transfer Agreement, Masthercell Global transferred all of its equity interests of OBI and the Korean Subsidiary to lossOrgenesis Inc in exchange for one dollar ($1.00). The Transfer Agreement also contained agreements made with respect to certain intercompany loans. The Company accounted for the year:

Year ended
November 30,
2018
in thousands
Segment subtotal performance(12,743)
Stock-based compensation(4,364)
Financial expenses, net(2,938)
Net gain on remeasurement of previously equity interest in Atvio and CureCell to acquisition date fair value4,509
Transaction expenses related to GPP agreement(1,500)
Share in losses of associated companies(731)
Loss before income tax(17,767)

Segment data for the year ended November 30, 2017 isTransfer Agreement as follows:

        Corporate    
     CT  and    
  CDMO  Business  Eliminations  Consolidated 
  (in thousands) 
Revenues from external customers$ 11,484 $ - $ (1,395)$ 10,089 
Cost of revenues (6,356)    638  (5,718)
Gross profit 5,128  -  (757) 4,371 
Research and development expenses, net    (2,517) 757  (1,760)
Operating expenses (4,699) (3,335)    (8,034)
Operating profit 429  (5,852) -  (5,423)

Adjustments to presentation of segment Adjusted EBITa transaction with non-controlling interest.

F-23



     Depreciation and amortization (2,720) (6)
Segment performance (2,291) (5,858)

Reconciliation of segment performance to loss for the year:Year ended
November 30,
2017
in thousands
Segment performance(8,149)
   Stock-based compensation(3,364)
   Financial income (expenses), net(950)
   Share in losses of associated company(1,214)
   Loss before income tax$ (13,677)

Geographic, Product and Customer Information

     Substantially all the Company's revenues and long-lived assets are located in Belgium through its subsidiary, MaSTherCell. Net revenues from single customers from the CDMO segment that exceed 10% of total net revenues are:

  Year Ended  Year Ended 
  November 30,  November 30, 
  2018  2017 
  (in thousands) 
Customer A$ 2,338 $ 4,115 
Customer B$ 4,374 $ 2,837 
Customer C$ - $ 2,055 
Customer D$ 5,236 $ - 
Customer E$ 2,242 $ - 

     The CDMO business has substantially diversified revenues by source signing contracts with biotech companies in their respective cell-based therapy field.

NOTE 65PROPERTY, PLANTS AND EQUIPMENT

The following table represents the components of property, plants and equipment:

  November 30, 
  2018  2017 
  (in thousands) 
Cost:      
Production facility$ 11,413 $ 6,246 
Office furniture and computers 1,875  353 
Lab equipment 3,292  2,039 
  16,580  8,638 
Less – accumulated depreciation (4,679) (3,534)
Total$ 11,901 $ 5,104 

SCHEDULE OF COMPONENTS OF PROPERTY, PLANTS AND EQUIPMENT

  December 31, 
  2020  2019 
  (in thousands) 
Cost:        
Production facility $2,801  $2,481 
Office furniture and computers  697   606 
Lab equipment  1,483   656 
Advance payment  281   - 
Subtotal  5,262   3,743 
Less – accumulated depreciation  (2,189)  (1,438)
Total $3,073  $2,305 

Depreciation expense for the years ended November 30, 2018December 31, 2020 and 2017 was $711December 31, 2019 were $ 705 thousand and $1,096$634 thousand, respectively.

Property, plants and equipment, net by geographical location were as follows:

SCHEDULE OF PROPERTY, PLANT AND EQUIPMENT BY GEOGRAPHICAL LOCATION

  December 31, 
  2020  2019 
  (in thousands) 
       
Belgium $358  $- 
Korea  839   983 
Israel  1,386   1,322 
U.S.  490   - 
Total $3,073  $2,305 

F-25

NOTE 76INTANGIBLE ASSETS AND GOODWILL

F-24


Changes in the carrying amount of the Company’s goodwill in our CDMO platform for the years ended November 30, 2018December 31, 2020 and 20172019 are as follows:

(in thousands)
Goodwill as of November 30, 2016$ 9,584
Translation differences1,100
Goodwill as of November 30, 201710,684
Goodwill as acquired4,918
Translation differences(437)
Goodwill as of November 30, 2018$ 15,165

SCHEDULE OF GOODWILL

  (in thousands) 
Goodwill as of December 31, 2018 $4,942 
Goodwill as acquired, (Koligo) see note 4  - 
Translation differences  (130)
Goodwill as of December 31, 2019 $4,812 
Goodwill as of December 31, 2019 $4,812 
Goodwill as acquired, (Koligo) see note 4  3,704 
Translation differences  229 
Goodwill as of December 31, 2020 $8,745 

Goodwill Impairment

     The Company reviews goodwill

See Note 2(m) for impairment annually and whenever events or changes in circumstances indicate the carrying amount of goodwill may not be recoverable. The Company performed a quantitative two-step assessment forCompany’s goodwill impairment for each reporting unit.analysis.

Reporting Unit of MaSTherCell’ s Goodwill

     As part of the first step of the two-step impairment test, the Company compared the fair value of the reporting unit to her carrying value and determined that the carrying amount of the unit do not exceed her fair value. The Company estimated the fair value of the unit by using an income approach based on discounted cash flows. The assumptions used to estimate the fair value of the Company’s reporting unit were based on expected future cash flows and an estimated terminal value using a terminal year growth rate based on the growth prospects for each reporting unit. The Company used an applicable discount rate which reflected the associated specific risks for the reporting unit future cash flows.

     Key assumptions used to determine the estimated fair value include: (a) expected cash flow for the five-year period following the testing date (including market share, sales volumes and prices, costs to produce and estimated capital needs); (b) an estimated terminal value using a terminal year growth rate of 2% determined based on the growth prospects; and (c) a discount rate of 15%. Based on the Company’s assessment as of November 30, 2018 and 2017, respectively, the carrying amount of its reporting unit does not exceed its fair value.

     A decrease in the terminal year growth rate of 1% or an increase of 1% to the discount rate would reduce the fair value of the reporting unit by approximately $1,126 thousand and $1,961 thousand, respectively. These changes would not result in an impairment. A decrease in the terminal year growth rate and an increase in the discount rate of 1% would reduce the fair value of the reporting unit by approximately $2,899 thousand. These changes would not result in an impairment.

Reporting Unit of Atvio’s Goodwill and Reporting Unit of CureCell’s Goodwill

     As of the year ended November 30, 2018 the Company elected to preform qualitative assessment. Due to the fair value measurement that was performed on June 28, 2018 as part of the business combination. Based on the Company’s assessment as of November 30, 2018, the carrying amount of its reporting unit does not exceeds its fair value.

Other Intangible Assets

Other intangible assets consisted of the following:

  November 30,  November 30, 
  2018  2017 
  (In thousands) 
Gross Carrying Amount:      
   Know How$ 20,344 $ 17,998 
   Backlog 117  - 

F-25SCHEDULE OF OTHER INTANGIBLE ASSETS



 December 31, 
 2020  2019 
 (in thousands) 
Gross Carrying Amount:        
Know How $3,170  $2,991 
Customer relationships 1,274  369   886   895 
Brand name 1,359  1,418 
 23,094  19,785 
Accumulated amortization 6,394  4,734 
Kyslecel Technology  9,340   - 
IPR&D  641   - 
Subtotal  14,037   3,886 
Less – Accumulated amortization  (1,014)  (538)
Net carrying amount of other intangible assets$ 16,700 $ 15,051  $13,023  $3,348 

Intangible assetassets amortization expenses were approximately $1.9$478 thousand and $1.8 and million$430 thousand for the years ended November 30, 2018December 31, 2020 and 2017,December 31, 2019, respectively.

Estimated aggregate amortization expenses for the five succeeding years ending November 30on December 31thst are as follows:

SCHEDULE OF ESTIMATED AGGREGATE AMORTIZATION EXPENSES

  2021  2022 to 2025 
  (in thousands) 
Amortization expenses $965  $3,910 

  2019  2020 to 2023 
  (in thousands) 
Amortization expenses$ 2,086 $ 7,929 
F-26

NOTE 8– 7 – CONVERTIBLE LOANS

a. SCHEDULE OF LONG TERM CONVERTIBLE LOANS

a.Long term convertible loans outstanding as of December 31, 2020 and December 31, 2019 are as follows:

Principal

Amount

  

Issuance

Year

 

Interest

Rate

  Maturity Period Exercise Price  

BCF

 
(in thousands)       (Years)      
Convertible Loans Outstanding as of December 31, 2020    
$1,000  2018  2% 3  7.00(1)  71 
 9,500  2019  6%-8% 2-5  7.00(2)  - 
 250  2020  8% 2  7.00(3)  - 
$10,750                 
                   
Convertible Loans Outstanding as of December 31, 2019 
$1,500  2018  2% 3  7.00(1)  124 
 11,400  2019  6%-8% 2-5  7.00(2)  - 
$12,900                 

Convertible Loans repaid during the year ended December 31, 2020 
               

Principal

Amount

  

Issuance

Year

 

Interest

Rate

  Maturity Period Exercise Price  

BCF

 
                   
 500  2018  2% 0.87 $7   53 
 500  2019  6% 0.28  7   - 
 1,400  2019  8% 0.76  7   - 
 2,400                 

Apart from the items mentioned below there were no repayments of convertible loans outstanding as of November 30, 2018during the fiscal years ended December 31, 2019 and 2017 are as follows:December 31, 2020. In addition, there were no conversions during the fiscal years ended December 31, 2019 and December 31, 2020.

                    Shares and Warrants 
                    Issued Upon Conversion 
                 Accumulated  Shares  Warrant 
                 Interest Up to     s 
Principal Issuance  Interest  Maturity  Exercise     Conversion     (5)
Amount Year  Rate   Period  Price  BCF Date       
(in                (in thousands)       
thousands)       (Years)                
Convertible Loans Outstanding as of November 30, 2018                
                         
                         
$ 1,250 2018  2%  3 $7(1) 114 $ -  -  - 

Convertible Loans converted during the year ended November 30, 2018          
                         
220 2018  6%  2 $6.24  87 $ 2  35,543  35,543 
500 (2) 2018  6%  0.5 $6.24  106  4  80,756  80,756 
5,050 2017  6%  2 $6.24  2,311  235  846,961  846,961 
798 (3) 2017  6%  0.5-1.7 $6.24  81  40  134,372  34,269 
1,388 2016  6%  2 $6.24  251  132  243,443  243,443 
100 2014  6%/24%(4) 0.5 $4.8  85  81  37,662  - 
8,056               $ 494  1,378,737  1,240,972 
                         
Convertible Loans repaid during the year ended November 30, 2017             
                         
$ 400 (6) 2017  6%  0.25 $6.24     -  -  - 

     (1)

(1)The holders, at their option, may convert the outstanding principal amount and accrued interest under this note into a total of 148,838 shares and 148,838three-year warrants to purchase up to an additional 148,838 shares of the Company’s common stock at a per share exercise price of $7. In the initial two years, the holders have the right to convert the outstanding principal amount and accrued interest into shares of capital stock of Hemogenyx-Cell or Immugenyx, LLC according under the relevant note agreement, subsidiaries of Hemogenyx Pharmaceuticals Plc, at a price per share based on a pre-money valuation of Hemogenyx-Cell or Immugenyx, LLC of $12 million and $8 million, respectively, pursuant to the collaboration agreement with Hemogenyx Pharmaceuticals Plc and Immugenyx, LLC. As of December 31, 2020, the loans are presented in current maturities of convertible notes in the balance sheet (See Notes 11(c) and 11(d).

(2)The holders, at their option, may convert the outstanding principal amount and accrued interest under this note into a total of 1,443,734 shares and 1,053,503three-year warrants to purchase up to an additional 1,053,503 shares of the Company’s common stock at a per share exercise price of $7. As of December 31, 2020, $2,500 thousand of the principal amount is included in current maturities of convertible loans in the balance sheet and the remainder in long-term convertible loans. See also Notes 7(b), 7(c), 7(e), 7(f) and 7(g).

(3)The holders, at their option, may convert the outstanding principal amount and accrued interest under this note into a total of 38,559 shares at a per share exercise price of $7. As of December 31, 2020, all the principal amount is included in long-term convertible loans in the balance sheet See also Notes 7(h).

b. During April 2019, the Company entered into a convertible loan agreement with an offshore investor for an aggregate amount of $500 thousand into the U.S. Subsidiary. The investor, at its option, may convert the outstanding principal amount and accrued interest under this note into a total of 178,570 shares and 178,570 three-year warrants to purchase up to an additional 178,570 shares of the Company’s common stock at a per share exercise price of $7. In$7.00; or into shares of the initial two years,U.S. Subsidiary at a valuation of the holders haveU.S. Subsidiary of $50 million. During February 2020 the rightcompany repaid this convertible loan to the investor in full.

F-27

c. During May 2019, the Company entered into a private placement subscription agreement with an investor for $5 million. The lender shall be entitled, at any time prior to or no later than the maturity date, to convert the outstanding principal amount, and accrued interest into units of (1) shares of capitalcommon stock of Hemogenyx-Cell or Immugenyx, LLC according under the relevant note agreement, subsidiaries of Hemogenyx Pharmaceuticals Plc,Company at a conversion price per share based on a pre-money valuation of Hemogenyx-Cell or Immugenyx, LLC of $12 millionequal to $7.00 and $8 million, respectively, pursuant to the collaboration agreement with Hemogenyx Pharmaceuticals Plc and Immugenyx, LLC. As of November 30, 2018, the loans are presented in long term convertible notes in the balance sheet. See Note 11(f) and 11(g).

F-26


(2) On the issuance date of the note the Company issued to certain investors 40,064 three-year warrants to purchase up to an equal number of additional one shareshares of the Company’s common stock at a price of $7.00 per share.

The transaction costs were approximately $497 thousand, out of which $97 thousand are stock-based compensation due to issuance of warrants.

d. In May 2019, the Company had agreed to enter into a 6% convertible loan agreement with an investor for an aggregate amount of $5 million. The lender shall be entitled, at any time prior to or no later than the maturity date, to convert the outstanding amount, into units of (1) shares of stock of the Company at a conversion price per share exercise price of $6.24.

     (3) On the issuance date of the note the Company issuedequal to certain investors 145,509 three-year$7.00 and (2) warrants to purchase up to an equal number of additional one shareshares of the Company’s common stock at a per share exercise price of $6.24.

     (4) The Company failed to reimburse$7.00 per share. As of the date of the filing of this Annual Report on Form 10-K, the loan had not yet been received by the Company.

e. In June 2019, the Company entered into private placement subscription agreements with investors for an aggregate amount of $2 million. The lenders shall be entitled, at any time prior to or no later than the maturity date, thereforeto convert the interest expenses increaseoutstanding amount, into units of (1) shares of common stock of the Company at a conversion price per share equal to loan had$7.00 and (2) warrants to purchase an equal number of additional shares of the Company’s common stock at a default interestprice of 24%$7.00 per share.

f. During October 2019, the Company entered into a Private Placement Subscription Agreement and Convertible Credit Line Agreement (collectively, the “Credit Line Agreements”) with four non-U.S. investors (the “Lenders”), pursuant to which the Lenders furnished to the Company access to an aggregate $5.0 million credit line (which consists of $1.25 million from each Lender) (collectively, the “Credit Line”). Pursuant to the Credit Line Agreements, the Company is entitled to draw down an aggregate of $1 million (consisting of $250 thousand from each Lender) of the Credit Line in each of October 2019 and November 2019. In each of December 2019, January 2020 and February 2020, the Company may draw down an additional aggregate of $1 million (consisting of $250 thousand from each Lender), until the total amount drawn down under the Credit Line reaches an aggregate of $5 million (consisting of $1.25 million from each Lender), subject to the approval of the Lenders.

Pursuant to the terms of the agreement.Credit Line Agreements and the Notes, the total loan amount, and all accrued but unpaid interest thereon, shall become due and payable on the second anniversary of the Effective Date (the “Maturity Date”). The Maturity Date may be extended by each Lender in its sole discretion and shall be in writing signed by the Company and the Lender. Interest on any amount that has been drawn down under the Credit Line accrues at a per annum rate of eight percent (8%). At any time prior to or on the Maturity Date, by providing written notice to the Company, each of the Lenders is entitled to convert its respective drawdown amounts and all accrued interest, into shares of the Company’s common stock, par value $0.0001 per share (the “Common Stock”), at a conversion price equal to $7.00 per share.

     (5)

Furthermore, upon the drawdown of $500 thousand from each Lender and, together with the other Lenders, a drawdown of an aggregate of $2 million under the Credit Line, the existing warrants of the Lenders to purchase shares of Common Stock shall be amended to extend their exercise date to June 30, 2021 and the Company will issue to each of the Lenders warrants to purchase 50,000 shares of Common Stock at an exercise price of $7.00 per share. The warrant,new warrants will be exercisable for a period of three (3) years from the Effective Date. During October 2019, such drawdown was reached and the warrants were issued. The modification of the existing warrants in the amount of $145 thousands was recorded against the accumulated deficit and the value of the new warrants in the amount of $370 thousands was offset against the convertible loan amount.

The lender shall be entitled, at any time prior to or no later than the maturity date, to convert the outstanding amount, into units of shares of common stock of the Company at a conversion price per share equal to $7.00.

As at December 31, 2019, the Company had received $3.65 million from the Convertible Credit Line investment comprised of $1.15 million from one investor, $1 million from a second investor, and $750 thousand from two of the other lenders.

F-28

The transaction costs were approximately $145 thousand.

During the year ended December 2020 the company repaid principal amount of $2,400 thousand and a total interest amount of $372 thousand to certain of the credit line investors.

g. In December 2019, the Company entered into private placement subscription agreements with investors for an aggregate amount of $250 thousand. The lenders shall be entitled, at any time prior to or no later than the maturity date, to convert the outstanding amount, into units of 1 share of common stock of the Company at a conversion price per share equal to $7.00 and warrants to purchase 183,481additional shares of the Company’s common stock at a price of $7.00 per share. The fair value of the warrants was $124 thousand using the fair value of the shares on the grant date.

h. On January 2, 2020, the Company entered into private placement subscription agreements with investors for an aggregate amount of $250 thousand of convertible loans. The lenders shall be entitled, at any time prior to or no later than the maturity date, to convert the outstanding amount, into shares of Common Stock of the Company at a conversion price per share equal to $7.00. In addition, the Company granted the investors 151,428 warrants to purchase an equal number of additional shares of Common Stock at a per share exercise price of $6.24.$7.00 per share.

     (6)

i. In December 2018, the Company entered into a Controlled Equity Offering Sales Agreement, or Sales Agreement, with Cantor Fitzgerald & Co., or Cantor, pursuant to which the Company may offer and sell, from time to time through Cantor, shares of its common stock having an aggregate offering price of up to $25.0 million. The principal amountCompany will pay Cantor a commission rate equal to 3.0% of the aggregate gross proceeds from each sale. Shares sold under the Sales Agreement will be offered and accrued interest were repaidsold pursuant to the Company’s Shelf Registration Statement on Form S-3 (Registration No. 333-223777) that was declared effective by the CompanySecurities and Exchange Commission on March 7, 2017. The fair value of28, 2018, or the shares as of March 7, 2017, was $494 thousandShelf Registration Statement, and was recorded as financial expenses.

b. On February 27, 2017,a prospectus supplement and accompanying base prospectus that the Company filed with the Securities and Admiral Ventures Inc. (“Admiral”) entered into an agreement resolving the payment of convertible loan received in prior years and owed to Admiral. Under the terms of the settlement agreement, Admiral extended the maturity date to June 30,Exchange Commission on December 20, 2018. The Company agreed to pay to Admiral, on or before March 1, 2017, between $0.3 million and $1.5 million. Further, beginning April 2017, the Company agreed to make a monthly paymenthas not yet sold any shares of $125 thousand on account of remaining unpaid balance, and also agreed to remit additional payments under the term of the agreement. The Company accounted for the above changes as a modification of the old debt.

     During the year ended November 30, 2017, the Company repaid $1,875 thousand on account of the principal amount and accrued interest. In January 2018, the Company repaid the remaining of accrued interest in total amount of $179 thousand. In 2018 and 2017 the Company was in arrears in its payment obligations under such agreement therefore, the Company issued to Admiral 120,193 units as forbearance fees accordingcommon stock pursuant to the terms of the agreement. Each unit consisting of one share of the Company’s common Stock and one three-year warrant exercisable into an additional share of common stock at a per share exercise price of $6.24. The fair value of the units was recorded as financial expenses during the year ended November 30, 2018 and 2017 in total amount of $179 and 983thousand, respectively, out of which $434 thousand reflect the fair value of the warrants using the Black-Scholes valuation model.Sales Agreement.

c.

j. On November 2, 2016, the Company entered into unsecured convertible note agreements with accredited or offshore investors for an aggregate amount of NIS 1 million ($280 thousand). The loan bears a monthly interest rate of 2%2% and mature on May 1, 2017, unless converted earlier. On April 27, 2017 and November 2, 2017, the Company entered into extension agreements through November 2, 2017 and May 2, 2018, respectivelyrespectively.

F-27


In March 2018, the investor submitted a notice of its intention to convert into shares of the Company'sCompany’s common stock the principal amount and accrued interest of approximately $383$383 thousand outstanding. A related party of such investor at the same time, exercised warrants issued in November 2016 to purchase shares of the Company'sCompany’s Common Stock. The exercise price of the warrants and conversion price were fixed at $0.52$0.52 per share (pre-reverse stock split implemented by the Company in November 2017). There is a significant disagreement between the Company and these two entities as to the number of shares of Common Stock issuable to these entities, and they contend that the number of shares of Common Stock issuable to them should not consider the reverse stock split. The Company rejects these contentions in their entirety and, based on the advice of specially retained counsel, believes that these claims are without legal merit and not made in good faith. The Company intends to vigorously defend its interests and pursue other avenues of legal address. Through its counsel, the Company has advised these entities that unless they withdraw their request within a specified period, the Company will cancel the above referenced agreements and these parties’ right to receive any shares of the Company’s Common Stock. In April 2018, the Company withdrew the agreements and deposited the shares in total amount of 107,985 issued under those agreements and the principal amount and accrued interest of the loan in escrow account. The deposit of the principal amount and accrued interest presented as restricted cash in the balance sheet as of November 30, 2018.December 31, 2020.

d. In January 2017, MaSTherCell repaid all but one of its bondholders (originally issued on September 14, 2014), and the aggregate payment amounted to $1.7 million (€1.5 million). On January 17, 2017, the remaining bondholder agreed to extend the duration of his Convertible bond until March 21, 2017. In consideration for the extension, the Company issued to the bondholder warrants to purchase 8,569 shares of the Company’s Common Stock, exercisable over a three-year period at a per share exercise price of $6.24. The fair value of those warrants as of the date of grant was $20 thousand using the Black-Scholes valuation model.

F-29

     On March 20, 2017, the remaining bondholder converted his convertible bonds into 40,682 shares of the Company’s Common Stock (See also note 13(c)).NOTE 8 – LOANS

NOTE 9 – LOANS

a.Terms of Long-term Loans

  Principal        Year of     November 30, 
  Amount  Grant Year  Interest Rate  Maturity  2018  2017 
  (in thousands)           (in thousands) 
Long-term loan (*)1,400  2012  4.05%  2022 $ 700 $ 899 
Long-term loan1,000  2013  6%-7.5%  2023  869  977 
Long-term loan790  2012-2016  5.5%-6%  2021-2024  461  620 
             $ 2,030 $ 2,496 
Current portion of loans payable           (368) (378)
             $ 1,662 $ 2,118 

(*) The loan has a business pledge on the Company’s assets at the same value.

b.Terms of Short-term Loans

SCHEDULE OF LOANS

       December 31, 
  Currency Interest Rate  2020  2019 
       (in thousands) 
Short term loans KRW  3.61% $-  $260 
Short term loans KRW  6.00%  -   131 
Short term loans USD  1.00%  145   - 
        $145  $391 

NOTE 9 – LEASES

The Company leases research and Current Portiondevelopment facilities, equipment and offices under finance and operating leases. For leases with terms greater than 12 months, the Company record the related asset and obligation at the present value of Long-Term Loanslease payments over the term. Many of the leases include rental escalation clauses, renewal options and/or termination options that are factored into the determination of lease payments when appropriate.

The Company’s leases do not provide a readily determinable implicit rate. Therefore, the Company estimated the incremental borrowing rate to discount the lease payments based on information available at lease commencement.

Manufacturing facilities

        November 30, 
  Currency  Interest Rate  2018  2017 
        (in thousands) 
Current portion of loans payable Euro  4.05% $ 168 $ 169 
Current portion of loans payable Euro  6%-7.5%  67  70 
Current portion of loans payable Euro  5.5%-6%  133  139 
Short term loans KRW  3.02%, 4.15%  279  - 
       $ 647 $ 378 

The Company leases space for its manufacturing facilities in Israel under operating lease agreements. The leasing contracts are for a period of 3 - 5 years.

Research and Development facilities

The Company leases space for its research and development facilities in South Korea under an operating lease agreement. The leasing contracts are for a period of 25 years.

Offices

The Company leases space for offices in Israel under operating leases. The leasing contracts are valid for terms of 5 years. These contracts are considered as operational leasing and under operating lease right-of-use assets.

Lease Position

The table below presents the lease-related assets and liabilities recorded on the balance sheet.

SCHEDULE OF LEASE-RELATED ASSETS AND LIABILITIES

  December 31, 2020 
Assets    
Operating Leases    
Operating lease right-of-use assets $1,474 
     
Finance Leases    
Property, plants and equipment, gross  99 
Accumulated depreciation  (17)
Property and equipment, net $82 
     
Liabilities    
Current liabilities    
Current maturities of operating leases $485 
Current maturities of long-term finance leases $19 
     
Long-term liabilities    
Non-current operating leases $1,020 
Long-term finance leases $64 
     
Weighted Average Remaining Lease Term    
Operating leases   3.4 years 
Finance leases   4.2 years 
     

Weighted Average Discount Rate

    
Operating leases  6.7%
Finance leases  2.0%

F-30

Lease Costs

The table below presents certain information related to lease costs and finance and operating leases during the year ended December 31, 2020.

SCHEDULE OF LEASE COSTS

  

Year ended

December 31,

2020

 
    
Operating lease cost: $547 
     
Finance lease cost:    
Amortization of leased assets  17 
Interest on lease liabilities  3 
Total finance lease cost $20 

The table below presents supplemental cash flow information related to leases during the year ended December 31, 2020:

SCHEDULE OF SUPPLEMENTAL CASHFLOW INFORMATION

  

Year ended

December 30,

2020

 
  (in Thousands) 
Cash paid for amounts included in the measurement of leases liabilities:    
Operating leases $515 
Finance leases $42 
     
Right-of-use assets obtained in exchange for lease obligations:    
Operating leases $967 
Finance leases  366 

Undiscounted Cash Flows

The table below reconciles the undiscounted cash flows for each of the first five years and total of the remaining years to the finance lease liabilities and operating lease liabilities recorded on the balance sheet.

SCHEDULE OF FINANCE LEASE LIABILITIES AND OPERATING LEASE LIABILITIES

  

Operating

Leases

  Finance
Leases
 
Year ended December 31,        
2021 $526  $20 
2022  528   20 
2023  342   20 
2024  188   20 
2025  59   4 
Total minimum lease payments  1,643   84 
Less: amount of lease payments representing interest  (138)  (1)
Present value of future minimum lease payments  1,505   83 
Less: Current leases obligations  (485)  (19)
Long-term leases obligations $1,020  $64 

F-31

Right-of-use assets by geographical location were as follows:

SCHEDULE OF RIGHT-OF-USE ASSETS BY GEOGRAPHICAL LOCATION

  December 31, 
  2020  2019 
  (in thousands) 
       
Korea $683  $145 
Israel  496   580 
U.S.  295   - 
Total $1,474  $725 

NOTE 10 - COMMITMENTS

a.Maryland Technology Development Corporation

See Note 11 for additional commitments for funding of the ventures of the company.

a.Maryland Technology Development Corporation

On June 30, 2014, the Company’s U.S. Subsidiary entered into a grant agreement with Maryland Technology Development Corporation (“TEDCO”). TEDCO was created by the Maryland State Legislature in 1998 to facilitate the transfer and commercialization of technology from Maryland’s research universities and federal labs into the marketplace and to assist in the creation and growth of technology-based businesses in all regions of the State. Under the agreement, TEDCO paid to the U.S Subsidiary an amount of $406$406 thousand (the “Grant”). On June 21, 2016 TEDCO has approved an extension until June 30, 2017. Through November 30, 2018, the Company utilized $356 thousand from the grant and recorded it as a deduction of research and development expenses in the statement of comprehensive loss.

F-28


b.Department De La Gestion Financiere Direction De L’analyse Financiere (“DGO6”)

b..Department De La Gestion Financiere Direction De L’analyse Financiere (“DGO6”)

(1) On March 20, 2012, MaSTherCell was awarded an investment grant from the DGO6 of Euro 1.2 million. This grant is related to the investment in the production facility with a coverage of 32% of the investment planned. As of November 30, 2018, the DGO6 transferredthe entire amount to MaSTherCell .

     (2) On November 17, 2014, the Belgian Subsidiary, received the formal approval from the DGO6 for a Euro 2 million ($2.4 million) support program for the research and development of a potential cure for Type 1 Diabetes. The financial support was composed of Euro 1,085 thousand (70%(70% of budgeted costs) grant for the industrial research part of the research program and a further recoverable advance of Euro 930 thousand (60%(60% of budgeted costs) of the experimental development part of the research program. In December 2014, the Belgian Subsidiary received advance payment of Euro 1,209 thousand under the grant. The grants are subject to certain conditions with respect to the Belgian Subsidiary’s work in the Walloon Region. In addition, the DGO6 is also entitled to a royalty upon revenue being generated from any commercial application oftheof the technology. In 2017 the Company received by the DGO6 final approval for Euro 1.8 million costs invested in the project out of which Euro 1.2 million foundedfunded by the DGO6. As of November 30, 2018,December 31, 2020, the Company repaid to the DGO6 a total amount of $34$118 thousand (Euro 3096 thousand) and amount of $152$106 thousand was recorded in other payables.

     (3) ln

(2) In April 2016, the Company's Belgian Subsidiary received the formal approval from DGO6 for a Euro 1.3 million ($1.5 million) support program for the development of a potential cure for Type 1 Diabetes. The financial support was awarded to the Belgium Subsidiary as a recoverable advance payment at 55%55% of budgeted costs, or for a total of Euro 717 thousand ($800 thousand). The grant will be paid over the project period. On December 19, 2016, theThe Belgian Subsidiary received advance payment of Euro 359 438 thousand ($374 537 thousand). Up through November 30, 2018,December 31, 2020, an amount of Euro 303 358 thousand ($437 thousand) was recorded as deduction of research and development expenses and an amount of $64 Euro 80 thousand was recorded as advance payments on account of grant.

     b.

F-32

(3) On October 8, 2016, the Belgian Subsidiary received the formal approval from the DGO6 for a Euro 12.3 million ($12.8 million) support program for the GMP production of AIP cells for two clinical trials that will be performed in Germany and Belgium. The project will be heldconducted during a period of three years commencing January 1, 2017. The financial support is awarded to the Belgium subsidiary at 55% 55% of budgeted costs, a total of Euro 6.8 million ($7 million). The grant will be paid over the project period. On December 19, 2016, the Belgian Subsidiary received a first payment of Euro 1.7 million ($1.82 million). Up through November 30, 2018,December 31, 2020, an amount of $1.1Euro 1.7 million was recorded as deduction of research and development expenses and an amount of $847Euro 53 thousand was recorded as advance paymentsreceivable on account of grant.

c.Israel-U.S Binational Industrial Research

(4) In December 2020, the Belgian Subsidiary received the formal approval from DGO6 for a Euro 2.9 million ($3.5 million) support program for research on Dermatitis Treatments and Development Foundation (“BIRD”)Wound Healing Using Cell Regenerative Technologies. The financial support was awarded to the Belgium Subsidiary as a recoverable advance payment at 60% of budgeted costs, or for a total of Euro 1.7 million ($2.1 million). The grant will be paid over the project period. The Belgian Subsidiary received an advance payment of Euro 301 thousand ($366 thousand) in December 2020. The research program is to be started in 2021.

c.Israel-U.S. Binational Industrial Research and Development Foundation (“BIRD”)

On September 9, 2015, the Israeli Subsidiary entered into a pharma Cooperation and Project Funding Agreement (CPFA) with BIRD and Pall Corporation, a U.S. company. BIRD will giveawarded a conditional grant of $400$400 thousand each (according to terms defined in the agreement), for a joint research and development project for the use Autologous Insulin Producing (AIP) Cells for the Treatment of Diabetes (the “Project”). The Project started on March 1, 2015. Upon the conclusion of product development, the grant shall be repaid at the rate of 5%5% of gross sales. The grant will be used solely to finance the costs to conduct the research of the project during a period of 18 months starting on March 1, 2015.To date the Israeli Subsidiary received $200 thousand under the grant.2015. On July 28, 2016, BIRD approved an extension tillfor the project period until May 31, 2017 and the final report was submitted to BIRD.

     Up through November 30, 2018, an amount As of $359 thousand was recorded as deduction of research and development expenses and $159 thousand as a receivable on account of grant.

d.Korea-Israel Industrial Research and Development Foundation (“KORIL”)

     On March 14, 2016, the Israel subsidiary, entered into a collaboration agreement with CureCell, initially for the purpose of applying a grant from KORIL for pre-clinical and clinical activities related to the commercialization of the Israel subsidiary AIP cell therapy product in Korea. The Parties agreed to carry out at their own expenses and their respective commitments under the work plan approved by KORIL and any additional work plan to be agreed betweenDecember 31, 2020, the Israeli Subsidiary and CureCell. The Israeli Subsidiary will own sole rights to any intellectual property developed from the collaboration which is derivedreceived a total amount of $299 thousand under the Israeli Subsidiary’s AIP cell therapy product, information licensed from THM. Subject to obtaininggrant and the requisite approval needed to commence commercialization in Korea, the Israel subsidiary has agreed to grant to CureCell, or a fully owned subsidiary thereof, under a separate sub-license agreement an exclusive sub-license to the intellectual property underlying the Company’s API product solely for commercialization of the Israel subsidiary products in Korea. As part of any such license, CureCell has agreed to pay annual license fees, ongoing royalties based on net sales generated by CureCell and its sublicensees, milestone payments and sublicense fees. Under the agreement, CureCell is entitled to share in the net profits derived by the Israeli Subsidiary from world-wide sales (except for sales in Korea) of any product developed as a result of the collaboration with CureCell. Additionally, CureCellproject was given the first right to obtain exclusive commercialization rights in Japan of the AIP product, subject to CureCell procuring all the regulatory approvals required for commercialization in Japan. As of November 30, 2018, none of the requisite regulatory approvals for conducting clinical trials had been obtained.completed.

F-29


d.Korea-Israel Industrial Research and Development Foundation (“KORIL”)

On May 26, 2016, the Israeli Subsidiary and CureCellthe Korean Subsidiary entered into a pharma Cooperation and Project Funding Agreement (CPFA) with KORIL. KORIL will give a conditional grant of up to $400$400 thousand each (according to terms defined in the agreement), for a joint research and development project for the use of AIP Cells for the Treatment of Diabetes (the “Project”). The Project started on June 1, 2016. Upon the conclusion of product development, the grant shall be repaid at the yearly rate of 2.5%2.5% of gross sales. The grant will be used solely to finance the costs to conduct the research of the project during a period of 18 months starting. On July 26, 2018 KORIL approved extension for the project period till May 31, 2019.2019 and was further extended to May 2020. During 2019, the grant was assigned to Cure Therapeutics from the Korean Subsidiary. As of November 30, 2018, TheDecember 31, 2020, the Israeli Subsidiary and CureCellthe Korean Subsidiary received total amount of $440$440 thousand under the grant.  Up through November 30, 2018, The Israeli Subsidiary recorded an amount of $279 thousand as a deduction of research and development expenses and $119 thousand as a receivable on account of grant, and CureCell recorded $134 thousand as advance payments on account of grant.

e.BIRD Secant

e.BIRD Secant

On July 30, 2018, Orgenesis Inc and AtvioOBI entered into a collaboration agreement with Secant Group LLC (“Secant”). Under the agreement, Secant will engineer and prototype 3D scaffolds based on novel biomaterials and technologies involving bioresorbable polymer microparticles, while AtvioOBI will provide expertise in cell coatings, cell production, process development and support services. Under the agreement, Orgenesis is authorized to utilize the jointly developed technology for its autologous cell therapy platform, including its Autologous Insulin Producing (“AIP”) cell technology for patients with Type 1 Diabetes, acute pancreatitis and other insulin deficient diseases. In the beginning of 2018, AtvioOBI entered into a Cooperation and Project Funding Agreement (CPFA) with BIRD and Secant. BIRD will give a conditional grant up to $450$450 thousand each to support the joint project (according to terms defined in the agreement).

As of November 30, 2018, AtvioDecember 31, 2020, OBI received a total amount of $125$425 thousand under the grant. Up through November 30, 2018,For the year ended December 31, 2020, an amount of $275$28 thousand was recorded as deduction of research and development expenses and $163 thousand as a receivable on account of grant.expenses.

f.Lease Agreement

     The Company leases office space for its CDMO facilities and  research and development facilities in Belgium, Korea, Israel and the United States of America under several lease agreements.  The expiration dates of the lease agreements for the facilities in each country are as follow:

Country
Expire Date
BelgiumFebruary 29, 2020 and March 31, 2030
IsraelJuly 31, 2023
KoreaJuly 14, 2020
United States*F-33

*The company signed a new lease agreement during January 2019, see Note 21(f)

Future minimum lease commitments under non-cancelable operating lease agreements are as follows:

2019$783 
2020 626 
2021 and thereafter 3,504 
Total$4,913 

F-30


NOTE 11 – COLLABORATION AND LICENSE AGREEMENTS

a.Adva Biotechnology Ltd.

a.Adva Biotechnology Ltd.

On January 28, 2018, the Company and Adva Biotechnology Ltd. (“Adva”), entered into a Master Services Agreement (“MSA”), under which the Company and/or its affiliates are to provide certain services relating to development of products to Adva, as may be agreed between the parties from time to time. Under the MSA, the Company undertook to provide Adva with in kind funding in the form of materials and services having an aggregate value of $750approximately $760 thousand at the Company’s own cost in accordance with a project schedule and related mutually acceptable project budget. The Company entered into an agreement with Atvio,Orgenesis Biotech Israel (previously Atvio), to fulfill its obligations pursuant this MSA. As of November 30, 2018,MSA and it completed its contractual obligations under the Company incurred a total expense of $361 thousand.contract during 2019.

In consideration for and subject to the fulfillment by the Company of such in-kind funding commitment, Adva agreed that upon completion of the development of the products, the Company and/or its affiliates and Adva shall enter into a supply agreement pursuant to which for a period of eight (8) years following execution of such supply agreement, the Company and/or its affiliates (as applicable) is entitled (on a non-exclusive basis) to purchase the products from Adva at a specified discount pricing from their then standard pricing. The Company and/or its affiliates were also granted a non-exclusive worldwide right to distribute such products, directly or through any of their respective contract development and manufacturing organization (CDMO) service centers during such term. indirectly. The MSA shall remain in effect for 10 years unless earlier terminated in accordance with its terms.

b.Tel Hashomer Medical Research, Infrastructure and Services Ltd(“THM”).

b.Tel Hashomer Medical Research, Infrastructure and Services Ltd (“THM”).

On February 2, 2012, the Company’s Israeli Subsidiary entered into a licensing agreement with THM. According to the agreement, the Israeli Subsidiary was granted a worldwide, royalty bearing, exclusive license to trans-differentiation of cells to insulin producing cells, including the population of insulin producing cells, methods of making this population, and methods of using this population of cells for cell therapy or diabetes treatment developed by Dr. Sarah Ferber of THM.

As consideration for the license, the Israeli Subsidiary will pay the following to THM:

1)

A royalty of 3.5%3.5% of net sales;

2)

16%16% of all sublicensing fees received;

3)

An annual license fee of $15$15 thousand, which commenced on January 1, 2012 and shall be paid once every year thereafter. The annual fee is non-refundable, but it shall be paid each year against the royalty noted above, to the extent that such are payable, during that year; and

4)

Milestone payments as follows:

a.

a.

$50 thousand on the date of initiation of phasePhase I clinical trials in human subjects;

b.

b.

$50 thousand on the date of initiation of phasePhase II clinical trials in human subjects;

c.

c.

$150 thousand on the date of initiation of phasePhase III clinical trials in human subjects;

d.

d.

$750 thousand on the date of initiation of issuance of an approval for marketing of the first product by the FDA; and

e.

e.

$2 million when worldwide net sales of Products (as defined in the agreement) have reached the amount of $150$150 million for the first time, (the “Sales Milestone”).

As of November 30, 2018,December 31, 2020, the Israeli Subsidiary had not reached any of these milestones.

F-31


In the event of closing of an acquisition of all of the issued and outstanding share capital of the Israeli Subsidiary and/or consolidation of the Israeli Subsidiary or the Company into or with another corporation (“Exit”), the THM shall be entitled to choose whether to receive from the Israeli Subsidiary a one-time payment based, as applicable, on the value of either 463,651 shares of common stock of the Company at the time of the Exit or the value of 1,000 shares of common stock of the Israeli Subsidiary at the time of the Exit.

F-34

c. Mircod LimitedHemogenyx Pharmaceuticals PLC.

     On June 18, 2018, the Company and Mircod Limited, a company formed under the laws of Cyprus (“Mircod”) entered into a Collaboration and License Agreement (the “Mircod Collaboration Agreement”) for the research, development and commercialization of potential key technologies related to biological sensing for the Company's clinical development and manufacturing projects (the “Development Project”).

     Under the Mircod Collaboration Agreement, all results of such collaboration (“Project Results”) shall be jointly owned by Mircod and the Company. The Company was granted an exclusive, worldwide sub licensable license under Mircod’s right in such Project Results to use and commercialize Project Results in consideration for a royalty of 5% of net sales (as defined in the Collaboration Agreement) of products incorporating Project Results.

     Subject to completion of the Development Project, Mircod and the Company are to negotiate and enter into a manufacturing and supply agreement under which Mircod is to manufacture and supply products incorporating the Project Results and, at the Company’s request, to provide support and maintenance service for such products. If for whatever reason the parties fail to enter into such manufacturing and supply agreement within 90 days of the completion of the Development Project or if Mircod is unable to perform such services, the Company is entitled to manufacture the products, in which event Mircod will be entitled to a payment of $80,000 and royalties on Net Sales are to increase to 8% of Net Sales. As of November 30, 2018, the Developments Project has not completed.

d. HekaBio K.K

     On July 10, 2018, the Company and HekaBio K.K. (“HB”), a corporation organized under the laws of Japan entered into a Joint Venture Agreement (the “HB JVA”) pursuant to which the parties will collaborate in the clinical development and commercialization of regeneration and cell and gene therapeutic products (hereinafter the “Products”) in Japan (the “Project”). The parties intend to pursue the joint venture through a newly established Japanese company (hereinafter the “JV Company”) which the Company by itself, or together with a designee, will hold a 49% participating interest therein, with the remaining 51% participating interest being held by HB. HB will fund, at its sole expense, all costs associated with obtaining the requisite regulatory approvals for conducting clinical trials, as well as performing all clinical and other testing required for market authorization of the Products in Japan.

     Under the JVA, each party may invest up to $10 million, which may take the form of a loan, if required, as determined by the steering committee. The terms of such investment, if any, will be on terms mutually agreeable to the parties, provided that the minimum pre-money valuation for any such investment shall not be less than $10 million. Additionally, HB was granted an option to affect an equity investment in the Company of up to $15 million within the next 12 months on mutually agreeable terms. If such investment is in fact consummated, the Company agreed to invest in the JV Company by way of a convertible loan an amount equal to HB’s pro-rata participating interest in the JV Company, which initially will be at 51%. Such loan may then be converted by the Company into share capital of the JV company at an agreed upon formula for determining JV Company valuation which in no event shall be less than $10 million. Under the JVA, the Company can require HB to sell to the Company its participating (including equity) interest in the JV Company in consideration for the issuance of the Company’s common stock based on an agreed upon formula for determining JV Company valuation which in no event shall be less than $10 million.

     In addition, under the JVA, the Company shall grant the JV Company an exclusive license to certain intellectual property of the Company as may be required for the JV Company to develop and commercialize the Products in Japan. In consideration of such license, the JV Company shall pay the Company, in addition to other payments, royalties at the rate of 10% of the JV Company’s net sales of Products.

F-32


     It was further agreed that the JV Company shall grant the Company (and its affiliates) a non-exclusive, worldwide (other than Japan), royalty-free and fully paid-up license to use and practice, for any purpose, new inventions, discoveries and intellectual property rights that are generated by and/or on behalf of HB and/or the JV Company in connection with the Project.

     On October 3, 2018, the Company entered into a License Agreement with the JV Company pursuant to the JVA pertaining to the licenses described above.

     Apart from the above, as of November 30, 2018, no activity had begun in the said JVand no investments were made therein.

e. Image Securities Ltd.

     On July 11, 2018, the Company and Image Securities Ltd., a corporation with its registered office in Grand Cayman, Grand Cayman Islands (“India Partner”) entered into a Joint Venture Agreement (the “India JVA”) pursuant to which the parties will collaborate in the development and/or marketing, clinical development and commercialization of cell therapy products in India (the “Cell Therapy Products”). The India Partner will collaborate with a network of healthcare facilities and a healthcare infrastructure as well as financial partners to advance the development and commercialization of the cell therapy products.

     The India JVA becomes effective upon the consummation of an equity investment by the India Partner in the Company of $5 million within 15 days of the execution of the India JVA through the purchase of units of the Company securities at a per unit purchase price payable into the Company of $6.24, with each unit comprised of one share of the Company and three-year warrant for the acquisition of an additional common share at a per share exercise price of $6.24. Following the consummation of such equity investment in the Company, on October 18, 2018, the Company entered into a convertible loan agreement with the India Joint Venture company (“India JV”) pursuant to which the Company agreed to invest $5 million into the India JV. The loan is convertible into equity capital of the India JV at an agreed upon formula for determining India JV valuation. The investment in the Company by the India Partner was the consummation of the previously disclosed private placement subscription agreement entered into in December 2016 between the Company and an affiliate of the India Partner pursuant to which the closing of such subscription agreement was by the terms thereof delayed until terms comprising the India JV were mutually agreed to. As of November 30, 2018, the Company has advanced $1 million to the JV Company under the convertible loan agreement, the loan will bear interest of 6% per annum and the outstanding amount (principal and interest) will be payable after two years. The loan was presented in the balance sheet as loan to related party.

     Under the India JVA, the India Partner agreed to invest in the JV $10 million within 12 months of the incorporation of the India JV. If for whatever reason such investment is not made by the India Partner within such time, then the Company is authorized to convert its above-referenced loan into 50% of the equity capital of the India JV on a fully diluted basis, provided that if the pre-money valuation of the India JV is then independently determined to be less than $5 million, then such conversion to be effected in the basis of such valuation. Apart from the above, as of November 30, 2018, no activity had begun.

     As part of the agreement, the Indian joint venture will procure consulting services from the Company in the amount of $1 million per month – See also Note 21(c).

f. Hemogenyx Pharmaceuticals PLC.

On October 18, 2018, the Company and Hemogenyx Pharmaceuticals PLC., a corporation with its registered office in the United Kingdom and Hemogenyx-Cell (“H-Cell"H-Cell”), a corporation with its registered office in Belgium (together “Hemo”) which, who are engaged in the development of cell replacement bone marrow therapy technology, entered into a Collaboration Agreement (the “Hemo agreement”Agreement”) pursuant to which the parties will collaborate in the funding, of the continued development, of, and commercialization of the Hemo technology via the Hemo group companies.Hemo. Pursuant to the Hemo agreement the Company and Hemogenyx LLC (“Hemo-LLC”) (a wholly owned USAUS subsidiary of Hemo) entered into a loan agreement on November 7, 2018 according to which the Company agreed to loan Hemo-LLC not less than $1 $1 million by way of a convertible loan. On November 25, 2018 the Company and Hemo entered into a License and Distribution agreement according to which Company received the worldwide rights to market the products under the agreement in consideration for the payment of a 12%12% royalty all subject to the terms of the agreement. On November 25, 2018, the Company and H-Cell signed an Exclusive Manufacturing agreement according to which the Company will receive the exclusive right to manufacture certain of H-Cell products.

F-33


     As of November 30, During 2018 and 2020 the Company advanced $0.5 $0.75 million and $0.25 million, respectively, to Hemo as a convertible loan and an additional $0.25 million was advanced in December 2018 (see Note 21(b)). Thethe entire loan as of November 30, 2018, in the amount of $0.5 million, was charged to expenseexpenses under ASC 730-10-50 and 20-50 and presented as research and development costs.

See Note 8 (a).7.

g. Immugenyx LLC.

d.Immugenyx LLC.

On October 16, 2018, the Company and Immugenyx LLC., a corporation with its registered office in the USA (“Immu”) which, who is engaged in the development of technology related to the production and use of humanized mice entered into a Collaboration Agreement (the “Immu agreement”Agreement”) pursuant to which the parties will collaborate in the funding, of the continued development, of, and commercialization of the Immu technology. Pursuant to the agreement, the Company received the worldwide rights to market the products under the agreement in consideration for the payment of a 12%12% royalty all subject to the terms of the agreement. Pursuant to the Immu agreement the Company and Immu entered into a loan agreement on November 7, 2018 according to which the Company agreed to loan Immu not less than US$1 Million by way of a convertible loan. As of November 30,During 2018 and 2020 the Company advanced $0.5 $0.75 million and $0.25 million, respectively, to Immu as a convertible loan and an additional $0.25 million was advanced in December 2018 (see Note 21(b)). Thethe entire loan as of November 30, 2018, in the amount of $0.5 million, was charged to expenseexpenses under ASC 730-10-50 and 20-50 and presented as research and development costs. See also Note 8 (a).

h.BG Negev Technologies and Applications (“BGN”).

e.BG Negev Technologies and Applications (“BGN”).

On August 2, 2018, the Company’s USAU.S. Subsidiary entered into a licensing agreement with BGN. According to the agreement, the USAU.S. Subsidiary was granted a worldwide, royalty bearing, exclusive license to develop and commercialize a novel alginate scaffold technology for cell transplantation focused on autoimmune diseases.

On November 25, 2018, the Company’s USAU.S. Subsidiary entered into a further licensing agreement with BGN. According to the agreement, the USAU.S. Subsidiary was granted a worldwide, royalty bearing, exclusive license to develop and commercialize technology directed to RAFT modification of polysaccharides and use of a bioreactor for supporting cell constructs.

As consideration for the licenses, the USAU.S. Subsidiary will pay royalties of between 2%4% and 4%7% (subject to rate reductions to 5% and 4%, respectively, in specific circumstances) of net sales of the licensed product, sub-license fees of between 15% and 20%20% of sub-license income received, license fees of $10,000$10,000 per year per license, and milestone and budget payments according to agreeagreed upon work plans to BGN.

i.Collaboration agreement

F-35

f.Collaboration Agreement with Tarus Therapeutics, Inc.

     During 2018,On February 27, 2019, the Company and CureTarus Therapeutics Inc., a Delaware corporation, (“Tarus”) entered into a Collaboration Agreement (the “Tarus Agreement”) for the collaboration in the funding, development and commercialization of certain technologies, products and patents of Tarus in the areas of therapeutics for cancer and other diseases in the field of cell therapies and their combination with checkpoint inhibitors comprised of Adenosine Receptor Antagonists. Under the terms of the Tarus Agreement and subject to final due diligence and approved financing of the Company, the Company and/or one or more qualified investors (the “Investors”) shall advance to Tarus a convertible loan in an amount of not less than $1,750 thousand and up to $3,000 thousand (the “Loan Agreement”). As of December 31, 2020, the loan agreements have not been concluded, nor has any financing been made to Tarus. As part of such Loan Agreement, and subject to approval by the board of directors of the Company, the Investors shall have the right, within two years of the date of the Loan Agreement, to convert the outstanding convertible loan into either (i) shares of Tarus at a price per share based on a pre- money valuation of $12,500 thousand or (ii) shares of the Company’s common stock at a price per share set in accordance with an approved financing of the Company, with such terms as approved by the Company in its sole discretion. In the event the Investors elect to convert into shares of the Company’s common stock, the Company shall have the right upon notice to Tarus to receive the same number of shares of capital stock of Tarus that the Investors would have received had the Investors converted their convertible loans into shares of Tarus. Further, as part of the Loan Agreement, the Company shall advance to Tarus up to $500 thousand within fourteen days of execution of the Loan Agreement. Subject to the closing of the Loan Agreement, the Company and/or the Investors shall have an option, exercisable by sending written notice to Tarus at any time through the second anniversary of the closing of the Loan Agreement, to invest additional funds in an amount of up to $1,250 thousand and not less than $500 thousand in Tarus. The Company will also have the right to appoint and/or replace one member of board of directors of Tarus. Upon and subject to the execution of a definitive development and manufacturing agreement between the Company and Tarus (“Manufacturing and Supply Agreement”), the Company, or one or more of its affiliates, shall manufacture and supply to Tarus and any of its affiliates, licensees, assignees of interest all requirements for all cell therapy elements of any combination therapy incorporating the technology of Tarus. Following the conclusion of the clinical development stage of each product emanating from the technology of Tarus, the cell therapy component of any such product borne out of the technology of Tarus shall be exclusively supplied by the Company under the Manufacturing and Supply Agreement. If the Company and Tarus fail to sign such Manufacturing and Supply Agreement for any given Tarus product, Tarus shall pay the Company an amount equal to four percent (4%) of gross revenues derived by Tarus from such Tarus products.

Apart from the above, there was no activity in the Tarus collaboration.

g.Sponsored Research and Exclusive License Agreement with Columbia University

Effective April 2, 2019, the Company and The Trustees of Columbia University in the City of New York, a New York corporation, (“Columbia”) entered into a Sponsored Research Agreement (the “SRA”) whereby the Company will provide financial support for studying the utility of serological tumor marker for tumor dynamics monitoring. Under the terms of the SRA, the Company shall pay $300 thousand per year for three years, or for a total of $900 thousand, with payments of $150 thousand due every six months.

Effective April 2, 2019, the Company and Columbia entered into an Exclusive License Agreement (the “Columbia License Agreement”) whereby Columbia granted to the Company an exclusive license to discover, develop, manufacture, sell, and otherwise distribute certain product in the field of cancer therapy. In consideration of the licenses granted under the Columbia License Agreement, the Company shall pay to Columbia (i) a royalty of 5% of net sales of any product sold which incorporates a licensed Columbia patent and (ii) 2.5% of net sales of other products. In addition, the Company shall pay a flat $100 thousand fee to Columbia upon the achievement of each regulatory milestone.

h.IRB Approval for Liver Cell Collection

On April 29, 2019, the Company received Institutional Review Board (“IRB”) approval to collect liver biopsies from patients at Rambam Medical Center located in Haifa, Israel for a planned study to confirm the suitability of liver cells for personalized cell replacement therapy for patients with insulin-dependent diabetes resulting from total or partial pancreatectomy. The liver cells are intended to be bio-banked for potential future clinical use.

The goal of the proposed study, entitled “Collection of Human Liver Biopsy and Whole Blood Samples from Type 1 Diabetes Mellitus (T1DM), Total or Partial Pancreatectomy Patients for Potential use as an Autologous Source for Insulin Producing Cells in Future Clinical Studies,” is to confirm the suitability of the liver cells for personalized cell replacement therapy, as well as eligibility of patients to participate in a future clinical study, as defined by successful AIP cell production from their own liver biopsy. The secondary objective of the study is to evaluate patients’ immune response to AIPs based on the patient’s blood samples and followed by subcutaneous implantation into the patients’ arm which would represent the first human trial. The Company has developed a novel technology based on technology licensed from Tel Hashomer Medical Research Infrastructure and Services Ltd., utilizing liver cells as a source for AIP cells as replacement therapy for islet transplantation.

During the study, liver samples will be collected and then processed and stored in specialized, clinical grade, tissue banks for potential clinical use. The propagated cells will be maintained in a tissue bank and are intended to be utilized in a future clinical study, in which the cells will be transdifferentiated and administered back to the patients as a potential treatment. This personalized autologous process will be performed under our POC platform in which the patient liver samples are processed, cryopreserved and potentially re-injected, all in the medical center under clinical grade/GMP level conditions.

F-36

In June 2019, the Company received additional Institutional Review Board (“IRB”) approval to collect liver biopsies from patients at a leading medical center in USA for a planned study to confirm the suitability of liver cells for personalized cell replacement therapy for patients with insulin-dependent diabetes resulting from total pancreatectomy (the granted Orphan Drug Designation indication). The liver cells are intended to be bio-banked at the New York Blood Center, NYC for potential future clinical use. In October 2019, a liver sample from the first recruited patient was collected and processed and stored at the New York Blood Center, NYC in specialized, clinical grade, tissue banks for potential clinical use.

i.FDA Approval for Orphan Drug Designation for AIP Cells

On June 11, 2019, the FDA granted Orphan Drug Designation for the Company’s AIP cells as a cell replacement therapy for the treatment of severe hypoglycemia-prone diabetes resulting from total pancreatectomy (“TP”) due to chronic pancreatitis. The incidence of diabetes following TP is 100%, resulting in immediate and lifelong insulin-dependence with the loss of both endogenous insulin secretion and that of the counter-regulatory hormone, glucagon. Glycemic control after TP is notoriously difficult with conventional insulin therapy due to complete insulin dependence and loss of glucagon-dependent counter-regulation. Patients with this condition experience both severe hyperglycemic and hypoglycemic episodes.

j.Regents of the University of California

In December 2019, the Company and the Regents of the University of California (“University”) entered into a joint research agreement in the field of therapies and processing technologies according to an agreed upon work plan. According to the agreement, the Company will pay the University royalties of up to 5% (or up to 20% of sub-licensing sales) in the event of sales that includes certain types of University owned IP.

k.Caerus Therapeutics Inc (a related party)

In October 2019, the Company and Caerus Therapeutics (“Caerus”), a Virginia company, concluded a license agreement whereby Caerus granted the Company an exclusive license to all Caerus IP relating to Advance Chemeric Antigen Vectors for Targeting Tumors for the development and/or commercialization of therapiescertain licensed products. In consideration for the License granted to the Company under this Agreement, the Company shall pay Caerus feasibility fees (including the grant to purchase 70,000 options in the Company, annual maintenance fees and royalties of sales of up to 5% and up to 18% of sub-license fees. Expenses in the amount of approximately $200 thousand including the fair value of the options granted were recorded as research and development expenses. The Company also has the right to instruct Caerus to transfer the license, development, development results and any other rights and licenses granted to the Company to a joint venture (“JV”) in which Company shall have a 51% controlling ownership stake in the JV Entity. Upon Company’s election of such option, the development shall be carried out by Caerus for the JV and the royalty, sublicense fees and annual maintenance fee shall be terminated. Company may provide requisite funding for the JV Entity as determined by the Company and Caerus.

l.Extracellular Vesicle (“EV”) Technology License

During the third quarter of 2020, the Company purchased the IP and related EV technology from a service provider (the “Service Provider”) pursuant to an EV agreement (the “EV agreement”). According to the EV agreement, the Service Provider sold to the Company all of its rights in the EV technology that it had produced, in the amount of $500 thousand, to be paid in installments over the next 12 months from September 2020. The $500 thousand was recorded in R&D expenses. In addition, the Service Provider granted the Company an exclusive worldwide license to use the EV IP technology for any purpose.

F-37

m.Tamir Biotechnology acquisition

Included in the purchased assets of the Tamir Biotechnology Inc acquisition (See Note 4) was the assumption by the Company of a worldwide license to a private company of certain Tamir technologies in the field of treatment, amelioration, mitigation or prevention of diseases or conditions of the eye and its adnexa in return for certain development and sales milestone payments to be paid to Tamir. This license fee and the right to receive future milestone payments (of up to $11 million assuming that certain milestones are reached) and royalties (of up to $35 million based on liver and NK cells. The agreement will be governednet sales milestones), were assumed by a joint steering committee and carried outthe Company in accordanceconnection with the projects' work plans. UnderTamir Purchase Agreement together with a less than 10% share interest. To date, no milestones have been reached.

n.Tissue Genesis, LLC (“Tissue Genesis”)

Included in the plan, each partyKoligo acquisition (See Note 4) were the assets of Tissue Genesis. The Company is committed to paying the previous owners of Tissue Genesis up to $500 thousand upon the achievement of certain performance milestones and earn-out payments on future sales provided that in no event will generally bear its own sharethe aggregate of expenses. As of November 30, 2018,the earn-out payments exceed $4 million. To date, no milestones have been reached.

o.Joint venture agreements

Additionally, the Company has incurred $1.2 millionentered into joint venture agreements (“JVAs”) with its joint venture partners (Company and partner are referred to as “parties”) to facilitate the collaboration in the field of researchCGT development and development expenses in relation to the project. As part of the agreement, Cure Therapeutics has subcontractedCompany’s worldwide POCare network. The provisos and the table below summarize the major agreements. CGT and POCare activities covered by the JVAs include the development, marketing, clinical development, and contract manufacturing activates to CureCell, for which service revenuecommercialization of $1 million has been recognized.the Company’s and / or partner’s products within defined territories. The extent of the collaboration is set out in each agreement.

Unless otherwise stated in the table below the JVAs include the following provisos (“Provisos”):

1.The incorporation of a joint venture entity (“JVE”) in which the Company will hold between 49% and 50 % of the equity.
2.The partner will manage the joint venture activities until the JVE is incorporated.
3.The JVE will be managed by a steering committee consisting of 3 members which will act as the entity’s board of directors. The Company is entitled to appoint 1 member, the partner is entitled to appoint 1 member, and Company and partner will jointly appoint the third member.
4.The Company has the right to exercise a call option to acquire the partner’s share in the JVE based on the occurrence of certain events and according to an agreed upon mechanism.
5.The funding of the parties’ investment in the joint venture share may be made in the form of cash investment and / or in-kind services. The Company’s cash investment may be in the form of additional shares, a convertible loan, and/or procured services.
6.Each of the parties may agree to provide additional funding to the JVE to cover the operation costs and such additional funding may be in the form of in-kind contributions. The Company’s investments may be made in the form of a cash investment for additional shares, a convertible loan, and/or procured services. Procured services refer to certain services that the Company has engaged the partner or the JVE to provide the Company with, in support of Company’s activity. All results of these procured services shall be owned by Company.
7.As appropriate, the parties will grant to the JVE an exclusive or nonexclusive, sublicensable, royalty-bearing, right and license to the relevant party’s background IP as required solely to manufacture, distribute and market and sell the party’s products within the territory. Each party shall receive royalties in an amount of ten percent (10%) of the net sales generated by the JVE and/or its sublicensees.
8.Once the JVE is profitable, the Company will be entitled (in addition to any of its rights as the holder of the JVE) to an additional share of fifteen percent (15%) of the JVE’s GAAP profit after tax, over and above all rights granted pursuant to Company’s participating interest in the JVE.

F-38

Name of party (and country of origin)TerritoryNotes
Theracell Advanced BiotechnologyGreece, Turkey, Cyprus, Israel and Balkans(1)
Broaden Bioscience and Technology CorpCertain projects in China and the Middle East

Mircod LLC

(US)

Russia(2)
Image Securities FZC (UAE) (a related party)India
Cure TherapeuticsKorea and Japan
Kidney Cure LtdWorldwide(3)
Sescom LtdWorldwide(4)

Educell D.O.O

(Slovenia)

Croatia, Serbia and Slovenia

Med Centre for Gene and Cell Therapy FZ-LLC

(UAE)

UAE

Mida Biotech B.V.

(Netherlands)

Netherlands, Lithuania, Spain, Switzerland, Germany, Belgium or any other countries within West Europe(5)

First Choice International Company, Inc

Panama and certain other Latin American countries(6)
KinerjaPay CorpSingapore(7)
SBH Sciences IncWorldwide(8)
HekaBio KKJapan(9)

(1)The Theracell JVE was incorporated in Greece under the name of Theracell Laboratories Ltd. (See Note 12).
(2)Under the Mircod JVA, provisos 7 and 8 do not apply. Subject to payment by the Company ORGS of the contribution amount, the JVA will grant Company an exclusive, perpetual, irrevocable, royalty free and fully paid up and sublicensable license to use the Project IP for research and development and for the manufacturing, processing, supplying, and use of products based on point of care manufacturing and/or processing of treatments for patients and for use in hospitals, medical centers and academic institution settings solely outside the territory. The parties also, following proviso 6, concluded a convertible loan agreement pursuant to which Company shall lend Mircod up to $5 million based upon a development plan to be agreed upon. The loan bears simple interest in the amount of 6% annually. As at December 31, 2020, the development plan had not been finalized and no transfers under the loan agreement were made.
(3)Pursuant to the Kidney Cure JVA, the parties will collaborate in the (i) implementation of a point-of-care strategy; (ii) assessment of the options for development and manufacture of various cell-based types (including kidney derived cells, MSC cells, exosomes, gene therapies) development; and (iii) development of protocols and tests for kidney therapies (the “Project”). Provisos 7 and 8 do not apply to the Kidney Cure JVA. The Kidney Cure JVE was incorporated in Switzerland under the name of Butterfly Biosciences Sarl (See Note 12).
(4)Under the Sescom JVA, the parties will collaborate in the field of the assessment of relevant tools and technologies to be used in the Company’s information security system (the “ISS”); (ii) the implementation of the ISS within the Company and in the Company’s point-of-care network; and (iii) the operation and maintenance of the ISS. Provisos 7 and 8 do not apply to this JVA. Company has agreed to provide the Sescom JVE with: (a) a non-exclusive, not transferable and non-sublicensable worldwide royalty-free license to use its background IP to the extent required for carrying out certain activities by the Sescom JVE; and (b) access to its point-of-care network and relevant data to be used for the certain activities.
(5)Under the Mida JVA, commencing January 1, 2022 and thereafter Mida shall have the right to sell to Company its then issued and outstanding shares in the JVA, and if the JVA was not yet set up, its assets, contracts and liabilities relating to the project, for a consideration to be agreed between the parties in good faith, provided that such consideration is not lower than $500 thousand.
(6)Under the First Choice JVA, each party shall, subject to fulfilment of the party’s JVA, grant the Panama JV Entity an exclusive license to certain intellectual property of the part to develop and commercialize the party’s products in the territory, subject to minimum sales obligations. In consideration of such license, the Panama JV shall pay the relevant part royalties at the rate of 15% of the Panama JVE net sales of party’s products sold in the territory.

F-39

(7)No activities have taken place since the JVA was signed. According to the JVA, Company was eligible to receive 51% of the equity and 10% royalties on sales of products. The steering committee was to compromise 5 members of which Company could appoint 2, and a third member to be an industry expert, to be appointed by Orgenesis. The JVA did not include the proviso 8.
(8)Pursuant to the SBH JVA the parties will collaborate in the field of gene and cell therapy development, process and services of bio-exosome therapy products and services in the areas of diabetes, liver cells and skin applications, including wound healing. The SBH JVE has not yet been incorporated. According to the JVA, the board of directors of the SBH JVE shall be comprised of three directors with one appointed by SBH and two appointed by the Company. All intellectual property conceived or developed resulting from the business of the SBH JV Entity, that is not SBH’s or the Company’s background intellectual property, shall be owned exclusively by the SBH JV Entity, although the Company shall be granted the right to exclusively license any intellectual property arriving from the development activities of the SBH JV Entity, or exclusively distribute products based thereon. Provisos 7 and 8 do not apply to the SBH JVA.
During the third quarter of 2019, the Company transferred $50 thousand to SBH. Apart from the above, there was no material activity in the SBH Collaboration and the SBH JV entity had not been incorporated as at December 31, 2020.
(9)During the third quarter of 2020, the Company and HB agreed to terminate the license agreement. As of December 31, 2020, no activity had begun in the said JV and no investments were made therein.

NOTE 12 – INVESTMENTS IN ASSOCIATES, NET

a. On May 10, 2016,

a.Theracell Laboratories Private Company

During October 2020, the Company and AtvioTheracell, pursuant to the Greek JVA (See Note 11) incorporated the Greek JVA entity known as Theracell Laboratories Private Company (“TLABS”). The Theracell Project activities will be run through TLABS. The Company and Theracell each hold a 50% participating interest in TLABS.

b.Butterfly Biosciences Sarl

During October 2020, the Company and Kidney Cure, pursuant to the Kidney Cure JVA (See Note 11) incorporated the KC JV Entity known as Butterfly Biosciences Sarl (“BB”) in Switzerland. BB will be involved in the (i) implementation of a point-of-care strategy; (ii) assessment of the options for development and manufacture of various cell-based types (including kidney derived cells, MSC cells, exosomes, gene therapies) development; and (iii) development of protocols and tests for kidney therapies (the “BB Project”). The Company holds a 49% participating interest on BB and Kidney Cure holds the remaining 51%.

c.The table below sets forth a summary of the changes in the investments for the year ended December 31, 2020:

SCHEDULE OF CHANGES IN INVESTMENTS

  December 30, 
  2020 
  (In thousands) 
    
Opening balance $- 
Investments during the period  69 
Share in net income of associated companies  106 
Ending balance $175 

F-40

NOTE 13 – EQUITY

a.Financings

On January 20, 2020, the Company entered into joint venture agreement,a Securities Purchase Agreement (the “January Purchase Agreement”) with certain investors pursuant to which the parties agreed to collaborateCompany issued and sold, in the field of the CDMO in Israel (the “Atvio JVA”). The parties pursued the joint venture through Atvio, the Company had 50% participating interest therein in any and all rights and obligations and in any and all profits and losses. Atvio's operations commenced in September 2016.

F-34


The Company remitted to Atvio $1 million under the terms of the Atvio JVA to defray the costs associated with the setting up and the maintenance of the GMP facility. The Company’s funding was made by way of a convertible loan to Atvio, which could have been convertible at the Company’s option at any time. The Company concluded that, based on the terms of the agreement, it had the ability to exercise significant influence in Atvio, but had no control. Therefore, the investment was accounted for under the equity method. In addition, at any time following the first anniversary year of the Effective Date the Company had the option to require the Atvio’s shareholders to transfer to the Company the entirety of their interest in Atvio for the consideration specified in the agreement. Within three years from the Effective Date, the Atvio’s shareholders had the option to require the Company to purchase from Atvio's shareholders their entire interest in Atvio for the consideration based on Atvio's valuation mechanism as specified in the agreement. The above-mentioned options were accounted as derivatives and measured at fair value and was presented in the balance sheet in " put option derivative " line item (See Note 17). On June 28, 2018 the Company exercised its call options in Atvio, see also Note 4.

b. On March 14, 2016, Orgenesis Inc. and CureCell entered into a joint venture agreement (the “CureCell JVA”), pursuant to which the parties are collaborating in the field of the CDMO in Korea.

     Under the CureCell JVA, CureCell had procured, at its sole expense, a GMP facility and appropriate staff in Korea for the manufacture of the cell therapy products. The Company had to share with CureCell the Company’s know-how in the field of cell therapy manufacturing All obligations were fulfilled by the parties and each party had 50% from the participating interest and in any and all profits and losses of the joint venture. The Company remitted to CureCell $2.1 million under the terms of the CureCell JVA. On June 28, 2018 the Company exercised its call options in CureCell, see also Note 4.

c. The table below sets forth a summary of the changes in the investments for the years ended November 30, 2018 and 2017:

  November 30, 
  2018  2017 
  (In thousands) 
       
Opening balance$ 1,321 $ (12)
Reclass with short-term receivables (795) 118 
Investments during the period -  2,429 
Share in losses (731) (1,214)
Reductions due to the acquisition of CureCell and Atvio – see also Note 4 205  - 
 $ - $ 1,321 

NOTE 13 – EQUITY

a.Share Capital

     The Company’s common shares were traded on the OTCQB Venture Market under OTC Market Group’s OTCQB tier under the symbol “ORGS”. On March 13, 2018, the Company's common stock began to be listed and traded on the Nasdaq Capital Market under the symbol “ORGS.”

b.Financings

     (1) In January 2017, the Company entered into definitive agreements with an institutional investor for the private placement of 2,564,115 units of the Company’s securities for aggregate subscription proceeds to the Company of $16 million at $6.24 price per unit. Each unit is comprised of one share of the Company’s Common Stock and a one warrant, exercisable over a three-years period from the date of issuance, to purchase one additional share(the “Offering”), 2,200,000 shares of Common Stock at a purchase price of $4.20per share exercise price of $6.24 (“Unit”(the “Shares”). The subscription proceeds have been paid to the Company on a periodic basis through October 2018.

F-35


     In July 2018, the Company entered into definitive agreements with assignees of the aforementioned institutional investor whereby these assignees remitted $4.6 million in respect of the units available under the original subscription agreement that have not been subscribed for, entitling such investors to 702,307 units, with each unit being comprised of (i) one share of the Company's common stock and (ii) one three-year warrantwarrants to purchase up to an additional one share1,000,000 shares of the Company’s common stockCommon Stock at a per sharean exercise price of $6.24.$5.50 per share (the “Warrants”) which are exercisable between June 2021 and January 2023. The Company received gross proceeds of approximately $9.24 million before deducting related offering expenses in the amount of $0.8 million.

     During

b.Tamir Biotechnology, Inc.

For the year ended November 30, 2018 and 2017acquisition of Tamir, see Note 4.

As aggregate consideration for the investor and the assignees remitted toacquisition, the Company $11.5 and $4.5 million, and the Company issued 1,813,687 and 721,160 Units, respectively.

     As of November 30, 2018, 550,481 shares have not been issued and therefore the Company has recorded $2.3paid $2.5 million in receiptscash and issued an aggregate of 3,400,000 shares (the “Shares”) of Common Stock to Tamir resulting in a total consideration of $20.2 million based on account of shares to be allotted in the statement of equity.

     In connection therewith, during the year ended November 30, 2018 and 2017, the Company had transaction costs of approximately $328 and $225 thousand, respectively, out of which $121 and $253 thousand are stock-based compensation expenses due to issuance of warrants and shares. See also Note 15(d).

     (2) During the year ended November 30, 2018, the Company entered into definitive agreements with accredited and other qualified investors relating to a private placement of 1,237,642 units. Each unit is comprised of (i) one share of the Company’s common stockshare price at the closing date. $59 thousand and (ii) three-year warrant to purchase up to an additional one share of the Company’s Common Stock at a per share exercise price of $6.24, for aggregate proceeds to the Company of approximately $7.7 million.

     The transaction costs were approximately $349 thousand, out of which $125 thousand340,000 Shares are stock-based compensation due to issuance of warrants. See also 15(d).

     (3) During the year ended November 30, 2018, investors exercised 136,646 warrants into 136,646 shares of the Company’s Common Stock, for aggregate proceeds of $853 thousand.

c.Contingent Shares

     According to the share exchange agreement signed during 2015, the former shareholders of MaSTherCell received a “consideration of shares” of Orgenesis Inc. in exchange of their shares in MaSTherCell. At the time of MaSTherCell’s acquisition by the Company, there was outstanding convertible bonds issued by MaSTherCellbeing held in an amountescrow account for a period of $1.8 million (Euro 1.6 million). Under the share exchange agreement in case MaSTherCell is repaying the principal amount and the accrued interest18 months from closing to secure indemnification obligations of the convertible bonds, the former shareholders will give back to the Company a portion of the consideration shares. To that effect, the number of consideration shares to be released back to the Company, shall be determined by dividing the subscription amount of the outstanding convertible bonds plus interest owed thereunder (converted into USD according to the currency exchange rate applicable on the day of conversion) by the consideration and by applying the resulting quotient to actual total number of consideration shares received by former shareholder of MaSTherCell.

     During January 2017 MaSTherCell repaid all but one of its bondholders and the aggregate payment amounted to $1.7 million (Euro 1.5 million). AccordingTamir pursuant to the terms of the release back pursuantTamir Purchase Agreement. The share price was $5.26 at the share exchange agreement,day of the closing.

c.Koligo Therapeutics Inc.

For the acquisition of Koligo, see Note 4.

Pursuant to the terms of the Merger Agreement, at the Effective Time, the shares of capital stock of Koligo that were issued and outstanding immediately prior to the Effective Time were automatically cancelled and converted into the right to receive, subject to customary adjustments, an aggregate of 2,063,713 shares of Company returned to treasury a total of 263,148 shares. These sharescommon stock which have been retired and cancelled. See also Note 8(d)issued to Koligo’s accredited investors (with certain non-accredited investors being paid solely in cash in the amount of approximately $20 thousand). In addition, we issued 66,910 shares to Maxim Group LLC for advisory services in connection with the Merger.

d.WarrantsWarrants

A summary of the Company'sCompany’s warrants granted to investors and as findersfinder’s fees as of November 30, 2018December 31, 2020, and 2017December 31, 2019 and changes for the yearsperiods then ended is presented below:

  2018  2017 
     Weighted     Weighted 
     Average     Average 
     Exercise     Exercise 
  Number of  Price  Number of  Price 
  Warrants $   Warrants $  
Warrantsoutstanding at the beginning of the year 2,609,864  6.26  1,620,993  6.25 
Changes during the year:            
     Issued 4,488,854  6.27  1,144,647  6.32 
     Exercised (136,646) 6.24  -    
     Expired (382,414) 6.10  (155,776) 6.24 
     Cancelled** (66,667) 6.24  -    
Warrantsoutstanding and exercisable at end of the year* 6,512,991  6.27  2,609,864  6.26 

SCHEDULE OF WARRANTS ACTIVITY

  December 31, 
  2020  2019 
  

Number of

Warrants

  

Weighted

Average

Exercise Price

$

  

Number of

Warrants

  

Weighted

Average

Exercise Price

$

 

Warrants outstanding at the

beginning of the period

  6,010,087   6.35   6,286,351   6.29 
Changes during the period:                
Issued  1,344,606   5.64   471,980   6.95 
Expired  (284,452)  6.53   (748,244)  6.24 
Warrants outstanding and exercisable at end of the period** 7,070,241   6.20   6,010,087   6.35 

*As of December 31, 2020 and December 31, 2019, there are no warrants that are subject to exercise price adjustments.

F-41

 * As

e.Treasury shares

A summary of November 30, 2018,the Company’s treasury shares purchased as of December 31, 2020 and 2017, 769,411 and 1,066,691 warrants respectively, are subject to exercise price adjustmentschanges for the period then ended is presented below:

**see also Note 15(d).SCHEDULE OF TREASURY SHARES

  December 31, 
  2020 
  

Number of

Treasury Shares

  

Weighted

Average

Price Paid

$

 
Treasury Shares at the beginning of the period  -   - 
Changes during the period:        
Purchased  55,309   4.47 
Shares at end of the period  53,309   4.47 

F-36



  Year Ended 
  November 30, 
  2018  2017 
  (in thousands, 
  except per share data) 
Basic:      
   Net loss attributable to Orgenesis Inc$18,291 $12,367 
   Adjustment of redeemable non-controlling interest to redemption amount 884  - 
   Net loss attributable to Orgenesis Inc. for loss per share$ 19,637 $ 12,367 
   Weighted average number of common shares outstanding 13,374,103  9,679,964 
   Basic loss per common share$ 1.43 $ 1.28 
Diluted:      
   Net loss attributable to Orgenesis Inc. for loss per share$ 19,175  12,367 
   Changes in fair value of embedded derivative and interest expenses on convertible note -  392 
   Loss for the year$ 19,175 $ 12,759 
       
   Weighted average number of shares used in the computation of basic loss per share 13,374,103  9,679,964 
   Number of dilutive shares related to convertible note -  34,288 
   Weighted average number of common shares outstanding 13,374,103  9,714,252 
       
Diluted loss per common share$ 1.43 $ 1.31 

NOTE 14 – LOSSINCOME (LOSS) PER SHARE

The following table sets forth the calculation of basic and diluted loss per share for the periods indicated:

SCHEDULE OF BASIC AND DILUTED LOSS PER SHARE

  2020  2019 
  Year ended December 31, 
  2020  2019 
  (in thousands, except per share data) 
Basic and diluted:        
Net loss from continuing operations attributable to Orgenesis Inc. $95,088  $22,490 
         
Net (income) loss from discontinued operations attributable to Orgenesis Inc. for loss per share  (96,198)  1,631 
Adjustment of redeemable non-controlling interest to redemption amount  (5,160)  4,095 
Basic: Net income (loss) available to common stockholders  (101,358)  5,726 
         
Net (income) loss attributable to Orgenesis Inc. for loss per share  (6,270)  28,216 
         
Weighted average number of common shares outstanding  21,320,314   15,907,995 
Loss per common share from continuing operations $4.46  $1.41 
Net (income) loss common share from discontinued operations $(4.75) $0.36 
Net (income) loss per share $(0.29) $1.77 

For the year ended November 30, 2018,December 31, 2020, and December 31, 2019, all outstanding convertible notes, options and warrants have been excluded from the calculation of the diluted net loss per share since their effect was anti-dilutive.

     For the year ended November 30, 2017, all Diluted loss per share does not include 10,212,789 shares underlying outstanding options and warrants and 1,057,7851,630,857 shares upon conversion of convertible notes have been excluded fromloans for the calculationyear ended December 31, 2020, because the effect of their inclusion in the diluted net loss per share since their effect wascomputation would be anti-dilutive.

F-42

NOTE 15– 15 – STOCK-BASED COMPENSATION

a.Global Share Incentive PlanGlobal Share Incentive Plan

On May 11, 2017, the annual meeting of the Company’s stockholders approved the 2017 Equity Incentive Plan (the “2017 Plan”) under which, the Company had reserved a pool of 1,750,000 shares of the Company’s common stock, which may be issued at the discretion of the Company'sCompany’s board of directors from time to time. Under this Plan, each option is exercisable into one share of common stock of the Company. The options may be exercised after vesting and in accordance with the vesting schedule that will be determined by the Company'sCompany’s board of directors for each grant. The maximum contractual life term of the options is 10 years. At the Company’s annual meeting of stockholders on November 26, 2019 the Company’s stockholders approved an amendment to increase the number of shares authorized for issuance of awards under the Company’s 2017 Equity Incentive Plan from 1,750,000 shares to an aggregate of 3,000,000 shares of Common Stock. As of November 30, 2018,December 31, 2020, total options granted under this plan are 1,040,942,1,362,133 and the total options that are available for grants under this plan are 709,058.1,724,966.

F-37


On May 23, 2012, the Company'sCompany’s board of directors adopted the Global Share Incentive Plan 2012 (the “2012 Plan"Plan”) under which, the Company had reserved a pool of 1,000,000 shares of the Company’s common stock, which may be issued at the discretion of the Company'sCompany’s board of directors from time to time. Under this plan, each option is exercisable into one share of common stock of the Company. The options may be exercised after vesting and in accordance with the vesting schedule that will be determined by the Company'sCompany’s board of directors for each grant. The maximum contractual life term of the options is 10 years. As of November 30, 2018,December 31, 2020, total options granted under this plan are 699,991,1,183,182 and the total options that are available for grants under this plan are 300,009.248,024.

b.Options Granted to Employees and DirectorsOptions Granted to Employees and Directors

Below is a table summarizing all of the options grants to employees and Directors made during the years ended November 30, 2018,December 31, 2020, and 2017:December 31, 2019:

 Year ofNo. of optionsExercise priceVesting periodFair value at grantExpiration
 grantgranted  (in thousands)period
Employees2018762,400$4.42-$8.91vest immediately-4 years$4,23310 years
Directors2018113,800$5.991 year$50710 years
Directors2017166,668$4.802 years$55810 years
Employees2017525,005$4.8,$7.2vest immediately-4 years$1,91510 years

SCHEDULE OF EMPLOYEE STOCK OWNERSHIP PLAN DISCLOSURES

  Year Ended No. of options
granted
  Exercise price  Vesting period 

Fair value at grant

(in thousands)

  Expiration
period
Employees December 31, 2020  531,450   $2.99-$6.84  Quarterly over a period of two years $1,312  10 years
Directors December 31, 2020  145,050   $2.99-$4.7  96% on the one-year anniversary, and the remaining 4% in three equal instalments on the first, second and third year anniversaries $377  10 years
Employees December 31, 2019  94,500   $3.14-$5.07  Quarterly over a period of two years $322  10 years
Directors December 31, 2019  50,000  $2.99  One-year anniversary $103  10 years

The fair value of each stock option grant is estimated at the date of grant using a Black Scholes option pricing model. The volatility is based on historical volatility of the Company, by statistical analysis of the weekly share price for past periods based on expected term. The expected option term is calculated using the simplified method,as the Company concludes that its historical share option exercise experience does not provide a reasonable basis to estimate its expected option term.

The fair value of each option grant is based on the following assumptions:

SCHEDULE OF STOCK OPTIONS, VALUATION ASSUMPTIONS

  Year Ended December 31, 
  2020  2019 
Value of one common share $2.99-$6.84  $2.99-$5.07 
Dividend yield  0%  0%
Expected stock price volatility  80%-86%  83%-88%
Risk free interest rate  0.36%-1.71%  1.45%-2.47%
Expected term (years)  5.50-6.00   5.38-5.56 

  Year Ended November 30,
 20182017
Value of one common share$4.42-$8.7$4.68,$7.2
Dividend yield0%0%
Expected stock price volatility88%-98%93.8%-95.4%
Risk free interest rate2.33%-3.2%1.89%-1.76%
Expected term (years)4.13-105
F-43

A summary of the Company'sCompany’s stock options granted to employees and directors as of November 30, 2018December 31, 2020 and 2017 and changes for the years then endedDecember 31, 2019 is presented below:

  2018  2017 
     Weighted     Weighted 
     Average     Average 
     Exercise     Exercise 
  Number of  Price  Number of  Price 
  Options $   Options $  
Options outstanding at the beginning of the year 1,605,055  3.11  978,853  1.92 
Changes during the year:            
     Granted 876,200  7.13  691,673  5.28 
     Expired (61,463) 5.26  (38,106) 7.58 
     Forfeited (43,365) 4.68  (27,365) 4.8 
Options outstanding at end of the year 2,376,427  4.51  1,605,055  3.11 
Options exercisable at end of the year 1,504,542  2.91  1,135,107  2.57 

F-38



SCHEDULE OF STOCK OPTIONS ACTIVITY

  Year Ended December 31 
  2020  2019 
  

Number of

Options

  

Weighted

Average

Exercise Price

$

  

Number of

Options

  

Weighted

Average

Exercise

Price

$

 
Options outstanding at the beginning of the period  2,465,522   4.44   2,376,427   4.51 
Changes during the period:                
Granted  676,500   3.74   144,500   4.15 
Exercised  -  -   -   - 
Expired  (11,876)  7.88   (16,750)  6.01 
Forfeited  (57,042)  4.52   (38,655)  7.11 
Cancelled  (155,437)  8.38   -   - 
Options outstanding at end of the period  2,917,667   4.05   2,465,522   4.44 
Options exercisable at end of the period  2,299,937   4.03   2,112,567   4.21 

The following table presents summary information concerning the options granted and exercisable to employees and directors outstanding as of November 30, 2018:December 31, 2020 (in thousands, except per share data):

     Weighted          
     Average  Aggregate     Aggregate 
Exercise Number of  Remaining  Intrinsic  Number of  Exercisable 
Price Outstanding  Contractual  Value  Exercisable  Options 
$ Options  Life $   Options  Value $ 
        (in thousands)     (in thousands) 
0.0012 462,015  4.78  2,776  462,015  1 
0.012 278,191  3.18  1,669  278,191  3 
4.42 50,000  9.02  79  37,500  166 
4.8 524,999  8.03  635  440,104  2,112 
5.99 384,050  9.35  8  32,750  196 
6 33,334  5.67  -  33,334  200 
7.2 83,334  8.51  -  83,334  600 
8.36 250,000  9.58  -     - 
8.43 160,994  9.46  -  14,492  156 
8.91 30,500  9.55  -  3,813  34 
9 20,834  4.63  -  20,834  188 
9.48 58,908  3.61  -  58,908  558 
10.2 39,268  3.51  -  39,267  401 
  2,376,427  7.15  5,167  1,504,542  4,598 

SCHEDULE OF STOCK OPTIONS EXERCISABLE

Exercise

Price

$

  

Number of

Outstanding

Options

  

Weighted Average

Remaining

Contractual

Life

  

Aggregate

Intrinsic

Value

$

  

 

 

Number of

Exercisable

Options

  

Aggregate

Exercisable

Options

Value $

 
         (in thousands)     (in thousands) 
 0.0012   230,189   3.64   1,036   230,189   0 
 0.012   510,017   1.09   2,289   510,017   6 
 2.99   445,013   9.15   672   174,208   521 
 3.14   3,750   6.27   5   1,875   6 
 4.42   50,000   6.93   4   50,000   221 
 4.5   34,000   8.47   -   23,938   108 
 4.6   185,300   9.96   -   -   - 
 4.7   6,250   9.03   -   -   - 
 4.8   483,337   5.94   -   483,337   2,320 
 5.07   53,250   8.08   -   39,750   202 
 5.1   63,000   9.68   -   7,875   40 
 5.99   352,550   7.26   -   290,488   1,740 
 6   16,667   3.59   -   16,667   100 
 6.84   17,000   9.38   -   4,250   29 
 7.2   83,334   6.43   -   83,334   600 
 8.36   250,001   7.50   -   250,001   2,090 
 8.91   15,000   7.46   -   15,000   134 
 9   20,834   2.54   -   20,834   187 
 9.48   58,908   1.52   -   58,908   558 
 10.2   39,267   1.42   -   39,267   401 
     2,917,667   5.98   4,006   2,299,937   9,263 

Costs incurred with respect to stock-based compensation for employees and directors for the years ended November 30, 2018December 31, 2020 and 2017December 31, 2019 were $2,426 $1,470 thousand and $1,536 $2,107 thousand, respectively.respectively, out of which $450 thousand and $360 thousand related to options granted to employees of Masthercell Global, respectively, and presented as part of net loss from discontinued operations in the consolidated statements of comprehensive loss. As of November 30, 2018,December 31, 2020, there was $3,783 thousand$1,594 thousands of unrecognized compensation costs related to non-vested employees and directors stock options, to be recorded over the next 3.89 2.02 years.

F-44

c.Options Granted to Consultants and service providers

c.Options Granted to Consultants and service providers

Below is a table summarizing all the compensation granted to consultants and service providers during the years ended November 30, 2018December 31, 2020 and 2017:December 31, 2019 and for the one-month period ended December 31, 2019:

     Fair value at 
 Year ofNo. of optionsExercise grantExpiration
 grantgrantedpriceVesting period(in thousands)period
Non-employees2018102,763$4.42-$8.34vest immediately-4 years$44410 years
Non-employees201716,668$4.8Quarterly over a period of one year$6810 years

SCHEDULE OF STOCK OPTIONS GRANTED TO CONSULTANTS

  
Year of grant
 
No. of options
granted
  
Exercise price
  Vesting period 

Fair value at grant

(in thousands)

  
Expiration
period
Non-employees
 2020
  62,500  $2.99-$6.84  Quarterly over a period of two years $209  10 years
Non-employees
 2019
  128,336  $ 3.14-$7  Vest immediately-5 years $394  10 years

 

The fair value of options granted during 20182020 and 20172019 to consultants and service providers, was computed using the Black-Scholes model. The fair value of each stock option grant is estimated at the date of grant using a Black Scholes option pricing model. The volatility is based on historical volatility of the Company, by statistical analysis of the weekly share price for past periods based on the expected term period, the expected term is the contractual term of each grant. The expenses are subsequently adjusted to fair value at the end of each reporting period until such options vest, and the fair value of such instruments, as adjusted, is expensed over the related vesting period.

The underlying data used for computing the fair value of the options are as follows:

F-39



  Year Ended November 30,  
   2018  2017  
Value of one common share $4.42-$8.34  $3.6-$7.44 
Dividend yield 0%  0% 
Expected stock price volatility 91%-95%  87%-95% 
Risk free interest rate 2.33%-3.20%  1.19%-1.89% 
Expected term (years) 9.79-10  4-5 

SCHEDULE OF STOCK OPTIONS, VALUATION ASSUMPTIONS

  Year Ended December 31, 
  2020  2019 
Value of one common share $ 2.99-$6.84  $3.14-$5.07 
Dividend yield  0%  0%
Expected stock price volatility  86%-89%   89%-92%
Risk free interest rate  0.73%-1.12%   1.52%-2.62%
Expected term (years)  10   10 

A summary of the status of theCompany’s stock options granted to consultants and service providers as of November 30, 2018,December 31, 2020, and 2017 and changes for the years then endedDecember 31, 2019 is presented below:

  2018  2017 
     Weighted     Weighted 
     Average     Average 
     Exercise     Exercise 
  Number of  Price  Number of  Price 
  Options $   Options $  
Options outstanding at the beginning of the year 399,380  7.47  441,621  6.24 
Changes during the year:            
   Granted 102,763  4.92  16,668  4.80 
   Expired -  -  (58,909) 8.28 
   Forfeited (15,500) -  -  - 
   Cancelled (16,669) 7.02       
Options outstanding at end of the year 469,974  5.75  399,380  7.47 
Options exercisable at end of the year 436,640  5.75  379,712  5.76 

SCHEDULE OF STOCK OPTIONS ACTIVITY

  Year Ended December 31, 
  2020  2019 
  

Number of

Options

  

Weighted

Average

Exercise

Price

$

  

Number of

Options

  

Weighted

Average

Exercise

Price

$

 
Options outstanding at the
beginning of the year
  598,310   5.76   469,974   5.75 
Changes during the year:                
Granted  62,500   3.97   128,336   5.65 
Exercised  (83,334)  3.60   -   - 
Forfeited  (8,335)  5.99   -   - 
Cancelled  (20,000)  5.30   -   - 
Options outstanding at end of the year  549,141   5.89   598,310   5.76 
Options exercisable at end of the year  450,972   6.28   539,515   5.88 

F-45

 

The following table presents summary information concerning the options granted and exercisable to consultants and service providers outstanding as of November 30, 2018December 31, 2020 (in thousands, except per share data):

     Weighted          
     Average  Aggregate  Number of  Aggregate 
Exercise Number of  Remaining  Intrinsic  Exercisable  Exercisable 
Price Outstanding  Contractual  Value*  Options  Options 
$ Options  Life $      Value $ 
        (in thousands)     (in thousands) 
3.36 136,775  7.41  362  136,775  460 
3.6 83,334  7.25  201  83,334  300 
4.42 10,325  9.02  16  10,325  46 
4.8 16,667  8.03  20  16,667  80 
5.3 35,000  9.79  25  20,000  106 
5.99 25,005  9.9     13,337  80 
6 90,000  5.67     90,000  540 
6.4 8,333  9.13     1,667  11 
7.32 8,334  3.98     8,334  61 
8.34 8,600  9.61     8,600  72 
11.52 8,334  4.35     8,334  96 
16.8 39,267  3.37     39,267  660 
  469,974  6.83  624  436,640  2,512 

SCHEDULE OF STOCK OPTIONS EXERCISABLE

Exercise

Price

$

  

Number of

Outstanding

Options

  

Weighted

Average

Remaining

Contractual

Life

  

Aggregate

Intrinsic

Value*

$

  

Number of

Exercisable

Options

  

Aggregate

Exercisable

Options

Value $

 
         (in thousands)     (in thousands) 
 2.99   35,000   9.22   53   -   - 
 3.14   15,000   8.91   20   -   - 
 3.36   136,775   5.32   156   136,775   460 
 4.09   25,000   8.76   10   25,000   102 
 4.42   10,325   6.93   1   10,325   46 
 4.5   13,335   8.53   -   -   - 
 4.6   20,000   9.96   -   -   - 
 4.8   16,668   5.94   -   16,668   80 
 5.07   5,000   8.19   -   1,000   5 
 5.3   15,000   7.70   -   15,000   80 
 5.99   16,670   7.81   -   16,670   100 
 6   90,000   3.59   -   90,000   540 
 6.84   7,500   9.38   -   -   - 
 7   70,000   8.83   -   70,000   490 
 7.32   8,334   1.89   -   8,334   61 
 8.34   8,600   7.52   -   8,600   72 
 8.43   8,333   7.05   -   4,999   42 
 11.52   8,334   2.26   -   8,334   96 
 16.8   39,267   1.28   -   39,267   660 
     549,141   6.18   240   450,972   2,834 

Costs incurred with respect to options granted to consultants and service providers for the yearyears ended November 30, 2018December 31, 2020 and 2017 was $331December 31, 2019 were $113 thousand and $322$330 thousand, respectively. As of November 30, 2018,December 31, 2020, there was $157 thousand$231 thousands of unrecognized compensation costs related to non-vested consultants and service providers, to be recorded over the next 3.134.58 years.

d.Warrants and Shares Issued to Non-Employees

F-40The fair value of Common Stock issued was the share price of the shares issued at the day of grant.


d.Warrants

1) On January 20, 2020, the Company entered into a Securities Purchase Agreement (the “January Purchase Agreement”) with certain investors pursuant to which the Company issued and Shares Issuedsold, in a private placement (the “Offering”), 2,200,000 shares of Common Stock at a purchase price of $4.20 per share (the “Shares”) and warrants to Non-Employeespurchase up to 1,000,000 shares of Common Stock at an exercise price of $5.50 per share (the “Warrants”) which are exercisable between June 2021 and January 2023. The Company received gross proceeds of approximately $9.2 million before deducting related offering expenses in the amount of $0.8 million. The fair value of those warrants as of the date of grant using the Black-Scholes valuation model was $1,911 thousand.

     1)

2) On January 2, 2020, the Company entered into private placement subscription agreements with investors for an aggregate amount of $250 thousand of convertible loans. The lenders shall be entitled, at any time prior to or no later than the maturity date, to convert the outstanding amount, into shares of Common Stock of the Company at a conversion price per share equal to $7.00. In addition, the Company granted the investors 151,428 warrants to purchase an equal number of additional shares of the Company’s Common Stock at a price of $7.00 per share. The fair value of those warrants as of the date of grant using the Black-Scholes valuation model was $210 thousand.

3) During the year ended December 31, 2020, the Company granted to several consultants 193,178 warrants each exercisable between $3.14 and $5.34 per share for three years. The fair value of those options as of the date of grant using the Black-Scholes valuation model was $378 thousand, out of which $350 thousand is related to 179,428 warrants granted as a success fee with respect to the issuance of the convertible notes and private Investment.

F-46

4) During the year ended December 31, 2019, the Company granted to several consultants 88,499 warrants each exercisable between $4.3 and $7.00 per share for three years. The fair value of those options as of the date of grant using the Black-Scholes valuation model was $155 thousand, out of which $97 thousand is related to 57,142 warrants granted as a success fee with respect to the issuance of the convertible notes.

5) In September 2019, the Company entered into an investor relation services, marketing and related services agreement. Under the terms of the agreement, the Company agreed to issue the consultant 40,174 shares of restricted common stock, of which the first 20,087 shares will be held in escrow by the Company until the six months anniversary of the agreement and 20,087 shares will be issued on the six months anniversary of the agreement to be held in escrow by the company until the one-year anniversary of the agreement. The fair value of the shares was $178 thousand using the fair value of the shares on the grant date. $96 and 82 thousand was recognized during the year ended December 31, 2020 and December 31, 2019, respectively.

6) In March 2019, the Company issued First Choice 525,000 shares of Common Stock. The value of Common Stock issued in the amount of $2.6 million were charged to research and development expenses during the year ended December 31, 2019.

7) In December 2018, the Company entered into an investor relation services, marketing and related services agreement. Under the terms of the agreement, the Company agreed to issue the consultant 10,000 shares of restricted common stock, of which the first 2,500 shares vested on the signing date, and 7,500 shares are to vest monthly over 3 months commencing January 2019. As of December 31, 2019, 10,000 shares were fully vested. The fair value of the shares was $51 thousand using the fair value of the shares on the vesting dates. $37 thousand was recognized during the year ended December 30, 2019.

8) In December 2018, the Company entered into a separate investor relations services, marketing and related services agreement. Under the terms of the agreement, the Company agreed to issue the consultant 40,000 shares of restricted common stock, of which the first 6,667 shares vested on the signing date, and 33,333 shares vested monthly over five months commencing January 2019. As of December 31, 2019, 40,000 shares were fully vested. The fair value of the shares was $200 thousand using the fair value of the shares at the vesting dates. $163 thousand was recognized during the year ended December 30, 2019.

9) During the year ended November 30, 2018, the Company granted to several consultants 78,782 warrants each exercisable between $6.24$6.24 and $15.41$15.41 per share for three years. The fair value of those warrants as of the date of grant using the Black-Scholes valuation model was $350$350 thousand. The warrants granted as a success fee with respect to private placement and the issuance of convertible loans.

     2) In December 2017, the Company entered into investor relations services, marketing and related services agreements. Under the terms of the agreement, the Company agreed to grant the consultants a total of 195,000 shares of restricted common stock, out of which the first 50,000 shares will vest after 30 days from the signing date, and 145,000 shares are to vest monthly over 15 months commencing February 2018. As of November 30, 2018, 140,000 shares were vested. The fair value of the shares as of the date of grant was $1,439 thousand.

     3)10) In January 2018, the Company entered into a consulting agreement with a financial advisor for a period of one year. Under the terms of the agreement, the consultant was entitled to receive $60$60 thousand and 19,000 units of the Company securities. Each unit is comprised of (i) one share of the Company’s common stock and (ii) a three-yearthree-year warrant to purchase up to an additional one share of the Company’s Common Stock at a per share exercise price of $6.24.$6.24. The fair value of the units as of the date of grant was $171$171 thousand, out of which $62$62 thousand reflect the fair value of the warrants using the Black-Scholes valuation model. In July 2018, the board approved an additional issuance of 6,629 shares and three-yearthree-year warrants to purchase up to 6,629 shares of the Company’s Common Stock at a per share exercise price of $6.24.$6.24. The fair value of the units as of the date of grant was $88$88 thousand.

     4) During the year ended November 30,

11) In December 2017, the Company grantedentered into investor relations services, marketing and related services agreements. Under the terms of the agreement, the Company agreed to severalgrant the consultants 53,148 warrants each exercisable at $6.24 or $10.20 per share for three years.a total of 195,000 shares of restricted common stock, out of which the first 50,000 shares will vest after 30 days from the signing date, and 145,000 shares are to vest monthly over 15 months commencing February 2018. As of December 31, 2019, all shares were vested. The fair value of those optionsthe shares as of the date of grant usingwas $1,439 thousand.

12) During the Black-Scholes valuation model was $211 thousand, outtwelve months ended December 31, 2020, the Company issued 270,174 shares of which $169 thousand is relatedcommon stock to 38,001 warrants granted as a success fee with respect to the issuanceservice providers. As of the convertible notes.December 31, 2020, 30,000 shares have additional restrictions on transfer until such services have been provide.

F-47

NOTE 16 – TAXES

a.

a.Corporate taxation in the U.S.

The corporate U.S.

     The applicable corporate Federal Income tax rate forapplicable to the Company and the U.S.its US subsidiaries is 21% following the U.S. Tax Cuts and Jobs Act (the “TCJA”), excluding state tax and local tax. On December 22, 2017, the TCJA was signed into law, which among other changes reduced the federal corporate income tax rate from 35% to 21%, effective January 1, 2018..

As of November 30, 2018,December 31, 2020, the Company has an accumulated tax loss carryforward of approximately $19 $ 18 million (as of November 30,December 31, 2019, approximately $34million).

For U.S. federal income tax purposes, net operating losses (“NOLs”) arising in tax years beginning after December 31, 2017, approximately $12.8 million). Under U.S.the Internal Revenue Code of 1986, as amended (the “Code”) limits the ability to utilize NOL carryforwards to 80% of taxable income in tax laws,years beginning after December 31, 2020. In addition, NOLs arising in tax years ending after December 31, 2017 can be carried forward indefinitely, but carryback is generally prohibited. NOLs generated in tax years beginning before January 1, 2018 will not be subject to certain limitations,the taxable income limitation, and NOLs generated in tax years ending before January 1, 2018 will continue to have a two-year carryback and twenty-year carryforward period. Deferred tax losses expire 20 yearsassets for NOLs will need to be measured at the applicable tax rate in effect when the NOL is expected to be utilized. The changes in the carryforward/carryback periods as well as the new limitation on use of NOLs may significantly impact the Company’s valuation allowance assessments for NOLs generated after the year in which incurred. UtilizationDecember 31, 2017.

In addition, utilization of the U.S. net operating lossesNOLs may be subject to substantial annual limitation under Section 382 of the Code due to an “ownership change” within the "changemeaning of Section 382(g) of the Code. An ownership change, subjects pre-ownership change NOLs carryforwards to an annual limitation, which significantly restricts the ability to use them to offset taxable income in ownership"periods following the ownership change. In general, the annual use limitation equals the aggregate value of the Company’s stock at the time of the ownership change multiplied by a specified tax-exempt interest rate.

On March 27, 2020, the Coronavirus Aid, Relief and Economic Security Act (the “CARES Act”) was enacted into law. The CARES Act is aimed at providing emergency relief and health care for individuals and businesses affected by the COVID-19 pandemic. The CARES Act, among other things, includes provisions related to refundable payroll tax credits, deferral of the employer portion of social security payments, expanded net operating loss application, modifications to the net interest deduction limitations, and technical corrections to tax depreciation methods for qualified improvement property. The CARES act allowed the Company to utilize 100% of NOLs arising in tax years after December 31, 2017. The Company assess all other provisions of the Internal Revenue Code of 1986CARES Act and similar state provisions. The annual limitation may result innotes no other material impact to the expiration of net operating losses before utilization.Company.

b.Corporate taxation in IsraelCorporate taxation in Israel

The Israeli Subsidiaries are taxed in accordance with Israeli tax laws. The corporate tax ratesrate applicable to 20182020 and 20172019 are 23% and 24% respectively..

As of November 30, 2018,December 31, 2020, the Israeli Subsidiaries has an accumulated tax loss carryforward of approximately $7.3$11 million (as of November 30, 2017,December 31, 2019, approximately $5.8$10 million). Under the Israeli tax laws, carryforward tax losses have no expiration date.

c.Corporate taxation in BelgianCorporate taxation in Belgium

The Belgian SubsidiariesSubsidiary are taxed according to Belgian tax laws. The corporate tax raterates applicable to 2020, 2019- 2018 2019 are 25% and 2017 are 25%29.58%, 29.58% and 34%.respectively.

F-41


As of November 30, 2018,December 31, 2020, the Belgian SubsidiariesSubsidiary has an accumulated tax loss carryforward of approximately $5.9 $ 8 million (€5.2 6 million), (as of November 30, 2017 $15.8 December 31, 2019 $6 million). Under the Belgian tax laws there is are limitation on accumulated tax loss carryforward deductions of Euro 1 million per year.

F-48

d.Corporate taxation in KoreaCorporate taxation in Korea

The basic Korean corporate tax rates are currently: 10% on the first KRW 200 million of the tax base, 20% up to KRW 20 billion, , 22% up to KRW 300 billion and 25% for tax base above KRW 300 billion. In addition, the local income tax rate is 1% on the first KRW 200 million of taxable income, 2% on taxable income over KRW 200 million up to KRW 20 billion, 2.2% of taxable income over KRW 20 billion up to 300 billion and 2.5% on taxable income over KRW 300 billion.

As of November 30, 2018, CureCellDecember 31, 2020, the Korean subsidiary has an accumulated tax loss carryforward of approximately $3.2 $ 4 million (KRW 3,421 3,813 million), (as of December 31, 2019, approximately $3 million). Under the Korean tax laws accumulated tax loss can be carry forwarded for 15 years.

e.Deferred TaxesDeferred Taxes

The following table presents summary of information concerning the Company’s deferred taxes as of the periodsyears ending November 30, 2018December 31, 2019 and 2017December 31, 2019 (in thousands):

  November 30, 
  2018  2017 
  (U.S dollars in thousands) 
Net operating loss carry forwards$8,868 $ 11,893 
Research and development expenses 2,915  1,065 
Employee benefits 181  180 
Property and equipment (47) (61)
Deferred income (117) (292)
Intangible assets (4,142) (5,117)
Less: Valuation allowance (9,235) (8,358)
Net deferred tax liabilities$ (1,702)$ (690)

SCHEDULE OF DEFERRED TAX ASSETS

  2020  2019 
  December 31, 
  2020  2019 
  (U.S. dollars in thousands) 
Net operating loss carry forwards $9,606  $14,033 
Research and development expenses  1,684   1,358 
Equity compensation  2,747   - 
Employee benefits  252   228 
Leases asset  533   - 
Lease liability  (324)  - 
Intangible assets  (2,863)  (737)
Other  297   (1)
Less: Valuation allowance  (11,932)  (14,939)
Net deferred tax liabilities $-  $(58)

Realization of deferred tax assets is contingent upon sufficient future taxable income during the period that deductible temporary differences and carry forwards losses are expected to be available to reduce taxable income. As the achievement of required future taxable income is not considered more likely than not achievable, the Company and all its subsidiaries except MaSTherCell, Atvio and CureCellthe Korean Subsidiary (previously CureCell) have recorded full valuation allowance.

The changes in valuation allowance are comprised as follows:

  Year Ended November 30, 
  2018  2017 
  (U.S dollars in thousands) 
Balance at the beginning of year$ (8,358)$ (5,151)
Additions during the year (877) (3,207)
Balance at end of year$ (9,235)$ (8,358)

f.Reconciliation of the Theoretical Tax Expense to Actual Tax Expense

SCHEDULE OF VALUATION ALLOWANCE, ACTIVITY

  December 31, 
  2020  2019 
  (U.S dollars in thousands) 
Balance at the beginning of year $(14,939) $(10,254)
Change during the year  3,007   (4,685)
Balance at end of year $(11,932) $(14,939)

f.Reconciliation of the Theoretical Tax Expense to Actual Tax Expense

The main reconciling item between the statutory tax rate of the Company and the effective rate is the provision for full valuation allowance with respect to tax benefits from carry forward tax losses and changes in cooperate tax rate in the U.S and Belgium.losses.

g.Uncertain Tax Provisions

g.Uncertain Tax Provisions

ASC Topic 740, “Income Taxes” requires significant judgment in determining what constitutes an individual tax position as well as assessing the outcome of each tax position. Changes in judgment as to recognition or measurement of tax positions can materially affect the estimate of the effective tax rate and consequently, affect the operating results of the Company. As of November 30, 2018,December 31, 2020, the Company has not accrued a provision for uncertain tax positions.

F-42


NOTE 17 - FAIR VALUE PRESENTATION

     The Company measures fair value and discloses fair value measurements for financial assets and liabilities. Fair value is based on the price that would be received to sell an asset or paid to transfer a liability in an orderly transaction between market participants at the measurement date. The accounting standard establishes a fair value hierarchy that prioritizes observable and unobservable inputs used to measure fair value into three broad levels, which are described below:

  • Level 1: Quoted prices (unadjusted) in active markets that are accessible at the measurement date for assets or liabilities. The fair value hierarchy gives the highest priority to Level 1 inputs.
  • Level 2: Observable inputs that are based on inputs not quoted on active markets but corroborated by market data.
  • Level 3: Unobservable inputs are used when little or no market data is available. The fair value hierarchy gives the lowest priority to Level 3 inputs.

     In determining fair value, the Company utilizes valuation techniques that maximize the use of observable inputs and minimize the use of unobservable inputs, to the extent possible, and considers credit risk in its assessment of fair value.

     As of November 30, 2017, the Company’s liabilities that are measured at fair value and classified as level 3 fair value are as follows (in thousands):

November 30,
2017
Level 3
Embedded derivatives convertible loans *(37)
CALL/PUT option derivative(339)F-49

* The embedded derivative is presented in the Company's balance sheets on a combined basis with the related host contract (the convertible loans).

The fair value is determined by using a Black-Scholes Model.NOTE 17 – REVENUES

Disaggregation of Revenue

The following table presentsdisaggregates the assumptions that were used forCompany’s revenues by major revenue streams.

SCHEDULE OF DISAGGREGATION OF REVENUE

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Revenue stream:        
POC and hospital services $6,068  $3,109 
Cell process development services  1,584   790 
Total $7,652  $3,899 

POC development services are the models asresult of November 30,2017:

      EmbeddedPut Option
      DerivativeDerivative
Fair value of shares of common stock$ 4.38 
Expected volatility77%54%
Discount on lack of marketability-12%
Risk free interest rate1.21%-1.39%1.44%
Expected term (years)0.17-0.420.5
Expected dividend yield0% 

agreements between Company and its partners (See Note 11). The table below sets forth a summaryCompany provides certain services in support of the changes in the fair value of the Company’s financial liabilities classified as Level 3 for the year ended November 30, 2018:

  Embedded  Put Option 
  Derivatives  Derivative 
       
Balance at beginning of the period$ 37 $ (339)
Repayment (14) - 
Changes in fair value during the period (23) 49 
Option disposal -  290 
Balance at end of the period$ - $ - 

F-43



(*) There were no transfers to Level 3 during the twelve months ended November 30, 2018.

     The table below sets forth a summary of the changes in the fair value of the Company’s financial liabilities classified as Level 3 for the year ended November 30, 2017:

  Embedded  Convertible  Put Option 
  Derivatives  Bonds  Derivative 
          
Balance at beginning of the year$ 240 $ 1,818 $ 273 
Repayment (876) (1,827) - 
Changes in fair value during the period 662  22  (612)
Translation adjustments 11  (13) - 
Balance at end of the year$ 37 $ - $ (339)

(*) There were no transfers to Level 3 during the twelve months ended November 30, 2017.

partners’ clinical activity. The Company has performed a sensitivity analysissigned Master Services Agreements with joint venture partners in the aggregate amount of over $38 million for services to be provided from 2021 to 2022.

A breakdown of the resultsrevenues per customer what constituted at least 10% of revenues is as follows:

SCHEDULE OF BREAKDOWN OF REVENUES PER CUSTOMER

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
Revenue earned:        
Customer A $2,857  $1,420 
Customer B  1,577   - 
Customer C – related party  1,475   1,270 
Customer D  1,412   857 

Contract Assets and Liabilities

Contract assets are mainly comprised of trade receivables net of allowance for the Put Option Derivative fair value as of November 30, 2017 with the following parameters:doubtful debts, which includes amounts billed and currently due from customers.

The activity for trade receivables is comprised of:

SCHEDULE OF ACTIVITY FOR TRADE RECEIVABLES

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
       
Balance as of beginning of period $1,831  $129 
Acquisition of Koligo  228   - 
Additions  6,997   2,079 
Collections  (5,982)  (364)
Exchange rate differences  11   (13)
Balance as of end of period $3,085  $1,831 

  Base -50%  Base  Base+50% 
  (in thousands) 
Sensitivity analysis due to changes in the assumptions expected volatility$ 335 $ 339 $ 379 
Sensitivity analysis due to changes in Atvio's FV 252  339  440 
F-50

The activity for contract liabilities is comprised of:

SCHEDULE OF ACTIVITY FOR CONTRACT LIABILITIES

  Year Ended December 31, 
  2020  2019 
  (in thousands) 
       
Balance as of beginning of period $325  $56 
Additions  597   1,126 
Realizations*  (862)  (854)
Exchange rate differences  (1)  (3)
Balance as of end of period $59  $325 

*Out of which $ 325 thousand were realized from the beginning of the period for the year ended December 31, 2020.

NOTE 18 –COST OF RESEARCH AND DEVELOPMENT AND RESEARCH AND DEVELOPMENT SERVICES, NET

SCHEDULE OF RESEARCH AND DEVELOPMENT EXPENSES NET

 Year Ended December 31, 
 Year Ended November 30,  2020  2019 
 2018  2017  (in thousands) 
 (in thousands)      
Total expenses$ 7,386 $ 3,326  $84,182  $14,826 
Less grants (922) (848)  (196)  (812)
Total$ 6,464 $ 2, 478  $83,986  $14,014 

NOTE 19– 19 – FINANCIAL EXPENSES, NET

  Year ended November 30, 
  2018  2017 
  (in thousands) 
Decrease in fair value of warrants and financial liabilities measured at fair value$48 $ (902)
Stock-based compensation related to warrants granted due to issuance of credit facility 180  1,497 
Interest expense on convertible loans 2,753  1,233 
Foreign exchange loss, net 129  562 
Other income 7  57 
Total$ 3,117 $ 2,447 

SCHEDULE OF FINANCIAL EXPENSES

  2020  2019 
  Year Ended December 31, 
  2020  2019 
  (in thousands) 
       
Increase in fair value of warrants and financial liabilities measured at fair value $-  $63 
Interest expense on convertible loans  1,254   498 
Foreign exchange loss, net  160   395 
Other income  (353)  (113)
Total $1,061  $843 

NOTE 20- 20 – RELATED PARTIES TRANSACTIONS

a.Related Parties presented in the consolidated statements of comprehensive lossRelated Parties presented in the consolidated statements of comprehensive loss

SCHEDULE OF RELATED PARTY TRANSACTIONS

  Year ended December 31, 
  2020  2019 
  (in thousands) 
Continuing operations:        
Stock-based compensation expenses to executive officers $221  $898 
Stock-based compensation expenses to Board Members* $209  $414 
Compensation of executive officers $1,321  $812 
Management and consulting fees to Board Members $264  $233 
Revenues from customer $1,475  $1,270 
Cost of research and development and research and development services, net $4,772  $- 
Financial income $169  $112 

*Does not include $192 thousand for the year ended December 31, 2019 related to Stock Based Compensation expenses for options exercisable at an exercise price of $7.00 per share into 70,000 ordinary shares held by Caerus Therapeutics LLC for which the director does not have beneficial control.

F-44



  For the year ended November 30,  
  2018  2017 
  (in thousands) 
Management and consulting fees to Board Members$ 52 $ 25 
Stock Based Compensation expenses to Board Members 304  393 
Compensation of executive officers 1,119  419 
Stock Based Compensation expenses to executive officers 1,479  821 
Interest Expenses on convertible loan from director$ 13 $ 55 
F-51

b.Related Parties presented in the consolidated balance sheets

  

Year ended

December 31,

 
  2019 
  (in thousands) 
Discontinued operations:    
Stock-based compensation expenses to executive officers $76 
Compensation of executive officers $685 

b.Related Parties presented in the consolidated balance sheets

SCHEDULE OF RELATED PARTIES PRESENTED IN CONSOLIDATED BALANCE SHEETS

  December 31, 
  2020  2019 
  (in thousands) 
Continuing operations:        
Executive officers’ payables $170  $1,251 
Non-executive directors’ payable $13  $202 
Loan to Related Party $-  $2,623 
Accounts receivable, net $744  $- 
Contract liabilities $-  $230 

  Year ended November 30, 
  2018  2017 
     (in thousands) 
Convertible Loan from director$ - $ 167 
Executive officers’ payables 1,164  358 
Loan to Related Party. See Note 11(e) 1,007  - 
Non-executive directors payable$ 41 $ 316 
F-52

NOTE 21 - SUBSEQUENT EVENTS

a. In December 2018, the Company entered into unsecured convertible loan agreements with accredited or offshore investors for an aggregate amount of $250 thousand.  The loans bear an annual interest rate of 2% and mature in three years unless converted earlier under the same terms as the convertible loans presented in Note 8(a).

b. During December 2018, the Company advanced a total of $0.5 million to Hemogenyx Pharmaceuticals PLC and Immugenyx PLC.  See Notes 11(f) and 11(g).

c. On February 4, 2019, the Indian Joint Venture transferred the first payment of $1 million for services under the India JVA.  See also Note 11(e).

d. In December 2018, the Company entered into a Controlled Equity Offering Sales Agreement (“Sales Agreement”) with Cantor Fitzgerald & Co.(“Cantor”) pursuant to which the Company may offer and sell, from time to time through Cantor, shares of the Company’s common stock having an aggregate offering price of up to $25.0 million.  The Company will pay Cantor a commission rate equal to 3.0% of the aggregate gross proceeds from each sale.  The Company has not yet sold any shares pursuant to the Sales Agreement.

e. During December 2018, the Company received authorization from the Direction des Programmes de recherche in Belgium that based on a program and budget of $1.5 million in the field of gene-therapy research for diabetes 1 treatment, the Company could receive up to $ 350 thousand by June 2021.

f. During January 2019, Masthercell Global executed a lease agreement for production facilities in the United States.  Under the terms of the agreement, Masthercell Global leased approximately 32,000 square feet for 180 months. Masthercell Global advanced $1.6 million on account of a security deposit, tenant improvement allowance and prepaid base rent.  The annual rental increases over the lease period from approximately $726 thousand to $962 thousand.

g. In December 2018 and January 2019, the Board of Directors of Masthercell Global approved option grants for the purchase of 61,111 options in Masthercell Global under the Masthercell Global option plan to Masthercell Global executives, at an exercise price of $13.52.

F-45