Table of Contents

UNITED STATES

SECURITIES AND EXCHANGE COMMISSION

WASHINGTON, DC 20549

FORM 10-Q

(Mark One)

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

For the quarterly period ended June 30, 20212022

Or

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

Commission file number: 001-36020

Onconova Therapeutics, Inc.

(Exact name of registrant as specified in its charter)

DeDelawarelaware

    

22-3627252

(State or other jurisdiction of

(I.R.S. Employer

incorporation or organization)

Identification No.)

12 Penns Trail, Newtown, PA

18940

(Address of principal executive offices)

(Zip Code)

Registrant’s telephone number, including area code: (267) 759-3680

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   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   No

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

Non-accelerated filer

Smaller reporting company

Emerging growth company

If an emerging growth company, indicate by check mark 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 is a shell company (as defined in Rule 12b-2 of the Act).   Yes   No

The number of outstanding shares of the registrant’s Common Stock, par value $0.01 per share, as of August 4, 20211, 2022 was 15,781,040.20,895,563.

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

Title of each class

    

Trading Symbol(s)

    

Name of each exchange on which registered

Common Stock, par value $.01 per share

ONTX

The Nasdaq Stock Market LLC

Table of Contents

ONCONOVA THERAPEUTICS, INC.

 

TABLE OF CONTENTS FOR QUARTERLY REPORT ON FORM 10-Q

FOR THE QUARTER ENDED JUNE 30, 20212022

 

Page

PART I — FINANCIAL INFORMATION

Item 1. Financial Statements (Unaudited)

3

Condensed Consolidated Balance Sheets

3

Condensed Consolidated Statements of Operations

4

Condensed Consolidated Statements of Comprehensive Loss

5

Consolidated Statement of Stockholders’ Equity (Deficit)

6

Condensed Consolidated Statements of Cash Flows

7

Notes to Condensed Consolidated Financial Statements

8

Item 2. Management’s Discussion and Analysis of Financial Condition and Results of Operations

2119

Item 3. Quantitative and Qualitative Disclosures About Market Risk

3231

Item 4. Controls and Procedures

3231

PART II — OTHER INFORMATION

Item 1. Legal Proceedings

32

Item 1A. Risk Factors

32

Item 2. Unregistered Sales of Equity Securities and Use of Proceeds

3332

Item 3. Defaults Upon Senior Securities

3332

Item 4. Mine Safety Disclosures

3432

Item 5. Other Information

3432

Item 6. Exhibits

3433

SIGNATURES

3534

All common stock, equity, share and per share amounts have been retroactively adjusted to reflect a one-for-fifteen reverse stock split which was effective May 20, 2021.

2

Table of Contents

PART I — FINANCIAL INFORMATION

Item 1. Financial Statements

Onconova Therapeutics, Inc.

Condensed Consolidated Balance Sheets

June 30, 

December 31, 

2021

2020

Assets

(unaudited)

Current assets:

Cash and cash equivalents

$

43,709,000

$

19,025,000

Receivables

 

27,000

 

37,000

Prepaid expenses and other current assets

 

457,000

 

722,000

Total current assets

 

44,193,000

 

19,784,000

Property and equipment, net

 

45,000

 

52,000

Other non-current assets

 

140,000

 

150,000

Total assets

$

44,378,000

$

19,986,000

Liabilities and stockholders’ equity

Current liabilities:

Accounts payable

$

4,290,000

$

4,833,000

Accrued expenses and other current liabilities

 

2,983,000

 

4,962,000

Deferred revenue

 

226,000

 

226,000

Total current liabilities

 

7,499,000

 

10,021,000

Warrant liability

530,000

321,000

Deferred revenue, non-current

 

3,356,000

 

3,469,000

Total liabilities

 

11,385,000

 

13,811,000

Commitments and contingencies

Stockholders’ equity:

Preferred stock, $0.01 par value, 5,000,000 authorized at June 30, 2021 and December 31, 2020, NaN issued and outstanding at June 30, 2021 and December 31, 2020

 

 

Common stock, $0.01 par value, 125,000,000 and 250,000,000 authorized at June 30, 2021 and December 31, 2020, 15,781,040 and 12,396,219 shares issued and outstanding at June 30, 2021 and December 31, 2020

 

158,000

 

124,000

Additional paid in capital

 

470,335,000

 

434,593,000

Accumulated deficit

 

(437,502,000)

 

(428,556,000)

Accumulated other comprehensive income

 

2,000

 

14,000

Total stockholders’ equity

 

32,993,000

 

6,175,000

Total liabilities and stockholders’ equity

$

44,378,000

$

19,986,000

June 30, 

December 31, 

2022

2021

Assets

(unaudited)

Current assets:

Cash and cash equivalents

$

46,533,000

$

55,070,000

Receivables

 

28,000

 

28,000

Prepaid expenses and other current assets

 

1,472,000

 

332,000

Total current assets

 

48,033,000

 

55,430,000

Property and equipment, net

 

31,000

 

38,000

Other non-current assets

 

10,000

 

10,000

Total assets

$

48,074,000

$

55,478,000

Liabilities and stockholders’ equity

Current liabilities:

Accounts payable

$

3,003,000

$

2,757,000

Accrued expenses and other current liabilities

 

3,231,000

 

3,132,000

Deferred revenue

 

226,000

 

226,000

Total current liabilities

 

6,460,000

 

6,115,000

Deferred revenue, non-current

 

3,130,000

 

3,243,000

Total liabilities

 

9,590,000

 

9,358,000

Commitments and contingencies

Stockholders’ equity:

Preferred stock, $0.01 par value, 5,000,000 shares authorized, NaN issued and outstanding at June 30, 2022 and December 31, 2021

 

 

Common stock, $0.01 par value, 125,000,000 shares authorized, 20,895,563 shares issued and outstanding at June 30, 2022 and December 31, 2021

 

209,000

 

209,000

Additional paid in capital

 

491,181,000

 

490,644,000

Accumulated deficit

 

(452,865,000)

 

(444,719,000)

Accumulated other comprehensive loss

 

(41,000)

 

(14,000)

Total stockholders’ equity

 

38,484,000

 

46,120,000

Total liabilities and stockholders’ equity

$

48,074,000

$

55,478,000

See accompanying notes to condensed consolidated financial statements.

3

Table of Contents

Onconova Therapeutics, Inc.

Condensed Consolidated Statements of Operations (unaudited)

Three Months Ended June 30, 

Six Months Ended June 30, 

Three Months Ended June 30, 

Six Months Ended June 30, 

    

2021

    

2020

    

2021

    

2020

    

    

2022

    

2021

    

2022

    

2021

    

Revenue

$

57,000

$

56,000

$

113,000

$

108,000

$

57,000

$

57,000

$

113,000

$

113,000

Operating expenses:

General and administrative

 

2,850,000

 

2,594,000

 

5,067,000

 

4,401,000

 

2,139,000

 

2,850,000

 

4,325,000

 

5,067,000

Research and development

 

1,852,000

 

4,801,000

 

3,789,000

 

8,171,000

 

2,038,000

 

1,852,000

 

4,040,000

 

3,789,000

Total operating expenses

 

4,702,000

 

7,395,000

 

8,856,000

 

12,572,000

 

4,177,000

 

4,702,000

 

8,365,000

 

8,856,000

Loss from operations

 

(4,645,000)

 

(7,339,000)

 

(8,743,000)

 

(12,464,000)

 

(4,120,000)

 

(4,645,000)

 

(8,252,000)

 

(8,743,000)

Change in fair value of warrant liability

 

427,000

 

(56,000)

 

(209,000)

 

(119,000)

 

 

427,000

 

 

(209,000)

Other (loss) income, net

 

(13,000)

 

 

6,000

 

96,000

Other income, net

 

96,000

 

(13,000)

 

106,000

 

6,000

Net loss

$

(4,231,000)

$

(7,395,000)

$

(8,946,000)

$

(12,487,000)

$

(4,024,000)

$

(4,231,000)

$

(8,146,000)

$

(8,946,000)

Net loss per share, basic and diluted

$

(0.27)

$

(0.65)

$

(0.59)

$

(1.14)

$

(0.19)

$

(0.27)

$

(0.39)

$

(0.59)

Basic and diluted weighted average shares outstanding

 

15,780,863

 

11,303,508

 

15,201,719

 

10,996,624

 

20,904,085

 

15,780,863

 

20,904,085

 

15,201,719

See accompanying notes to condensed consolidated financial statements.

4

Table of Contents

Onconova Therapeutics, Inc.

Condensed Consolidated Statements of Comprehensive Loss (unaudited)

Three Months Ended June 30, 

Six Months Ended June 30, 

Three Months Ended June 30, 

Six Months Ended June 30, 

    

2021

    

2020

    

2021

    

2020

    

    

2022

    

2021

    

2022

    

2021

    

Net loss

$

(4,231,000)

$

(7,395,000)

$

(8,946,000)

$

(12,487,000)

$

(4,024,000)

$

(4,231,000)

$

(8,146,000)

$

(8,946,000)

Other comprehensive income (loss), net of tax:

Other comprehensive (loss) income, net of tax:

Foreign currency translation adjustments, net

 

4,000

 

7,000

 

(12,000)

 

1,000

 

(20,000)

 

4,000

 

(27,000)

 

(12,000)

Other comprehensive income (loss), net of tax

4,000

7,000

(12,000)

1,000

Other comprehensive (loss) income, net of tax

(20,000)

4,000

(27,000)

(12,000)

Comprehensive loss

$

(4,227,000)

$

(7,388,000)

$

(8,958,000)

$

(12,486,000)

$

(4,044,000)

$

(4,227,000)

$

(8,173,000)

$

(8,958,000)

See accompanying notes to condensed consolidated financial statements.

5

Table of Contents

Onconova Therapeutics, Inc.

Consolidated Statement of Stockholders’ Equity (Deficit) (unaudited)

Three Month Periods Ended June 30, 2021 and 2020

Three Month Periods Ended June 30, 2022 and 2021

Accumulated

Accumulated

Additional

other

Additional

other

Common Stock

Paid in

Accumulated

comprehensive

Common Stock

Paid in

Accumulated

comprehensive

    

Shares

    

Amount

    

Capital

    

deficit

    

(loss) income

    

Total

Balance at March 31, 2022

20,895,563

$

209,000

$

490,940,000

$

(448,841,000)

$

(21,000)

$

42,287,000

Net loss

 

 

 

 

(4,024,000)

 

 

(4,024,000)

Other comprehensive loss

 

 

 

 

 

(20,000)

 

(20,000)

Stock-based compensation

 

 

 

241,000

 

 

 

241,000

Balance at June 30, 2022

20,895,563

$

209,000

$

491,181,000

$

(452,865,000)

$

(41,000)

$

38,484,000

    

Shares

    

Amount

    

Capital

    

deficit

    

(loss) income

    

Total

Balance at March 31, 2021

15,779,160

$

158,000

$

470,268,000

$

(433,271,000)

$

(2,000)

$

37,153,000

15,779,160

$

158,000

$

470,268,000

$

(433,271,000)

$

(2,000)

$

37,153,000

Net loss

 

 

 

 

(4,231,000)

 

 

(4,231,000)

 

 

 

 

(4,231,000)

 

 

(4,231,000)

Other comprehensive loss

 

 

 

 

 

4,000

 

4,000

 

 

 

 

 

4,000

 

4,000

Exercise of stock options

1,776

0

7,000

7,000

1,776

 

 

7,000

 

 

 

7,000

Stock-based compensation

 

 

 

60,000

 

 

 

60,000

 

 

 

60,000

 

 

 

60,000

Shares issued in connection with reverse stock split

104

104

 

 

 

 

 

Balance at June 30, 2021

15,781,040

$

158,000

$

470,335,000

$

(437,502,000)

$

2,000

$

32,993,000

15,781,040

$

158,000

$

470,335,000

$

(437,502,000)

$

2,000

$

32,993,000

Balance at March 31, 2020

11,161,072

$

111,000

$

429,752,000

$

(408,491,000)

$

(24,000)

$

21,348,000

Net loss

 

 

 

 

(7,395,000)

 

 

(7,395,000)

Other comprehensive loss

 

 

 

 

 

7,000

 

7,000

Stock-based compensation

 

 

 

92,000

 

 

 

92,000

Issuance of common stock upon exercise of warrants

367,425

4,000

1,576,000

1,580,000

Issuance of common stock upon exercise of pre-funded warrants

83,333

1,000

1,000

Balance at June 30, 2020

11,611,830

$

116,000

$

431,420,000

$

(415,886,000)

$

(17,000)

$

15,633,000

Six Month Periods Ended June 30, 2021 and 2020

Six Month Periods Ended June 30, 2022 and 2021

Accumulated

Accumulated

Additional

other

Additional

other

Common Stock

Paid in

Accumulated

comprehensive

Common Stock

Paid in

Accumulated

comprehensive

    

Shares

    

Amount

    

Capital

    

deficit

    

income (loss)

    

Total

    

Shares

    

Amount

    

Capital

    

deficit

    

income (loss)

    

Total

Balance at December 31, 2021

20,895,563

$

209,000

$

490,644,000

$

(444,719,000)

$

(14,000)

$

46,120,000

Net loss

 

 

 

 

(8,146,000)

 

 

(8,146,000)

Other comprehensive loss

 

 

 

 

 

(27,000)

 

(27,000)

Stock-based compensation

 

 

 

537,000

 

 

 

537,000

Balance at June 30, 2022

20,895,563

$

209,000

$

491,181,000

$

(452,865,000)

$

(41,000)

$

38,484,000

Balance at December 31, 2020

12,396,219

$

124,000

$

434,593,000

$

(428,556,000)

$

14,000

$

6,175,000

12,396,219

$

124,000

$

434,593,000

$

(428,556,000)

$

14,000

$

6,175,000

Net loss

 

 

 

 

(8,946,000)

 

 

(8,946,000)

 

 

 

 

(8,946,000)

 

 

(8,946,000)

Other comprehensive income

 

 

 

 

 

(12,000)

 

(12,000)

Other comprehensive loss

 

 

 

 

 

(12,000)

 

(12,000)

Exercise of stock options

4,642

24,000

24,000

4,642

 

 

24,000

 

 

 

24,000

Stock-based compensation

 

 

 

125,000

 

 

 

125,000

 

 

 

125,000

 

 

 

125,000

Shares issued in connection with reverse stock split

104

104

 

 

 

 

 

Issuance of common stock, net

3,220,075

32,000

35,115,000

35,147,000

3,220,075

 

32,000

 

35,115,000

 

 

 

35,147,000

Issuance of common stock upon exercise of warrants

160,000

2,000

478,000

480,000

160,000

2,000

478,000

480,000

Balance at June 30, 2021

15,781,040

$

158,000

$

470,335,000

$

(437,502,000)

$

2,000

$

32,993,000

15,781,040

$

158,000

$

470,335,000

$

(437,502,000)

$

2,000

$

32,993,000

Balance at December 31, 2019

7,411,157

$

74,000

$

414,917,000

$

(403,399,000)

$

(18,000)

$

11,574,000

Net loss

 

 

 

 

(12,487,000)

 

 

(12,487,000)

Other comprehensive loss

 

 

 

 

 

1,000

 

1,000

Stock-based compensation

 

 

 

185,000

 

 

 

185,000

Issuance of common stock, net

1,844,168

18,000

9,044,000

9,062,000

Issuance of common stock upon exercise of warrants

2,273,172

23,000

7,274,000

7,297,000

Issuance of common stock upon exercise of pre-funded warrants

83,333

1,000

1,000

Balance at June 30, 2020

11,611,830

$

116,000

$

431,420,000

$

(415,886,000)

$

(17,000)

$

15,633,000

See accompanying notes to condensed consolidated financial statements.

6

Table of Contents

Onconova Therapeutics, Inc.

Condensed Consolidated Statements of Cash Flows (unaudited)

Six Months Ended June 30, 

Six Months Ended June 30, 

    

2021

    

2020

    

    

2022

    

2021

    

Operating activities:

Net loss

$

(8,946,000)

$

(12,487,000)

$

(8,146,000)

$

(8,946,000)

Adjustment to reconcile net loss to net cash used in operating activities:

Depreciation and amortization

 

7,000

 

5,000

 

7,000

 

7,000

Change in fair value of warrant liabilities

209,000

119,000

209,000

Stock compensation expense

 

125,000

 

185,000

 

537,000

 

125,000

Changes in assets and liabilities:

Receivables

 

10,000

 

57,000

 

 

10,000

Prepaid expenses and other current assets

 

265,000

 

(70,000)

 

(1,140,000)

 

265,000

Other assets

10,000

10,000

Accounts payable

 

(543,000)

 

877,000

 

246,000

 

(543,000)

Accrued expenses and other current liabilities

 

(1,979,000)

 

(418,000)

 

99,000

 

(1,979,000)

Deferred revenue

 

(113,000)

 

(112,000)

 

(113,000)

 

(113,000)

Net cash used in operating activities

 

(10,955,000)

 

(11,844,000)

 

(8,510,000)

 

(10,955,000)

Investing activities:

Payments for purchase of property and equipment

(15,000)

Net cash used in investing activities

 

 

(15,000)

Financing activities:

Proceeds from the sale of common stock and warrants, net of costs

35,147,000

9,062,000

35,147,000

Proceeds from the exercise of warrants

480,000

7,298,000

Proceeds from the exercise of common warrants

480,000

Proceeds from the exercise of stock options

 

24,000

 

 

 

24,000

Net cash provided by financing activities

 

35,651,000

 

16,360,000

 

 

35,651,000

Effect of foreign currency translation on cash

 

(12,000)

 

1,000

 

(27,000)

 

(12,000)

Net increase in cash and cash equivalents

 

24,684,000

 

4,502,000

Net (decrease) increase in cash and cash equivalents

 

(8,537,000)

 

24,684,000

Cash and cash equivalents at beginning of period

 

19,025,000

 

22,726,000

 

55,070,000

 

19,025,000

Cash and cash equivalents at end of period

$

43,709,000

$

27,228,000

$

46,533,000

$

43,709,000

See accompanying notes to condensed consolidated financial statements.

7

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements

(Unaudited)

1. Nature of Business

The Company

Onconova Therapeutics, Inc. (the “Company”) was incorporated in the State of Delaware on December 22, 1998 and commenced operations on January 1, 1999. The Company’s headquarters are located in Newtown, Pennsylvania. The Company is a clinical-stage biopharmaceutical company focused on discovering and developing novel products for patients with cancer. The Company has proprietary molecularly targeted anti-cancer agents designed to disrupt specific cellular pathways that are important for cancer cell proliferation. The Company believes that the product candidates in its pipeline have the potential to be efficacious in a variety of cancers with unmet medical need. The Company currently has the following 2 clinical-stage programs: 1. ON 123300narazaciclib (ON 123300), a multi-targeted kinase inhibitor in solid tumors;tumors and hematological malignancies as a single agent or in combination with other anti-cancer therapies; and 2. oral rigosertib administered alone or in combination with PD-1 inhibitors for the treatment of KRAS-mutated solid tumors. During 2012, Onconova Europe GmbH was established as a wholly owned subsidiary of the Company for the purpose of further developing business in Europe.

The Company has entered into several license and collaboration agreements. In 2011, the Company entered into a license agreement, as subsequently amended, with SymBio Pharmaceuticals Limited (“SymBio”), which grants SymBio certain rights to commercialize rigosertib in Japan and Korea. In December 2017, the Company entered into a license and collaboration agreement with HanX Biopharmaceuticals, Inc. (“HanX”) for the further development, registration and commercialization of ON 123300 in greater China. ON 123300 is a preclinical compound which the Company believes has the potential to overcome the limitations of current generation CDK 4/6 inhibitors. Under the terms of the agreement, the Company received an upfront payment, and will receive regulatory and commercial milestone payments, as well as royalties on Chinese sales. The key feature of the collaboration is that HanX provides all funding required for Chinese IND enabling studies performed for Chinese Food and Drug Administration IND approval, which was received in January 2020. The Company and HanX also intended for these studies to comply with the FDA standards for IND approval. Accordingly, such studies were used by the Company for an IND filing with the US FDA in November 2020. The FDA Study May Proceed letter was issued in December 2020. The Company maintains global rights outside of China. On March 2, 2018, the Company entered into a License, Development and Commercialization Agreement (the “Pint License Agreement”) with Pint International SA (which, together with its affiliate Pint Pharma GmbH, are collectively referred to as “Pint”). Under the terms of the agreement, the Company granted Pint an exclusive, royalty-bearing license, with the right to sublicense, under certain Company patent rights and know-how, to develop and commercialize any pharmaceutical product containing rigosertib in all uses of rigosertib in certain Latin American countries. In May 2019, the Company entered into a License and Collaboration Agreement (the “HanX License Agreement”) with HanX. Under the terms of the HanX License Agreement, the Company granted HanX an exclusive, royalty-bearing license, with the right to sublicense, under certain Company patent rights and know-how, to develop and commercialize any pharmaceutical product (the “HanX Product”) containing rigosertib in all uses of rigosertib or the HanX Product in human therapeutic uses in the People’s Republic of China, Hong Kong, Macau and Taiwan (the “HanX Territory”). In connection with the HanX License Agreement, the Company also entered into a Securities Purchase Agreement with each of HanX and Abundant New Investments Ltd. (“Abundant”), an affiliate of HanX (each, a “Securities Purchase Agreement” and together, the “Securities Purchase Agreements”). HanX did not fulfill its obligations under the HanX License Agreement and in January 2020, in accordance with the terms of the HanX License Agreement, the HanX License Agreement was deemed to be void ab initio. Upon this termination, the rights to HanX Product in the HanX Territory reverted to the Company in accordance with the terms of the HanX License Agreement. In addition, the Securities Purchase Agreements terminated automatically effective upon the termination of the HanX License Agreement in accordance with the Securities Purchase Agreements. In November 2019, the Company entered into a Distribution, License and Supply Agreement (the “Knight License Agreement”) with Knight Therapeutics Inc. (“Knight”). Under the terms of the Knight License Agreement, the Company granted Knight (i) a non-exclusive, royalty-bearing license, with the right to sublicense, under certain Company patent rights and know-how, to develop and manufacture any product (the “Knight Licensed Product”) containing rigosertib for Canada (and Israel, should Knight exercise its option as set forth in the Knight License Agreement) (the “Knight Territory”) and in human uses (the

8

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

“Field”), and (ii) an exclusive, royalty-bearing license, with the right to sublicense, under certain Company patent rights and know-how, to commercialize the Knight Licensed Product in the Knight Territory and in the Field. Knight has also agreed to obtain from the Company all of its requirements of the Knight Licensed Products for the Knight Territory, and the Company has agreed to supply Knight with all of its requirements of the Knight Licensed Products. In December 2019, the Company entered into a Distribution, License and Supply Agreement (the “STA License Agreement”) with Specialised Therapeutics Asia Pte. Ltd. (“STA”). Under the terms of the STA License Agreement, the Company granted STA (i) a non-exclusive, royalty -bearing license, with the right to sublicense, under certain Company patent rights and know-how, to develop and manufacture any product (the “STA Licensed Product”) containing rigosertib for Australia and New Zealand (the “STA Territory”) and in human uses (the “Field”), and (ii) an exclusive, royalty-bearing license, with the right to sublicense, under certain Company patent rights and know- how, to commercialize the STA Licensed Product in the STA Territory and in the Field. STA has also agreed to obtain from the Company all of its requirements of the STA Licensed Products for the STA Territory, and the Company has agreed to supply STA with all of its requirements of the STA Licensed Products.

On May 20, 2021, the Company amended its certificate of incorporation to effect a one-for-fifteen reverse stock split of its common stock. All common stock, equity, share and per share amounts in the financial statements and notes have been retroactively adjusted to reflect this one-for-fifteen reverse stock split.

On May 20, 2021, the Company amended its certificate of incorporation to decrease the number of authorized shares of common stock par value $0.01 per share from 250,000,000 to 125,000,000.

125,000,000, and to effect a one-for-fifteen reverse stock split of its common stock. All common stock, equity, share and per share amounts in the financial statements and notes have been retroactively adjusted to reflect this one-for-fifteen reverse stock split.

Liquidity

The Company has incurred recurring operating losses since inception. For the six months ended June 30, 2021,2022, the Company incurred a net loss of $8,946,000$8,146,000 and as of June 30, 20212022 the Company had generated an accumulated deficit of $437,502,000.$452,865,000. The Company anticipates operating losses to continue for the foreseeable future due to, among other things, costs related to research, development of its product candidates and its preclinical programs, strategic alliances and its administrative organization. At June 30, 2021,2022, the Company had cash and cash equivalents of $43,709,000.$46,533,000. The Company will require substantial additional financing to fund its ongoing clinical trials and operations, and to continue to execute its strategy.

On January 11, 2021, the Company closed on an offering of common stock. The Company issued 1,303,408 shares of common stock and net proceeds were approximately $8.5 million. On February 16, 2021, the Company closed on an offering of common stock. The Company issued 1,916,667 shares of common stock and net proceeds were approximately $26.7 million.

Following the unsuccessful conclusion of the INSPIRE trial, the Company has taken steps to reduce its cash expenditures. From September 2020 to December 2020, the Company implemented a workforce reduction of employees in research and development who were primarily focused on preparing the NDA for the use of rigosertib in higher risk MDS. In total, 10 employees were terminated, representing approximately 43% of the Company’s workforce. A severance related charge of approximately $1,207,000, which includes a non-cash charge of approximately $29,000 related to the accelerated vesting of outstanding stock options, was recorded in the year ended December 31, 2020. The accrued severance balance remaining at June 30, 2021 was $177,000 and is included in accrued expenses and other liabilities on the balance sheet. It will be paid in periodic amounts through September 2021. On October 30, 2020, the Company notified its landlord of its intention to not renew its office space lease. The lease expired in February 2021 and was modified to a month-to-month lease for a portion of the space. The lease terminated in June 2021 and the Company has relocated to temporary office space with all employees working remotely.

9

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

The Company has and may continue to delay, scale-back, or eliminate certain of its research and development activities and other aspects of its operations until such time as the Company is successful in securing additional funding. The Company is exploring various dilutive and non-dilutive sources of funding, including equity financings, strategic alliances, business development and other sources. The future success of the Company is dependent upon its ability to obtain additional funding. There can be no assurance, however, that the Company will be successful in obtaining such funding in sufficient amounts, on terms acceptable to the Company, or at all. The Company believes that its cash and cash equivalents will be sufficient to fund its ongoing trials and business operations for more than eighteentwelve months from the date of this filing.

COVID-19

While the Company is not aware of a material impact from the novel coronavirus disease (“COVID-19”) pandemic through June 30, 2021,2022, the full extent to which COVID-19 will directly or indirectly impact the Company’s business, results of operations and financial condition, including manufacturing, clinical trials and research and development costs, depends on future developments that are highly uncertain at this time.

8

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

2. Summary of Significant Accounting Policies

Basis of Presentation

The condensed consolidated financial statements are prepared in conformity with accounting principles generally accepted in the United States (“GAAP”) for interim financial information. Certain information and footnotes normally included in financial statements prepared in accordance with GAAP have been condensed or omitted pursuant to the rules and regulations of the Securities and Exchange Commission (the “SEC”). The financial statements include the consolidated accounts of the Company and its wholly-owned subsidiary, Onconova Europe GmbH. All significant intercompany transactions have been eliminated.

Unaudited Interim Financial Information

The accompanying condensed consolidated balance sheet as of June 30, 2021,2022, the condensed consolidated statements of operations and comprehensive loss for the three and six months ended June 30, 20212022 and 2020,2021, the consolidated statements of stockholders’ equity (deficit) for the three and six months ended June 30, 20212022 and 20202021 and the condensed consolidated statements of cash flows for the six months ended June 30, 20212022 and 20202021 are unaudited. The interim unaudited condensed consolidated financial statements have been prepared on the same basis as the annual audited consolidated financial statements and, in the opinion of management, reflect all adjustments, which include only normal recurring adjustments, necessary for the fair statement of the Company’s financial position as of June 30, 2021,2022, the results of its operations for the three and six months ended June 30, 20212022 and 2020,2021, and its cash flows for the six months ended June 30, 20212022 and 2020.2021. The financial data and other information disclosed in these notes related to the three and six months ended June 30, 20212022 and 20202021 are unaudited. The results for the three and six months ended June 30, 20212022 are not necessarily indicative of results to be expected for the year ending December 31, 2021,2022, any other interim periods, or any future year or period. These unaudited condensed consolidated financial statements should be read in conjunction with the audited consolidated financial statements and the notes thereto for the year ended December 31, 20202021 included in the Company’s annual report on Form 10-K filed with the SEC on March 18, 2021.21, 2022.

All common stock, equity, share and per share amounts in the financial statements and notes have been retroactively adjusted to reflect a one-for-fifteen reverse stock split which was effective May 20, 2021.

10

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

Segment Information

Operating segments are defined as components of an enterprise about which separate discrete information is available for evaluation by the chief operating decision maker, or decision-making group, in deciding how to allocate resources and in assessing performance. The Company views its operations and manages its business in one1 segment, which is the identification and development of oncology therapeutics.

Significant Accounting Policies

The Company’s significant accounting policies are disclosed in the audited consolidated financial statements for the year ended December 31, 20202021 included in the Company’s annual report on Form 10-K filed with the SEC on March 18, 2021.21, 2022. Since the date of such financial statements, there have been no changes to the Company’s significant accounting policies.

Fair Value Measurements

The carrying amounts reported in the accompanying consolidated financial statements for cash and cash equivalents, accounts payable, and accrued liabilities approximate their respective fair values because of the short-term nature of these accounts. The fair value of the warrant liability is discussed in Note 7, “Fair Value Measurements.”

9

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

Recent Accounting Pronouncements

In June 2016, the FASB issued new guidance on the accounting for credit losses on financial instruments. The guidance was amended in November 2019. The new guidance introduces an expected loss model for estimating credit losses, replacing the incurred loss model. The new guidance also changes the impairment model for available-for-sale debt securities, requiring the use of an allowance to record estimated credit losses (and subsequent recoveries). The guidance is effective for the Company in fiscal years beginning after December 15, 2022, and interim periods within those years, with early adoption permitted. The Companyguidance is evaluatingnot expected to have a material effect on the impact of the adoption of the standard on its consolidated financial statements.Company.

3. Revenue

The Company’s revenue during the three and six months ended June 30, 20212022 and 20202021 was from its license and collaboration agreement with SymBio.

Three Months Ended June 30, 

Six Months Ended June 30, 

Three Months Ended June 30, 

Six Months Ended June 30, 

    

2021

    

2020

    

2021

    

2020

    

    

2022

    

2021

    

2022

    

2021

    

Symbio

Upfront license fee recognition over time

$

57,000

$

56,000

$

113,000

$

112,000

$

57,000

$

57,000

$

113,000

$

113,000

Supplies

(4,000)

$

57,000

$

56,000

$

113,000

$

108,000

11

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

Deferred revenue is as follows:

Symbio

Symbio

    

Upfront Payment

    

Upfront Payment

Deferred balance at December 31, 2020

$

3,695,000

Deferred balance at December 31, 2021

$

3,469,000

Recognition to revenue

113,000

(113,000)

Deferred balance at June 30, 2021

$

3,582,000

Deferred balance at June 30, 2022

$

3,356,000

4. Net Loss Per Share of Common Stock

The following potentially dilutive securities outstanding at June 30, 20212022 and 20202021 have been excluded from the computation of diluted weighted average shares outstanding, as they would be antidilutive (reflects the number of common shares as if the dilutive securities had been converted to common stock):

June 30, 

June 30, 

    

2021

    

2020

    

2022

    

2021

Warrants

 

511,202

 

1,457,479

 

491,586

 

512,202

Stock options

 

60,431

 

69,640

 

850,553

 

43,026

 

571,633

 

1,527,119

 

1,342,139

 

555,228

5. Warrants

Common Stock warrants are accounted for in accordance with applicable accounting guidance provided in ASC Topic 815, Derivatives and Hedging - Contracts in Entity’s Own Equity (ASC Topic 815), as either derivative liabilities or as equity instruments depending on the specific terms of the warrant agreement.

10

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

5. Warrants

Common Stock warrants are accounted for in accordance with applicable accounting guidance provided in ASC Topic 815, Derivatives and Hedging - Contracts in Entity’s Own Equity (ASC Topic 815), as either derivative liabilities or as equity instruments depending on the specific terms of the warrant agreement. Some of the Company’s warrants are classified as liabilities because in certain circumstances they could require cash settlement.

Warrants outstanding and warrant activity (reflects the number of common shares as if the warrants were converted to common stock) for the six months ended June 30, 20212022 is as follows:

Balance

Balance

Exercise

Expiration

December 31, 

Warrants

Warrants

Warrants

June 30, 

Description

    

Classification

    

Price

    

Date

    

2020

    

Issued

    

Exercised

    

Expired

    

2021

Non-tradable warrants

 

Liability

$

2,587.50

 

July 2021

 

430

 

 

 

 

430

Tradable warrants

 

Liability

$

1,107.00

 

July 2021

 

14,187

 

 

 

 

14,187

Non-tradable pre-funded warrants

 

Equity

$

2.25

 

July 2023

 

26

 

 

 

 

26

Non-tradable warrants

Equity

$

24.00

December 2022

26,189

26,189

Non-tradable warrants

Equity

$

211.50

March 2021

333

(333)

Non-tradable warrants

Equity

$

317.25

March 2021

556

(556)

Non-tradable warrants

Equity

$

116.8425

June 2021

1,000

(1,000)

Non-tradable pre-funded warrants

Equity

$

2.25

none

3,522

3,522

Non-tradable warrants

Equity

$

24.00

December 2022

120,407

120,407

Non-tradable pre-funded warrants

Equity

$

2.25

none

4,974

4,974

Non-tradable warrants

Equity

$

30.00

September 2023

7,306

7,306

Non-tradable warrants

Equity

$

3.00

November 2024

409,500

(160,000)

249,500

Non-tradable warrants

Equity

$

6.54375

December 2024

16,953

16,953

Non-tradable warrants

Equity

$

6.75450

December 2024

46,263

46,263

Non-tradable warrants

Equity

$

6.77850

December 2023

29,968

29,968

681,614

(160,000)

(1,889)

519,725

The tradable warrants which expired in July 2021 were issued in connection with a financing transaction completed in August 2016. Subsequent to the closing of that financing transaction, the Company executed a one-for-fifteen reverse stock split in September 2018. Subsequently, the Company executed a one-for-fifteen reverse stock split in May 2021. As a result, each of the 3,192,140 warrants is exercisable for 1/225 of one share of common stock at an exercise price of $4.92 per warrant. The table above shows the number of shares of common stock which could be

12

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

obtained by the exercise of all of the outstanding warrants, 14,187; and shows the exercise price for 225 of the warrants, $1,107.00.

Balance

Balance

Exercise

Expiration

December 31, 

Warrants

Warrants

Warrants

June 30, 

Description

    

Classification

    

Price

    

Date

    

2021

    

Issued

    

Exercised

    

Expired

    

2022

Non-tradable pre-funded warrants

 

Equity

$

2.25

 

July 2023

 

26

 

 

 

 

26

Non-tradable warrants

Equity

$

24.00

December 2022

26,189

26,189

Non-tradable pre-funded warrants

Equity

$

2.25

none

3,522

3,522

Non-tradable warrants

Equity

$

24.00

December 2022

120,407

120,407

Non-tradable pre-funded warrants

Equity

$

2.25

none

4,974

4,974

Non-tradable warrants

Equity

$

30.00

September 2023

7,306

7,306

Non-tradable warrants

Equity

$

3.00

November 2024

244,500

244,500

Non-tradable warrants

Equity

$

6.54375

December 2024

16,953

16,953

Non-tradable warrants

Equity

$

6.75450

December 2024

46,263

46,263

Non-tradable warrants

Equity

$

6.77850

December 2023

29,968

29,968

500,108

500,108

6. Balance Sheet Detail

Prepaid expenses and other current assets:

June 30, 

December 31, 

June 30, 

December 31, 

    

2021

    

2020

    

2022

    

2021

Research and development

$

42,000

$

189,000

$

1,050,000

$

15,000

Manufacturing

 

144,000

 

90,000

 

31,000

 

29,000

Insurance

 

80,000

 

263,000

 

85,000

 

253,000

Other

 

191,000

 

180,000

 

306,000

 

35,000

$

457,000

$

722,000

$

1,472,000

$

332,000

Property and equipment:

June 30, 

December 31, 

June 30, 

December 31, 

    

2021

    

2020

    

2022

    

2021

Property and equipment

$

70,000

$

70,000

$

70,000

$

70,000

Accumulated depreciation

 

(25,000)

 

(18,000)

 

(39,000)

 

(32,000)

$

45,000

$

52,000

$

31,000

$

38,000

Accrued expenses and other current liabilities:

June 30, 

December 31, 

June 30, 

December 31, 

    

2021

    

2020

    

2022

    

2021

Research and development

$

1,849,000

$

2,541,000

$

1,968,000

$

1,759,000

Employee compensation

 

990,000

 

2,239,000

 

1,054,000

 

1,217,000

Professional fees

144,000

182,000

209,000

156,000

$

2,983,000

$

4,962,000

$

3,231,000

$

3,132,000

7. Fair Value Measurements

At both June 30, 2022 and December 31, 2021, the Company had 0 financial assets and liabilities measured at fair value on a recurring basis.

11

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

Fair value is defined as the exchange price that would be received for an asset or paid to transfer a liability (an exit price) in the principal or most advantageous market for the asset or liability in an orderly transaction between market participants on the measurement date.

The Company utilizes a valuation hierarchy for disclosure of the inputs to the valuations used to measure fair value. This hierarchy prioritizes the inputs into three broad levels as follows. Level 1 inputs are quoted prices (unadjusted) in active markets for identical assets or liabilities. Level 2 inputs are quoted prices for similar assets and liabilities in active markets or inputs that are observable for the asset or liability, either directly or indirectly through market corroboration, for substantially the full term of the financial instrument. Level 3 inputs are unobservable inputs based on the Company’s own assumptions used to measure assets and liabilities at fair value. A financial asset or liability’s classification within the hierarchy is determined based on the lowest level input that is significant to the fair value measurement.

On January 5, 2016,During 2021, the Company entered into a securities purchase agreement (the “Securities Purchase Agreement”) with an institutional investor providing for the issuancehad tradable warrants and sale by the Company of 861 shares of Common Stock,non-tradable warrants that were classified as liabilities and measured at a purchase price of $2,137.50 per share and warrants to purchase up to 430 shares of Common Stock

13

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

(the “Warrants”) for aggregate gross proceeds of $1,840,000. The Company has classified the warrants as a liability (see Note 5). The estimated fair value using the Black-Scholes pricing model was approximately $0 at June 30, 2021 and December 31, 2020. These warrants expired in July 2021.

On July 29, 2016 the Company closed on a Rights Offering, issuing 16,000 shares of Common Stock, 14,187 Tradable Warrants and 2,918 Pre-Funded Warrants.recurring basis. The Tradable Warrants are exercisable for a period of five years for one share of Common Stock at an exercise price of $1,107 per share. After the one-year anniversary of issuance, the Company may redeem the Tradable Warrants for $0.015 per Tradable Warrant if the volume weighted average price of its Common Stock is above $2,767.50 for each of 10 consecutive trading days. The Company has classified the Tradable Warrants as a liability (see Note 5). The Tradable Warrants have beentradable warrants were listed on the Nasdaq Capital Market since issuance and the Company regularly monitors the trading activity.Market. The Company has determined that an active and orderly market for the Tradable Warrants hastradable warrants developed and that the Nasdaq Capital Market price iswas the best indicator of fair value of the warrant liability. The quoted market price was used to determine the fair value. The fair value at December 31, 2020of the non-tradable warrants was estimated using the Black-Scholes pricing model. All of these tradable and June 30, 2021. Thesenon-tradable warrants expired in July 2021. During the three and six months ended June 30, 2021, there was a decrease (increase) in the fair value of the warrant liability of $427,000 and ($209,000), respectively.

The following fair value hierarchy table presents information about the Company’s financial assets and liabilities measured at fair value on a recurring basis as of June 30, 2021 and December 31, 2020:

Fair Value Measurement as of:

June 30, 2021

December 31, 2020

    

Level 1

    

Level 2

    

Level 3

    

Balance

    

Level 1

    

Level 2

    

Level 3

    

Balance

Tradable warrants liability

$

530,000

$

$

$

530,000

$

321,000

$

$

$

321,000

Non-tradable warrants liability

Total

$

530,000

$

$

$

530,000

$

321,000

$

$

$

321,000

There were no transfers between levels in any of the periods reported.

8. Stock-Based Compensation

The 2018 Omnibus Incentive Compensation Plan (the “2018 Plan”) was unanimously approved by the Company’s Board of Directors on May 24, 2018 and was approved by the Company’s stockholders on June 27, 2018.

Under the 2018 Plan, the Company may grant incentive stock options, non-qualified stock options, stock awards, stock units, stock appreciation rights and other stock-based awards to employees, non-employee directors and consultants, and advisors. The maximum aggregate number of shares of the Company’s common stock that may be issued under the 2018 Plan is 26,823.

The 2018 Plan was amended and restated following unanimous approval of the Company’s Board of Directors on April 24, 2019 and was approved by the Company’s shareholders on June 17, 2019. The amended 2018 Plan (the “Amended Plan”) allowed for an additional 39,300 shares of the Company’s common stock that may be issued under the Amended Plan with respect to awards made on and after June 17, 2019.

The 2021 Incentive Compensation Plan (the “2021 Plan”) was unanimously approved by the Company’s shareholders on July 30, 2021. Upon stockholders’ approval of the 2021 Plan, no further awards will be made under the amended 2018 Plan. Under the 2021 Plan, the Company may grant incentive stock options, non-qualified stock options, stock awards, stock units, stock appreciation rights and other stock-based awards to employees, non-employee directors and consultants, and advisors. The maximum aggregate number of shares of the Company’s common stock that may be issued under the 2021 Plan is 1,300,000. At June 30, 2021,2022, there were 5,205258,337 shares available for future issuance.

Stock-based compensation expense includes stock options granted to employees and non-employees and has been reported in the Company’s statements of operations and comprehensive loss in either research and development expenses or general and administrative expenses depending on the function performed by the optionee. NaN net tax benefits related to the stock-based compensation costs have been recognized since the Company’s inception. The

1412

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

Company recognized stock-based compensation expense related to stock options and restricted stock units as follows for the three and six months ended June 30, 20212022 and 2020:2021:

Three Months Ended June 30, 

Six Months Ended June 30, 

Three Months Ended June 30, 

Six Months Ended June 30, 

    

2021

    

2020

    

2021

    

2020

    

    

2022

    

2021

    

2022

    

2021

    

General and administrative

$

43,000

$

46,000

$

99,000

$

91,000

$

72,000

$

43,000

$

259,000

$

99,000

Research and development

17,000

46,000

26,000

94,000

169,000

17,000

278,000

26,000

$

60,000

$

92,000

$

125,000

$

185,000

$

241,000

$

60,000

$

537,000

$

125,000

A summary of stock option activity for the six months ended June 30, 20212022 is as follows:

    

Options Outstanding

    

Options Outstanding

Weighted

Weighted

Weighted-

Average

Weighted-

Average

Shares

Average

Remaining

Aggregate

Average

Remaining

Aggregate

Available

Number

Exercise

Contractual

Intrinsic

Number

Exercise

Contractual

Intrinsic

    

for Grant

    

of Shares

    

Price

    

Term (in years)

    

Value

    

    

of Shares

    

Price

    

Term (in years)

    

Value

Balance, December 31, 2020

 

12,339

57,939

$

368.10

 

8.38

$

Balance, December 31, 2021

 

452,999

$

20.71

 

9.42

$

Authorized

Granted

 

(21,000)

21,000

$

7.80

9.94

 

21,636

 

491,825

$

1.75

9.66

 

Exercised

 

(4,642)

$

4.65

8.47

 

 

0

$

0

$

Forfeitures

 

13,866

(13,866)

$

940.63

7.11

 

Balance, June 30, 2021

 

5,205

60,431

$

124.00

 

8.73

$

Vested or expected to vest, June 30, 2021

 

58,925

$

124.00

 

8.73

$

Exercisable at June 30, 2021

 

24,261

$

297.34

 

7.87

$

Forfeitures/adjustments

 

(94,271)

$

3.37

 

Balance, June 30, 2022

 

850,553

$

11.67

 

9.26

$

Exercisable at June 30, 2022

 

40,302

$

179.88

 

6.51

$

The Company accounts for all stock-based payments made to employees, non-employees and directors using an option pricing model for estimating fair value. Accordingly, stock-based compensation expense is measured based on the estimated fair value of the awards on the date of grant, net of forfeitures. Compensation expense is recognized for the portion that is ultimately expected to vest over the period during which the recipient renders the required services to the Company using the straight-line single option method.

The Company uses the Black-Scholes option-pricing model to estimate the fair value of stock options at the grant date. The Black-Scholes model requires the Company to make certain estimates and assumptions, including estimating the fair value of the Company’s Common Stock, assumptions related to the expected price volatility of the Common Stock, the period during which the options will be outstanding, the rate of return on risk-free investments and the expected dividend yield for the Company’s stock.

As of June 30, 2021,2022, there was $102,000$2,166,000 of unrecognized compensation expense related to the unvested stock options which is expected to be recognized over a weighted-average period of approximately 2.33 years.

2.29 years.

The weighted-average assumptions underlying the Black-Scholes calculation of grant date fair value of stock options include the following:

Six months ended June 30, 

    

2022

    

2021

 

    

Risk-free interest rate

 

2.01

%  

1.02

%  

 

Expected volatility

 

121.49

%  

123.32

%  

 

Expected term

 

5.85

years  

6.25

years

 

Expected dividend yield

 

0

%  

0

%  

 

Weighted average grant date fair value

$

1.51

$

0.25

1513

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

The weighted-average assumptions underlying the Black-Scholes calculation of grant date fair value include the following:

���

Six months ended June 30, 

    

2021

    

2020

 

    

Risk-free interest rate

 

1.02

%  

0.45

%  

 

Expected volatility

 

123.32

%  

105.14

%  

 

Expected term

 

6.25

years  

6.00

years

 

Expected dividend yield

 

0

%  

0

%  

 

Weighted average grant date fair value

$

3.80

$

3.75

The weighted-average valuation assumptions were determined as follows:

Risk-free interest rate: The Company based the risk-free interest rate on the interest rate payable on U.S. Treasury securities in effect at the time of grant for a period that is commensurate with the assumed expected option term.

Expected term of options: Due to its lack of sufficient historical data, the Company estimates the expected life of its employee stock options using the “simplified” method, as prescribed in Staff Accounting Bulletin (SAB) No. 107, whereby the expected life equals the arithmetic average of the vesting term and the original contractual term of the option.

Expected stock price volatility: Expected volatility is based on the historical volatility of the Company’s Common Stock since its IPO in July 2013.Stock.

Expected annual dividend yield: The Company has never paid, and does not expect to pay, dividends in the foreseeable future. Accordingly, the Company assumed an expected dividend yield of 0.0%.

Estimated forfeiture rate: The Company’s estimated annual forfeiture rate on stock option grants was 4.14% in 2021 and 2020, based on the historical forfeiture experience.

On August 2, 2021, the compensation committee of the board of directors approved restricted stock unit grants to the Company’s employees (“2021 RSU”). An aggregate of 104,700 service-based RSUs were issued at a grant date fair value of $5.19. The 2021 RSU awards will be settled in stock, vest 33% on each of the first and second anniversary of the date of grant, and vest 34% on the third anniversary of the date of grant. The 2021 RSU awards were granted under the 2021 Plan. There were no vesting events, expirations, forfeitures, or cancelations of the 2021 RSUs during the period. On February 7, 2022, the compensation committee of the board of directors approved restricted stock unit grants to the Companies employees (“2022 RSU”). An aggregate of 148,343 service-based RSUs were issued at a grant date fair value of $1.82. The 2022 RSU awards will be settled in stock, vest 33% on each of the first and second anniversary of the date of grant, and vest 34% on the third anniversary of the date of grant. The 2021 RSU and 2022 RSU awards were granted under the 2021 Plan. There were no vesting events, expirations, or cancelations of the 2021 RSU or 2022 RSU during the period. There were forfeitures of 31,033 RSUs during the six months ended June 30, 2022. At June 30, 2022, the unrecognized compensation cost related to unvested service-based RSUs was $568,000, which will be recognized over the remaining service period.

GrantsGrants of PSUs and SARs

On July 9, 2020, the compensation committee of the board of directors and the board approved a cash bonus program of cash-settled stock appreciation right (“2020 SAR”) awards and cash-settled performance stock unit (“2020 PSU”) awards to the Company’s employees. An aggregate of 2020 SAR awards with respect to 256,713 shares of common stock and 2020 PSU awards with respect to 124,220 shares of common stock were granted to the Company’s employees. The 2020 SAR awards will be settled in cash, vest 33% on the first anniversary of the date of grant, and the remaining 67% monthly over the next 24 months, have a per-share base amount of $8.40, which was the closing sales price of a share of the Company’s common stock on the grant date, and are in all cases subject to the terms and conditions of the Company’s form of SAR award agreement.

The 2020 PSUSAR awards vest 50% uponare cash-settled and were granted outside of the submission of a new drug application (“NDA”) to the U.S. FDA for rigosertib in higher-risk myelodysplastic syndromes (“HR-MDS”) and 50% upon U.S. FDA approval of rigosertib for HR-MDS. The 2020 PSU awards have a maximum value of $21.60 per share. The maximum price per share is the per-share value based on the Company’s market capitalization at $250 million2018 Plan and the Company’s outstanding shares of common stock, which was 11,611,829 shares on July 9, 2020. In all cases, the 2020 PSU awards are subject to the terms and conditions of the Company’s form of PSU award agreement.2021 Plan.

1614

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

The 2020 PSU awards vest 50% upon the submission of a new drug application (“NDA”) to the U.S. FDA for rigosertib in higher-risk myelodysplastic syndromes (“HR-MDS”) and 50% upon U.S. FDA approval of rigosertib for HR-MDS. The 2020 PSU awards have a maximum value of $21.60 per share. The maximum price per share is the per-share value based on the Company’s market capitalization at $250 million and the Company’s outstanding shares of common stock, which was 11,611,829 shares on July 9, 2020. In all cases, the 2020 PSU awards are subject to the terms and conditions of the Company’s form of PSU award agreement. The 2020 PSU awards are cash-settled and were granted outside of the 2018 Plan and the 2021 Plan.

In addition, on July 9, 2020, based on the recommendation of the compensation committee, the board approved a change in the non-employee director compensation policy that would provide for an annual SAR award (“2020 Director SAR”) with respect to 8,333 shares of common stock for each of the Company’s non-employee directors. No other changes to the non-employee director compensation policy were approved and, on July 9, 2020, the Board approved the initial 8,333 2020 Director SAR award to each of the non-employee directors for an aggregate total of 58,333 2020 Directors SAR awards granted. The 2020 Director SAR awards vest on the first anniversary of grant subject to the director’s continued service and will be settled in cash, have a per-share base amount of $8.40, and are in all cases subject to the terms and conditions of the Company’s form of 2020 Director SAR award agreement.

Each SAR subject to a 2020 SAR award represents the right to a cash payment equal to the excess, if any, of (i) the fair market value of each underlying share of the Company’s common stock, determined on the date of exercise of the SAR minus (ii) the base amount. Pursuant to the terms of the SAR awards, in no event may the cash payment for each SAR exceed $13.20, which is the maximum price per share of $21.60, minus the base amount of $8.40, subject to adjustment in accordance with the terms of the Stock Appreciation Right Award Agreement. The maximum price per share is the per-share value based on the Company’s market capitalization at $250 million and the Company’s outstanding shares of common stock, which was 11,611,829 shares on July 9, 2020.

On February 17, 2021, the compensation committee of the board of directors and the board approved a cash bonus program of cash-settled stock appreciation right (“2021 SAR”) awards and cash-settled performance stock unit (“2021 PSU”) awards to the Company’s employees. An aggregate of 2021 SAR awards with respect to 100,000 shares of common stock and 2021 PSU awards with respect to 100,000 shares of common stock were granted to the Company’s employees. The 2021 SAR awards will be settled in cash, vest 33% on the first anniversary of the date of grant, and the remaining 67% monthly over the next 24 months, have a per-share base amount of $22.65, which was the closing sales price of a share of the Company’s common stock on the grant date, and are in all cases subject to the terms and conditions of the Company’s form of SAR award agreement. Each SAR subject to a 2021 SAR award represents the right to a cash payment equal to the excess, if any, of (i) the fair market value of each underlying share of the Company’s common stock, determined on the date of exercise of the 2021 SAR minus (ii) the base amount. Pursuant to the terms of the 2021 SAR awards, in no event may the cash payment for each SAR exceed $15.45, which is the maximum price per share of $38.10, minus the base amount of $22.65, subject to adjustment in accordance with the terms of the Stock Appreciation Right Award Agreement. The maximum price per share is the per-share value based on the Company’s market capitalization at $600 million and the Company’s outstanding shares of common stock, which was 15,767,492 shares on February 17, 2021. The 2021 SAR awards are cash-settled and were granted outside of the 2018 Plan and the 2021 Plan.

The 2021 PSU awards vest 20% upon the initiation of a new clinical program with an in-licensed compound, 20% for reaching the recommended Phase 2 dose for any compound, 20% for the first patient enrolled in the expansion cohort of the Phase 1 ON123300 clinical trial, 20% for the first patient enrolled in a registrational study for any compound, and 20% for the topline data of a registrational study for any compound. The 2021 PSU awards have a maximum value of $38.10 per share. The maximum price per share is the per-share value based on the Company’s approximate market capitalization at $600$600 million and the Company’s outstanding shares of common stock, which was 15,767,492 shares on February 17, 2021. In all cases, the 2021 PSU awards are subject to the terms and conditions of the Company’s form of PSU award agreement.

1715

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

Company’s form of PSU award agreement. The 2021 PSU awards are cash-settled and were granted outside of the 2018 Plan and the 2021 Plan.

The fair value of the 20212021and 2020 SARs granted has been estimated using the Black-Scholes option-pricing model with the following weighted-average assumptions:

    

Six months ended

 

June 30, 2021

 

Risk-free interest rate

 

0.95

%

Expected volatility

 

129.79

%

Expected term

 

6.5 years

Expected dividend yield

 

0

%

Weighted average grant date fair value

$

0.62

As of June 30, 2022

Risk-free interest rate

3.01

%  

Expected volatility

133.44

%  

Expected term

5.14

years  

Expected dividend yield

0

%  

During the three months ended June 30, 2022 and 2021, the Company recognized $15,000 and $476,000 related to the SARs and PSUs, respectively. During the six months ended June 30, 2022 and 2021, the Company recognized $1,000 of reversals of compensation expense and $521,000 of compensation expense related to the SARs and PSUs. IncludedPSUs, respectively. The expense recognized in the 2021 periods includes approximately $442,000 of compensation expense related to SARs is $442,000 of expense resulting from the exercise of 2020 SARs during February 2021. As of June 30, 2021,2022, the SARs and PSUs liability was $102,000$63,000 and is included in accrued expenses. As of June 30, 2021,2022, there was $53,000$85,000 of unrecognized compensation cost related to the 2020 SARs and PSUs and $664,000 of unrecognized compensation cost related to the 2021 SARs and PSUs.

9. Research Agreements

The Company has entered into various licensing and right-to-sublicense agreements with educational institutions for the exclusive use of patents and patent applications, as well as any patents that may develop from research being conducted by such educational institutions in the field of anticancer therapy, genes and proteins. Results from this research have been licensed to the Company pursuant to these agreements. Under one of these agreements with Temple University (“Temple”), the Company is required to make annual maintenance payments to Temple and royalty payments based upon a percentage of sales generated from any products covered by the licensed patents, with minimum specified royalty payments. As 0 sales had been generated through June 30, 20212022 under the licensed patents, the Company has not incurred any royalty expenses related to this agreement. In addition, the Company is required to pay Temple a percentage of any sublicensing fees received by the Company.

10. Related-Party Transactions

The Company entered into a research agreement, as subsequently amended, with the Mount Sinai School of Medicine (“Mount Sinai”), with which a former member of its board of directors and a stockholder is affiliated. The agreement expired in June 2020 and was not renewed. The board member left the Company’s board in August 2020. Mount Sinai is undertaking research on behalf of the Company on the terms set forth in the agreements. Mount Sinai, in connection with the Company, will prepare applications for patents generated from the research. Results from all projects will belong exclusively to Mount Sinai, but the Company will have an exclusive option to license any inventions. Payments to Mount Sinai under this research agreement for the three months ended June 30, 2021 and 2020 were $0 and $77,000, respectively, and for the six months ended June 30, 2021 and 2020 were $0 and $201,000, respectively. At both June 30, 2021 and December 31, 2020, the Company had $77,000 payable to Mount Sinai under this agreement.

11. Securities Registrations and Sales Agreements

January 2020 Offering

On December 31, 2019, the Company entered into definitive securities purchase agreements with institutional investors for the issuance and sale in a registered direct offering of 1,844,168 shares of the Company's common stock at an offering price of $5.4225 per share.

18

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

Pursuant to the December 2019 HCW Engagement Letter, HCW agreed to serve as exclusive placement agent for the offering. In connection with the offering, the Company paid HCW an aggregate cash fee equal to 7.0% of the gross proceeds in the offering, management fee equal to 1.0% of the gross proceeds raised in the offering, $85,000 for non-accountable expenses; and $10,000 for clearing fees. The Company also issued to HCW or its designees placement agent warrant to purchase up to 92,208 shares of common stock at an exercise price of $6.7785 per share. The placement agent warrants are immediately exercisable and will expire on December 31, 2023.

The net proceeds to the Company from the offering, after deducting HCW's placement agent fees and expenses and other estimated offering expenses payable by the Company were approximately $9.0 million and were received in January 2020.

The offering was pursuant to a prospectus dated December 28, 2017, and a prospectus supplement dated as of December 31, 2019 filed in connection with a takedown from the Company's shelf registration statement on Form S-3 (File No. 333-221684). The offering closed on January 3, 2020.

January 7, 2021 Offering

On January 7, 2021, the Company entered into a purchase agreement with certain institutional and accredited investors for the sale of an aggregate of 1,303,408 shares of the Company’s common stock, at a purchase price of $6.675 per share.

Under the purchase agreement, subject to certain exceptions, the Company is prohibited from effecting or entering into an agreement to effect any “variable rate transactions” as defined in the purchase agreement for a period of five years following the closing of the offering.

In connection with the offering, pursuant to the purchase agreement we reimbursed Lincoln Park Capital Fund, LLC, as the lead investor (“Lincoln Park”), an aggregate of $100,000 for expenses incurred in connection with the offering, including any due diligence expenses and legal fees. Furthermore, pursuant to the purchase agreement, we have granted Lincoln Park certain rights to participate at fair value with other investors in up to 50% of the amount of any

16

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

future offerings of common stock or securities exercisable for or convertible into common stock that the Company seeks to complete within one year after the closing of the offering, other than a firm commitment public offering.

The net proceeds to the Company from the offering, after deducting Lincoln Park’s expenses and other estimated offering expenses payable by the Company were approximately $8.5 million.

The shares sold in the offering were offered and sold by the Company directly to the investors, without a placement agent, underwriter, broker or dealer, pursuant to an effective shelf registration statement on Form S-3 (File No. 333-237844) declared effective by the SEC on May 18, 2020, and the base prospectus contained therein. The offering closed on January 11, 2021.

February 10, 2021 Offering

On February 10, 2021, the Company entered into an underwriting agreement with Guggenheim Securities, LLC, , as representative of several underwriters, for the public offering of 1,666,667 shares of the Company’s common stock, at a public offering price of $15.00 per share. Under the terms of the underwriting agreement, the Company granted the underwriters an option, exercisable for 30 days, to purchase up to an additional 250,000 shares of common stock at the same price. The option was exercised prior to closing.

19

Table of Contents

Onconova Therapeutics, Inc.

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

In connection with the offering, the Company paid the underwriters a cash fee equal to 6% of the gross proceeds in the offering and $100,000 in legal fees and expenses.

The net proceeds to the Company from the offering, including exercise of the underwriters’ option, were approximately $26.7 million, after deducting fees and estimated offering expenses payable by the Company.

The offering was made pursuant to a registration statement (No. 333-237844) on Form S-3, which was initially filed by the Company with the SEC on April 24, 2020, amended on Form S-3/A that was filed with the SEC on May 15, 2020, and was declared effective by the SEC on May 18, 2020. The offering closed on February 16, 2021.

12. Subsequent EventAugust 2021 Equity Distribution Agreement

On JulyAugust 20, 2021, the Company entered into an Equity Distribution Agreement (the “Equity Distribution Agreement”) with Piper Sandler & Co. (“Piper Sandler”) under which the Company may offer and sell, from time to time at its sole discretion, shares of the Company’s common stock, with aggregate gross sales proceeds of up to $25.0 million through an “at the market” equity offering program under which Piper Sandler is the sales agent.

Under the Equity Distribution Agreement, the Company has the right to set the parameters for the sale of shares, including the number of shares to be issued, the time period during which sales are requested to be made, limitations on the number of shares that may be sold in any one trading day and any minimum price below which sales may not be made. Subject to the terms and conditions of the Equity Distribution Agreement, Piper Sandler may sell the shares by methods deemed to be an “at the market” offering as defined in Rule 415 promulgated under the Securities Act of 1933, as amended, including sales made through The Nasdaq Capital Market or any other trading market for our common stock. The Equity Distribution Agreement provides that Piper Sandler is entitled to compensation for its services equal to 3.0% of the gross proceeds of any shares of common stock sold through Piper Sandler under the Equity Distribution Agreement. The Company has no obligation to sell any shares under the Equity Distribution Agreement, and may at any time suspend solicitation and offers under the Equity Distribution Agreement. Through June 30, 2021, stockholdersapproved2022, the Company sold 109,523 shares under the agreement at a weighted average price of $5.32 per share. Net proceeds after commissions and offering expenses were approximately $0.5 million. There were 0 shares sold by the Company under the agreement during the six months ended June 30, 2022.

17

Table of Contents

Onconova Therapeutics, Inc. 2021 Incentive Compensation Plan (the ”2021Plan”)

Notes to Condensed Consolidated Financial Statements (Continued)

(Unaudited)

The shares are issued pursuant to the Company’s shelf registration statement on Form S-3 (File No. 333-237844). The Company filed a prospectus supplement, dated August 20, 2021 Plan is a successorwith the Securities and Exchange Commission in connection with the offer and sale of the shares pursuant to the Onconova Therapeutics, Inc. 2018 Omnibus Incentive Compensation Plan,Equity Distribution Agreement.

September 2021 Offering

On September 23, 2021, the Company entered into an underwriting agreement with Guggenheim Securities, LLC, as representative of several underwriters, for the public offering of 5,000,000 shares of the Company’s common stock, at a public offering price of $4.20 per share. Under the terms of the underwriting agreement, the Company granted the underwriters an option, exercisable for 30 days, to purchase up to an additional 750,000 shares of common stock at the same price. The option was not exercised.

In connection with the offering, the Company paid the underwriters a cash fee equal to 6% of the gross proceeds in the offering and $100,000 in legal fees and expenses.

The net proceeds to the Company from the offering, including exercise of the underwriters’ option, were approximately $19.5 million, after deducting fees and estimated offering expenses payable by the Company.

The offering was made pursuant to a registration statement (No. 333-237844) on Form S-3, which was initially filed by the Company with the SEC on April 24, 2020, amended on Form S-3/A that was filed with the SEC on May 15, 2020, and restated.was declared effective by the SEC on May 18, 2020. The 2021 Plan makes available 1,300,000 new shares for incentive compensation.offering closed on September 28, 2021.

2018

Table of Contents

Item 2. 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 should be read in conjunction with interim unaudited condensed consolidated financial statements contained in Part I, Item 1 of this quarterly report, and the audited consolidated financial statements and notes thereto for the year ended December 31, 20202021 and the related Management’s Discussion and Analysis of Financial Condition and Results of Operations, both of which are contained in our annual report on Form 10-K filed with the SEC on March 18, 2021.21, 2022. As used in this report, unless the context suggests otherwise, “we,” “us,” “our,” “the Company” or “Onconova” refer to Onconova Therapeutics, Inc. and its consolidated subsidiaries.

All common stock, equity, share and per share amounts have been retroactively adjusted to reflect a one-for-fifteen reverse stock split which was effective May 20, 2021.

Cautionary Note Regarding Forward-Looking Statements

This quarterly report on Form 10-Q includes forward-looking statements. We may, in some cases, use terms such as “believes,” “estimates,” “anticipates,” “expects,” “plans,” “intends,” “may,” “could,” “might,” “will,” “should,” “approximately” or other words that convey uncertainty of future events or outcomes to identify these forward-looking statements. Forward-looking statements appear in a number of places throughout this report and include statements regarding our intentions, beliefs, projections, outlook, analyses or current expectations concerning, among other things, our ongoing and planned preclinical development and clinical trials, the timing of and our ability to make regulatory filings and obtain and maintain regulatory approvals for our product candidates, protection of our intellectual property portfolio, the degree of clinical utility of our products, particularly in specific patient populations, our ability to develop commercial and manufacturing functions, expectations regarding clinical trial data, our results of operations, cash needs, financial condition, liquidity, collaborations, partnerships, prospects, growth and strategies, the industry in which we operate and the trends that may affect the industry or us.

By their nature, forward-looking statements involve risks and uncertainties because they relate to events, competitive dynamics and industry change, and depend on the economic circumstances that may or may not occur in the future or may occur on longer or shorter timelines than anticipated. Although we believe that we have a reasonable basis for each forward-looking statement contained in this report, we caution you that forward-looking statements are not guarantees of future performance and that our actual results of operations, financial condition and liquidity, and the development of the industry in which we operate may differ materially from the forward-looking statements contained in this report. In addition, even if our results of operations, financial condition and liquidity, and events in the industry in which we operate are consistent with the forward-looking statements contained in this report, they may not be predictive of results or developments in future periods.

Actual results could differ materially from our forward-looking statements due to a number of factors, including risks related to:

our need for additional financing for our INSPIRE trialclinical-stage programs, continued product development and other operations, and our ability to obtain sufficient funds on acceptable terms when needed, and our plans and future needs to scale back operations if adequate financing is not obtained;

our estimates regarding expenses, future revenues, capital requirements and needs for additional financing;

the success and timing of our preclinical studies and clinical trials, including site initiation and patient enrollment, and regulatory approval of protocols for future clinical trials;

our ability to enter into, maintain and perform collaboration agreements with other pharmaceutical companies, for funding and commercialization of our clinical product candidates or preclinical compounds, and our ability to achieve certain milestones under those agreements;

2119

Table of Contents

the difficulties in obtaining and maintaining regulatory approval of our product candidates, and the labeling under any approval we may obtain;

our plans and ability to develop, manufacture and commercialize our product candidates;

our failure to recruit or retain key scientific or management personnel or to retain our executive officers;

the size and growth of the potential markets for our product candidates and our ability to serve those markets;

regulatory developments in the United States and foreign countries;

the rate and degree of market acceptance of any of our product candidates;

obtaining and maintaining intellectual property protection for our product candidates and our proprietary technology;

the successful development of our commercialization capabilities, including sales and marketing capabilities;

recently enacted and future legislation and regulation regarding the healthcare system;

the success of competing therapies and products that are or become available;

our ability to maintain the listing of our securities on a national securities exchange;

the potential for third party disputes and litigation;

the performance of third parties, including contract research organizations (“CROs”) and third-party manufacturers; and

the impact of the novel coronavirus disease, COVID-19, to global economy and capital markets, and to our business and our financial results.

Any forward-looking statements that we make in this report speak only as of the date of such statement, and we undertake no obligation to update such statements to reflect events or circumstances after the date of this report or to reflect the occurrence of unanticipated events. Comparisons of results for current and any prior periods are not intended to express any future trends or indications of future performance, unless expressed as such, and should only be viewed as historical data.

You should also read carefully the factors described in the “Risk Factors” in our most recent annual report on Form 10-K, and quarterly reports on Form 10-Q, to better understand significant risks and uncertainties inherent in our business and underlying any forward-looking statements. As a result of these factors, actual results could differ materially and adversely from those anticipated or implied in the forward-looking statements in this report and you should not place undue reliance on any forward-looking statements.

Overview

We are a clinical-stage biopharmaceutical company focused on discovering and developing novel products for patients with cancer. We have proprietary molecularly targeted anti-cancer agents designed to disrupt specific cellular pathways that are important for cancer cell proliferation. We believe that the product candidates in our pipeline have the potential to be efficacious in a variety of cancers with unmet medical need. We have the following two clinical-stage programs: 1. ON 123300, multi-kinasenarazaciclib (ON 123300), a multi-targeted kinase inhibitor in solid tumors;tumors and hematological malignancies as a single agent or in combination with other anti-cancer therapies; and 2. rigosertib administered alone or in combination with PD-1 inhibitors for the treatment of solid tumors. We are currently evaluating potential compounds for in-licensing opportunities.

2220

Table of Contents

Our net losses were $8.9$8.1 million and $12.5$8.9 million for the six months ended June 30, 20212022 and 2020,2021, respectively. As of June 30, 2021,2022, we had an accumulated deficit of $437.4$452.9 million. We expect to incur significant expenses and operating losses for the foreseeable future as we continue the development of, and seek regulatory approval for, our product candidates, even if milestones under our license and collaboration agreements may be met. As of June 30, 2021,2022, we had $43.7$46.5 million in cash and cash equivalents.

On January 12, 2021, we closed on an offering of common stock. We issued 1,303,408 shares of common stock. Netstock and net proceeds were approximately $8.5 million.

On February 16, 2021, we closed on an offering of common stock. We issued 1,916,667 shares of common stock. Netstock and net proceeds were approximately $26.7 million. On September 28, 2021, we closed on an offering of common stock. We issued 5,000,000 shares of common stock and net proceeds were approximately $19.5 million.

On August 20, 2021, we entered into an at-the-market equity distribution agreement for the sale of up to $25.0 million of common stock. Through June 30, 2022, we sold 109,523 shares under the agreement at a weighted average price of $5.32 per share. Net proceeds after commissions and offering expenses were approximately $0.5 million.

On May 20, 2021, we amended our certificate of incorporation to effect a one-for-fifteen reverse stock split of our common stock. All common stock, equity, share and per share amounts in the financial statements and notes have been retroactively adjusted to reflect the reverse stock split.

On May 20, 2021, we amended our certificate of incorporation to decrease the number of authorized shares of common stock from 250,000,000 to 125,000,000.

We believe that our cash and cash equivalents of $43.7$46.5 million, at June 30, 2021,2022, will be sufficient to fund our operations and ongoing trials for more than eighteenat least 18 months from the date of this filing. We do not have a recurring source of revenue to fund our operations and will need to raise additional funds to continue to develop and apply for regulatory approval for our drug candidates.

We are exploring various sources of funding for development and applying for regulatory approval of our research compounds as well as for our ongoing operations. If we raise additional funds through strategic collaborations and alliances or licensing arrangements with third parties, which may include existing collaboration partners, we may have to relinquish valuable rights to our technologies or product candidates or grant licenses on terms that are not favorable to us. There can be no assurance, however, that we will be successful in obtaining such financing in sufficient amounts, on terms acceptable to us, or at all. In addition, there can be no assurance that we will obtain approvals necessary to market our product candidates or achieve profitability or sustainable, positive cash flow. If we are unable to successfully raise sufficient additional capital, through future financings or through strategic and collaborative arrangements, we will not have sufficient cash to fund our ongoing trials and operations.

Product Candidates / Compounds

ON 123300Narazaciclib (ON 123300) — Differentiated Multi-Kinase Inhibitor Targeting CDK4/6

We believe based on data from preclinical studies, that ON 123300 has the potential to overcome the limitations of the current generation of approved cyclin dependent kinase (CDK 4/6) inhibitors. Pursuant to a license agreement with Temple University dated January 1, 1999 as amended March 21, 2013, we licensed compounds including ON 123300our product candidate narazaciclib from Temple University. ON 123300 monolactate (ON 123300)Narazaciclib is a novel multimulti-targeted kinase inhibitor that targets both CDK4/targeting cyclin-dependent kinases (CDK) 2, 4, 6, and 9, AMPK related protein kinase 5 (ARK5), and colony-stimulating factor 1 receptor (CSF1R) at low nM concentrations as well as other tyrosine kinases believed to drive tumor proliferation.cell proliferation, survival and metastasis. As an apoptotic and antiproliferative agent, narazaciclib modulates the levels and activities of regulatory proteins of the cell cycle, including cyclin D1 and inhibits retinoblastoma (Rb) protein binding. Narazaciclib inhibits cancer cell growth and suppresses deoxyribonucleic acid (DNA) synthesis by preventing CDK-mediated G1-S phase transition, followed by tumor cell death by induction of mitochondria-mediated apoptosis. We believe, based on data from preclinical studies, that narazaciclib has the potential to overcome the limitations of the current generation of approved cyclin dependent kinase (CDK) 4/6 inhibitors. The below table depicts the half-maximal in vitro inhibitory concentration (IC50) of ON 123300narazaciclib palbociclib, ribociclib and Palbociclib, which

21

Table of Contents

abemaciclib. IC50 is a quantitative measure indicating the concentration of each drug needed to inhibit, in vitro, these listed kinases by 50%. We believe our CDK inhibitor is differentiated from other agents in the market or in development due to its multi-targeted kinase inhibition profile.

Narazaciclib

Palbociclib

Ribociclib

Abemaciclib

Sponsor

Onconova

Pfizer

Novartis

Lilly

CDK Family

CDK4/cyclin D1

2

2

3

0.8

CDK6/cyclin D1

0.6

0.8

6.0

0.6

CDK1/cyclin A

2190

>10,000

>10,000

270

CDK2/cyclin E

69

2300

>10,000

130

CDK9/T1

48

630

390

7

Other Kinases

CSF1R

0.7

>10,000

>10,000

>10,000

ARK 5/NUAK 1

5

1,400

1,540

773

FLT3

6.0

496

753

72

23Source: Reaction Biology 2021

TableIn addition to CDK 4/6, narazaciclib also inhibits ARK5 (NUAK1) with high potency with a 50% inhibitory concentration (IC50) of Contents

    

ON 123300 

    

PALBOCICLIB 

Kinase

IC50 (nM)

IC50 (nM)

CDK 4/cyclin D1

 

3.87

 

5.36

CDK 6/cyclin D1

 

9.82

 

3.76

ARK 5

 

4.95

 

>5,000.00

FLT3

 

12.22

 

>10,000.00

FYN

 

11.09

 

>10,000.00

FMS

 

10.00

 

>10,000.00

PDGFRβ

 

26.00

 

>10,000.00

FGFR1

 

26.00

 

>10,000.00

ABL

 

53.32

 

>10,000.00

PI3K-δ

 

144.00

 

>10,000.00

-ON 123300 Investigator Brochure v1.

One such tyrosine4.95 nM (Report EPR-123300-001 and Reddy 2014) while palbociclib, ribociclib, and abemaciclib do not. The equilibrium dissociation constant (Kd) value of narazaciclib binding to ARK5 was found to be 19 nM, while a known NUAK1 specific inhibitor (HTH-015-01) was 790 nM. In addition, using a cellular based assay that measures kinase activity in intact cells, NanoBret technology, it was determined that narazaciclib inhibited ARK5 alsowith an IC50 value of 30 nM, while 2 published inhibitors, HTH-015-01 and WZ4003, had IC50 values of >10,000 nM. ARK5 (also known as NUAK1, regulatesNUAK1) is a member of the AMP-activated protein kinase (AMPK) catalytic subunit family and functions as a key regulator of cellular energy homeo-stasis (Lui 2012). ARK5 has been shown to be important in a number of cancer cell regulated survival pathways such as regulating AKT dependent cell survival, cell metabolism through c-MYC activity, tumor cell survival under oxidative stress and tumor cell migration (perhaps involved with metastases) through inhibition of cellular metabolism.(Faisal, 2020, Lui, 2012, Port, 2018). The combination of CDK and ARK5 inhibitors in the same molecular entity is proposed to have a differentiated effect on cancer cells by simultaneously inhibiting both cell cycle (cytostatic) and cellular metabolism (cytotoxic) pathways through CDK and ARK5, respectively.

Narazaciclib also inhibits CSF1R with IC50 values between 0.7 to 10 nM (Unpublished data and Reddy 2014). The Kd value of narazaciclib binding to CSF1R was determined to be 0.7 nM. In addition, using a cellular based assay that measures kinase activity in intact cells, NanoBret technology, it was determined that narazaciclib inhibited CSF1R with an IC50 value of 0.7 nM. The ability of narazaciclib to bind and inhibit CSF1R at low nanomolar values, in both in vitro and cell-based assays suggests that this compound may have an impact in cancers with a dependence on CSF1R signaling.

We and our partner, HanX Biopharmaceuticals, Inc. (“HanX”), have recently initiated phase 1 dose escalation clinical studies to begin evaluating the safety, tolerability and PK of narazaciclib in order to establish a recommended phase 2 dose (RP2D). Once we establish the RP2D, we intend to progress clinical development to determine whether thesethe findings from preclinical studies may translate to clinical activity or potential clinical benefit in cancer patients.

In certain in vitro models, the kinase inhibitory profile of ON 123300narazaciclib had the highesthigh activity against CDK4, CDK6, ARK5, FGFR1,CSF1R, PDGFRß and PI3K-δ, all of which are associated with the growth, survival and metastasis of human tumor cells (Reddy, 2014). In an in vitro investigation of ON 123300narazaciclib against a broad spectrum of human tumor cell lines, ON 123300narazaciclib displayed potent antiproliferative activity, with 50% growth inhibitory concentrations (GI50) ranging from 0.02 µM to 1.5 µM. In these in vitro models, ON 123300narazaciclib exhibited a broad range of activity against a wide spectrum of cell lines of both hematological origin (lymphoma, leukemia and myeloma) as well as solid tumors derived from multiple organ sites. Studies on drug-resistant human tumor cell lines suggested that ON 123300narazaciclib is not a multidrug resistance gene (mdr1) substrate and may be active against drug-resistant tumor cell lines (IBv.1 2020; Reddy, 2014). The activity of ON 123300narazaciclib does not appear to be affected by the overexpression of MDR-1 and induced apoptosis in

22

Table of Contents

both ibrutinib-sensitive and ibrutinib-resistant patient derived cells (Divakar, 2016). The ability of ON 123300narazaciclib to inhibit the CDK4/6/RB1 pathway has also been shown in pre-clinical testing of mantle cell lymphoma (Divakar, 2016), multiple myeloma (Perumal, 2016), various breast cancer subtypes (Reddy 2014) and colorectal cancer (IBv.1 2020)(IBv.2 2022).

The effectiveness of first-generation non-selective CDK inhibitors (Selicilib/roscovitine and Alvocidib/ flavopiridol) in early trials was limited due to toxicities (Blachly 2013). Second-generation compounds (palbociclib ribociclib and abemaciclib)ribociclib) specifically inhibit CDK4 and 6, thereby inhibiting retinoblastoma (RB) protein phosphorylation. Abemaciclib is a multi-targeted kinase CDK4/6 inhibitor with low nano molar activity against CDK4/6. The second generation CDK4/6 inhibitors have substantially improved clinical outcomes for patients with hormonal-receptor (HR) positive metastatic breast cancer (Hortobagyi 2018, Sledge 2017, Finn 2016). Several CDK4/6 inhibitors (palbociclib, ribociclib and abemaciclib) have been approved and are now standard of care either alone (abemaciclib) or in combination with anti-estrogen therapy for patients with HR-positive, HER2-negative metastatic breast cancer. Another CDK4/6 inhibitor has recently been approved, trilaciclib, in the supportive care space, for the prevention of myelosuppression following chemotherapy.

In December 2017, we entered into a license and collaboration agreement with HanX, a company focused on development of novel oncology products, for the further development, registration and commercialization in China of ON 123300.narazaciclib. Under the terms of the agreement, we received an upfront payment, and will receive regulatory and commercial milestone payments, as well as royalties on any future Chinese sales if the drug is approved. The key feature of the 2017 collaboration was that HanX provided all funding required for the Chinese Investigational New Drug

24

Table of Contents

Application (a “IND”) thereby enabling the studies necessary in order to seek IND approval by the National Medical Products Administration (Chinese FDA). In the fourth quarter of 2019, HanX filed an IND with the Chinese FDA which was approved on January 6, 2020. We and HanX also intended for these studies underlying the Chinese IND approval, to meet the US Food and Drug Administration (“FDA”) standards for IND approval. Accordingly, such studies were used by us for an IND filing with the US FDA. In September 2020, a Phase 1 Study with ON123300narazaciclib in cancer patients was initiated in China. We maintain global rights to the study and study data outside of China.

Our IND submission to the US FDA was submitted in November 2020 and the FDA Study May Proceed letter was issued in December 2020. Enrollment into the US phase 1 study (Study 19-01) commenced in May 2021. Enrollment in the firstfourth dose cohort of the Phase 1 solid tumor study of ON 123300narazaciclib is complete with no dose limiting toxicities (DLT’s) observed. The secondfifth dose cohort is currently open for enrollment.ongoing. The study will assess the safety, tolerability and pharmacokinetics of ON 123300narazaciclib administered orally at increasing doses starting at 40 mg daily for consecutive 28-day cycles in patients (n=36) with relapsed/refractory advanced cancer, including but not limited to, patients with breast cancer that is resistant to approved second generation CDK 4/6 inhibitors as well as patients diagnosed with advanced Non-Hodgkin’s lymphoma. cancer.

In partnership with HanX, a complementary Phase 1 dose escalation study (Study HX301) for patients with advanced relapsed/refractory cancer has been initiated in China at three sites and the first patient was enrolled on September 15, 2020. TheIn this study HX301 (narazaciclib) is dosed every day for 21 days followed by 7 days off therapy in each 28 -day cycle. In China, the first twofour dose cohorts have been completed and the third dosecompleted. The fifth cohort is ongoing. No dose limiting toxicities have been observed to date. enrolling patients at 200 mg per day.

Collectively, once completed, these two Phase 1 studies are expected to provide preliminary safety data regardingand the safety profile of ON 123300recommended Phase 2 dose and potentially preliminary efficacy signals in patients with advanced cancer.

Positive preclinical data was announced at the American Associationschedule for Cancer Research (AACR) annual meeting, which took place April 1-5, 2017 in Washington, DC, for ON 123300. We believe our CDK inhibitor is differentiated from other agents in the market or in development due to its multi-kinase inhibition.

narazaciclib as a single agent.

Retinoblastoma (Rb) protein is a master regulator of cell division and is critical to several cellular processes including senescence, self-renewal, replication and apoptosis (Engel, 2015). It is believed that loss or inactivation of Rb by CDKs leads to malignant cell formation and occurs in the pathogenesis of mostsome cancers. In a preclinical Retinoblastoma (Rb) positive xenograft model for breast cancer, ON 123300narazaciclib activity was shown to be similar to palbociclib (Pfizer’s Ibrance ®). Moreover, based on the same preclinical model, ON 123300narazaciclib may have the potential advantage of reduced neutropenia when compared to palbociclib. Whereas both compounds resulted in decreased RBC and platelet counts in this preclinical model system, palbociclib was found to have a more prominent and statistically significant (P< 0.05)0.01) inhibitory effect on neutrophil counts when compared to ON 123300.narazaciclib. These results would need to be replicated in clinical trials.

23

Table of Contents

In vitro studies compared the growth inhibitory activity of ON 123300narazaciclib and palbociclib in breast cancer RB null cell lines, with mutated or deleted RB, which demonstrated resistance to palbociclib but retainedwhile maintaining sensitivity towards ON 123300 (IBv.1 2020)narazaciclib (IBv.2 2022). Further analysesStudies using mantle cell lymphoma cells indicated that ON 123300narazaciclib was able to induce cell death via induction of apoptosis by inhibiting the AKT/PI3KPI3K/mTOR pathway while palbociclib treatment was only able to induce cell cycle arrest due to the inhibition of CDK4/6 (Divakar, 2016). ON 123300Narazaciclib treatment was associated with the presence of several apoptotic markers (PARP, caspase 3, caspase 7 and caspase 9) and ON 123300narazaciclib (but not palbociclib) led to the generation of apoptotic cells. Overall, apoptosis following ON 123300narazaciclib exposure has been observed in the following cell lines: breast cancer (IBv.1 2020,(IBv.2 2022, Reddy, 2014), mantle cell lymphoma (Divakar, 2016), multiple myeloma (Perumal, 2016) and colorectal cancer (IBv.1 2020)(IBv.2 2022).

In addition to CDK4/6 and PI3 Kinase ON 123300 may inhibitpathways, narazaciclib inhibits several other kinases in vitro including ARK5 (NUAK1) (IC50 of 4.95 nM) (IBv.1 2020,(IBv.2 2022, Reddy, 2014) while palbociclib does not. ARK5 is a member of the AMP -activated protein kinase (AMPK) family and is thought to function as a key regulator of cellular energy homeo-stasis (Liu, 2012) and is important in a number of cancer cell survival pathways. Overexpression of ARK5 is associated with poor prognosis in hepatocellular carcinomacancer (Cui, 2013), ovarian cancerscancer (Phippen, 2016), colorectal cancer (Port, 2018) and glioblastoma (Lu, 2013). ARK5 is involved in the increased invasiveness, migration and metastatic potential of breast cancer cells (Chang, 2012), colorectal cancer (Kusakai, 2004), gastric cancer (Chen, 2017), and multiple myeloma (Suzuki et al., 2005). ON 123300Narazaciclib inhibits ARK5 resultingwhich may result in down regulation of the mTOR/MYC/RB1 pathways leading to cell cycle arrest and apoptosis.

25

Table of Contents

Because ARK5 activity is now recognized as cruciala component in promoting cancer cell migration and invasion (Kusaki, 2004) the effect of ON 123300narazaciclib treatment may have an impact on cell migration and wound healing.metastasis. In certain in vitro models, ON 123300narazaciclib was able to inhibit the percent migration of U87 cells in a concentration- dependent manner. The time and concentrations that were tested did not result in cell death but did inhibit cell division at the higher concentrations (IBv.1 2020)(IBv.2 2022). The ability of ON 123300narazaciclib to inhibit cell migration was compared to palbociclib using a wound healing model. Triple negative cancer cell migration was inhibited for 72 hours in the presence of ON 123300narazaciclib but not in the presence of palbociclib (IBv.1 2020)(IBv.2 2022).

The pathogenesis and progression of a number of cancers, including breast cancerand multiple myeloma, is linked to C-Myc expression(Li, 2003) which is subsequentlywas dependent on ARK5 activity.activity (Liu, 2012) and calcium dependent metabolism (Monteverde, 2018). The inhibition of ARK5 has been shown to be lethal in MYC overexpressing tumors (Liu, 2012)2012, Perumal, 2016) and targeting ARK5 in the inhibitory profile of ON 123300narazaciclib has the potential to overcome the emergence of resistance to CDK4/6 inhibitors due to the loss of retinoblastoma function and C-Myc overexpression. Preclinical studies with tumor cell lines suggest that several malignancies including HR-positive breast cancer, colorectal carcinoma, hepatocellular carcinoma, mantle cell lymphoma and multiple myeloma, may be clinically responsive to ON 123300narazaciclib exposure (Reddy, 2014, Divakar, 2016, Perumal, 2016). Furthermore, ON 123300narazaciclib has been tested in fivefour murine xenograft models (breast cancer, including triple negative disease, colorectal cancer, mantle cell lymphoma and multiple myeloma) and was found to have on-target activity and be non-toxic to the animals (Reddy, 2014; Divakar, 2016; Perumal, 2016; and IBv.1 2020)IBv.2 2022).

CSF1R is in the class III kinase receptors that include c-Kit, platelet-derived growth factor receptor (PDGFR) alpha, and FLT3. CSF1R has 2 high affinity binding ligands, colony stimulating factor 1 (CSF-1), also known as macrophage colony-stimulating factor (M-CSF) and interleukin 34 (IL-34). CSF-1 is important for the differentiation and proliferation of myeloid progenitor cells into macrophages, monocytes, dendritic cells, and osteoclasts. Macrophages play an important role in the pathogenesis of not only tumor growth but multiple other diseases such as inflammatory diseases and bone metabolism. High levels of CSF-1 are critical for the recruitment of tumor associated macrophages (TAMs), predominantly the immunosuppressive phenotype (M2). They are the main inflammatory immune cells in the tumor microenvironment and are involved in tumor immunosuppression, angiogenesis, invasion, and metastasis.

Overexpression of CSF-1 or CSF1R is associated with tumor aggressiveness and poor prognosis. Inhibiting the signaling pathway of CSF1R provides a method to reduce the number of M2 macrophages/TAMs within the tumor microenvironment and thus improve anti-tumor immunological therapy. Recent studies have found that CSF-1/CSF1R axis blockade can improve the efficiency of immune checkpoint inhibitors, especially programmed death-ligand 1 inhibitors.

24

Table of Contents

Cancer cells can lose RB function through mutation and become resistant or insensitive to palbociclib. Generally, second generation agents have not been shown to be suitable for single agent therapy and must typically be used in combination with hormonal therapy.therapy in the treatment of HR+/HER2- mBC. In addition, the rate of disease progression that occurs, especially in patients with visceral disease (Hortobagyi 2018), may benefit from the novel inhibitory effects of ON 123300.narazaciclib. This hypothesis needs to be proven in a clinical trial.

Unfortunately, several mechanisms of acquired resistance are emerging with the approved CDK4/6 inhibitors leading to progression in patients with breast cancerHR+/HER2- mBC (Spring, 2019; Knudsen, 2020). Therefore, the unmet medical need supports development of the next (third) generation CDK4/6 inhibitors in advanced HR+/HER- breast cancer.mBC. The inhibitory effect of ON 123300narazaciclib may provide a therapeutic strategy to optimize efficacy of CDK 4/6 inhibition and reduce the emergence of resistance.

resistance and/or provide clinical benefit for patients with progression on palbociclib, ribociclib and/or abemaciclib.

We believe ON 123300narazaciclib has a favorable kinase inhibitory profile in comparison to the approved CDK4/6 inhibitors (palbociclib, ribociclib, and abemaciclib) and highest single agent cytotoxicitymay result in both tumorigenic and safety benefits (Perumal, 2016, Divakar, 2016).

Based on data from continuous dosing studies in rats and monkeys the safety profile of ON 123300narazaciclib is anticipated to be similar tobetter than the approved CDK4/6 inhibitors with myelosuppression and gastrointestinal toxicity being most common. Management of these adverse events is expected to follow that used for the approved CDK 4/6 inhibitors. We believe that the proposed mechanism of action of ON 123300,narazaciclib, the unmet medical need of the advanced cancers potentially targeted by ON 123300narazaciclib and the anticipated safety profile of ON 123300narazaciclib as seen in pre-clinical studies, support conducting Phase 1 clinical studies.

Clinical development of ON 123300narazaciclib for both breast cancer as well as other solid tumors and hematological malignancies in clinical trials is warranted based on the preclinical in vitro studies as well as the xenograft models. Onconova plans to advance testing whether ON 123300narazaciclib will demonstrate improved activity and/or safety in patients with advanced malignancies.

Oral Rigosertib and PD-1 Combination in KRAS-Mutated Cancers

We are currently supporting investigator-initiated studies (ISS) that are exploring the use of rigosertib for cancers driven by mutated Ras genes including a Phase 1/2a study of rigosertib in combination with a PD-1 inhibitor for patients with progressive K-Ras mutated non-small cell lung cancer (NSCLC). The NSCLC study is open and continues to enroll patients. The objectives of this study are to identify the recommended Phase 2 dose (RP2D) for future studies and characterize the safety profile of the combination treatment. ResultsFinal results of the Phase 1 portion of the study are expected in 2021.late 2022. On June 28, 2021, we announced an update regarding this NSCLC study, with an expansion of the trial underway at the highest dose in the current

26

Table protocol. To date, one patient with a dose limiting toxicity of Contents

protocol, and continuedhyponatremia has been observed. Continued dose escalation planned with a protocol amendmentis being considered as we believe the maximum tolerated dose has not been reached. In addition, preliminary efficacy data support the preclinical observation of rigosertib augmenting the response to checkpoint inhibition (CPI) in patients who had previously failed all standard of care treatment, including CPI. We look forward to the presentation of preliminaryInterim data presented at the upcoming 3rd Annual RAS Targeted Drug Development Summit taking(September 21-23, 2021), demonstrated two partial responses and one stable disease out of seven evaluable patients, or a clinical benefit rate of 43% (3/7). The three patients with clinical benefit harbored different KRAS mutations; suggesting that patients with a variety of KRAS mutations may have the potential to respond to the novel combination including rigosertib. An abstract with updated information was submitted to the European Society of Medical Oncology (ESMO) meeting scheduled to take place in September 21-23, 2021, and at a future major medical meeting as the data mature.

2022. We believe this supports further investigation of rigosertib in combination with CPI in KRAS mutated NSCLC.

On June 17, 2021, we announced a publication in Molecular Cancer (Yan, C., Saleh, N., Yang, J. et al. Novel induction of CD40 expression by tumor cells with RAS/RAF/PI3K pathway inhibition augments response to checkpoint blockade. Mol Cancer 20, 85; 2021) demonstratingwhich demonstrated that rigosertib synergistically combined with CPI to improveimproved tumor growth inhibition and survival in a murine melanoma model that did not respond to CPI alone. Rigosertib’sIt was postulated

25

Table of Contents

that rigosertib’s anti-cancer activity was due to its ability to reverse immunosuppressive tumor microenvironments. ThisWe believe this pre-clinical data support the clinical evaluation of rigosertib in combination with checkpoint inhibitors. We anticipate, additional investigator initiated studies in RAS driven cancers in combination with PD-1 inhibitors, includinga CPI in metastatic melanoma.

melanoma that has progressed on CPI therapy and we expect an ISS for continued development in the area will be open for enrollment in mid-2022.

Rigosertib as monotherapy

An investigator-initiatedBased on rigosertib’s activity against another important cell cycle pathway, PLK-1 (Antanasova, 2019), a Phase 1b/2 studyISS with rigosertib monotherapy in patients with advanced squamous cell carcinoma associated with recessive dystrophic epidermolysis bullosa (RDEB-SCC) hasis enrolling patients. As we disclosed in December 2021 early preliminary data from an investigator-initiated Phase 2 open label trial of rigosertib monotherapy in advanced squamous cell carcinoma complicating recessive dystrophic epidermolysis bullosa (RDEB- SCC) were presented at the Austrian Society of Dermatology and Venerology Annual Conference 2021, which took place from November 25 – 27, 2021 and more recently at the World Congress on Rare Skin Diseases which took place in Paris, June 7-9, 2022.

RDEB is an ultra-rare condition with high unmet medical need caused by a lack of type VII collagen protein expression. Type VII collagen protein is responsible for anchoring the skin’s inner layer to its outer layer, and its absence leads to extreme skin fragility and chronic wound formation in RDEB patients. Over time, many of these patients develop squamous cell carcinomas (SCCs) that typically arise in areas of chronic skin wounding and inflammation. Preclinical investigations demonstrated overexpression of polo like kinase 1 (PLK1) in RDEB-associated SCC tumor cells. These tumors show a highly aggressive, early metastasizing course, making them the primary cause of death for these patients, with a cumulative risk of death of 70% and 78.7% by age 45 and 55, respectively (Mellerio, 2016), (Fine, 2016). These neoplasms show limited response rates of mostly short duration to conventional chemo- and radiotherapy as well as targeted therapy with epidermal growth factor and tyrosine kinase inhibitors (Mellerio, 2016), (Stratigos, 2020).

Data from the recent presentations are from a patient with a history of multiple, unresectable SCCs that were unresponsive to prior treatments including cemiplimab. Results showed that intravenously administered rigosertib had an acceptable safety profile and that the patient experienced sustained clinical and histological complete remission of all lesions without signs of metastatic disease following 13 treatment cycles. The patient remains in complete remission and on drug, and the trial remains ongoing. A second patient was recently enrolled, its first patient.. Ahowever, the patient was removed from the study by the Principal Investigator due to noncompliance. Additional patients are anticipated.

Though the trial’s currently available safety and efficacy data are from only a single patient, the investigators believe they represent an encouraging finding that warrants further study. In addition, the investigators and we believe the data generated in preclinical study is also currently investigatingmodels that suggest rigosertib’s activity against PLK1 have now been preliminarily supported in the clinic and suggest that rigosertib may play a role in clear cell renal carcinoma (ccRCC).

other more common cancers driven by PLK1.

Rare Disease Program in “RASopathies”

Based on the mechanism of action data published in the journal Cell in 2016, we initiated a collaborative development program focusing on a group of rare diseases with a well- defined molecular basis in expression or defects involving the Ras effector pathways. Since RASopathies are rare congenital diseases affecting young children, we embarked on a multifaceted collaborative program involving patient advocacy, government and academic organizations. RASopathies are usually caused by germline mutations in genes that alter the RAS subfamily and mitogen-activated protein kinases (MAPK) that control signal transduction and are among the most common genetic syndromes. Together, this group of diseases can impact more than 1 in 1,000 individuals, according to RASopathies.Net.

The NCI has conducted preclinical studies with cell lines from two pediatric solid tumors (rhabdomyosarcoma and neuroblastoma), including xenograft models. For both tumor cell lines, in vitro rigosertib exposure was associated with reduced cell viability associated with destabilization of microtubules, mitotic arrest and apoptosis. In a rhabdomyosarcoma xenograft model, rigosertib treatment delayed time to tumor progression and prolonged survival in the animals treated with rigosertib. (Kowalczyk, 2020).

26

Studies using leukemia cells from the rare childhood RASopathy, known as Juvenile Myelomonocytic Leukemia (JMML), have been conducted. In preliminary in vitro studies performed at Notable Labs, JMML cell killing was observed following rigosertib exposure. Murine xenograft studies performed at the UniversityTable of California, San Francisco and funded through the Leukemia Lymphoma Society, evaluated rigosertib in this Ras-mutated disease. Further studies with JMML and rigosertib are under consideration.Contents

Critical Accounting Policies and Significant Judgments and Estimates

This management’s discussion and analysis of our financial condition and results of operations is based on our interim unaudited consolidated financial statements, which have been prepared in accordance with GAAP. The preparation of these financial statements requires us to make estimates and judgments that affect the reported amounts of assets, liabilities, revenues and expenses and the disclosure of contingent assets and liabilities in our consolidated financial statements. On an ongoing basis, we evaluate our estimates and judgments, including those related to accrued expenses, revenue recognition, deferred revenue and stock-based compensation. We base our estimates on historical experience, known trends and events and various other factors that we believe to be reasonable under the circumstances, the results of which form the basis for making judgments about the carrying values of assets and liabilities that are not readily apparent from other sources. Actual results may differ from these estimates under different assumptions or

27

Table of Contents

conditions.  There have been no significant changes in our critical accounting policies and estimates as discussed in our annual report on Form 10-K filed with the SEC on March 18, 2021.21, 2022.

The full extent to which COVID-19 will directly or indirectly impact our business, results of operations and financial condition, including expenses and manufacturing, clinical trials and research and development costs, depends on future developments that are highly uncertain at this time.

Results of Operations

Comparison of the Three Months Ended June 30, 20212022 and 20202021

Three Months Ended June 30, 

Three Months Ended June 30, 

    

2021

    

2020

    

Change

    

2022

    

2021

    

Change

Revenue

$

57,000

$

56,000

$

1,000

$

57,000

$

57,000

$

Operating expenses:

 

  

 

  

 

  

 

  

 

  

 

  

General and administrative

 

2,850,000

 

2,594,000

 

(256,000)

 

2,139,000

 

2,850,000

 

711,000

Research and development

 

1,852,000

 

4,801,000

 

2,949,000

 

2,038,000

 

1,852,000

 

(186,000)

Total operating expenses

 

4,702,000

 

7,395,000

 

2,693,000

 

4,177,000

 

4,702,000

 

525,000

Loss from operations

 

(4,645,000)

 

(7,339,000)

 

2,694,000

 

(4,120,000)

 

(4,645,000)

 

525,000

Change in fair value of warrant liability

 

427,000

 

(56,000)

 

483,000

 

 

427,000

 

(427,000)

Other income (expense), net

 

(13,000)

 

 

(13,000)

Other income, net

 

96,000

 

(13,000)

 

109,000

Net loss

$

(4,231,000)

$

(7,395,000)

$

3,164,000

$

(4,024,000)

$

(4,231,000)

$

207,000

Revenues

Revenues increased by $1,000, or 2%, for the three months ended June 30,2022 were consistent with 2021, when comparedand were due to the same period in 2020 becauserecognition of a clinical supply creditdeferred revenue from SymBio in the 2020 period.our collaboration with SymBio.

General and administrative expenses

General and administrative expenses increased by $0.3decreased $0.7 million, or 10%26%, to $2.1 million for the three months ended June 30, 2022 from $2.9 million for the three months ended June 30, 2021 from $2.62021. This decrease was caused by $0.9 million for the three months ended June 30, 2020. The increase was attributable primarily to $0.8 million of expenses fordecrease in investor relations, proxy solicitation, and fees related to our special meeting by proxy in the 2021 period, and also to $0.1 million higher insurance expenses. These increases werewhich did not occur in 2022. This decrease was mostly partially offset by $0.3a $0.2 million lower commercialization expensesincrease in salary costs and bonus accruals in the 2021 period.

The details of our general and administrative expenses are:

Three Months Ended June 30, 

    

2021

    

2020

Professional & consulting fees

$

423,000

$

708,000

Stock based compensation

86,000

46,000

Personnel related

 

772,000

 

692,000

Commercial

359,000

Public company costs

 

1,324,000

 

550,000

Insurance & other

245,000

239,000

$

2,850,000

$

2,594,000

Research and development expenses

Research and development expenses decreased by $2.9 million, or 61%, to $1.9 million for the three months ended June 30, 2021 from $4.8 million for the three months ended June 30, 2020. This decrease was caused primarily by $2.0 million lower clinical development and consulting expenses on the INSPIRE program in the 2021 period, and also by $0.7 million lower personnel and stock compensation expense during the 2021 period, following reductions in our

28

Table of Contents

workforce completed in the third and fourth quarter of 2020 and $0.2 million lower manufacturing and development expenses related to rigosertib.

The details of our research and development expenses are:

Three Months Ended June 30, 

    

2021

    

2020

Preclinical & clinical development

$

594,000

$

2,002,000

Personnel related

 

296,000

 

993,000

Manufacturing, formulation & development

 

553,000

 

739,000

Stock based compensation

 

7,000

 

47,000

Consulting fees

 

402,000

 

1,020,000

$

1,852,000

$

4,801,000

Change in fair value of warrant liability

The fair value of the warrant liability decreased $427,000 for the three months ended June 30, 2021, compared to an increase of $56,000 for the three months ended June 30, 2020. This change was caused by a decrease in the 2021 period of the fair market value of the warrants issued in our rights offering in 2016. These warrants expired in July 2021.

Other income (expense), net

Other income (expense), net, was a $13,000 loss for the three months ended June 30, 2021 and $0 for the three months ended June 30, 2020.  The change of $13,000 was due to $6,000 lower net interest income in the 2021 period and $7,000 higher foreign currency exchange losses in the 2021 period.

Comparison of the Six Months Ended June 30, 2021 and 2020

Six Months Ended June 30, 

    

2021

    

2020

    

Change

Revenue

$

113,000

$

108,000

$

5,000

Operating expenses:

 

  

 

  

 

  

General and administrative

 

5,067,000

 

4,401,000

 

(666,000)

Research and development

 

3,789,000

 

8,171,000

 

4,382,000

Total operating expenses

 

8,856,000

 

12,572,000

 

3,716,000

 

Loss from operations

 

(8,743,000)

 

(12,464,000)

 

3,721,000

 

Change in fair value of warrant liability

 

(209,000)

 

(119,000)

 

(90,000)

 

Other income (expense), net

 

6,000

 

96,000

 

(90,000)

 

Net loss

$

(8,946,000)

$

(12,487,000)

$

3,541,000

Revenues

Revenues increased by $5,000, or 5%, for the six months ended June 30, 2021 when compared to the same period in 2020 because of a clinical supply credit from SymBio in the 2020 period.

General and administrative expenses

General and administrative expenses increased by $0.7 million, or 15%, to $5.1 million for the six months ended June 30, 2021 from $4.4 million for the six months ended June 30, 2020. The increase was attributable primarily to $1.1 million of expenses for investor relations, proxy solicitation, and fees related to our special meeting by proxy in the 2021 period, to $0.1 million higher personnel related costs, and also to $0.2 million higher insurance expenses. These increases were partially offset by $0.3 million lower professional and consulting fees, and $0.4 million of commercial expenses during the 20202022 period.

2927

Table of Contents

The details of our general and administrative expenses are:

Six Months Ended June 30, 

Three Months Ended June 30, 

    

2021

    

2020

    

2022

    

2021

Professional & consulting fees

$

941,000

$

1,202,000

$

417,000

$

423,000

Stock based compensation

120,000

90,000

72,000

43,000

Personnel related

 

1,573,000

 

1,420,000

 

919,000

 

772,000

Commercial

423,000

Public company costs

 

1,855,000

 

738,000

 

372,000

 

1,367,000

Insurance & other

578,000

528,000

359,000

245,000

$

5,067,000

$

4,401,000

$

2,139,000

$

2,850,000

Research and development expenses

Research and development expenses decreasedincreased by $4.4$0.2 million, or 54%10%, to $3.8$2.0 million for the sixthree months ended June 30, 20212022 from $8.2$1.8 million for the sixthree months ended June 30, 2020.2021. This decreaseincrease was caused primarily by $3.6$0.3 million lower clinical developmenthigher salary costs and consulting expenses on the INSPIRE programbonus accruals in the 20212022 period and also by $0.8$0.2 million lower personnel andhigher stock compensation expense during the 2021 period, following reductions2022 period. These increases were partially offset by a $0.3 million decrease in our workforce completed inmanufacturing costs related to the thirdtiming of narazaciclib drug substance and fourth quarter of 2020.drug product manufacturing.

The details of our research and development expenses are:

Six Months Ended June 30, 

Three Months Ended June 30, 

    

2021

    

2020

    

    

2022

    

2021

Preclinical & clinical development

$

1,160,000

$

3,911,000

$

574,000

$

584,000

Personnel related

 

1,117,000

 

1,849,000

 

645,000

 

296,000

Manufacturing, formulation & development

 

708,000

 

799,000

 

272,000

 

553,000

Stock based compensation

 

16,000

 

94,000

 

169,000

 

17,000

Consulting fees

 

788,000

 

1,518,000

 

378,000

 

402,000

$

3,789,000

$

8,171,000

$

2,038,000

$

1,852,000

Change in fair value of warrant liability

The fair value of the warrant liability increased $0.2was reduced to $0 during the third quarter of 2021, following the expiration of the underlying tradable warrants. The change in the fair value of the warrant liability was $0.4 million during the three months ended June 30, 2021 based on the change in market value of the tradable warrants in the 2021 period.

Other income, net

Other income, net, was income of $96,000 and expense of $13,000 for the three months ended June 30, 2022 and 2021, respectively. The change of $109,000 was due to $32,000 higher foreign currency exchange gain and $77,000 higher interest income in the 2022 period.

28

Table of Contents

Comparison of the Six Months Ended June 30, 2022 and 2021

Six Months Ended June 30, 

    

2022

    

2021

    

Change

Revenue

$

113,000

$

113,000

$

Operating expenses:

 

  

 

  

 

  

General and administrative

 

4,325,000

 

5,067,000

 

742,000

Research and development

 

4,040,000

 

3,789,000

 

(251,000)

Total operating expenses

 

8,365,000

 

8,856,000

 

491,000

 

Loss from operations

 

(8,252,000)

 

(8,743,000)

 

491,000

 

Change in fair value of warrant liability

 

 

(209,000)

 

209,000

 

Other income, net

 

106,000

 

6,000

 

100,000

 

Net loss

$

(8,146,000)

$

(8,946,000)

$

800,000

Revenues

Revenues for 2022 were consistent with 2021, and were due to the recognition of deferred revenue from our collaboration with SymBio.

General and administrative expenses

General and administrative expenses decreased $0.8 million, or 15% to $4.3 million for the six months ended June 30, 2021, compared to an increased of $0.12022, from $5.1 million for the six months ended June 30, 2020.2021. This changedecrease was caused a $1.3 million decrease in investor relations, proxy solicitation, and fees related to our special meeting by proxy in 2021 which did not occur in 2022. This decrease was partially offset by a larger$0.4 million increase in payroll expenses and bonus accruals in the 20212022 period ofand $0.1 million higher stock compensation expense in the fair market value of the warrants issued in our rights offering in 2016.2022 period.

The details of our general and administrative expenses are:

Six Months Ended June 30, 

    

2022

    

2021

Professional & consulting fees

$

893,000

$

941,000

Stock based compensation

259,000

99,000

Personnel related

 

1,916,000

 

1,573,000

Public company costs

 

563,000

 

1,876,000

Insurance & other

694,000

578,000

$

4,325,000

$

5,067,000

Other income (expense), netResearch and development expenses

Other income (expense)Research and development expenses increased by $0.2 million, or 7%, net, was income of $6,000to $4.0 million for the six months ended June 30, 2022 from $3.8 million for the three months ended June 30, 2021. This increase was caused primarily by $0.3 million higher manufacturing costs related to narazaciclib drug substance and drug product manufacturing and $0.3 million higher stock based compensation expense in the 2022 period. These increases were partially offset by a $0.4 million decrease in clinical development and consulting expenses on the INSPIRE program in the 2022 period.

29

Table of Contents

The details of our research and development expenses are:

Six Months Ended June 30, 

    

2022

    

2021

    

Preclinical & clinical development

$

811,000

$

1,150,000

Personnel related

 

1,228,000

 

1,117,000

Manufacturing, formulation & development

 

1,020,000

 

708,000

Stock based compensation

 

278,000

 

26,000

Consulting fees

 

703,000

 

788,000

$

4,040,000

$

3,789,000

Change in fair value of warrant liability

The fair value of the warrant liability was reduced to $0 during the third quarter of 2021, following the expiration of the underlying tradable warrants. The change in the fair value of the warrant liability was an increase of $0.2 million during the six months ended June 30, 2021 based on the change in market value of the tradable warrants in the 2021 period.

Other income, net

Other income, net, was $106,000 and $96,000$6,000 for the six months ended June 30, 2020.2022 and 2021, respectively. The change of $90,000$100,000 was due to $103,000 lower net$20,000 higher foreign currency exchange gain and $80,000 higher interest income in the 2021 period due to lower average cash balances, partially offset by $13,000 lower foreign currency exchange losses and other items in the 20212022 period.

Liquidity and Capital Resources

Since our inception, we have incurred net losses and experienced negative cash flows from our operations. We incurred net losses of $8.9$8.1 million and $12.5$8.9 million for the six months ended June 30, 20212022 and 2020,2021, respectively. Our operating activities used $11.0$8.5 million and $11.8$11.0 million of net cash during the six months ended June 30, 20212022 and 2020,2021, respectively. At June 30, 2021,2022, we had an accumulated deficit of $437.5$452.9 million, working capital of $36.7$41.6 million, and cash and cash equivalents of $43.7$46.5 million. We believe that our cash and cash equivalents as of June 30, 2021,2022, will be sufficient to fund our operations and ongoing trials for more than eighteenat least 18 months from the date of this filing.

30

Table of Contents

Cash Flows

The following table summarizes our cash flows for the six months ended June 30, 20212022 and 2020:2021:

Six Months Ended June 30, 

Six Months Ended June 30, 

    

2021

    

2020

    

2022

    

2021

Net cash (used in) provided by:

 

  

 

  

 

  

 

  

Operating activities

$

(10,955,000)

$

(11,844,000)

$

(8,510,000)

$

(10,955,000)

Investing activities

 

 

(15,000)

 

 

Financing activities

 

35,651,000

 

16,360,000

 

 

35,651,000

Effect of foreign currency translation

 

(12,000)

 

1,000

 

(27,000)

 

(12,000)

Net increase in cash and cash equivalents

$

24,684,000

$

4,502,000

Net increase (decrease) in cash and cash equivalents

$

(8,537,000)

$

24,684,000

Net cash used in operating activities

Net cash used in operating activities was $8.5 million for the six months ended June 30, 2022 and consisted primarily of a net loss of $8.2 million, including $0.5 million of noncash stock-based compensation expense. Changes in operating assets and liabilities resulted in a net decrease in cash of $0.9 million. Significant changes in operating assets and liabilities included an increase in prepaid expenses and other current assets of $1.2 million, an increase in accounts payable of $0.3 million and decrease in accrued liabilities of $0.1 million due to timing of invoices and payments to our

30

Table of Contents

vendors, and a decrease in deferred revenue of $0.1 million due to recognition of the unamortized portion of the upfront payment under our collaboration agreement with SymBio.

Net cash used in operating activities was $11.0 million for the six months ended June 30, 2021 and consisted primarily of a net loss of $8.9 million, including an increase in the fair value of warrant liability of $0.2 million, and $0.1 million of both noncash stock-based compensation and depreciation expense. Changes in operating assets and liabilities resulted in a net decrease in cash of $2.3 million. Significant changes in operating assets and liabilities included a decrease in prepaid expenses and other current assets of $0.3 million and a decrease in accounts payable and accrued liabilities of $2.5 million due to timing of invoices and payments to our vendors, and a decrease in deferred revenue of $0.1 million due to recognition of the unamortized portion of the upfront payment under our collaboration agreement with SymBio.

Net cash used in operating activities was $11.8 million for the six months ended June 30, 2020 and consisted primarily of a net loss of $12.5 million, including an increase in the fair value of warrant liability of $0.1, and $0.2 million of both noncash stock-based compensation and depreciation expense. Changes in operating assets and liabilities resulted in a net increase in cash of $0.3 million. Significant changes in operating assets and liabilities included an increase in accounts payable and accrued liabilities of $0.4 million due to timing of invoices and payments to our vendors, and a decrease in deferred revenue of $0.1 million due to recognition of the unamortized portion of the upfront payment under our collaboration agreement with SymBio.

Net cash used in investingprovided by financing activities

There waswere no cash used in investingflows from financing activities during the six months ended June 30, 2021. Net cash used in investing activities was $15,000 related to computer equipment during the six months ended June 30, 2020.

Net cash provided by financing activities

2022. Net cash provided by financing activities was $35.7 million and $16.4 million for the six months ended June 30, 2021 and 2020, respectively.  The net cash provided by financing activities in the 2021 period resultedresulting from proceeds received from the sales of common stock and the exercise of warrants. The net cash provided by financing activities in the 2020 period of $16.4 million resulted from the sale of common stock and the exercise of warrants.

Operating and Capital ExpenditureMaterial Cash Requirements

We believe that our cash and cash equivalents of $43.7$46.5 million at June 30, 2021,2022, will be sufficient to fund our operations and ongoing trials for more than eighteenat least 18 months from the date of this filing. The consolidated financial statements do not include any adjustments relating to recoverability and classification of recorded asset amounts or the amounts and classification of liabilities that might be necessary should we be unable to continue in existence.

We have not achieved profitability since our inception and we expect to continue to incur net losses for the foreseeable future. We expect our net cash expendituresexpended in 20212022 to be lowerslightly higher than they were in 2020, due primarily2021. We expect clinical trial costs to the INSPIRE study being completed in 2020, reductions inincrease as we focus on our workforce during 2020, and having an earlier clinical stage

31

Table compound, narazaciclib, and increased headcount in our clinical and regulatory groups. We would also expect an increase in costs for any completed potential in-licensing, the timing of Contents

which would be determined by the timing of any potential in-licensing. We enter into contracts in the normal course of business with third-party contract organizations for clinical trials, preclinical studies, manufacturing and other services and products for operating purposes. These contracts generally provide for termination following a certain period after notice and therefore less expensive to develop, pipeline in 2021. The nature, design, size, and cost of further studies will depend in large part on the outcome of ongoing studies, discussions with regulators, and the potential in-license of any additional compounds or product candidates.

we believe that, currently, our non-cancelable obligations under these agreements are not material.

For additional risks, please see “Risk Factors” in Part II of this report and in previously disclosed in our most recent annual report on Form 10-K.

Item 3. Quantitative and Qualitative Disclosures About Market Risk

As a smaller reporting company, the Company is not required to provide the information otherwise required by this Item.

Item 4. Controls and Procedures

Managements’ Evaluation of our Disclosure Controls and Procedures

Our management, with the participation of our principal executive and principal financial officers, evaluated the effectiveness of our disclosure controls and procedures as of June 30, 2021.2022. The term “disclosure controls and procedures,” as defined in Rules 13a-15(e) and 15d-15(e) promulgated under the Securities Exchange Act of 1934, as amended (the “Exchange Act”), means controls and other procedures of a company that are designed to ensure that information required to be disclosed by a company in the reports that it files or submits under the Exchange Act is recorded, processed, summarized and reported, within the time periods specified in the SEC’s rules and forms. Disclosure controls and procedures include, without limitation, controls and procedures designed to ensure that information required to be disclosed by a company in the reports that it files or submits under the Exchange Act is

31

Table of Contents

accumulated and communicated to the company’s management, including its principal executive and principal financial officers, as appropriate to allow timely decisions regarding required disclosure. Based on the evaluation of our disclosure controls and procedures as of June 30, 2021,2022, our principal executive and principal financial officers concluded that, as of such date, our disclosure controls and procedures were effective.

Changes in Internal Control Over Financial Reporting

Our management, with the participation of our principal executive and principal financial officers, evaluated any changes in our internal control over financial reporting (as such term is defined in Rules 13a-15(f) and 15d-15(f) under the Exchange Act) that occurred during our most recently completed fiscal quarter. Based on that evaluation, our principal executive and principal financial officers concluded that no change in our internal control over financial reporting (as defined in Rules 13a-15(f) and 15d-15(f) under the Exchange Act) occurred during the fiscal quarter ended June 30, 20212022 that has materially affected, or is reasonably likely to materially affect, our internal control over financial reporting.

PART II — OTHER INFORMATION

Item 1. Legal Proceedings

We are not party to any pending material legal proceedings and are not aware of any such proceedings contemplated by governmental authorities.

Item 1A. Risk Factors

The followingIn addition to the information contained in this report, you should carefully consider the risk factor should be readfactors discussed in conjunction with the “RiskPart I, “Item 1A. Risk Factors” previously disclosed in our annual reportAnnual Report on Form 10-K filed with the SEC on March 18, 2021 and21, 2022 which could materially affect our Quarterlybusiness, financial condition or future results. The risks described in our Annual Report on Form 10-Q for10-K are not the quarter ended March 31, 2021.

32

Table of Contents

The COVID-19 pandemic, including new variantsonly risks we face. Additional risks and uncertainties not currently known to us or that we currently deem to be immaterial also may be more contagious, couldmaterially adversely impactaffect our business, including our clinical trials, drug manufacturing and nonclinical activities.

As the COVID-19 pandemic continues to spread around the globe, we may experience disruptions that could severely impact our business, clinical trials, drug manufacturing and nonclinical activities. These potential disruptions may include but are not limited to delays or difficulties in clinical site initiation and patient recruitment, patient withdrawals, postponement of planned clinical or preclinical studies, redirection of site resources from studies, study modification, suspension, or termination, the introduction of remote study procedures and modified informed consent procedures, study site changes, direct delivery of investigational products to patient homes requiring state licensing, study deviations or noncompliance, diversion of healthcare resources away from the conduct of clinical trials, including the diversion of hospitals serving as our clinical trial sites and hospital staff supporting the conduct of our clinical trials, delays in receiving approval from local regulatory authorities to initiate our planned clinical trials, and changes or delays in site monitoring. The foregoing may require that we consult with relevant review and ethics committees, IRBs, and the FDA. The foregoing may also impact the integrity of our study data. The effects of the COVID-19 pandemic may also increase the need for clinical trial patient monitoring and regulatory reporting of adverse effects.

The COVID-19 pandemic may also impact our ability to obtain supplies of our product candidates or other materials that may be necessary for the conduct of our development program. If any of our suppliers are adversely impacted by the COVID-19 pandemic or the restrictions resulting from the outbreak, if they cannot obtain the necessary supplies, or if such third parties need to prioritize other products or customers over us, including under the Defense Production Act, we may experience delays or disruptions in our supply chain, which could have a material and adverse impact on our business. Third party manufacturers may also need to implement measures and changes, or deviate from typical requirements because of the COVID-19 pandemic that may otherwise adversely impact our supply chains or the quality of the resulting products or supplies. Depending on the change, we may need to obtain FDA pre-approval or otherwise provide FDA with a notification of the change.

The pandemic could further impact our ability to interact with the FDA or other regulatory authorities and obtain any necessary inspections. Due to the potential impact of the COVID -19 outbreak on clinical trials, drug development, and manufacturing, FDA issued a number of guidance documents concerning how sponsors and investigators may address these challenges. FDA has also issued guidance on the development of products to treat COVID-19. FDA’s guidance is continually evolving.

The COVID-19 pandemic may also result in changes in laws and regulations. By example, in March 2020, the U.S. Congress passed the Coronavirus Aid, Relief, and Economic Security Act, or CARES Act, which includes various provisions regarding FDA drug shortage reporting requirements, as well as provisions regarding supply chain security, such as risk management plan requirements, and the promotion of supply chain redundancy and domestic manufacturing. This and any future changes in law may require that we change our internal processes and procedures to ensure continued compliance.

The COVID-19 pandemic continues to rapidly evolve. The extent to which the COVID-19 may impact our business, including our drug manufacturing, nonclinical activities, clinical trials and financial condition will depend on future developments, which are highly uncertain and cannot be predicted with confidence, such as the ultimate geographic spread of the disease, the duration of the pandemic, travel restrictions and social distancing in the United States and other countries, business closures and/or business disruptions and the effectiveness of actions taken in the United States and other countries to contain and treat the disease.

operating results.

Item 2. Unregistered Sales of Equity Securities and Use of Proceeds

Not applicable.

Item 3. Defaults Upon Senior Securities

Not applicable.

33

Table of Contents

Item 4. Mine Safety Disclosures

Not applicable.

Item 5. Other Information

Not applicable.

32

Table of Contents

Item 6. Exhibits

EXHIBIT INDEX

Exhibit

Number

    

Description

3.110.1

Certificate ofEmployment Agreement Amendment to the Tenth Amended and Restated Certificate of Incorporation ofbetween Onconova Therapeutics, Inc., as amended and Mark Guerin dated June 10, 2022 (Incorporated by reference to Exhibit 3.110.1 to the Company’s Current Report on Form 8-K filed on May 20, 2021).

3.2

Certificate of Amendment to the Tenth Amended and Restated Certificate of Incorporation of Onconova Therapeutics, Inc., as amended (Incorporated by reference to Exhibit 3.2 to the Company’s Current Report on Form 8-K filed on May 20, 2021).

10.1

Employment Agreement, dated June 14, 2021, by and between Onconova Therapeutics, Inc. and Mark Stephen Gelder,M.D.

10.2

Employment Agreement, dated March 9, 2021, by and between Onconova Therapeutics, Inc. and Abraham N. Oler13, 2022).

31.1

 

Rule 13a-14(a)/15d-14(a) Certifications of Principal Executive Officer

31.2

 

Rule 13a-14(a)/15d-14(a) Certifications of Principal Financial Officer

32.1

 

Section 1350 Certifications of Principal Executive Officer

32.2

 

Section 1350 Certifications of Principal Financial Officer

 

 

 

101.INS

 

XBRL Instance – The instance document does not appear in the Interactive Data File because its XBRL tags are embedded within the Inline XBRL 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 Labels Linkbase Document

101.PRE

 

XBRL Taxonomy Extension Presentation Linkbase Document

104

Cover Page Interactive Data File (formatted as Inline XBRL and contained in Exhibit 101)

3433

Table of Contents

SIGNATURES

Pursuant to the requirements 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.

 

ONCONOVA THERAPEUTICS, INC.

 

 

Dated: August 16, 202115, 2022

 

 

 

 

/s/ STEVEN M. FRUCMTMAN, M. D.

 

Steven M. Fruchtman, M.D.

 

President and Chief Executive Officer

 

(Principal Executive and Principal Operating Officer)

 

 

Dated: August 16, 202115, 2022

 

 

 

 

/s/ MARK GUERIN

 

Mark Guerin

 

Chief Operating Officer and Chief Financial Officer

 

(Principal Financial Officer)

3534