UNITED STATES
SECURITIES AND EXCHANGE COMMISSION
Washington, D.C. 20549

FORM 10-K
xANNUAL REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934
For the fiscal year ended December 31, 20142017
OR
¨
TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d)
OF THE SECURITIES EXCHANGE ACT OF 1934
For the transition period from __________           to             __________                    
Commission File Number:Number 001-32335
Halozyme Therapeutics, Inc.___________________________
HALOZYME THERAPEUTICS, INC.
(Exact name of registrant as specified in its charter)
___________________________
Delaware 88-0488686
(State or other jurisdiction of
incorporation or organization)
 
(I.R.S. Employer
Identification No.)
11388 Sorrento Valley Road,
San Diego, California
CA
 
92121
(Zip Code)
(Address of principal executive offices) (Zip Code)
(858) 794-8889
(Registrant’s Telephone Number, Including Area Code)telephone number, including area code)
Securities registered under Section 12(b) of the Act:
Title of Each Class Name of Each Exchange on Which Registered
Common Stock, $0.001 Par Value The NASDAQ Stock Market, LLC
Securities registered under Section 12(g) of the Act:
None
Indicate by check mark if the registrant is a well-known seasoned issuer, as defined in Rule 405 of the Securities Act.    x  Yes        ¨  No
Indicate by check mark if the registrant is not required to file reports pursuant to Section 13 or 15(d) of the Act.    ¨  Yes        x  No
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.    x  Yes        ¨  No
Indicate by check mark whether the registrant has submitted electronically and posted on its corporate Web site, if any, every Interactive Data File required to be submitted and posted 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 and post such files).    x  Yes        ¨  No
Indicate by check mark if disclosure of delinquent filers pursuant to Item 405 of Regulation S-K is not contained herein, and will not be contained, to the best of registrant’s knowledge, in definitive proxy or information statements incorporated by reference in Part III of this Form 10-K or any amendment to this Form 10-K. ox




Indicate by check mark whether the registrant is a large accelerated filer, an accelerated filer, a non-accelerated filer, or a smaller reporting company or emerging growth company. See the definitions of “large accelerated filer,” “accelerated filer” and, “smaller reporting companycompany”and “emerging growth company” in Rule 12b-2 of the Exchange Act. (Check one):
Large accelerated filer
x
Accelerated filer
o¨
  
Non-accelerated filer
o¨
Smaller reporting company
¨
Emerging growth company
¨
 
Smaller reporting company o
(Do not check if a smaller reporting 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 Exchange Act).    ¨  Yes        x    No
The aggregate market value of the voting and non-voting common equity held by non-affiliates of the registrant as of June 30, 20142017 was approximately $1.0$1.3 billion based on the closing price on the NASDAQ Global Select Market reported for such date. Shares of common stock held by each officer and director and by each person who is known to own 10% or more of the outstanding common stock have been excluded in that such persons may be deemed to be affiliates of the registrant. This determination of affiliate status is not necessarily a conclusive determination for other purposes.
AsThe number of February 24, 2015, there were 126,704,135outstanding shares of the registrant’s common stock, issued, $0.001 par value $0.001 per share, and outstanding.

was 143,076,590 as of February 13, 2018.
DOCUMENTS INCORPORATED BY REFERENCE
Portions of the registrant'sregistrant’s definitive Proxy Statement to be filed subsequent to the date hereof with the Securities and Exchange Commission pursuant to Regulation 14A in connection with the registrant’s 20152018 Annual Meeting of Stockholders are incorporated by reference into Part III of this Annual Report.
 




Table of ContentsHALOZYME THERAPEUTICS, INC.
INDEX
 
   
Page
Item 1.
Item 1A.
Item 1B.
Item 2.
Item 3.
Item 4.
 
Item 5.
Item 6.
Item 7.
Item 7A.
Item 8.
Item 9.
Item 9A.
Item 9B.
 
Item 10.
Item 11.
Item 12.
Item 13.
Item 14.
 
Item 15.
SIGNATURESItem 16.






This Annual Report on Form 10-K contains “forward-looking statements” within the meaning of the “safe harbor” provisions of Section 21E of the Securities Exchange Act of 1934, as amended, and Section 27A of the Securities Act of 1933, as amended. All statements, other than statements of historical fact, included herein, including without limitation those regarding our future product development and regulatory events and goals, product collaborations, our business intentions and financial estimates and anticipated results, are forward-looking statements. Words such as “expect,” “anticipate,” “intend,” “plan,” “believe,” “seek,” “estimate,” “think,” “may,” “could,” “will,” “would,” “should,” “continue,” “potential,” “likely,” “opportunity” and similar expressions or variations of such words are intended to identify forward-looking statements, but are not the exclusive means of identifying forward-looking statements in this Annual Report. Additionally, statements concerning future matters such as the development or regulatory approval of new products, enhancements of existing products or technologies, third party performance under key collaboration agreements, revenue and expense levels and other statements regarding matters that are not historical are forward-looking statements.
Although forward-looking statements in this Annual Report reflect the good faith judgment of our management, such statements can only be based on facts and factors currently known by us. Consequently, forward-looking statements are inherently subject to risks and uncertainties and actual results and outcomes may differ materially from the results and outcomes discussed in or anticipated by the forward-looking statements. Factors that could cause or contribute to such differences in results and outcomes include without limitation those discussed under the heading “Risk Factors” in Part I, Item 1A below, as well as those discussed elsewhere in this Annual Report. Readers are urged not to place undue reliance on these forward-looking statements, which speak only as of the date of this Annual Report. We undertake no obligation to revise or update any forward-looking statements in order to reflect any event or circumstance that may arise after the date of this Annual Report. Readers are urged to carefully review and consider the various disclosures made in this Annual Report, which attempt to advise interested parties of the risks and factors that may affect our business, financial condition, results of operations and prospects.
References to “Halozyme,” “the Company,” “we,” “us,” and “our” refer to Halozyme Therapeutics, Inc. and its wholly owned subsidiary, Halozyme, Inc., and Halozyme, Inc.'s’s wholly owned subsidiary,subsidiaries, Halozyme Holdings Ltd., Halozyme Royalty LLC, Halozyme Switzerland GmbH and Halozyme Switzerland Holdings GmbH. References to “Notes” refer to the Notes to Consolidated Financial Statements included herein (refer to Part II, Item 8).
PART I
Item 1.
Business
Overview
Halozyme Therapeutics, Inc. is a biotechnology company focused on developing and commercializing novel oncology therapies. We are seeking to translate our unique knowledge of the tumor microenvironment to create novel therapies that canhave the potential to improve cancer patient survival. Our research primarily focuses on human enzymes that alter the extracellular matrix and tumor microenvironment. The extracellular matrix is a complex matrix of proteins and carbohydrates surrounding the cell that provides structural support in tissues and orchestrates many important biological activities, including cell migration, signaling and survival. Over many years, we have developed unique technology and scientific expertise enabling us to pursue this target-rich environment for the development of therapies.
Our proprietary enzymes can beare used to facilitate the delivery of injected drugs and fluids, potentially enhancing the efficacy and the convenience of other drugs or can be used to alter abnormal tissue structures for potential clinical benefit. We have chosen to exploit our technology and expertise inusing a balanced waytwo pillar strategy that we believe enables us to modulate bothmanage risk and spendcost by: (1) developing our own proprietary products in therapeutic areas with significant unmet medical needs, with a focus on oncology, and (2) licensing our technology to biopharmaceutical companies to collaboratively develop products whichthat combine our technology with the collaborators'collaborators’ proprietary compounds.


The majority of our approved product and product candidates are based on rHuPH20, our patented recombinant human hyaluronidase enzyme. rHuPH20 is the active ingredient in our first commercially approved product, Hylenex® recombinant, and it works by temporarily breaksbreaking down hyaluronic acid (HA)hyaluronan (or HA), a naturally occurring substancecomplex carbohydrate that is a major component of the extracellular matrix in tissues throughout the body such as skin and cartilage. We believe this temporary degradation creates an opportunistic window for the improved subcutaneous delivery of injectable biologics, such as monoclonal antibodies and other large therapeutic molecules, as well as small molecules and fluids. We refer to the application of rHuPH20 to facilitate the delivery of other drugs or fluids as Enhanzeour ENHANZE® technology. rHuPH20 is alsoTechnology. We license the active ingredientENHANZE Technology to form collaborations with biopharmaceutical companies that develop or market drugs requiring or benefiting from injection via the subcutaneous route of administration.
We currently have ENHANZE collaborations with F. Hoffmann-La Roche, Ltd. and Hoffmann-La Roche, Inc. (Roche), Baxalta US Inc. and Baxalta GmbH (Baxalta Incorporated was acquired by Shire plc in June 2016) (Baxalta), Pfizer Inc. (Pfizer), Janssen Biotech, Inc. (Janssen), AbbVie, Inc. (AbbVie), Eli Lilly and Company (Lilly), Bristol-Myers Squibb Company (BMS) and Alexion Pharma Holding (Alexion). We receive royalties from two of these collaborations, including royalties from sales of one product from the Baxalta collaboration and two products from the Roche collaboration. Future potential revenues from the sales and/or royalties of our first commercially approved products, product Hylenex® recombinant.


candidates, and ENHANZE collaborations will depend on the ability of Halozyme and our collaborators to develop, manufacture, secure and maintain regulatory approvals for approved products and product candidates and commercialize product candidates.
1



Our proprietary development pipeline consists primarily of pre-clinical and clinical stage product candidates in oncology and diabetes.oncology. Our lead oncology program is PEGPH20 (PEGylatedpegvorhyaluronidase alfa (PEGPH20, PEGylated recombinant human hyaluronidase), a new molecular entity under developmentwe are developing in combination with currently approved cancer therapies as a candidate for the systemic treatment of tumors that accumulate HA. WhenWe have demonstrated that when HA accumulates in a tumor, it can cause higher pressure in the tumor, reducing blood flow into the tumor and with that, reduced access of cancer therapies to the tumor. PEGPH20 workshas been demonstrated in animal models to work by temporarily degrading HA surrounding some cancer cells and resultsresulting in reduced pressure and increased blood flow to the cancer withtumor thereby enabling increased amounts of anticancer treatments administered concomitantly gaining access to the tumor. WeThrough our efforts and efforts of our partners and collaborators, we are currently in Phase 3 clinical testing for PEGPH20 with ABRAXANE® (nab-paclitaxel) and gemcitabine in stage IV pancreatic ductal adenocarcinoma (“PDA”) (HALO 109-301), in Phase 1b clinical testing for PEGPH20 with KEYTRUDA® (pembrolizumab) in non-small cell lung cancer and gastric cancer (HALO 107-101), in Phase 1b/2 clinical testing for PEGPH20 with HALAVEN® (eribulin) in patients treated with up to two lines of prior therapy for HER2-negative metastatic pancreaticbreast cancer, (Study 109-202), and we have recently initiated ain Phase 1b/2 clinical trial in non-small cell lung cancer (Study 107-201). We have also been investigating Hylenex recombinanttesting for use as pre-treatmentPEGPH20 with Tecentriq® (atezolizumab) in patients with Type 1 diabetes using pumps.
Our recent receipt of Fast Track and Orphan Drug designationspreviously treated metastatic PDA, in Phase 1b/2 clinical testing for PEGPH20 new pre-clinical data further supporting the pan-tumor potentialwith Tecentriq in patients with gastric cancer and in Phase 1b/2 clinical testing for PEGPH20 with Tecentriq in patients with cholangiocarcinoma and investigator interest in both pancreatic and lunggall bladder cancer trials have confirmed PEGPH20 as our priority product candidate for investment. As a result of ongoing evaluations to confirm and focus on the highest value opportunities, we have made the decision to seek collaborations with third parties or explore other strategic alternatives in order to exploit our diabetes and dermatology programs.
Regarding Enhanze, we currently have collaborations with F. Hoffmann-La Roche, Ltd. and Hoffmann-La Roche, Inc. (Roche), Baxter Healthcare Corporation (Baxter), Pfizer Inc. (Pfizer) and Janssen Biotech, Inc. (Janssen), with one product approved in the U.S. and three products approved for marketing in Europe from which we are receiving royalties and several others at various stages of development.
Future revenues from the sales and/or royalties of our product candidates which have not been approved or have recently been approved will depend on the ability of Halozyme and our collaborators to develop, manufacture, secure regulatory approvals for and commercialize the product candidates. We have incurred net operating losses each year since inception, with an accumulated deficit of approximately $450.4 million as of December 31, 2014(HALO 110-101/MATRIX).
Our principal offices and research facilities are located at 11388 Sorrento Valley Road, San Diego, California 92121. Our telephone number is (858) 794-8889 and our e-mail address is info@halozyme.com. Our website address is www.halozyme.com. Information found on, or accessible through, our website is not a part of, and is not incorporated into, this Annual Report on Form 10-K. Our periodic and current reports that we filed with the SEC are available on our website at www.halozyme.com, free of charge, as soon as reasonably practicable after we have electronically filed such material with, or furnished them to, the SEC, including our annual reports on Form 10-K, quarterly reports on Form 10-Q, current reports on Form 8-K and any amendments to those reports. Further copies of these reports are located at the SEC'sSEC’s Public Reference Room at 100 F Street, N.W., Washington, D.C. 20549, and online at http://www.sec.gov.
Technology
The majority of our approved product and product candidates are based on rHuPH20, a patented recombinant human hyaluronidase enzyme. rHuPH20 temporarily breaks down HA - a naturally occurring substance that is a major component of the extracellular matrix in tissues throughout the body such as skin and cartilage. We believe this temporary degradation creates an opportunistic window for the improved subcutaneous delivery of injectable biologics, such as monoclonal antibodies and other large therapeutic molecules, as well as small molecules and fluids. The HA reconstitutes its normal density within several days and, therefore, we anticipate that any effect of rHuPH20 on the architecture of the subcutaneous space is temporary. rHuPH20 can thus be applied as a drug delivery platform to increase dispersion and absorption of other injected drugs and fluids that are injected under the skin or in the muscle thereby potentially enhancing efficacy or convenience. For example, rHuPH20 can behas been used to convert drugs that must be delivered intravenously into subcutaneous injections or to reduce the number of subcutaneous injections needed for effective therapy. We refer toWhen ENHANZE Technology is applied subcutaneously, the applicationrHuPH20 acts locally and has a tissue half-life of less than 15 minutes. HA at the local site reconstitutes its normal density within a few days and, therefore, we anticipate that any effect of rHuPH20 to facilitateon the deliveryarchitecture of other drugs or fluids as Enhanze technology. rHuPH20the subcutaneous space is also the active ingredient in our first commercially approved product, Hylenex recombinant.temporary.


Additionally, we are expanding our scientific work to develop other enzymes and agents that target the extracellular matrix'smatrix’s unique aspects, giving rise to potentially new molecular entities with a particular focus on oncology. For example, weWe are developing a PEGylated version of the rHuPH20 enzyme (PEGPH20), that lasts for an extended period in the bloodstream (half-life of one to two days), and may therefore better target solid tumors that accumulate HA by degrading the surrounding HA and reducing the interstitial fluid pressure within malignant tumors to allow better penetration by co-administered agents.


2



Strategy
During the fourth quarter of 2014,2017, we affirmedcontinued our strategy of focusing on developing our PEGPH20 product candidate for oncology as well as entering intopipeline and expanding our collaborations for EnhanzeENHANZE Technology. This business model allows for growth in which revenue garnered from collaboration products helpsto help fund our investment in PEGPH20 clinical development, with the goal of a future product approval that will support sustained growth.
Key aspects of our corporate strategy include the following:
Focus on our oncology pipeline. We are currently developing PEGPH20, our investigational new drug candidate, in multiple different tumors that accumulate high levels of HA. PEGPH20 is in phase 2Phase 3 development in metastatic pancreatic cancerstage IV PDA and in phase 1 development in Non-Small Cell Lung Cancer.multiple Phase 1b/2 studies for various tumor types. Over time, it is our goal to study additional types of cancer and to advance this program toward regulatory approval and commercial launch. In addition, we have a novel oncology preclinical asset.
Focus on Enhanze collaborations.our ENHANZE platform. We currently have foureight collaborations with three current product approvals and additional product candidates being evaluated forin development. We intend to work with our existing collaborators to expand our collaborations to add new targets and develop targets and product candidates under the terms of the operative agreements. In addition, we will continue our efforts to enter into new collaborations to further exploitderive additional value from our technology and derive value therefrom.proprietary technology.


Product and Product Candidates
We have one marketed proprietary product, and multiplethree partnered products, one proprietary product candidatescandidate targeting several indications in various stages of development.development, and one preclinical product candidate. The following table summarizes our proprietary product and product candidatescandidate as well as products and product candidates under development with our collaborators:


3




Proprietary Pipeline
Hylenex Recombinant (hyaluronidase human injection)
Hylenex recombinant is a formulation of rHuPH20 that has received FDAU.S. Food and Drug Administration (FDA) approval to facilitate subcutaneous fluid administration for achieving hydration, to increase the dispersion and absorption of other injected drugs and, in subcutaneous urography, to improve resorption of radiopaque agents. Hylenex recombinant is currently the number one prescribed branded hyaluronidase.
PEGPH20
We are developing PEGPH20 in combination with currently approved cancer therapies as a candidate for the systemic treatment of tumors that accumulate HA in combination with currently approved cancer therapies. 'PEG'HA. ‘PEG’ refers to the attachment of polyethylene glycol to rHuPH20, thereby creating PEGPH20. One of the novel properties of PEGPH20 is that it lasts for an extended duration in the bloodstream (one to two days) and, therefore, can be usedadministered systemically to maintain its therapeutic effect to treat systemic disease.
Cancer malignancies, including pancreatic, lung, breast, colongastric, and prostatebiliary tract cancers oftencan accumulate high levels of HA and therefore we believe that PEGPH20 has the potential to help patients with these types of cancer when used with certain currently approved cancer therapies. Among solid tumors, pancreatic ductal adenocarcinomaPDA has been reported to be associated with the highest frequency of HA accumulation.
Over 100,000 patients There are approximately 65,000 annual diagnoses of PDA in the U.S.United States and EU are diagnosed with pancreatic cancer annuallythe European Union, and are frequently diagnosed in late stagewe estimate that 35-40% have high levels of the disease. HA.


The pathologic accumulation of HA, along with other matrix components, creates a unique microenvironment for the growth of tumor cells compared to normal cells. We believe that depletingdegrading the HA component of the tumor microenvironment with PEGPH20 remodels the tumor microenvironment, resulting in tumor growth inhibition.inhibition in animal models. Removal of HA from the tumor microenvironment results in expansion of previously constricted blood vessels to allowallowing increased blood flow, potentially increasing the access of activated immune cells and factors in the blood into the tumor micro environment.microenvironment. If PEGPH20 is administered in conjunction with other anti-cancer therapies, the increase in blood flow may allow anti-cancer therapies to have greater access to the tumor, which may enhance the treatment effect of therapeutic modalities like chemotherapies, monoclonal antibodies and other agents.
Study Halo 109-201:We are developing PEGPH20 as a targeted therapy, for patients who have tumors with high levels of HA. We have a collaboration with Ventana Medical Systems Inc. (Ventana), a member of the Roche Group, to develop, and for Ventana to ultimately commercialize, a companion diagnostic assay for use with PEGPH20. The companion diagnostic assay is being used to identify high levels of HA in tumor biopsies, and may be the first diagnostic to target tumor-associated HA and possibly the first companion diagnostic assay in pancreatic cancer.
Pancreatic cancer indications:
HALO 109-201:
In January 2015, we presented the final results from StudyHALO 109-201, a multi-center, international open label dose escalation Phase 1b clinical study of PEGPH20 in combination with gemcitabine for the treatment of patients with stage IV metastatic pancreatic cancerPDA at the 2015 Gastrointestinal Cancers Symposium (also known as ASCO-GI meeting). This study enrolled 28 patients with previously untreated stage IV pancreatic ductal adenocarcinoma.PDA. Patients were treated with one of three doses of PEGPH20 (1.0, 1.6 and 3.0 µg/kg twice weekly for four weeks, then weekly thereafter) in combination with gemcitabine 1000 mg/m2 administered intravenously. In this study, the confirmed overall response rate (complete response + partial response confirmed on a second scan as assessed by an independent radiology review) was 29 percent (7 of 24 patients) for those treated at therapeutic dose levels of PEGPH20 (1.6 and 3.0 µg/kg). Median progression freeprogression-free survival (PFS) was 154 days (95% CI, 50-166) in the efficacy-evaluable population (n = 24). Among efficacy-evaluable patients with baseline tumor HA staining (n = 17), the median PFS in patients with high baseline tumor HA staining (6/17 patients) was substantially longer, 219 days, than in the patients with low baseline tumor HA staining (11/17 patients), 108 days. Median overall survival (OS) was 200 days (95% CI, 123-370) in the efficacy-evaluable population (n = 24). Among efficacy-evaluable patients with baseline tumor HA staining (n = 17), the median OS in patients with high baseline tumor HA staining (6/17 patients) was substantially longer, 395 days, than in the patients with low baseline tumor HA staining (11/17 patients), 174 days. The most common treatment-emergent adverse events (occurring in ≥ 15% of patients) were peripheral edema, muscle spasms, thrombocytopenia, fatigue, myalgia, anemia, and nausea. Thromboembolic (TE) events were reported in 8 patients (28.6%) and musculoskeletal events were reported in 21 patients (75%) which were generally grade 1/2 in severity.


HALO 109-202:
4



Study Halo 109-202:
In the second quarter of 2013, we initiated StudyHALO 109-202 (HALO-202), a Phase 2 multicenter randomized clinical trial evaluating PEGPH20 as a first-line therapy for patients with stage IV metastatic pancreatic cancer.PDA. The study was designed to enroll patients who would receive gemcitabine and nab-paclitaxel (ABRAXANE®(ABRAXANE®) either with or without PEGPH20. The primary endpoint is to measure the improvement in progression-free survivalPFS in patients receiving PEGPH20 plus gemcitabine and nab-paclitaxelABRAXANE (PAG arm) compared to those who are receiving gemcitabine and nab-paclitaxel alone.ABRAXANE alone (AG arm). In April 2014, after 146 patients had been enrolled, the trial was put on clinical hold by Halozyme and the FDA to assess a question raised by the Data Monitoring Committee regarding a possible difference in the thromboembolic eventTE events rate between the group of patients treated with PEGPH20, nab-paclitaxel and gemcitabine (PAG arm)in the PAG arm versus the group of patients treated with nab-paclitaxel and gemcitabine without PEGPH20 (AG arm).in the AG arm. This portion of the study and patients in this portion are now referred to as Stage 1. It should be noted that atAt the time of the clinical hold all patients remaining in the study continued on gemcitabine and nab-paclitaxel.ABRAXANE. In July 2014, the Study 109-202HALO-202 was reinitiated (Stage 2) under a revised protocol, which excludes patients that are expected to be at a greater risk for thromboembolic events,TE events. The revised protocol provides for thromboembolism prophylaxis of all patients in both arms of the study with low molecular weight heparin, and adds evaluation of the thromboembolic eventTE events rate in Stage 2 PEGPH20-treated patients compared with Stage 1 as a co-primary end point. Stage 2 of Study 109-202 is designed to enrollHALO-202 enrolled an additional 114133 patients, to add to the 146 patients already accrued in the clinical trial, with a 2:1 randomization for the PAG arm compared to AG.the AG arm.


In January 2015,March 2016, our partner Ventana received approval for an investigational device exemption (IDE) application from the FDA for our companion diagnostic test to enable patient selection in our Phase 3 Study HALO-301 of PEGPH20 in HA-High patients. Based on the cutpoint for the Ventana diagnostic, we disclosed initial efficacy and safety dataexpect approximately 35 to 40 percent of stage IV PDA patients to have HA-High tumors, similar to the previously reported interim results from an interim assessment of Stage 1 of Study 109-202. HALO-202 using the Halozyme prototype assay.
In January 2017, we announced topline results from the safety evaluable population, i.e. patients who had received at least one dosecombined analysis of study drugs in test or comparator arm, of 135 patients (74 in PAG arm, 61 in AG arm), the RECIST 1.1 Objective Response Rate,Stage 1 and Stage 2, and Stage 2 alone, based on patienta December 2016 data through April 9, 2014, was 34% (25/74 patients)cutoff. The combined analysis included 135 treated patients in Stage 1, of whom a total of 45 patients (24 in the PAG arm and 23%21 in the AG arm (14/61 patients) (P=0.17). Among the safety evaluablearm) were determined to have high HA, and 125 treated patients with baseline HA staining and at least one protocol specified post base line scan after two cyclesin Stage 2, of study drug to assess tumor response (62 patients), the objective response rate was 60%whom a total of 35 patients (24 in the PAG arm (21/35 patients), and 37%11 in the AG arm (10/27 patients) (P=0.09). In patients witharm) were determined to have high base line tumor staining for HAHA. This analysis of secondary and exploratory endpoints was conducted using the objective response rate was 71%Ventana companion diagnostic to prospectively identify high levels of HA. The key results showed in the PAG arm (12/17 patients),combined Stage 1 and 29%Stage 2 dataset:
The primary endpoint of PFS in the AG arm (5/17efficacy evaluable population (total of 231 patients) (P=0.016). To assesswas met with statistical significance with a median PFS data for all Stage 1 patients who had received at least one dose of drug and had a base line HA analysis (106 patients), data was evaluated through December 9, 2014 to allow for sufficient follow up duration. Per the description above, patients in the PAG arm all discontinued PEGPH20 in April and only a portion restarted PEGPH20 in July 2014 upon the lift of the clinical hold. The PFS in this population was 5.56.0 months in the PAG arm vs 4.8compared to 5.3 months in the AG arm, (p=0.086). In patientshazard ratio (HR) with high base line tumor staining for HA,a 95% confidence interval (CI): 0.73 (0.53, 1.00); p=0.048;
The secondary endpoint of PFS in the HA-High intent to treat population (total of 84 HA-High patients) was met with statistical significance with a median PFS wasof 9.2 months in the PAG arm (12/25 patients) vs 4.3compared to 5.2 months in the AG arm, (15/23 patients) (P=0.031).HR 0.51 (95% CI: 0.26, 1.00); p=0.048;
WeThe exploratory analysis of median OS was 11.5 months vs. 8.5 months in the PAG vs. AG arms, respectively. Factors potentially having an impact on these results include less aggressive disease among patients in the AG arm within the Stage 1 patient population, and 9 of the data monitoring committee for Study 109-202 continue24 patients in the PAG arm (approximately 40 percent) discontinued PEGPH20 treatment at the time of the clinical hold, resulting in many patients receiving AG alone in both arms.
In the Stage 2 cohort population, in a total of 35 HA-High patients, the key results showed:
Median PFS was 8.6 months in the PAG arm compared to closely monitor4.5 months in the occurrenceAG arm, hazard ratio of thromboembolic events0.63 (95% CI: 0.21, 1.93);
Median overall survival (OS) was 11.7 months in enrolled patients after the revisionPAG arm compared to 7.8 months in the protocol. AG arm, hazard ratio of 0.52 (95% CI: 0.22, 1.23);
The revised protocol includes pre-specified analyses to evaluateprimary safety endpoint of decreasing the rate of thromboembolicTE events in Stage 2 was also met with the rate of TE events reducing from 43 percent to 10 percent in the PAG arm and from 25 percent to 6 percent in the AG arm, following a protocol amendment that excluded patients at high risk of TE events and Study 109-202 may be halted again if the protocol changes do not result in a reduction of thromboembolic events in accordance with the pre-specified analysesintroduction of prophylaxis with low molecular weight heparin (enoxaparin) in Stage 2 of the study with the current 1mg/kg/day dose of enoxaparin prophylaxis given in both treatment arms of the study.
In June 2017, results from Study HALO-202 were presented at the ESMO World Congress of Gastrointestinal Cancer and the Annual Meeting of the American Society of Clinical Oncology (ASCO). HALO-202 is an ongoing study with an open database, and has completed enrollment. We continue to collect and receive data on both Stage 1 and Stage 2 patients. When the database is considered complete and locked, an updated analysis and Final Study Report will be generated.
HALO 109-301:
In March 2015, we met with the FDA to discuss both the interim efficacy and safety data from HALO-202, which included the potential risk profile including TE event rate. Based on the feedback from that meeting, we proceeded with HALO 109-301 (HALO-301), a Phase 3 clinical study of PEGPH20 in patients with stage IV PDA, using a design allowing for potential marketing application based on PFS (accelerated approval pathway) or OS. The study will enroll patients whose tumors accumulate high levels of HA measured using the Ventana companion diagnostic test. The FDA provided feedback on the current companion diagnostic approach and confirmed that an approved IDE is required for the Phase 3 study.


The use of PFS as the basis for marketing approval will be subject to the overall benefit and risk associated with PEGPH20 combined with gemcitabine and ABRAXANE therapy, including the:
Magnitude of the PFS treatment effect observed;
Toxicity profile; and
Interim OS data.
In June 2015, we received scientific advice/protocol assistance from the European Medicines Agency (EMA) regarding our Phase 3 study. The EMA agreed to the patient population, and the use of both PFS and OS as co-primary endpoints stating that OS is the preferred endpoint and that ultimate approval would require an overall positive benefit:risk balance.
In March 2016, we dosed the first patient in HALO-301, a Phase 3 multicenter randomized clinical trial evaluating PEGPH20 as a first-line therapy for patients with stage IV PDA. The study will evaluate the effects on PFS and OS of PEGPH20 with gemcitabine and ABRAXANE compared with gemcitabine and ABRAXANE alone in stage IV PDA patients. In September 2017, our independent Data Safety Monitoring Committee met to review ongoing safety data from the trial and informed us the study should proceed as planned. Approximately 235 sites in 22 countries located in North America, Europe, South America and Asia have been initiated to participate in the protocol.HALO-301 study. An interim analysis will be conducted for our first primary endpoint when we achieve the target number of PFS events. We project that the target number of PFS events will be achieved in the fourth quarter of 2018. At that time we project we will have enrolled approximately 500 patients.
SWOG Study S1313:S1313:
In October 2013, SWOG, a cancer research cooperative group of more than 4,000 researchers in over 500 institutions around the world, initiated a 144 patient Phase 1b/2 randomized clinical trial in some of their study centers, examining PEGPH20 in combination with modified FOLFIRINOX chemotherapy (mFOLFIRINOX) compared to mFOLFIRINOXmodified FOLFIRINOX treatment alone in patients with metastatic pancreatic adenocarcinomastage IV PDA (funded by the National Cancer Institute). ThisAs announced in March 2017, SWOG stopped enrollment in the Phase 1b/2 trial. While PEGPH20 is a targeted investigational therapy for patients with high levels of HA, the SWOG study was also placedenrolling patients irrespective of HA levels, referred to as an all-comer population. During a planned early futility analysis, SWOG’s independent Data Monitoring Committee found, based on clinical hold temporarilypreliminary data, that the addition of PEGPH20 given every two weeks to modified FOLFIRINOX in this all-comer population would be unlikely to demonstrate a statistically significant improvement in the primary endpoint of overall survival. SWOG further reported that a higher rate of death was observed in the PEGPH20 arm versus modified FOLFIRINOX alone. SWOG has stopped the study and continues its ongoing effort to collect and clean outstanding data. In January 2018, SWOG presented final data of the all-comers population at the timeASCO-GI conference. The median overall survival was 7.7 months for the PEGPH20 arm vs. 14.4 months in the modified FOLFIRINOX alone arm. Also, increased GI-toxicities and substantially shorter median treatment duration for modified FOLFIRINOX were reported for the PEGPH20 arm compared to the modified FOLFIRINOX alone arm. Collection of biopsy samples from participating sites, to the hold on Study 109-202. extent available, is ongoing to potentially enable an HA biomarker subgroup analysis. Our PEGPH20 studies and clinical collaborations in combination with agents other than modified FOLFIRINOX continue unchanged.
Clinical collaboration:
In September 2014, the FDA removed the clinical hold on patient enrollment and dosing ofOctober 2016, we announced that PEGPH20 in this SWOG cooperative study. The study has resumed under a revised protocol, and patient enrollment is continuing. As with Study 109-202, the occurrence of thromboembolic events will be closely monitoredincluded in enrolleda pancreatic cancer clinical trial initiative called Precision Promise, an initiative that aims to change the current treatment approach to pancreatic cancer by offering options to patients based on the molecular profile of their tumor. This is being accomplished through the Pancreatic Cancer Action Network leading a collaboration that brings together clinicians, researchers, and drug developers. Pancreatic Cancer Action Network continues to work to finalize the continuation of this study may be halted againtrial design and protocol which is expected to include a potential PEGPH20 trial arm or trial in accordance with event rate rules established in the protocol, or for other safety reasons.2019.
Other indications outside of pancreatic cancer:
Study HALO 107-201, PRIMAL Study107-101::
In November 2015, we initiated a Phase 1b study exploring the combination of PEGPH20 and KEYTRUDA®, an immuno-oncology agent in relapsed non-small cell lung cancer (NSCLC) and gastric cancer. In December 2014,2016, we identified a dose of PEGPH20, namely 2.2 ug/kg, to move into the dose expansion phase of the study with KEYTRUDA in combination with PEGPH20. We are enrolling both NSCLC and gastric cancer patients prospectively based on a patient being determined to be HA-High using


the Ventana companion diagnostic test. In September 2017, our standing Independent Data Monitoring Safety Committee met to review ongoing safety data from the trial and informed us that the study should proceed with study protocol modifications to exclude patients at risk and increase liver safety monitoring, after observing clinical and laboratory signs of hepato-biliary dysfunction.
Clinical collaborations:
In July 2015, we entered into a clinical collaboration agreement with Eisai Co., Ltd. (Eisai) to evaluate Eisai's HALAVEN® (eribulin) with PEGPH20 in HER2-negative metastatic breast cancer. In July 2016, the first patient was dosed in a Phase 1b/2 study for patients treated with up to two lines of prior therapy for HER2-negative metastatic breast cancer. In January 2018, the Phase 1b portion of the study closed enrollment. As a result of an Eisai portfolio decision, no further clinical development is planned on the Phase 2 portion of this study. Data analysis is ongoing and a submission of the results of this study to a scientific forum is expected in the second half of 2018. Halozyme and Eisai jointly share the costs to conduct this global study.
In November 2016, we entered into an agreement with Genentech, a member of the Roche Group, to collaborate on clinical studies to evaluate their cancer immunotherapy Tecentriq, an anti-PD-L1 monoclonal antibody, in combination with PEGPH20, in up to eight different tumor types. Genentech initiated a Phase 1b/2 clinical trial in patients with previously treated metastatic PDA in July 2017 and a Phase 1b/2 clinical trial in patients with gastric cancer in October 2017, as part of its Morpheus master protocol. We will supply PEGPH20 for the Genentech-funded studies. In October 2017, we initiated a Phase 1b/2 clinical trial to evaluateassess Tecentriq with PEGPH20 in second line in combination with docetaxel (Taxotere®) in Non-Small Cell Lung Cancer (NSCLC) patients. In this study, we expect to evaluate and identify the dose, dosing regimen and safety of PEGPH20 plus docetaxel in previously treated patients with NSCLC. Upon identification ofcholangiocarcinoma and gall bladder cancer (HALO 110-101/MATRIX). Genentech will supply Tecentriq for the dose and dosing regimen, we plan to expand the trial to evaluate safety and efficacy.


Halozyme sponsored study.
5



Study HALO 107-101, the immuno-oncology trial: In the second half of 2015, we plan to initiate an immuno-oncology trial, exploring the combination of PEGPH20 and an immune-oncology agent such as a PD1/PDL1 inhibitor. We expect to evaluate and identify the dose and safety of PEGPH20 plus the immune-oncology agent prior to embarking on expansion into multiple tumors including NSCLC and Gastric cancers.Regulatory
Regulatory:
In September 2014, theThe FDA has granted Fast Track designation for our program investigating PEGPH20 in combination with gemcitabine and nab-paclitaxel for the treatment of patients with metastatic pancreatic cancerstage IV PDA to demonstrate an improvement in overall survival.OS. The Fast Track designation process was developed by the FDA to facilitate the development and expedite the review of drugs to treat serious or life-threatening diseases and address unmet medical needs.
In October 2014, theThe FDA has granted Orphan Drug designation for PEGPH20 for the treatment of pancreatic cancer. The FDA Office of Orphan Products DevelopmentDevelopment’s mission is to advance the evaluation and development of products (drugs, biologics, devices, or medical foods) that demonstrate promise for the diagnosis and/or treatment of rare diseases or conditions. In December 2014,Similarly, the European Committee for Orphan Medicinal Products of the EMA designated PEGPH20 an orphan medicinal product for the treatment of pancreatic cancer.
In the first quarter of 2015,Other Pipeline Assets
HTI-1511: HTI-1511 is a novel antibody-drug conjugate (ADC) targeting epidermal growth factor receptor (EGFR) to treat solid tumors, including those with drug-resistant mutations. At this time, we plan to meet with the Health authorities in the U.S., and subsequently in Europe, to discuss a potential registration seeking study in stage IV metastatic pancreatic cancer patients whose tumors are determined to have high levels of HA accumulation. In the first half of 2015, we also plan to engage the health authorities fordiscontinued further development of a companion diagnostic agent for detection and qualification of hyaluronanthis program.
PEG-ADA2: PEGylated adenosine deaminase 2, or PEG-ADA2, is an immune checkpoint inhibitor that targets adenosine, which may accumulate to high levels in the tumor tissue of cancer patients.
In October 2014, we announced the issuance of U.S. Patent No. 8,846,034 claiming methods for selecting a subject for treatment of a hyaluronan-associated diseasemicroenvironment and has been linked to immunosuppression. We are currently in preclinical development with an anti-hyaluronan agent, such as PEGPH20, as well as diagnostic agents for the detectionPEG-ADA2 and quantification of hyaluronanare exploring potential collaboration or partnership interest in a biological sample in patients.
Ultrafast Insulin Program Evaluation of rHuPH20 and Hylenex in Patients with Diabetes
Two distinct opportunities exist for the use of rHuPH20 and Hylenex recombinant in diabetes. We intendthis program prior to seek to enter into collaborations with third parties or explore other strategic alternatives in order to exploit these opportunities.
The first opportunity, Continuous Subcutaneous Insulin Infusion (CSII), is focused on assessing the effects of pre-treating the insulin infusion site with Hylenex recombinant at the time of infusion site change (once every 3 days). Insulin pump therapy is growingmaking additional investments in the U.S. among patients with type 1 and type 2 diabetes and the focusdevelopment of our program is type 1. We believe that the pre-treatment of the infusion site with Hylenex recombinant could result in faster onset and shorter duration of insulin action.PEG-ADA2.
In the first quarter of 2013, we initiated CONSISTENT 1, a clinical trial evaluating the safety and efficacy of Hylenex recombinant and a new formulation of Hylenex when used as pretreatment of the insulin infusion site in patients with type 1 diabetes. With enrollment completed in the third quarter of 2013, the trial has enrolled 456 patients with type 1 diabetes who were randomized 3:1 to either rapid acting analog insulin (RAI) delivered by CSII with Hylenex recombinant pretreatment of the infusion site or standard CSII using RAI alone. Subjects randomized to the Hylenex recombinant group were administered 150 units of Hylenex recombinant once every three days through each new infusion cannula, immediately prior to initiation of insulin delivery. In June 2014, we presented a scientific poster highlighting the CONSISTENT 1 trial results at the 74th Scientific Sessions of the American Diabetes Association in San Francisco. Data reported in the presentation showed that the study's primary endpoint of non-inferiority for A1C at six months between the use of Hylenex and the new formulation of Hylenex in comparison to no pre-treatment, was met. In addition to other trial data, the poster also presented data indicating that there was a potential reduction in the rate of some types of hypoglycemic events associated with the use of the Hylenex formulations in comparison to no pre-treatment. The most common treatment related adverse event in the Hylenex recombinant groups was mild infusion site discomfort. With this exception, adverse events were similar across the treatment and control groups.


6



In parallel with our clinical activities, we have been in dialog with the FDA regarding the requirements for updating the Hylenex recombinant label in a manner that would support future promotion in this indication. While discussions with the FDA are ongoing, we have learned that the FDA will likely request additional clinical data for a label update, translating to potentially higher projected costs and longer time to market than we had originally anticipated. We are continuing to seek clarity with the FDA on what data will be required, if any. In addition, we determined in the fourth quarter 2014 that with all patients having completed 12 months on the trial, we did not need additional data contribution from the second year of the CONSISTENT 1 trial, and we stopped the trial and are currently summarizing the data for all patients treated for 12 months. After evaluating our options, we have decided that except for any costs in connection with closing out the CONSISTENT 1 trial, we do not intend to incur substantial additional expenses for the program.
The second opportunity, Multiple Daily Injection (MDI), is to combine rHuPH20 with an FDA approved RAI, e.g., insulin lispro (Humalog®) (Lispro-PH20), insulin aspart (Novolog®) (Aspart-PH20) and insulin glulisine (Apidra®) (Glulisine-PH20), (each such combination, analog-PH20), to seek to accelerate their action. A number of clinical trials investigating the various attributes of our product candidates have been completed, with the last study completed in 2011. Since then we have only conducted limited additional development work with this opportunity.
Data from two treatment studies - one in type 1 diabetes patients and one in type 2 diabetes patients has been published. Copies of these publications can be found at http://www.halozyme.com/Technology/Journals-Abstracts-And-Posters/default.aspx. Both studies met their primary endpoints of A1C non-inferiority and improved post-prandial glucose control compared to patients who were treated with RAI alone. Additionally, data from the type 1 diabetes treatment study indicated that Analog-PH20 formulations reduced hypoglycemia compared to RAI alone.
ENHANZE Collaborations
Roche Collaboration
In December 2006, we and Roche entered into ana collaboration and license agreement under which Roche obtained a worldwide exclusive license to develop and commercialize product combinations of rHuPH20 withand up to thirteen Roche target compounds (the Roche Collaboration). Roche initially had the exclusive right to apply rHuPH20 to only three pre-defined Roche biologic targets with the option to exclusively develop and commercialize rHuPH20 with ten additional targets. Roche had the right to exercise this option to identify additional targets for ten years. As of the ten year anniversary of the Roche Collaboration in December 31, 2014,2016, Roche hashad elected a total of five exclusiveeight targets, and retains the option to develop and commercialize rHuPH20 with three additional targets through the paymenttwo of annual license maintenance fees.which are exclusive.


In September 2013, Roche launched a SCsubcutaneous (SC) formulation of Herceptin (trastuzumab) (Herceptin SC) in Europe for the treatment of patients with HER2-positive breast cancer. This formulation utilizes our patented Enhanze technologyENHANZE Technology and is administered in two to five minutes, rather thancompared to 30 to 90 minutes with the standard intravenous form. Roche received European marketing approval for Herceptin SC in August 2013. The European Commission’s approval was based on data from Roche’s Phase 3 HannaH study which showed that the subcutaneous formulation of Herceptin was associated with comparable efficacy (pathological complete response, pCR) to Herceptin administered intravenously in women with HER2-positive early breast cancer and resulted in non-inferior trastuzumab plasma levels. Overall, the safety profile in both arms of the HannaH study was consistent with that expected from standard treatment with Herceptin and chemotherapy in this setting. No new safety signals were identified. Breast cancer is the most common cancer among women worldwide. Each year, about 1.4 million new cases of breast cancer are diagnosed worldwide, and over 450,000 women will die of the disease annually. In HER2-positive breast cancer, increased quantities of the human epidermal growth factor receptor 2 (HER2) are present on the surface of the tumor cells. This is known as “HER2 positivity” and affects approximately 15% to 20% of women with breast cancer. HER2-positive cancer is reported to be a particularly aggressive form of breast cancer.


Directed at the same target, Roche initiated a Phase 1 study of rHuPH20 with PERJETA® (pertuzumab) in patients with early breast cancer in March 2016.
7



In June 2014, Roche launched MabThera® SC in Europe for the treatment of patients with common forms of non-Hodgkin lymphoma (NHL). This formulation utilizes our patented Enhanze technologyENHANZE Technology and is administered in approximately five minutes compared to the approximately 2.51.5 to 4 hour infusion time for intravenous MabThera. The European Commission approved MabThera SC in March 2014. The European Commission's approval was based primarily on data from Roche's Phase 3 pivotal clinical studies, which was recently published in The Lancet Oncology. NHL is a type of cancerIn May 2016, Roche announced that affects lymphocytes (white blood cells)the EMA approved Mabthera SC to treat patients with chronic lymphocytic leukemia (CLL). NHL represents approximately 85% of all lymphomas diagnosed and was responsible for approximately 200,000 annual deaths worldwide in 2012. Lymphomas are a cancerIn June 2017, the FDA approved Genentech’s (a member of the lymphatic system (composedRoche Group) RITUXAN HYCELA™, a combination of lymph vessels, lymph nodesrituximab and organs) which helps to keeprHuPH20 (approved and marketed under the bodily fluid levels balanced and to defend the body against invasion by disease. Lymphoma develops when white blood cells (usually B-lymphocytes) in the lymph fluid become cancerous and begin to multiply and collect in the lymph nodes or lymphatic tissues such as the spleen. Some of these cells are released into the bloodstream and spread around the body, interfering with the body's production of healthy blood cells.
Additional information about the Phase 3 Herceptin SC and Phase 3 MabThera SC clinical trials canbrand in countries outside the U.S.), for CLL and two types of NHL, follicular lymphoma and diffuse large B-cell lymphoma.
In September 2017, we and Roche entered into an agreement providing Roche the right to develop and commercialize one additional exclusive target using our ENHANZE Technology (the 2017 Roche Collaboration). The upfront license payment may be found at www.clinicaltrials.govfollowed by event-based payments subject to Roche’s achievement of specified development, regulatory and www.roche-trials.com. Information available on these websites is not incorporated into this report.sales-based milestones. In addition, Roche will pay royalties to us if products under the collaboration are commercialized.
BaxterIn January 2018, Roche initiated a Phase 1 study of an undisclosed target with ENHANZE Technology.
Baxalta Collaboration
In September 2007, we and BaxterBaxalta entered into ana collaboration and license agreement under which BaxterBaxalta obtained a worldwide, exclusive license to develop and commercialize product combinations of rHuPH20 with GAMMAGARD LIQUID (HYQVIA) (the BaxterBaxalta Collaboration). GAMMAGARD LIQUID is a current Baxter product thatHYQVIA is indicated for the treatment of primary immunodeficiency disorders associated with defects in the immune system.
In OctoberMay 2013, the European Commission granted Baxalta marketing authorization in all EU Member States for the use of HYQVIA (solution for subcutaneous use) as replacement therapy for adult patients with primary and secondary immunodeficiencies. Baxalta launched HYQVIA in the first EU country in July 2013 and has continued to launch in additional countries.
In September 2014, Baxter announcedHYQVIA was approved by the launch and first shipments of Baxter’s HYQVIA productFDA for treatment of adult patients with primary immunodeficiency in the U.S. HYQVIA was approved by the FDA in September 2014 and is the first subcutaneous IGimmune globulin (IG) treatment approved for adult primary immunodeficiency patients with a dosing regimen requiring only one infusion up to once per month (every three to four weeks) and one injection site per infusion in most patients, to deliver a full therapeutic dose of IG. ThePrior to the approval of HYQVIA, the majority of primary immunodeficiency patients today receivereceived intravenous infusions in a doctor’s office or infusion center, and currentother subcutaneous IG treatments require weekly or bi-weekly treatment with multiple infusion sites per treatment. The FDA'sFDA’s approval of HYQVIA iswas a significant milestone for us as it representsrepresented the first U.S. approved BLA which utilizes our rHuPH20 platform.
In May 2013,2016, Baxalta announced that HYQVIA received a marketing authorization from the European Commission granted Baxter marketing authorizationfor a pediatric indication, which is being launched in all EU Member States for the use of HYQVIA (solution for subcutaneous use) as replacement therapy for adult patients withEurope to treat primary and certain secondary immunodeficiencies. Baxter launched HYQVIA in the first EU country in July 2013 and in additional EU countries in the second half of 2013 and in 2014.
Pfizer Collaboration
In December 2012, we and Pfizer entered into a collaboration and license agreement, under which Pfizer has the worldwide license to develop and commercialize products combining our rHuPH20 enzyme with Pfizer proprietary biologics directed to up to six targets in primary care and specialty care indications. Targets may be selected on an exclusive or non-exclusive basis. In September 2013, Pfizer has elected the fourth therapeutic targetfive targets on an exclusive basis. In December 2013, Pfizer announced that one of the targets is proprotein convertase subtilisin/kexin type 9, also known as PCSK9. The PCSK9 gene provides instructions for making a protein that helps regulate the amount of cholesterol in the bloodstream. Pfizer is also developing Rivipansel directed to another target under the collaboration to treat vaso-occlusive crisis in individuals with sickle cell disease.basis and returned two targets.


Janssen Collaboration
In December 2014, we and Janssen entered into a collaboration and license agreement, under which Janssen has the worldwide license to develop and commercialize products combining our rHuPH20 enzyme with Janssen proprietary biologics directed to up to five targets. Targets may be selected on an exclusive or non-exclusive basis. Janssen has elected one undisclosedCD38 as the first target on an exclusive basis. In November 2015, Janssen initiated dosing in a Phase 1b clinical trial evaluating subcutaneous delivery of DARZALEX (daratumumab), directed at CD38, using ENHANZE Technology, in multiple myeloma patients. In December 2016, Janssen announced results of the trial, which supported continued development of daratumumab with rHuPH20. In December 2017, Janssen announced data which demonstrated that subcutaneous administration of DARZALEX and rHuPH20 was well-tolerated, with rates of infusion reactions lower than those observed with IV administration of DARZALEX. Janssen has initiated four Phase 3 studies and one Phase 1 study of daratumumab combined with the ENHANZE Technology in patients with Amyloidosis, Smoldering Myeloma and Multiple Myeloma. A Phase 2 study of daratumumab combined with the ENHANZE Technology is planned in 2018 for patients with multiple myeloma.
AbbVie Collaboration
In June 2015, we and AbbVie entered into a collaboration and license agreement, under which AbbVie has the worldwide license to develop and commercialize products combining our rHuPH20 enzyme with AbbVie proprietary biologics directed to up to nine targets. Targets may be selected on an exclusive basis. AbbVie elected one target on an exclusive basis, TNF alpha, for which it has discontinued development and returned the target.
Lilly Collaboration
In December 2015, we and Lilly entered into a collaboration and license agreement, under which Lilly has the worldwide license to develop and commercialize products combining our rHuPH20 enzyme with Lilly proprietary biologics directed to up to five targets. Targets may be selected on an exclusive basis. Lilly has elected two targets on an exclusive basis and one target on a semi-exclusive basis. In August 2017, Lilly initiated a Phase 1 study of an investigational new therapy in combination with rHuPH20.
BMS Collaboration
In September 2017, we and BMS entered into a collaboration and license agreement, which became effective in November 2017, under which BMS has the worldwide license to develop and commercialize products combining our rHuPH20 enzyme with BMS immuno-oncology targets directed at up to eleven targets. Targets may be selected on an exclusive basis, with the exception of one co-exclusive target. BMS has designated multiple immuno-oncology targets including programmed death 1 (PD-1) and has an option to select additional targets within five years from the effective date.
Alexion Collaboration
In December 2017, we and Alexion entered into a collaboration and license agreement, under which Alexion has the worldwide license to develop and commercialize products combining our rHuPH20 enzyme with Alexion’s portfolio of products directed at up to four targets. Targets may be selected on an exclusive basis. Alexion elected two targets on an exclusive basis, including a C5 complement inhibitor and has an option to select two additional targets within five years from the effective date. Alexion plans to initiate a Phase 1 trial in 2018 to study a next-generation subcutaneous formulation of ALXN1210 (ALXN1210 SC) with ENHANZE.
For a further discussion of the material terms of our collaboration agreements, refer to Note 4, Collaborative Agreements, to our consolidated financial statements.


8



Customers
The following table indicates the percentage of total revenues in excess of 10% with any single customer:
 Year Ended December 31,
 2014 2013 2012
Roche57% 64% 45%
Janssen20% 
 
Baxter3% 10% 17%
Pfizer1% 4% 22%
 Year Ended December 31,
 2017 2016 2015
Roche38% 63% 42%
BMS32% 
 
Alexion13% 
 
Baxalta7% 12% 7%
Lilly
 6% 19%
AbbVie
 4% 17%
For additional information regarding our revenues from external customers, refer to Note 2, Summary of Significant Accounting Policies — Concentrations of Credit Risk, Sources of Supply and Significant CustomersCustomer.s, to our consolidated financial statements.
Patents and Proprietary Rights
Patents and other proprietary rights are essential to our business. Our success will depend in part on our ability to obtain patent protection for our inventions, to preserve our trade secrets and to operate without infringing the proprietary rights of third parties. Our strategy is to actively pursue patent protection in the U.S. and certain foreign jurisdictions for technology that we believe to be proprietary to us and that offers us a potential competitive advantage. Our patent portfolio includes 2033 issued patents in the U.S., 80more than 370 issued patents in Europe and other countries in the world and a number ofmore than 170 pending patent applications. In general, patents have a term of 20 years from the application filing date or earlier claimed priority date. Our issued patents will expire between 2022 and 2032. We have multiple patents and patent applications throughout the world pertaining to our recombinant human hyaluronidase and methods of use and manufacture, including an issued U.S. patent which expires in 2027 and an issued European patent which expires in 2024, which we believe cover the products and product candidates under our existing collaborations, Hylenex recombinant, PEGPH20 and our endocrinology product candidates. In addition, we have, under prosecution throughout the world, multiple patent applications that relate specifically to individual product candidates under development, the expiration of which can only be definitely determined upon maturation into our issued patents. We believe our patent filings represent a barrier to entry for potential competitors looking to utilize these hyaluronidases.
In addition to patents, we rely on unpatented trade secrets, proprietary know-how and continuing technological innovation. We seek protection of these trade secrets, proprietary know-how and innovation, in part, through confidentiality and proprietary information agreements. Our policy is to require our employees, directors, consultants, advisors, collaborators, outside scientific collaborators and sponsored researchers, other advisors and other individuals and entities to execute confidentiality agreements upon the start of employment, consulting or other contractual relationships with us. These agreements provide that all confidential information developed or made known to the individual or entity during the course of the relationship is to be kept confidential and not disclosed to third parties except in specific circumstances. In the case of employees and some other parties, the agreements provide that all inventions conceived by the individual will be our exclusive property. Despite the use of these agreements and our efforts to protect our intellectual property, there will always be a risk of unauthorized use or disclosure of information. Furthermore, our trade secrets may otherwise become known to, or be independently developed by, our competitors.
We also file trademark applications to protect the names of our products and product candidates. These applications may not mature to registration and may be challenged by third parties. We are pursuing trademark protection in a number of different countries around the world. There can be no assurances that our registered or unregistered trademarks or trade names will not infringe on rights of third parties or will be acceptable to regulatory agencies.


Research and Development Activities
Our research and development expenses consist primarily of costs associated with the development and manufacturing of our product candidates, compensation and other expenses for research and development personnel, supplies and materials, costs for consultants and related contract research, clinical trials, facility costs and amortization and depreciation. We charge all research and development expenses to operations as they are incurred. Our research and development activities are primarily focused on the development of our various product candidates.


9



Due to the uncertainty in obtaining the FDA and other regulatory approvals, our reliance on third parties and competitive pressures, we are unable to estimate with any certainty the additional costs we will incur in the continued development of our proprietary product candidates for commercialization. However, we expect our research and development expenses for PEGPH20 to increase as our program advances into additional tumors and later stages of clinical development.
Manufacturing
We do not have our own manufacturing facility for our product and product candidates, or their active pharmaceutical ingredient (API) or bulk forms, or the capability to package our product.products. We have engaged third parties to manufacture bulk rHuPH20, PEGPH20 and our product Hylenex recombinant.
We have existing supply agreements with contract manufacturing organizations Avid Bioservices, Inc. (Avid) and Catalent Indiana LLC (formerly Cook Pharmica LLC (Cook)LLC) (Catalent) to produce supplies of bulk rHuPH20. These manufacturers each produce bulk rHuPH20 under current Good Manufacturing Practices (cGMP) for clinical and commercial uses. Avid and CookCatalent currently produceproduces bulk rHuPH20 for use in Hylenex recombinant, product candidates and our other collaboration products and product candidates. Avid currently produces bulk rHuPH20 for use in collaboration products. We rely on their ability to successfully manufacture these batches according to product specifications. Cook has limited experience manufacturing bulk rHuPH20. In addition, we have been working to scale-up, validatevalidated and qualify Cookqualified a new facility operated by Avid as a manufacturer of bulk rHuPH20 for use in the productproducts and product candidates under the Roche collaboration. To date, Cook has not been submitted to the European regulatory authorities by Roche as an approved manufacturer for Herceptin SC and MabThera SC.Collaboration. It is essentialimportant for our business for CookCatalent and Avid to (i) retain their status as cGMP-approved manufacturing facilities; (ii) to successfully scale up bulk rHuPH20 production; and/or (iii) manufacture the bulk rHuPH20 required by us and our collaborators for use in our proprietary and collaboration products and product candidates. In addition to supply obligations, Avid and CookCatalent will also provide support for data and information used in the chemistry, manufacturing and controls sections for FDA and other regulatory filings.
We have a commercial manufacturing and supply agreement with Baxter, a cGMP-approved manufacturing facility, under which Baxter provides the final fill and finishing steps in the production process of Hylenex recombinant. Under our commercial manufacturing and supply agreement with Baxter, Baxter has agreed to fill and finish Hylenex recombinant product for us until December 31, 2017, subject to further extensions in accordance with the terms of the agreement. In June 2011, we entered into a services agreement with another third party manufacturer, Patheon Manufacturing Services, LLC (Patheon), for the technology transfer and manufacture, fill, finish or packaging of Hylenex recombinant. In October 2014, we received regulatory approval from the FDA for Patheon to manufacture Hylenex recombinant. In December 2014, we entered into a manufacturing services agreement with Patheon under which Patheon will provide the final fill and finishing steps in the production process of Hylenex recombinant. Under our commercial services agreement with Patheon, Patheon has agreed to fill and finish Hylenex recombinant product for us until December 31, 2019, subject to further extensions in accordance with the terms of the agreement. In 2015,addition, we will begin to transitionscaled up our manufacturing supplyof PEGPH20 with third party suppliers to Patheon to achieve higher capacitysupport additional clinical trials, including the Phase 3 trial, and lower cost of goods.ultimately, if approved, potential commercial supply.
Sales, Marketing and Distribution
HYLENEXHylenex Recombinant
Our commercial activities currently focus on Hylenex recombinant. We have a team of sales specialists that provide hospital and surgery center customers with the information about Hylenex recombinant and information needed to obtain formulary approval for, and support utilization of, Hylenex recombinant. Our commercial activities also include marketing and related services and commercial support services such as commercial operations, managed markets and commercial analytics. We also employ third-party vendors, such as advertising agencies, market research firms and suppliers of marketing and other sales support related services to assist with our commercial activities.
We sell Hylenex recombinant in the U.S. to wholesale pharmaceutical distributors, who sell the product to hospitals ambulatory surgery centers and other end-users.end-user customers. We have engagedengage Integrated CommercialCommercialization Solutions (ICS), a division of AmerisourceBergen Specialty Group, a subsidiary of AmerisourceBergen, to act as our exclusive distributor for commercial shipment and distribution of Hylenex recombinant to our customers in the United States. In addition to distribution services, ICS provides us with other key services related to logistics, warehousing, returns and inventory management, contract administration and chargebacks processing and accounts receivable management. In addition, we utilize third parties to perform various other services for us relating to regulatory


10



monitoring, including call center management, adverse event reporting, safety database management and other product maintenance services.
Competition
The pharmaceutical industry is characterized by rapidly advancing technologies, intense competition and a strong emphasis on proprietary therapeutics. We face competition from a number of sources, some of which may target the same indications as our product or product candidates, including large pharmaceutical companies, smaller pharmaceutical companies, biotechnology companies, academic institutions, government agencies and private and public research institutions, many of which have greater financial resources, drug development experience, sales and marketing capabilities, including larger, well established sales forces, manufacturing capabilities, experience in obtaining regulatory approvals for product candidates and other resources than us. We face competition not only in the commercialization of Hylenex recombinant, but also for the in-licensing or acquisition of additional product candidates, and the out-licensing of our Enhanze technology.ENHANZE Technology. In addition, our collaborators face competition in the commercialization of the product candidates for which the collaborators seek marketing approval from the FDA or other regulatory authorities.
HYLENEXHylenex Recombinant
Hylenex recombinant is currently the only FDA-approvedFDA approved recombinant human hyaluronidase on the market. The competitors for Hylenex recombinant include, but are not limited to, Bausch & LombValeant Pharmaceuticals International, Inc.'s FDA-approved’s product, Vitrase®, an ovine (ram) hyaluronidase, and Amphastar Pharmaceuticals, Inc.'s’s product, Amphadase®, a bovine (bull) hyaluronidase, which is not currently on the market.hyaluronidase. In addition, some commercial pharmacies compound hyaluronidase preparations for institutions and physicians even though compounded preparations are not FDA-approvedFDA approved products.
Government Regulations
The FDA and comparable regulatory agencies in foreign countries regulate the manufacture and sale of the pharmaceutical products that we have developed or currently are developing. The FDA has established guidelines and safety standards that are applicable to the laboratory and preclinical evaluation and clinical investigation of therapeutic products and stringent regulations that govern the manufacture and sale of these products. The process of obtaining regulatory approval for a new therapeutic product usually requires a significant amount of time and substantial resources. The steps typically required before a product can be introduced for human use include:
animal pharmacology studies to obtain preliminary information on the safety and efficacy of a drug; or
laboratory and preclinical evaluation in vitro and in vivo including extensive toxicology studies.
The results of these laboratory and preclinical studies may be submitted to the FDA as part of an IND application. The sponsor of an IND application may commence human testing of the compound 30 days after submission of the IND, unless notified to the contrary by the FDA.
The clinical testing program for a new drug typically involves three phases:
Phase 1 investigations are generally conducted in healthy subjects (in certain instances, Phase 1 studies that determine the maximum tolerated dose and initial safety of the product candidate are performed in patients with the disease);
Phase 2 studies are conducted in limited numbers of subjects with the disease or condition to be treated and are aimed at determining the most effective dose and schedule of administration, evaluating both safety and whether the product demonstrates therapeutic effectiveness against the disease; and
Phase 3 studies involve large, well-controlled investigations in diseased subjects and are aimed at verifying the safety and effectiveness of the drug.


11



Data from all clinical studies, as well as all laboratory and preclinical studies and evidence of product quality, are typically submitted to the FDA in a new drug application (NDA). The results of the preclinical and clinical testing of a biologic product candidate are submitted to the FDA in the form of a BLA, for evaluation to determine whether the product candidate may be approved for commercial sale. In responding to a BLA or NDA, the FDA may grant marketing approval or request additional information. If additional information is requested we may provide such information or deny thewithdraw our application. Although the FDA’s requirements for clinical trials are well established and we believe that we have planned and conducted our clinical trials in accordance with the FDA’s applicable regulations and guidelines, these requirements including requirements relating to testing the safety of drug candidates, may be subject to change as a result of recent announcements regarding safety problems with approved drugs. Additionally,change. Accordingly, we could be required to conduct additional trials beyond what we had planned due to the FDA’s safety and/or efficacy concerns or due to differing interpretations of the meaning of our clinical data.data or a change in the therapeutic landscape. (See Part I, Item 1A, Risk Factors.)
The FDA’s Center for Drug Evaluation and Research must approve an NDA and the FDA’s Center for Biologics Evaluation and Research must approve a BLA for a drug before it may be marketed in the United States. If we begin to market our proposed products for commercial sale in the U.S., any manufacturing operations that may be established in or outside the U.S. will also be subject to rigorous regulation, including compliance with cGMP. We also may be subject to regulation under the Occupational Safety and Health Act, the Environmental Protection Act, the Toxic Substance Control Act, the Export Control Act and other present and future laws of general application. In addition, the handling, care and use of laboratory animals are subject to the Guidelines for the Humane Use and Care of Laboratory Animals published by the National Institutes of Health.
Regulatory obligations continue post-approval, and include the reporting of adverse events when a drug is utilized in the broader patient population. Promotion and marketing of drugs is also strictly regulated, with penalties imposed for violations of FDA regulations, the Lanham Act and other federal and state laws, including the federal anti-kickback statute.
We currently intend to continue to seek, directly or through our collaborators, approval to market our products and product candidates in foreign countries, which may have regulatory processes that differ materially from those of the FDA. We anticipate that we will rely upon independent consultants to seek and gain approvals to market our proposed products in foreign countries or may rely on other pharmaceutical or biotechnology companies to license our proposed products. We cannot assure youguarantee that approvals to market any of our proposed products can be obtained in any country. Approval to market a product in any one foreign country does not necessarily indicate that approval can be obtained in other countries.
From time to time, legislation is drafted and introduced in Congress that could significantly change the statutory provisions governing the approval, manufacturing and marketing of drug products. In addition, FDA regulations and guidance are often revised or reinterpreted by the agency or reviewing courts in ways that may significantly affect our business and development of our product candidates and any products that we may commercialize. It is impossible to predict whether additional legislative changes will be enacted, or FDA regulations, guidance or interpretations changed, or what the impact of any such changes may be.
Segment Information
We operate our business as one segment, which includes all activities related to the research, development and commercialization of human enzymes.enzymes and other drug candidates. This segment also includes revenues and expenses related to (i) research and development activities conducted under our collaboration agreements with third parties and (ii) product sales of Hylenex recombinant. The chief operating decision-maker reviews the operating results on an aggregate basis and manages the operations as a single operating segment. We had no foreign based operations and noOur long-lived assets located in foreign countries had minimal book value as of and for the years ended December 31, 2014, 20132017 and 2012. Refer to the Notes for additional financial information regarding our operating segment.2016.
Executive Officers of the Registrant
Information concerning our executive officers, including their names, ages and certain biographical information can be found in Part III, Item 10, Directors, Executive Officers and Corporate Governance. This information is incorporated by reference into Part I of this report.


12



Employees
As of February 24, 2015,13, 2018, we had 153255 full-time employees. None of our employees are unionized and we believe our employee relations to be good.
Item 1A.Risk Factors
Item 1A.Risk Factors
Risks Related To Our Business
We have generated only limited revenuerevenues from product sales to date; we have a history of net losses and negative cash flow,flows, and we may never achieve or maintain profitability.
Relative to expenses incurred in our operations, we have generated only limited revenues from product sales, royalties, licensing fees, milestone payments, bulk rHuPH20 supply payments and research reimbursements to date and we may never generate sufficient revenues from future product sales, licensing fees and milestone payments to offset expenses. Even if we ultimately do achieve significant revenues from product sales, royalties, licensing fees, research reimbursements, bulk rHuPH20 supply payments and/or milestone payments, we expect to incur significant operating losses over the next few years. We have never been profitable,years and we may never become profitable.profitable on an extended basis. Through December 31, 2014,2017, we have incurred aggregate net losses of approximately $450.4 million.$522.4 million.
If our product candidates do not receive and maintain regulatory approvals, or if approvals are not obtained in a timely manner, such failure or delay would substantially impair our ability to generate revenues.
Approval from the FDA or equivalent health authorities is necessary to manufacture and market pharmaceutical products in the U.S. and the other countries in which we anticipate doing business have similar requirements. The process for obtaining FDA and other regulatory approvals is extensive, time-consuming, risky and costly, and there is no guarantee that the FDA or other regulatory bodies will approve any applications that may be filed with respect to any of our product candidates, or that the timing of any such approval will be appropriate for the desired product launch schedule for a product candidate. We and our collaborators attempt to provide guidance as to the timing for the filing and acceptance of such regulatory approvals, but such filings and approvals may not occur when we or our collaborators expect, or at all. The FDA or other foreign regulatory agency may refuse or delay approval of our product candidates for failure to collect sufficient clinical or animal safety data and require us or our collaborators to conduct additional clinical or animal safety studies which may cause lengthy delays and increased costs to our programs. For example, we have been in dialog with the FDA regarding the regulatory pathway and data requirements for updating the label for Hylenex recombinant for use in CSII but do not have clarity to date. However, we have learned that additional clinical data will likely be required for a label update. The lack of clarity has hampered our ability to develop and commercialize this opportunity and may impede our future ability to identify a strategic partner willing to invest in future development, registration and commercialization. In addition, the approval of Baxter'sBaxalta’s HYQVIA BLA in the U.S. was delayed until we and BaxterBaxalta provided additional preclinical data sufficient to address concerns regarding non-neutralizing antibodies to rHuPH20 that were detected in the registration trial. Although these antibodies have not been associated with any known adverse clinical effects, and the HYQVIA BLA was approved by the FDA in September 2014, we cannot assure you that they will not arise and have an adverse impact on future development of products which include rHuPH20, future sales of Hylenex recombinant, our ability to enter into collaborations, or be raised by the FDA or other health authorities in connection with testing or approval of products including rHuPH20.
Only three of our collaboration product candidates and one of our proprietary products have been approved for commercialization. We have no proprietary product candidates currently in the regulatory approval process. We and our collaborators may not be successful in obtaining approvals for any additional potential products in a timely manner, or at all. Refer to the risk factor titled “Our proprietary and collaboration product candidates or companion diagnostic assays may not receive regulatory approvals or their development may be delayed for a variety of reasons, including delayed or unsuccessful clinical trials, regulatory requirements or safety concerns” for additional information relating to the approval of product candidates.
Additionally, even with respect to products which have been approved for commercialization, in order to continue to manufacture and market pharmaceutical products, we or our collaborators must maintain our regulatory approvals. If we or any of our collaborators are unsuccessful in maintaining our regulatory approvals, our ability to generate revenues would be adversely affected.


13


We may need to raise additional capital in the future and there can be no assurance that we will be able to obtain such funds.

We may need to raise additional capital in the future to continue the development of our product candidates or for other current corporate purposes. Our current cash reserves and expected revenues during the next few years will not be sufficient for us to continue the development of our proprietary product candidates, to fund general operations and conduct our business at the level desired. In addition, if we engage in acquisitions of companies, products or technologies in order to execute our business strategy, we may need to raise additional capital. We may raise additional capital in the future through one or more financing vehicles that may be available to us including (i) the public offering of securities; (ii) new collaborative agreements; (iii) expansions or revisions to existing collaborative relationships; (iv) private financings; (v) other equity or debt financings; and/or (vi) monetizing assets.
In view of our stage of development, business prospects, the nature of our capital structure and general market conditions, if we are required to raise additional capital in the future, the additional financing may not be available on favorable terms, or at all. If additional capital is not available on favorable terms when needed, we will be required to raise capital on adverse terms or significantly reduce operating expenses through the restructuring of our operations or deferral of one or more product development programs. If we raise additional capital, a substantial number of additional shares may be issued, which may negatively affect our stock price and these additional shares will dilute the ownership interest of our current investors.
Use of our product candidates or those of our collaborators could be associated with side effects or adverse events.
As with most pharmaceutical products, use of our product candidates or those of our collaborators could be associated with side effects or adverse events which can vary in severity (from minor reactions to death) and frequency (infrequent or prevalent). Side effects or adverse events associated with the use of our product candidates or those of our collaborators may be observed at any time, including in clinical trials or when a product is commercialized, and any such side effects or adverse events may negatively affect our or our collaborators'collaborators’ ability to obtain or maintain regulatory approval or market our product candidates. Side effects such as toxicity or other safety issues associated with the use of our product candidates or those of our collaborators could require us or our collaborators to perform additional studies or halt development or salecommercialization of these product candidates or expose us to product liability lawsuits which will harm our business. We or our collaborators may be required by regulatory agencies to conduct additional animal or human studies regarding the safety and efficacy of our pharmaceutical product candidates which we have not planned or anticipated. Furthermore, there can be no assurance that we or our collaborators will resolve any issues related to any product related adverse events to the satisfaction of the FDA or any regulatory agency in a timely manner or ever, which could harm our business, prospects and financial condition. For example, in April 2014, a clinical hold was placed on patient enrollment and dosing of PEGPH20 in Study 202HALO-202 as a result of a possible difference in the thromboembolicTE event rate that had been observed at that time in the trial between the group of patients treated with PEGPH20 versus the group of patients treated without PEGPH20. The clinical hold was lifted by the FDA in June 2014, and we have resumedcompleted enrollment and dosing of PEGPH20continue to monitor ongoing patients who remain either on treatment or in follow-up on Study 202HALO-202 under a revised clinical protocol. We and the data monitoring committee for Study 202 continue to closely monitor the occurrence of thromboembolic events in enrolled patients after the protocol amendments. The continuation of Study 202 may be halted again if the protocol changes do not result in a reduction of thromboembolic events in accordance with event rate rules established in the protocol, or for other safety reasons.
If our contract manufacturers are unable to manufacture and supply to us bulk rHuPH20 or other raw materials in the quantity and quality required by us or our collaborators for use in our products and product candidates, our product development and commercialization efforts could be delayed or stopped and our collaborations could be damaged.
We have existing supply agreements with contract manufacturing organizations Avid Bioservices, Inc. (Avid) and Catalent Indiana LLC (formerly Cook Pharmica LLC (Cook)LLC) (Catalent) to produce bulk rHuPH20. These manufacturers each produce bulk rHuPH20 under current cGMP for clinical uses. Avid and CookCatalent currently produceproduces bulk rHuPH20 for use in Hylenex recombinant, product candidates and certain other collaboration products and product candidates. Avid currently produces bulk rHuPH20 for use in collaboration products. In addition to supply obligations, Avid and CookCatalent will also provide support for the chemistry, manufacturing and controls sections for FDA and other regulatory filings. We rely on their ability to successfully manufacture these batches according to product specifications. Cook has relatively limited experience manufacturing bulk rHuPH20. In addition, we have been working to scale-up, validate and qualify Cook as a manufacturer of bulk rHuPH20 for use in the product and product candidates under the Roche collaboration. To date, Cook has not been submitted to the European regulatory authorities by Roche as an approved manufacturer for Herceptin SC and MabThera SC. If Cook is unable to obtain its status as an approved European manufacturing facility, or if either Avid or Cook:Catalent: (i) is unable to retain its status as an FDA approved manufacturing facility; (ii) is unable to otherwise successfully scale up bulk rHuPH20 production to meet corporate or regulatory authority quality standards; or (iii) fails to manufacture and supply bulk rHuPH20 in the quantity and quality required by us or our collaborators for use in our proprietary and collaboration products and product candidates for any other reason, our business will be adversely affected. In addition, a significant change in such parties'parties’ or other third party manufacturers’ business or financial condition could adversely affect their abilities to fulfill their contractual obligations to us. We have not established, and may not be able to establish, favorable


arrangements with additional bulk rHuPH20 manufacturers and suppliers of the ingredients necessary to manufacture bulk rHuPH20 should the existing manufacturers and suppliers become unavailable or in the event that our existing manufacturers and suppliers are unable to adequately perform their responsibilities. We have attempted to mitigate the impact of a potential supply interruption through the establishment of excess bulk rHuPH20 inventory where possible, but there can be no assurances that this safety stock will be maintained or that it will be sufficient to address any delays, interruptions or other problems experienced by Avid and/or Cook.Catalent. Any delays, interruptions or other problems regarding the ability of Avid and/or CookCatalent to supply bulk rHuPH20 or the ability of other third party manufacturers, to supply other raw materials or ingredients necessary to produce our products on a timely basis could: (i) cause the delay of clinical trials or otherwise delay or prevent the regulatory approval of proprietary or collaboration product candidates; (ii) delay or prevent the effective commercialization of proprietary or collaboration products; and/or (iii) cause us to breach contractual obligations to deliver bulk rHuPH20 to our collaborators. Such delays would likely damage our relationship with our collaborators, under our key collaboration agreements, and they would have a material adverse effect on royalties and thus our business and financial condition.


14



If we or any party to a key collaboration agreement failsfail to perform material obligations under such agreement, or if a key collaboration agreement, is terminated for any reason, our business could significantly suffer.
We have entered into multiple collaboration agreements under which we may receive significant future payments in the form of milestone payments, target designation fees, maintenance fees and royalties. We are dependent on our collaborators to develop and commercialize product candidates subject to our collaborations in order for us to realize any financial benefits from these collaborations. Our collaborators may not devote the attention and resources to such efforts that we would to such efforts ourselves, change their clinical development plans, promotional efforts or simultaneously develop and commercialize products in competition to those products we have licensed to them. Any of these actions could not be visible to us immediately and could negatively impact the benefits and revenue we receive from such collaboration. In addition, in the event that a party fails to perform under a key collaboration agreement, or if a key collaboration agreement is terminated, the reduction in anticipated revenues could delay or suspend our product development activities for some of our product candidates, as well as our commercialization efforts for some or all of our products. Specifically, the termination of a key collaboration agreement by one of our collaborators could materially impact our ability to enter into additional collaboration agreements with new collaborators on favorable terms, if at all. In certain circumstances, the termination of a key collaboration agreement would require us to revise our corporate strategy going forward and reevaluate the applications and value of our technology.
Most of our current proprietary and collaboration products and product candidates rely on the rHuPH20 enzyme, and any adverse development regarding rHuPH20 could substantially impact multiple areas of our business, including current and potential collaborations, as well as proprietary programs.
rHuPH20 is a key technological component of Enhanze technologyENHANZE Technology and our most advanced proprietary and collaboration products and product candidates, including the current and future products and product candidates under our Roche, Pfizer, Janssen and BaxterENHANZE collaborations, our PEGPH20 program, our ultrafast insulin program, and Hylenex recombinant. If there is an adverse development for rHuPH20 (e.g., an adverse regulatory determination relating to rHuPH20, if we are unable to obtain sufficient quantities of rHuPH20, if we are unable to obtain or maintain material proprietary rights to rHuPH20 or if we discover negative characteristics of rHuPH20), multiple areas of our business, including current and potential collaborations, as well as proprietary programs would be substantially impacted. For example, elevated anti-rHuPH20 antibody titers were detected in the registration trial for Baxter'sBaxalta’s HYQVIA product as well as in a former collaborator’s product in a Phase 2 clinical trial with rHuPH20, but have not been associated, in either case, with any adverse events. We monitor for antibodies to rHuPH20 in our collaboration and proprietary programs, and although we do not believe at this time that the incidence of non-neutralizing anti-rHuPH20 antibodies in either the HYQVIA program or the former collaborator’s program will have a significant impact on our other proprietary and other collaboration product candidates, there can be no assurance that there will not be other such occurrences in the foregoing programs or our other programs or that concerns regarding these antibodies will not also be raised by the FDA or other health authorities in the future, which could result in delays or discontinuations of our development or commercialization activities or deter entry into additional collaborations with third parties.


We routinely evaluate, and may modify, our business strategy and our strategic focus to only a few fields or applications of our technology which may increase or decrease the risk for potential negative impact offrom adverse developments.
We routinely evaluate our business strategy, and may modify this strategy in the future in light of our assessment of unmet medical needs, growth potential, resource requirements, regulatory issues, competition, risks and other factors. As a result of these strategic evaluations, we may focus our resources and efforts on one or a few programs or fields and may suspend or reduce our efforts on other programs and fields. For example, in the third quarter of 2014, we decided to focus our resources on advancing PEGPH20 and expanding utilization of our Enhanze platform.ENHANZE Technology. While we believe these are applications with the greatest potential value, we have reduced the diversification of our programs and increased our dependence on the success of the areas we are pursuing. By focusing on one or a few areas, we increase the potential impact on us if one of those programs or product candidates does not successfully complete clinical trials, achievesachieve commercial acceptance or meetsmeet expectations regarding sales and revenue. Our decision to focus on one or a few programs may also reduce the value of programs that are no longer within our principal strategic focus, which could impair our ability to pursue collaborations or other strategic alternatives for those programs we are not pursuing.


15



Our proprietary and collaboration product candidates or companion diagnostic assays may not receive regulatory approvals or their development may be delayed for a variety of reasons, including delayed or unsuccessful clinical trials, regulatory requirements or safety concerns.
Clinical testing of pharmaceutical products is a long, expensive and uncertain process, and the failure or delay of a clinical trial can occur at any stage, including the patient enrollment stage. Even if initial results of preclinical and nonclinical studies or clinical trial results are promising, we or our collaborators may obtain different results in subsequent trials or studies that fail to show the desired levels of safety and efficacy, or we may not, or our collaborators may not, obtain applicable regulatory approval for a variety of other reasons. Preclinical, nonclinical, and clinical trials for any of our proprietary or collaboration product candidates or development of any collaboration companion diagnostic assays could be unsuccessful, which would delay or preclude regulatory approval and commercialization of the product candidates.candidates or companion diagnostic assays. In the U.S. and other jurisdictions, regulatory approval can be delayed, limited or not granted for many reasons, including, among others:
during the course of clinical studies, the final data may differ from initial reported data, and clinical results may not meet prescribed endpoints for the studies or otherwise provide sufficient data to support the efficacy of our product candidates;
clinical and nonclinical test results may reveal side effects, adverse events or unexpected safety issues associated with the use of our product candidates; for example, in April 2014, a clinical hold was placed on patient enrollment and dosing of PEGPH20 in Study 202HALO-202 as a result of a possible difference in the thromboembolicTE event rate that had been observed at that time in the trial between the group of patients treated with PEGPH20 versus the group of patients treated without PEGPH20. The clinical hold was lifted by the FDA in June 2014, and we have resumedcompleted enrollment and dosing of PEGPH20continue to monitor patients who remain either on treatment or in follow-up on Study 202HALO-202 under a revised clinical protocol;
completion of clinical trials may be delayed for a variety of reasons including the amount of time it may take to identify and enroll patients with high levels of HA in our target population, and the ability to procure drug supply required in clinical trial protocols;
clinical trial results may be negatively impacted if our companion diagnostic does not accurately identify patients most likely to respond to the therapy, including the level of HA in patients;
third parties, such as contract research organizations, upon whom we rely to help conduct and manage our clinical trials may not perform satisfactorily, fulfill their contractual obligations to us, meet expected deadlines or conduct our clinical trials in accordance with regulatory requirements or our stated protocols;
regulatory review may not find a product candidate safe or effective enough to merit either continued testing or final approval;
regulatory review may not find that the data from preclinical testing and clinical trials justifies approval;
regulatory authorities may require that we change our studies or conduct additional studies which may significantly delay or make continued pursuit of approval commercially unattractive; for example, we are currently in dialog but do not have clarity from the FDA regarding the data that we will need for a label change for Hylenex recombinant to be used in CSII;


a regulatory agency may reject our trial data or disagree with our interpretations of either clinical trial data or applicable regulations;
a regulatory agency may approve only a narrow use of our product or may require additional safety monitoring and reporting through Risk Evaluation and Mitigation Strategies (REMS) or conditions to assure safe use program;programs;
the cost of clinical trials required for product approval may be greater than what we originally anticipate, and we may decide to not pursue regulatory approval for such a product;
a regulatory agency may not approve our manufacturing processes or facilities, or the processes or facilities of our collaborators, our contract manufacturers or our raw material suppliers;
a regulatory agency may identify problems or other deficiencies in our existing manufacturing processes or facilities, or the existing processes or facilities of our collaborators, our contract manufacturers or our raw material suppliers;
a regulatory agency may change its formal or informal approval requirements and policies, act contrary to previous guidance, adopt new regulations or raise new issues or concerns late in the approval process; or
a product candidate may be approved only for indications that are narrow or under conditions that place the product at a competitive disadvantage, which may limit the sales and marketing activities for such product candidate or otherwise adversely impact the commercial potential of a product.
If a proprietary or collaboration product candidate or companion diagnostic assay is not approved in a timely fashion or obtained on commercially viable terms, or if development of any product candidate or a companion diagnostic assay is terminated due to difficulties or delays encountered in the regulatory approval process, it could have a material adverse impact on our business, and we would become more dependent on the development of other proprietary or collaboration product candidates and/or our ability to successfully acquire other products and technologies. There can be no assurances that any proprietary or collaboration product candidate or companion diagnostic assay will receive regulatory approval in a timely manner, or at all. For example, we are in dialog but have not reached agreement with the FDA regarding the requirements for updating the Hylenex recombinant label for use in CSII. There can be no assurance that we will be able to gain clarity as to the FDA’s requirements or that the requirements may be satisfied in a commercially feasible way, in which case our ability to enter into collaborations with third parties or explore other strategic alternatives to exploit this opportunity will be limited or may not be possible.


16



We anticipate that certain proprietary and collaboration products will be marketed, and perhaps manufactured, in foreign countries. The process of obtaining regulatory approvals in foreign countries is subject to delay and failure for the reasons set forth above, as well as for reasons that vary from jurisdiction to jurisdiction. The approval process varies among countries and jurisdictions and can involve additional testing. The time required to obtain approval may differ from that required to obtain FDA approval. Foreign regulatory agencies may not provide approvals on a timely basis, if at all. Approval by the FDA does not ensure approval by regulatory authorities in other countries or jurisdictions, and approval by one foreign regulatory authority does not ensure approval by regulatory authorities in other foreign countries or jurisdictions or by the FDA.
Our third party collaborators are responsible for providing certain proprietary materials that are essential components of our collaboration products and product candidates, and any failure to supply these materials could delay the development and commercialization efforts for these collaboration products and product candidates and/or damage our collaborations.
Our development and commercialization collaborators are responsible for providing certain proprietary materials that are essential components of our collaboration products and product candidates. For example, Roche is responsible for producing the Herceptin and MabThera required for its subcutaneous products and BaxterBaxalta is responsible for producing the GAMMAGARD LIQUID for its product HYQVIA. If a collaborator, or any applicable third party service provider of a collaborator, encounters difficulties in the manufacture, storage, delivery, fill, finish or packaging of the collaboration product or product candidate or component of such product or product candidate, such difficulties could (i) cause the delay of clinical trials or otherwise delay or prevent the regulatory approval of collaboration product candidates; and/or (ii) delay or prevent the effective commercialization of collaboration products. Such delays could have a material adverse effect on our business and financial condition.


We rely on third parties to manufacture, prepare, fill, finish and package our products and product candidates, and if such third parties should fail to perform, our commercialization and development efforts for our products and product candidates could be delayed or stopped.
We rely on third parties to store and ship bulk rHuPH20 on our behalf and to alsomanufacture, prepare, fill, finish, package, store and packageship our products and product candidates prior to their distribution.on our behalf. If we are unable to locate third parties to perform these functions on terms that are acceptable to us, or if the third parties we identify fail to perform their obligations, the progress of clinical trials could be delayed or even suspended and the commercialization of approved product candidates could be delayed or prevented. In addition, we are in the early stages of scaling up our manufacturing of PEGPH20 with a third party suppliers to support additional clinical trials, including a possible registration-enablingthe Phase 3 trial, and ultimately, if approved, potential commercial supply. If our contract manufacturer ismanufacturers are unable to successfully manufacture and supply PEGPH20, the progress of our clinical trials could be delayed or halted for a period of time.
If we are unable to sufficiently develop our sales, marketing and distribution capabilities or enter into successful agreements with third parties to perform these functions, we will not be able to fully commercialize our products.
We may not be successful in marketing and promoting our approved product, Hylenex recombinant, or any other products we develop or acquire in the future. Our sales, marketing and distribution capabilities are very limited. In order to commercialize any products successfully, we must internally develop substantial sales, marketing and distribution capabilities or establish collaborations or other arrangements with third parties to perform these services. We do not have extensive experience in these areas, and we may not be able to establish adequate in-house sales, marketing and distribution capabilities or engage and effectively manage relationships with third parties to perform any or all of such services. To the extent that we enter into co-promotion or other licensing arrangements, our product revenues are likely to be lower than if we directly marketed and sold our products, and any revenues we receive will depend upon the efforts of third parties, whose efforts may not meet our expectations or be successful. These third parties would be largely responsible for the speed and scope of sales and marketing efforts, and may not dedicate the resources necessary to maximize product opportunities. Our ability to cause these third parties to increase the speed and scope of their efforts may also be limited. In addition, sales and marketing efforts could be negatively impacted by the delay or failure to obtain additional supportive clinical trial data for our products. In some cases, third party collaborators are responsible for conducting these additional clinical trials, and our ability to increase the efforts and resources allocated to these trials may be limited. For example, in January 2011, we and Baxter mutually agreed to terminate the collaboration agreement for the marketing rights of Hylenex recombinant and the associated agreements.


17



If we or our collaborators fail to comply with regulatory requirements applicable to promotion, sale and manufacturing of approved products, regulatory agencies may take action against us or them, which could significantly harm our business.
Any approved products, along with the manufacturing processes, post-approval clinical data, labeling, advertising and promotional activities for these products, are subject to continual requirements and review by the FDA, state and foreign regulatory bodies. Regulatory authorities subject a marketed product, its manufacturer and the manufacturing facilities to continual review and periodic inspections. We, our collaborators and our respective contractors, suppliers and vendors, will be subject to ongoing regulatory requirements, including complying with regulations and laws regarding advertising, promotion and sales of drug products, required submissions of safety and other post-market information and reports, registration requirements, cGMP regulations (including requirements relating to quality control and quality assurance, as well as the corresponding maintenance of records and documentation), and the requirements regarding the distribution of samples to physicians and recordkeeping requirements. Regulatory agencies may change existing requirements or adopt new requirements or policies. We, our collaborators and our respective contractors, suppliers and vendors, may be slow to adapt or may not be able to adapt to these changes or new requirements.
In particular, regulatory requirements applicable to pharmaceutical products make the substitution of suppliers and manufacturers costly and time consuming. We have minimal internal manufacturing capabilities and are, and expect to be in the future, entirely dependent on contract manufacturers and suppliers for the manufacture of our products and for their active and other ingredients. The disqualification of these manufacturers and suppliers through their failure to comply with regulatory requirements could negatively impact our business because the delays and costs in obtaining and qualifying alternate suppliers (if such alternative suppliers are available, which we cannot assure) could delay clinical trials or otherwise inhibit our ability to bring approved products to market, which would have a material adverse effect on our business and financial condition. Likewise, if we, our collaborators and our respective contractors, suppliers and vendors involved in sales and promotion of our products do


not comply with applicable laws and regulations, for example off-label or false or misleading promotion, this could materially harm our business and financial condition.
Failure to comply with regulatory requirements may result in any of the following:
restrictions on our products or manufacturing processes;
warning letters;
withdrawal of the products from the market;
voluntary or mandatory recall;
fines;
suspension or withdrawal of regulatory approvals;
suspension or termination of any of our ongoing clinical trials;
refusal to permit the import or export of our products;
refusal to approve pending applications or supplements to approved applications that we submit;
product seizure;
injunctions; or
imposition of civil or criminal penalties.
We may wish to raise additional capital in the future and there can be no assurance that we will be able to obtain such funds.
We may wish to raise additional capital in the future to continue the development of our product candidates or for other current corporate purposes. Our current cash reserves and expected revenues during the next few years may not be sufficient for us to continue the development of our proprietary product candidates, to fund general operations and conduct our business at the level desired. In addition, if we engage in acquisitions of companies, products or technologies in order to execute our business strategy, we may need to raise additional capital. We may raise additional capital in the future through one or more financing vehicles that may be available to us including (i) the public or private issuance of securities; (ii) new collaborative agreements; and/or (iii) expansions or revisions to existing collaborative relationships.
In view of our stage of development, business prospects, the nature of our capital structure and general market conditions, if we are required to raise additional capital in the future, the additional financing may not be available on favorable terms, or at all. If additional capital is not available on favorable terms when needed, we will be required to raise capital on adverse terms or


18



significantly reduce operating expenses through the restructuring of our operations. If we raise additional capital, a substantial number of additional shares may be issued, and these shares will dilute the ownership interest of our current investors.
We currently have significant debt and failure by us to fulfill our obligations under the applicable loan agreements may cause the repayment obligations to accelerate.
In December 2013,2015, our subsidiaries, Halozyme, Inc. (Halozyme) and Halozyme Royalty LLC (Halozyme Royalty) entered into a credit agreement (the Credit Agreement) with BioPharma Credit Investments IV Sub, LP and Athyrium Opportunities II Acquisition LP (the Royalty-backed Lenders) pursuant to which we borrowed $150 million through Halozyme Royalty (the Royalty-backed Loan). The Royalty-backed Loan will be repaid primarily from a specified percentage of the royalty payments we receive under our collaboration agreements with Roche and Baxalta (the Royalty Payments).
The obligations of Halozyme Royalty under the Credit Agreement to repay the Royalty-backed Loan may be accelerated upon the occurrence of certain events of default under the Credit Agreement, including but not limited to:
if any payment of principal is not made within three days of when such payment is due and payable or otherwise made in accordance with the terms of the Credit Agreement;
if any representations or warranties made in the Credit Agreement or any other transaction document proves to be incorrect or misleading in any material respect when made;
if there occurs a default in the performance of affirmative and negative covenants set forth in the Credit Agreement or any other transaction document;
the failure by either Baxalta or Roche to pay material amounts owed under our collaboration agreements because of an actual breach or default by us under the collaboration agreements;
the voluntary or involuntary commencement of bankruptcy proceedings by either Halozyme or Halozyme Royalty and other insolvency related defaults;
any materially adverse effect on the binding nature of any of the transaction documents or the collaboration agreements with Baxalta and Roche; or
Halozyme ceases to own, of record and beneficially, 100% of the equity interests in Halozyme Royalty.
The Credit Agreement also contains covenants applicable to Halozyme and Halozyme Royalty, including certain visitation, information and audits rights granted to the collateral agent and the lenders and restrictions on the conduct of business, including continued compliance with the Baxalta and Roche collaboration agreements and specified affirmative actions regarding the escrow account established to facilitate payment of Royalty Payments to the Royalty-backed Lenders or other specified parties. The Credit Agreement also contains covenants solely applicable to Halozyme Royalty, including restrictions on incurring indebtedness, creating or granting liens, making acquisitions and making specified restricted payments. These covenants could make it more difficult for us to execute our business strategy.


In connection with the Royalty-backed Loan, Halozyme Royalty granted a first priority lien and security interest (subject only to permitted liens) in all of its assets and all real, intangible and personal property, including all of its right, title and interest in and to the Royalty Payments.
In June 2016, we entered into an Amended and Restateda Loan and Security Agreement (the Loan Agreement) with Oxford Finance LLC (Oxford) and Silicon Valley Bank (SVB) (collectively, the Lenders), amendingproviding a senior secured loan facility of up to an aggregate principal amount of $70 million, comprising a $55.0 million draw in June 2016 and restating in its entiretyan additional $15.0 million tranche, which we had the option to draw during the second quarter of 2017 and did not exercise. The initial proceeds were partially used to pay the outstanding principal and final payment owed on our originalprevious loan agreement with the Lenders, dated December 2012.Lenders. The Loan Agreement provided for an additional $20 million principal amount of new term loan, bringing the total term loan balance to $50 million. Theremaining proceeds are to be used for working capital and general business requirements. In January 2015, we entered into the Second Amendment to the Amended and RestatedThe Loan and Security Agreement and First Amendment to Disbursement Letter (the “Amendment”) with the Lenders, amending and restating the loan payment schedules of the Amended and Restated Loan and Security Agreement. The amended and restated term loan repayment schedule provides for interest only payments through January 2016, followed by consecutive equal monthly payments of principal and interest in arrears starting in February 2016 and continuing through the previously established maturity date of January 2018. The amended and restated term loan facility is secured by substantially all of the assets of the Company and its subsidiary, Halozyme, Inc., except that the collateral does not include any equity interests in Halozyme, Inc., any intellectual property (including all licensing, collaboration and similar agreements relating thereto), and certain other excluded assets. The Loan Agreement contains customary representations, warranties and covenants by us, which covenants limit our ability to convey, sell, lease, transfer, assign or otherwise dispose of certain of our assets; engage in any business other than the businesses currently engaged in by us or reasonably related thereto; liquidate or dissolve; make certain management changes; undergo certain change of control events; create, incur, assume, or be liable with respect to certain indebtedness; grant certain liens; pay dividends and make certain other restricted payments; make certain investments; make payments on any subordinated debt; and enter into transactions with any of our affiliates outside of the ordinary course of business or permit our subsidiaries to do the same. In addition, subject to certain exceptions, we are required to maintain with SVB our primary deposit accounts, securities accounts and commodities, and to do the same for our domestic subsidiary. Complying with these covenants may make it more difficult for us to successfully execute our business strategy.
The Loan Agreement also contains customary indemnification obligations and customary events of default, including, among other things, our failure to fulfill certain of our obligations under the Loan Agreement and the occurrence of a material adverse change which is defined as a material adverse change in our business, operations or condition (financial or otherwise), a material impairment of the prospect of repayment of any portion of the loan, or a material impairment in the perfection or priority of lender’sthe Lender’s lien in the collateral or in the value of such collateral.
Our ability to make payments on our debt will depend on our future operating performance and ability to generate cash and may also depend on our ability to obtain additional debt or equity financing. We will need to use cash to pay principal and interest on our debt, thereby reducing the funds available to fund our research and development programs, strategic initiatives and working capital requirements. If we are unable to generate sufficient cash to service our debt obligation, an event of default may occur. In the event of default by us under the Credit Agreement or the Loan Agreement, the Lenderslenders would be entitled to exercise their remedies thereunder, including the right to accelerate the debt, upon which we may be required to repay all amounts then outstanding under the Credit Agreement or the Loan Agreement which could harm our financial condition.
If proprietary or collaboration product candidates are approved for marketing but do not gain market acceptance, our business may suffer and we may not be able to fund future operations.
Assuming that our proprietary or collaboration product candidates obtain the necessary regulatory approvals for commercial sale, a number of factors may affect the market acceptance of these existing product candidates or any other products which are developed or acquired in the future, including, among others:
the price of products relative to other therapies for the same or similar treatments;
the perception by patients, physicians and other members of the health care community of the effectiveness and safety of these products for their prescribed treatments relative to other therapies for the same or similar treatments;
our ability to fund our sales and marketing efforts and the ability and willingness of our collaborators to fund sales and marketing efforts;
the degree to which the use of these products is restricted by the approved product label;
the effectiveness of our sales and marketing efforts and the effectiveness of the sales and marketing efforts of our collaborators;
the introduction of generic competitors; and


the extent to which reimbursement for our products and related treatments will be available from third party payors including government insurance programs (Medicare and Medicaid) and private insurers.


19



If these products do not gain market acceptance, we may not be able to fund future operations, including the development or acquisition of new product candidates and/or our sales and marketing efforts for our approved products, which would cause our business to suffer.
In addition, our proprietary and collaboration product candidates will be restricted to the labels approved by FDA and applicable regulatory bodies, and these restrictions may limit the marketing and promotion of the ultimate products. If the approved labels are restrictive, the sales and marketing efforts for these products may be negatively affected.
Developing and marketing pharmaceutical products for human use involves significant product liability risks for which we currently have limited insurance coverage.
The testing, marketing and sale of pharmaceutical products involves the risk of product liability claims by consumers and other third parties. Although we maintain product liability insurance coverage, product liability claims can be high in the pharmaceutical industry, and our insurance may not sufficiently cover our actual liabilities. If product liability claims were to be made against us, it is possible that the liabilities may exceed the limits of our insurance policy, or our insurance carriers may deny, or attempt to deny, coverage in certain instances. If a lawsuit against us is successful, then the lack or insufficiency of insurance coverage could materially and adversely affect our business and financial condition. Furthermore, various distributors of pharmaceutical products require minimum product liability insurance coverage before purchase or acceptance of products for distribution. Failure to satisfy these insurance requirements could impede our ability to achieve broad distribution of our proposed products, and higher insurance requirements could impose additional costs on us. In addition, since many of our collaboration product candidates include the pharmaceutical products of a third party, we run the risk that problems with the third party pharmaceutical product will give rise to liability claims against us.
Our inability to attract, hire and retain key management and scientific personnel could negatively affect our business.
Our success depends on the performance of key management and scientific employees with relevant experience. For example, in order to pursue our current business strategy, we will need to recruit and retain personnel experienced in oncology drug development which is a highly competitive market for talent. We depend substantially on our ability to hire, train, motivate and retain high quality personnel, especially our scientists and management team. Particularly in view of the small number of employees on our staff to cover our numerous programs and key functions, if we are unable to retain existing personnel or identify or hire additional personnel, we may not be able to research, develop, commercialize or market our products and product candidates as expected or on a timely basis and we may not be able to adequately support current and future alliances with strategic collaborators. Our use of domestic and international third-party contractors, consultants and staffing agencies also subjects us to potential co-employment liability claims.
Furthermore, if we were to lose key management personnel, we would likely lose some portion of our institutional knowledge and technical know-how, potentially causing a substantial delay in one or more of our development programs until adequate replacement personnel could be hired and trained. We currently have a severance policy applicable to all employees and a change in control policy applicable to senior executives. Other than for vesting provisions in equity based awards, we have not adopted any other policies or entered into any other agreements specifically designed to motivate officers or other employees to remain with us.
We do not have key man life insurance policies on the lives of any of our employees.


Our operations might be interrupted by the occurrence of a natural disaster or other catastrophic event.
Our operations, including laboratories, offices and other research facilities, are located in four buildings in San Diego, California. In addition, we have a satellite office in South San Francisco, California. We depend on our facilities and on our collaborators, contractors and vendors for the continued operation of our business. Natural disasters or other catastrophic events, interruptions in the supply of natural resources, political and governmental changes, wildfires and other fires, floods, explosions, actions of animal rights activists, earthquakes and civil unrest could disrupt our operations or those of our collaborators, contractors and vendors. Even though we believe we carry commercially reasonable business interruption and liability insurance, and our contractors may carry liability insurance that protect us in certain events, we may suffer losses as a result of business interruptions that exceed the coverage available under our and our contractors'contractors’ insurance policies or for which we or our contractors do not have coverage. Any natural disaster or catastrophic event could have a significant negative impact on our operations and financial results. Moreover, any such event could delay our research and development programs.


20



If we or our collaborators do not achieve projected development, clinical, regulatory or sales goals in the timeframes we publicly announce or otherwise expect, the commercialization of our products and the development of our product candidates may be delayed and, as a result, our stock price may decline, and we may face lawsuits relating to such declines.
From time to time, we or our collaborators may publicly articulate the estimated timing for the accomplishment of certain scientific, clinical, regulatory and other product development goals. The accomplishment of any goal is typically based on numerous assumptions, and the achievement of a particular goal may be delayed for any number of reasons both within and outside of our control. If scientific, regulatory, strategic or other factors cause us to not meet a goal, regardless of whether that goal has been publicly articulated or not, our stock price may decline rapidly. For example, the announcement in April 2014 of the temporary halting of our Phase 2 clinical trial for PEGPH20 caused a rapid decline in our stock price. Stock price declines may also trigger direct or derivative shareholder lawsuits. As with any litigation proceeding, the eventual outcome of any legal action is difficult to predict. If any such lawsuits occur, we will incur expenses in connection with the defense of these lawsuits, and we may have to pay substantial damages or settlement costs in connection with any resolution thereof. Although we have insurance coverage against which we may claim recovery against some of these expenses and costs, the amount of coverage may not be adequate to cover the full amount or certain expenses and costs may be outside the scope of the policies we maintain. In the event of an adverse outcome or outcomes, our business could be materially harmed from depletion of cash resources, negative impact on our reputation, or restrictions or changes to our governance or other processes that may result from any final disposition of the lawsuit. Moreover, responding to and defending pending litigation significantly diverts management'smanagement’s attention from our operations.
In addition, the consistent failure to meet publicly announced milestones may erode the credibility of our management team with respect to future milestone estimates.
Future acquisitions could disrupt our business and harm our financial condition.
In order to augment our product pipeline or otherwise strengthen our business, we may decide to acquire additional businesses, products and technologies. As we have limited experience in evaluating and completing acquisitions, our ability as an organization to make such acquisitions is unproven. Acquisitions could require significant capital infusions and could involve many risks, including, but not limited to, the following:
we may have to issue convertible debt or equity securities to complete an acquisition, which would dilute our stockholders and could adversely affect the market price of our common stock;
an acquisition may negatively impact our results of operations because it may require us to amortize or write down amounts related to goodwill and other intangible assets, or incur or assume substantial debt or liabilities, or it may cause adverse tax consequences, substantial depreciation or deferred compensation charges;
we may encounter difficulties in assimilating and integrating the business, products, technologies, personnel or operations of companies that we acquire;
certain acquisitions may impact our relationship with existing or potential collaborators who are competitive with the acquired business, products or technologies;
acquisitions may require significant capital infusions and the acquired businesses, products or technologies may not generate sufficient value to justify acquisition costs;


we may take on liabilities from the acquired company such as debt, legal liabilities or business risk which could be significant;
an acquisition may disrupt our ongoing business, divert resources, increase our expenses and distract our management;
acquisitions may involve the entry into a geographic or business market in which we have little or no prior experience; and
key personnel of an acquired company may decide not to work for us.
If any of these risks occurred, it could adversely affect our business, financial condition and operating results. There is no assurance that we will be able to identify or consummate any future acquisitions on acceptable terms, or at all. If we do pursue any acquisitions, it is possible that we may not realize the anticipated benefits from such acquisitions or that the market will not view such acquisitions positively.


21



Security breaches may disrupt our operations and harm our operating results.
The wrongful use, theft, deliberate sabotage or any other type of security breach with respect to any of our or any of our vendors and partners’ information technology storage and access systems could result in the disruption of our ability to use such systems or disclosure or dissemination of our proprietary and confidential information that is electronically stored, including research or clinical data, resulting in a material adverse impact on our business, operating results and financial condition. Our security and data recovery measures may not be adequate to protect against computer viruses, break-ins, and similar disruptions from unauthorized tampering with our electronic storage systems. Furthermore, any physical break-in or trespass of our facilities could result in the misappropriation, theft, sabotage or any other type of security breach with respect to our proprietary and confidential information, including research or clinical data or damage to our research and development equipment and assets. Such adverse effects could be material and irrevocable to our business, operating results and financial condition.
Risks Related To Ownership of Our Common Stock
Our stock price is subject to significant volatility.
We participate in a highly dynamic industry which often results in significant volatility in the market price of common stock irrespective of company performance. As a result, ourThe high and low sales prices of our common stock during the twelve months ended December 31, 20142017 were $18.18$21.13 and $6.88,$9.68, respectively. We expect our stock price to continue to be subject to significant volatility and, in addition to the other risks and uncertainties described elsewhere in this Annual Report on Form 10-K and all other risks and uncertainties that are either not known to us at this time or which we deem to be immaterial, any of the following factors may lead to a significant drop in our stock price:
the presence of competitive products to those being developed by us;
failure (actual or perceived) of our collaborators to devote attention or resources to the development or commercialization of product candidates licensed to such collaborator;
a dispute regarding our failure, or the failure of one of our third party collaborators, to comply with the terms of a collaboration agreement;
the termination, for any reason, of any of our collaboration agreements;
the sale of common stock by any significant stockholder, including, but not limited to, direct or indirect sales by members of management or our Board of Directors;
the resignation, or other departure, of members of management or our Board of Directors;
general negative conditions in the healthcare industry;
general negative conditions in the financial markets;
the failure, for any reason, to obtaincost associated with obtaining regulatory approval for any of our proprietary or collaboration product candidates;
the failure, for any reason, to secure or defend our intellectual property position;
for those products that are not yet approved for commercial sale, the failure or delay of applicable regulatory bodies to approve such products;
identification of safety or tolerability issues;
failure of clinical trials to meet efficacy endpoints;
suspensions or delays in the conduct of clinical trials or securing of regulatory approvals;


adverse regulatory action with respect to our and our collaborators’ products and product candidates such as clinical holds, imposition of onerous requirements for approval or product recalls;
our failure, or the failure of our third party collaborators, to successfully commercialize products approved by applicable regulatory bodies such as the FDA;
our failure, or the failure of our third party collaborators, to generate product revenues anticipated by investors;
disruptions in our clinical or commercial supply chains, including disruptions caused by problems with a bulk rHuPH20 contract manufacturer or a fill and finish manufacturer for any product or product candidate;
the sale of additional debt and/or equity securities by us;
our failure to obtain financing on acceptable terms;terms or at all; or
a restructuring of our operations.


22



Future transactions where we raise capital may negatively affect our stock price.
We are currently a “Well-Known Seasoned Issuer” and may file automatic shelf registration statements at any time with the SEC. In addition,February 2017, we currently have the ability to offer and sell additional equity, debt securities and warrants to purchase such securities, either individually or in units, underfiled an effective automatic shelf registration statement.statement on Form S-3 (Registration No. 333-216315) with the SEC. Sales of substantial amounts of shares of our common stock or other securities under our current or future shelf registration statements could lower the market price of our common stock and impair our ability to raise capital through the sale of equity securities. In the future, we may issue additional options, warrants or other derivative securities convertible into our common stock.
Our rights agreement and anti-takeoverAnti-takeover provisions in our charter documents and Delaware law may make an acquisition of us more difficult.
We are party to a Rights Agreement designed to deter abusive takeover tactics and to encourage prospective acquirers to negotiate with our board of directors rather than attempt to acquire us in a manner or on terms that our board deems unacceptable, which could delay or discourage takeover attempts that stockholders may consider favorable.
In addition, anti-takeoverAnti-takeover provisions in our charter documents and Delaware law may make an acquisition of us more difficult. First, our board of directors is classified into three classes of directors. Under Delaware law, directors of a corporation with a classified board may be removed only for cause unless the corporation'scorporation’s certificate of incorporation provides otherwise. Our amended and restated certificate of incorporation, as amended, does not provide otherwise. In addition, our bylaws limit who may call special meetings of stockholders, permitting only stockholders holding at least 50% of our outstanding shares to call a special meeting of stockholders. Our amended and restated certificate of incorporation, as amended, does not include a provision for cumulative voting for directors. Under cumulative voting, a minority stockholder holding a sufficient percentage of a class of shares may be able to ensure the election of one or more directors. Finally, our bylaws establish procedures, including advance notice procedures, with regard to the nomination of candidates for election as directors and stockholder proposals.
These provisions may discourage potential takeover attempts, discourage bids for our common stock at a premium over market price or adversely affect the market price of, and the voting and other rights of the holders of, our common stock. These provisions could also discourage proxy contests and make it more difficult for stockholders to elect directors other than the candidates nominated by our board of directors.
In addition, because we are incorporated in Delaware, we are governed by the provisions of Section 203 of the Delaware General Corporation Law, which may prohibit large stockholders from consummating a merger with, or acquisition of, us.
These provisions may deter an acquisition of us that might otherwise be attractive to stockholders.


Risks Related to Our Industry
Our products must receive regulatory approval before they can be sold, and compliance with the extensive government regulations is expensive and time consuming and may result in the delay or cancellation of product sales, introductions or modifications.
Extensive industry regulation has had, and will continue to have, a significant impact on our business. All pharmaceutical companies, including ours, are subject to extensive, complex, costly and evolving regulation by the health regulatory agencies including the FDA (and with respect to controlled drug substances, the U.S. Drug Enforcement Administration (DEA)) and equivalent foreign regulatory agencies and state and local/regional government agencies. The Federal Food, Drug and Cosmetic Act, the Controlled Substances Act and other domestic and foreign statutes and regulations govern or influence the testing, manufacturing, packaging, labeling, storing, recordkeeping, safety, approval, advertising, promotion, sale and distribution of our products. We are dependent on receiving FDA and other governmental approvals, including regulatory approvals in jurisdictions outside the United States, prior to manufacturing, marketing and shipping our products. Consequently, there is always a risk that the FDA or other applicable governmental authorities, including those outside the United States, will not approve our products or may impose onerous, costly and time-consuming requirements such as additional clinical or animal testing. Regulatory authorities may require that we change our studies or conduct additional studies, which may significantly delay or make continued pursuit of approval commercially unattractive. We are currently in dialog but do not have clarity fromFor example, the approval of Baxalta’s HYQVIA BLA was delayed by the FDA until we and Baxalta provided additional preclinical data sufficient to address concerns regarding non-neutralizing antibodies to rHuPH20 that were detected in the path for a labeling update to include key efficacyregistration trial. Although these antibodies have not been associated with any known adverse clinical effects, and safety data prior to initiating Hylenex recombinant for usethe HYQVIA BLA was approved by the FDA in CSII. There can be no assurance that we will be able to gain clarity as toSeptember 2014, the FDA's requirements. The FDA or other foreign regulatory agency may,


23



at any time, halt our and our collaborators'collaborators’ development and commercialization activities due to safety concerns. In addition, even if our products are approved, regulatory agencies may also take post-approval action limiting or revoking our ability to sell our products. Any of these regulatory actions may adversely affect the economic benefit we may derive from our products and therefore harm our financial condition.
Under certain of these regulations, we and our contract suppliers and manufacturers are subject to periodic inspection of our or their respective facilities, procedures and operations and/or the testing of products by the FDA, the DEA and other authorities, which conduct periodic inspections to confirm that we and our contract suppliers and manufacturers are in compliance with all applicable regulations. The FDA also conducts pre-approval and post-approval reviews and plant inspections to determine whether our systems, or our contract suppliers'suppliers’ and manufacturers'manufacturers’ processes, are in compliance with cGMP and other FDA regulations. If we, or our contract supplier, fail these inspections, we may not be able to commercialize our product in a timely manner without incurring significant additional costs, or at all.
In addition, the FDA imposes a number of complex regulatory requirements on entities that advertise and promote pharmaceuticals including, but not limited to, standards and regulations for direct-to-consumer advertising, off-label promotion, industry-sponsored scientific and educational activities, and promotional activities involving the internet.
We may be subject, directly or indirectly, to various broad federal and state healthcare laws. If we are unable to comply, or have not fully complied, with such laws, we could face civil, criminal and administrative penalties, damages, monetary fines, disgorgement, possible exclusion from participation in Medicare, Medicaid and other federal healthcare programs, contractual damages, reputational harm, diminished profits and future earnings and curtailment or restructuring of our operations, any of which could adversely affect our ability to operate.
Our business operations and activities may be directly, or indirectly, subject to various broad federal and state healthcare laws, including without limitation, anti-kickback laws, the Foreign Corrupt Practices Act, false claims laws, civil monetary penalty laws, data privacy and security laws, tracing and tracking laws, as well as transparency laws regarding payments or other items of value provided to healthcare providers. These laws may restrict or prohibit a wide range of business activities, including, but not limited to, research, manufacturing, distribution, pricing, discounting, marketing and promotion and other business arrangements. These laws may impact, among other things, our current activities with principal investigators and research subjects, as well as sales, marketing and education programs. Many states have similar healthcare fraud and abuse laws, some of which may be broader in scope and may not be limited to items or services for which payment is made by a government health care program.


Efforts to ensure that our business arrangements will comply with applicable healthcare laws may involve substantial costs. While we have adopted a healthcare corporate compliance program, it is possible that governmental and enforcement authorities will conclude that our business practices may not comply with current or future statutes, regulations or case law interpreting applicable fraud and abuse or other healthcare laws. If our operations or activities are found to be in violation of any of the laws described above or any other governmental regulations that apply to us, we may be subject to, without limitation, civil, criminal and administrative penalties, damages, monetary fines, disgorgement, possible exclusion from participation in Medicare, Medicaid and other federal healthcare programs, contractual damages, reputational harm, diminished profits and future earnings and curtailment or restructuring of our operations, any of which could adversely affect our ability to operate.
In addition, any sales of products outside the U.S. will also likely subject us to foreign equivalents of the healthcare laws mentioned above, among other foreign laws.
We may be required to initiate or defend against legal proceedings related to intellectual property rights, which may result in substantial expense, delay and/or cessation of the development and commercialization of our products.
We primarily rely on patents to protect our intellectual property rights. The strength of this protection, however, is uncertain. For example, it is not certain that:
we will be able to obtain patent protection for our products and technologies;
the scope of any of our issued patents will be sufficient to provide commercially significant exclusivity for our products and technologies;
others will not independently develop similar or alternative technologies or duplicate our technologies and obtain patent protection before we do; and


24



any of our issued patents, or patent pending applications that result in issued patents, will be held valid, enforceable and infringed in the event the patents are asserted against others.
We currently own or license several patents and also have pending patent applications applicable to rHuPH20 and other proprietary materials. There can be no assurance that our existing patents, or any patents issued to us as a result of our pending patent applications, will provide a basis for commercially viable products, will provide us with any competitive advantages, or will not face third party challenges or be the subject of further proceedings limiting their scope or enforceability. Any weaknesses or limitations in our patent portfolio could have a material adverse effect on our business and financial condition. In addition, if any of our pending patent applications do not result in issued patents, or result in issued patents with narrow or limited claims, this could result in us having no or limited protection against generic or biosimilar competition against our product candidates which would have a material adverse effect on our business and financial condition.
We may become involved in interference proceedings in the U.S. Patent and Trademark Office, or other proceedings in other jurisdictions, to determine the priority, validity or enforceability of our patents. In addition, costly litigation could be necessary to protect our patent position.
We also rely on trademarks to protect the names of our products (e.g. Hylenex recombinant). We may not be able to obtain trademark protection for any proposed product names we select. In addition, product names for pharmaceutical products must be approved by health regulatory authorities such as the FDA in addition to meeting the legal standards required for trademark protection and product names we propose may not be timely approved by regulatory agencies which may delay product launch. In addition, our trademarks may be challenged by others. If we enforce our trademarks against third parties, such enforcement proceedings may be expensive.
We also rely on trade secrets, unpatented proprietary know-how and continuing technological innovation that we seek to protect with confidentiality agreements with employees, consultants and others with whom we discuss our business. Disputes may arise concerning the ownership of intellectual property or the applicability or enforceability of these agreements, and we might not be able to resolve these disputes in our favor.


In addition to protecting our own intellectual property rights, third parties may assert patent, trademark or copyright infringement or other intellectual property claims against us. If we become involved in any intellectual property litigation, we may be required to pay substantial damages, including but not limited to treble damages, attorneys'attorneys’ fees and costs, for past infringement if it is ultimately determined that our products infringe a third party'sparty’s intellectual property rights. Even if infringement claims against us are without merit, defending a lawsuit takes significant time, may be expensive and may divert management'smanagement’s attention from other business concerns. Further, we may be stopped from developing, manufacturing or selling our products until we obtain a license from the owner of the relevant technology or other intellectual property rights. If such a license is available at all, it may require us to pay substantial royalties or other fees.
Patent protection for protein-based therapeutic products and other biotechnology inventions is subject to a great deal of uncertainty, and if patent laws or the interpretation of patent laws changes,change, our competitors may be able to develop and commercialize products based on our discoveries.
Patent protection for protein-based therapeutic products is highly uncertain and involves complex legal and factual questions. In recent years, there have been significant changes in patent law, including the legal standards that govern the scope of protein and biotechnology patents. Standards for patentability of full-length and partial genes, and their corresponding proteins, are changing. Recent court decisions have made it more difficult to obtain patents, by making it more difficult to satisfy the patentable subject matter requirement and the requirement of non-obviousness, have decreased the availability of injunctions against infringers, and have increased the likelihood of challenging the validity of a patent through a declaratory judgment action. Taken together, these decisions could make it more difficult and costly for us to obtain, license and enforce our patents. In addition, the Leahy-Smith America Invents Act (HR 1249) was signed into law in September 2011, which among other changes to the U.S. patent laws, changes patent priority from “first to invent” to “first to file,” implements a post-grant opposition system for patents and provides for a prior user defense to infringement. These judicial and legislative changes have introduced significant uncertainty in the patent law landscape and may potentially negatively impact our ability to procure, maintain and enforce patents to provide exclusivity for our products.


25



There also have been, and continue to be, policy discussions concerning the scope of patent protection awarded to biotechnology inventions. Social and political opposition to biotechnology patents may lead to narrower patent protection within the biotechnology industry. Social and political opposition to patents on genes and proteins and recent court decisions concerning patentability of isolated genes may lead to narrower patent protection, or narrower claim interpretation, for isolated genes, their corresponding proteins and inventions related to their use, formulation and manufacture. Patent protection relating to biotechnology products is also subject to a great deal of uncertainty outside the U.S., and patent laws are evolving and undergoing revision in many countries. Changes in, or different interpretations of, patent laws worldwide may result in our inability to obtain or enforce patents, and may allow others to use our discoveries to develop and commercialize competitive products, which would impair our business.
If third party reimbursement and customer contracts are not available, our products may not be accepted in the market.
Our ability to earn sufficient returns on our products will depend in part on the extent to which reimbursement for our products and related treatments will be available from government health administration authorities, private health insurers, managed care organizations and other healthcare providers.
Third-party payors are increasingly attempting to limit both the coverage and the level of reimbursement of new drug products to contain costs. Consequently, significant uncertainty exists as to the reimbursement status of newly approved healthcare products. Third party payors may not establish adequate levels of reimbursement for the products that we commercialize, which could limit their market acceptance and result in a material adverse effect on our revenues and financial condition.
Customer contracts, such as with group purchasing organizations and hospital formularies, will often not offer contract or formulary status without either the lowest price or substantial proven clinical differentiation. If, our products arefor example, Hylenex is compared to animal-derived hyaluronidases by these entities, it is possible that neither of these conditions will be met, which could limit market acceptance and result in a material adverse effect on our revenues and financial condition.


The rising cost of healthcare and related pharmaceutical product pricing has led to cost containment pressures that could cause us to sell our products at lower prices, resulting in less revenue to us.
Any of the proprietary or collaboration products that have been, or in the future are, approved by the FDA may be purchased or reimbursed by state and federal government authorities, private health insurers and other organizations, such as health maintenance organizations and managed care organizations. Such third party payors increasingly challenge pharmaceutical product pricing. The trend toward managed healthcare in the U.S., the growth of such organizations, and various legislative proposals and enactments to reform healthcare and government insurance programs, including the Medicare Prescription Drug Modernization Act of 2003, could significantly influence the manner in which pharmaceutical products are prescribed and purchased, resulting in lower prices and/or a reduction in demand. Such cost containment measures and healthcare reforms could adversely affect our ability to sell our products.
In March 2010, the U.S. adopted the Patient Protection and Affordable Care Act, as amended by the Health Care and Education Reconciliation Act (the Healthcare Reform Act)PPACA). This law substantially changes the way healthcare is financed by both governmental and private insurers, and significantly impacts the pharmaceutical industry. The Healthcare Reform ActPPACA contains a number of provisions that are expected to impact our business and operations, in some cases in ways we cannot currently predict. Changes that may affect our business include those governing enrollment in federal healthcare programs, reimbursement changes, fraud and abuse and enforcement. These changes will impact existing government healthcare programs and will result in the development of new programs, including Medicare payment for performance initiatives and improvements to the physician quality reporting system and feedback program.
Additional provisions of the Healthcare Reform ActPPACA may negatively affect our revenues in the future. For example, the Healthcare Reform ActPPACA imposes a non-deductible excise tax on pharmaceutical manufacturers or importers that sell branded prescription drugs to U.S. government programs that we believe will impact our revenues from our products. In addition, as part of the Healthcare Reform Act'sPPACA’s provisions closing a funding gap that currently exists in the Medicare Part D prescription drug program, we will also be required to provide a 50% discount on branded prescription drugs dispensed to beneficiaries under this prescription drug program. Recently, Congress and the current administration have proposed and taken various steps to revise, repeal or delay implementation of, various aspects of the Healthcare Reform Act. We expect that the Healthcare Reform ActPPACA, as it may be amended, and other healthcare reform measures that may be adopted in the future could have a material adverse effect on our industry generally and on our ability to maintain or increase our product sales or successfully commercialize our product candidates orand could limit or eliminate our future spending on development projects.


26



Furthermore, individual states have become increasingly aggressive in passing legislation and implementing regulations designed to control pharmaceutical product pricing, including price or patient reimbursement constraints, discounts, restrictions on certain product access, importation from other countries and bulk purchasing. Legally mandated price controls on payment amounts by third party payors or other restrictions could negatively and materially impact our revenues and financial condition. We anticipate that we will encounter similar regulatory and legislative issues in most other countries outside the U.S.
We face intense competition and rapid technological change that could result in the development of products by others that are superior to our proprietary and collaboration products under development.
Our proprietary and collaboration products have numerous competitors in the U.S. and abroad including, among others, major pharmaceutical and specialized biotechnology firms, universities and other research institutions that have developed competing products. The competitors for Hylenex recombinant include, but are not limited to, Valeant Pharmaceuticals International, Inc.’s FDA-approved product, Vitrase®, an ovine (ram) hyaluronidase, and Amphastar Pharmaceuticals, Inc. For our ultrafast insulin’s product, candidates, such competitors may include Biodel Inc.Amphadase®, Eli Lily, Sanofi Aventis, Novo Nordisk Inc. and Mannkind Corporation.a bovine (bull) hyaluronidase. For our PEGPH20 product candidate, such competitors may include major pharmaceutical and specialized biotechnology firms. These competitors may develop technologies and products that are more effective, safer, or less costly than our current or future proprietary and collaboration product candidates or that could render our technologies and product candidates obsolete or noncompetitive. Many of these competitors have substantially more resources and product development, manufacturing and marketing experience and capabilities than we do. In addition, many of our competitors have significantly greater experience than we do in undertaking preclinical testing and clinical trials of pharmaceutical product candidates and obtaining FDA and other regulatory approvals of products and therapies for use in healthcare.


Item 1B.Unresolved Staff Comments
None.
Item 2.Properties
Our administrative offices and research facilities are currently located in San Diego, California. We lease an aggregate of approximately 76,000 square feet of office and research space for a monthly rent expensespace. In addition, we have an office in South San Francisco, California, where we lease approximately 10,000 square feet of approximately $145,000, net of costs and property taxes associated with the operation and maintenance of the subleased facilities.office space. We believe the current space isour facilities are adequate for our immediate needs.current and near-term needs, and, if necessary, we will be able to locate additional facilities as needed.
Item 3.Legal Proceedings
From time to time, we may be involved in disputes, including litigation, relating to claims arising out of operations in the normal course of our business. Any of these claims could subject us to costly legal expenses and, while we generally believe that we have adequate insurance to cover many different types of liabilities, our insurance carriers may deny coverage or our policy limits may be inadequate to fully satisfy any damage awards or settlements. If this were to happen, the payment of any such awards could have a material adverse effect on our consolidated results of operations and financial position. Additionally, any such claims, whether or not successful, could damage our reputation and business. We currently are not a party to any legal proceedings, the adverse outcome of which, in management’s opinion, individually or in the aggregate, would have a material adverse effect on our consolidated results of operations or financial position.
Item 4.Mine Safety Disclosures
Not applicable.


27



PART II
Item 5.Market for Registrant’s Common Equity, Related Stockholder Matters and Issuer Purchases of Equity Securities
Market Information
Our common stock is listed on the NASDAQ Global Select Market under the symbol “HALO.” The following table sets forth the high and low sales prices per share of our common stock during each quarter of the two most recent fiscal years:
 2014 2013 2017 2016
 High Low High Low High Low High Low
First Quarter 
$18.18
 
$11.28
 
$8.59
 
$5.14
 
$15.20
 
$9.68
 
$17.51
 
$6.96
Second Quarter 
$12.97
 
$6.88
 
$8.49
 
$5.03
 
$15.05
 
$11.51
 
$12.33
 
$7.70
Third Quarter 
$10.70
 
$8.58
 
$12.15
 
$6.51
 
$17.62
 
$11.41
 
$12.75
 
$8.43
Fourth Quarter 
$10.00
 
$7.51
 
$16.36
 
$9.33
 
$21.13
 
$16.58
 
$14.38
 
$8.18
On February 24, 2015,13, 2018, the closing sales price of our common stock on the NASDAQ Global Select Market was $15.00$17.84 per share. As of February 24, 2015,13, 2018, we had approximately 17,00021,200 stockholders of record.record and beneficial owners of our common stock.
Dividends
We have never declared or paid any dividends on our common stock. We currently intend to retain available cash for funding operations; therefore, we do not expect to pay any dividends on our common stock in the foreseeable future. In addition, the provisions of our Loan Agreementborrowing arrangements limit, among other things, our ability to pay dividends and make certain other payments. Any future determination to pay dividends on our common stock will be at the discretion of our board of directors and will depend upon, among other factors, our results of operations, financial condition, capital requirements, contract restrictions, business prospects and other factors our board of directors may deem relevant.


28



Stock Performance Graph and Cumulative Total Return
Notwithstanding any statement to the contrary in any of our previous or future filings with the SEC, the following information relating to the price performance of our common stock shall not be deemed to be “filed” with the SEC or to be “soliciting material” under the Securities Exchange Act of 1934, as amended, or the Exchange Act, and it shall not be deemed to be incorporated by reference into any of our filings under the Securities Act of 1933, as amended, or the Exchange Act, except to the extent we specifically incorporate it by reference into such filing.
The graph below compares Halozyme Therapeutics, Inc.’s cumulative five-year total shareholder return on common stock with the cumulative total returns of the NASDAQ Composite Index and the NASDAQ Biotechnology Index. The graph tracks the performance of a $100 investment in our common stock and in each of the indexes (with the reinvestment of all dividends) from December 31, 20092012 to December 31, 2014. In addition, we have provided supplemental information showing the comparative returns for an additional period through February 24, 2015.2017. The historical stock price performance included in this graph is not necessarily indicative of future stock price performance.
12/31/200912/31/201012/31/201112/31/201212/31/201312/31/20142/24/201512/31/201212/31/201312/31/201412/31/201512/31/201612/31/2017
Halozyme Therapeutics, Inc.$100$135$162$114$255$164$256$100$223$144$258$147$302
NASDAQ Composite$100$118$117$137$193$221$232$100$140$161$172$187$243
NASDAQ Biotechnology$100$115$129$171$284$381$421$100$166$223$249$196$239


29



Item 6.Selected Financial Data
The selected consolidated financial data set forth below as of December 31, 20142017 and 2013,2016, and for the fiscal years ended December 31, 2014, 20132017, 2016 and 2012,2015, are derived from our audited consolidated financial statements included elsewhere in this report. This information should be read in conjunction with those consolidated financial statements, the notes thereto, and with “Management’s Discussion and Analysis of Financial Condition and Results of Operations.” The selected consolidated financial data set forth below as of December 31, 2012, 20112015, 2014 and 2010,2013, and for the fiscal years ended December 31, 20112014 and 2010,2013, are derived from our audited consolidated financial statements that are contained in reports previously filed with the SEC, not included herein.
Summary Financial Information
  Year Ended December 31,
Statement of Operations Data: 2017 2016 2015 2014 2013
  (in thousands, except for per share amounts)
Total revenues $316,613
 $146,691
 $135,057
 $75,334
 $54,799
Net income (loss) $62,971
 $(103,023) $(32,231) $(68,375) $(83,479)
Net income (loss) per share, basic $0.46
 $(0.81) $(0.25) $(0.56) $(0.74)
Net income (loss) per share, diluted $0.45
 $(0.81) $(0.25) $(0.56) $(0.74)
Shares used in computing net income (loss) per share, basic 136,419
 127,964
 126,704
 122,690
 112,805
Shares used in computing net income (loss) per share, diluted 139,068
 127,964
 126,704
 122,690
 112,805
  Year Ended December 31,
Statement of Operations Data: 
2014(1)
 
2013(2)
 
2012(3)
 
2011(4)
 2010
  (in thousands, except for per share amounts)
Total revenues $75,334
 $54,799
 $42,325
 $56,086
 $13,624
Net loss $(68,375) $(83,479) $(53,552) $(19,770) $(53,242)
Net loss per share, basic and diluted $(0.56) $(0.74) $(0.48) $(0.19) $(0.56)
Shares used in computing net loss per share, basic and diluted 122,690
 112,805
 111,077
 102,566
 94,358
 
 As of December 31, As of December 31,
Balance Sheet Data: 2014 2013 2012 2011 2010 2017 2016 2015 2014 2013
 (in thousands) (in thousands)
Cash and cash equivalents and available-for-sale marketable securities $135,623
 $71,503
 $99,501
 $52,376
 $82,756
 $469,214
 $204,981
 $108,339
 $135,623
 $71,503
Working capital $136,990
 $70,293
 $111,682
 $46,236
 $73,655
 $379,044
 $201,947
 $109,315
 $136,990
 $70,293
Total assets $165,977
 $101,793
 $134,728
 $65,759
 $91,345
 $519,945
 $261,515
 $181,789
 $165,977
 $101,793
Deferred revenue $54,634
 $53,143
 $43,846
 $40,884
 $58,094
 $60,865
 $44,618
 $53,223
 $54,634
 $53,143
Long-term debt, net $49,860
 $49,772
 $29,662
 
 
 $125,140
 $199,228
 $27,971
 $49,860
 $49,772
Total liabilities $124,625
 $121,783
 $85,875
 $54,858
 $70,994
 $311,579
 $293,996
 $138,790
 $124,625
 $121,783
Stockholders’ equity (deficit) $41,352
 $(19,991) $48,854
 $10,900
 $20,351
 $208,366
 $(32,481) $42,999
 $41,352
 $(19,991)
______________ 
(1)Revenues in 2014 included a $15.0 million license fee from the Janssen Collaboration.
(2)
Revenues in 2013 reflected increases in supply of bulk rHuPH20 to Roche and product sales of Hylenex recombinant, which was relaunched in December 2011.
(3)Revenues in 2012 included $9.5 million in license fees from the Pfizer Collaboration.
(4)
Revenues in 2011 included $18.0 million in license fees from collaboration agreements with ViroPharma Incorporated and Intrexon Corporation and $18.1 million related to recognition of unamortized deferred prepaid product-based payments and unamortized deferred upfront payment in connection with the termination of the collaboration with Baxter for the marketing rights of Hylenex recombinant in July 2011.


30



Item 7.
Management’s Discussion and Analysis of Financial Condition and Results of Operation
In addition to historical information, the following discussion contains forward-looking statements that are subject to risks and uncertainties. Actual results may differ substantially from those referred to herein due to a number of factors, including but not limited to risks described in the Part I, Item 1A, Risks Factors, and elsewhere in this Annual Report. References to “Notes” are Notes included in our Notes to Consolidated Financial Statements.
Overview
Halozyme is seeking to translate our unique knowledge of the tumor microenvironment to create novel therapies that can improve cancer survival. Our research focuses on human enzymes that alter the extracellular matrix and tumor microenvironment. The extracellular matrixTherapeutics, Inc. is a complex matrix of proteinsbiotechnology company focused on developing and carbohydrates surrounding the cell that provides structural support in tissues and orchestrates many important biological activities, including cell migration, signaling and survival. Over many years, we have developed unique technology and scientific expertise enabling us to pursue this target-rich environment for the development ofcommercializing novel oncology therapies.
Our proprietary enzymes can beare used to facilitate the delivery of injected drugs and fluids, potentially enhancing the efficacy and the convenience of other drugs or can be used to alter abnormal tissue structures for potential clinical benefit. We have chosen to exploit our technology and expertise inusing a balanced waytwo pillar strategy that we believe enables us to modulate bothmanage risk and spendcost by: (1) developing our own proprietary products in therapeutic areas with significant unmet medical needs, with a focus on oncology, and (2) licensing our technology to biopharmaceutical companies to collaboratively develop products whichthat combine our technology with the collaborators'collaborators’ proprietary compounds.
The majority of our approved product and product candidates are based on rHuPH20, our patented recombinant human hyaluronidase enzyme. rHuPH20 temporarily breaks down hyaluronic acid (HA), a naturally occurring substance that is a major component of the extracellular matrix in tissues throughout the body such as skin and cartilage. We believe this temporary degradation creates an opportunistic window for the improved subcutaneous delivery of injectable biologics, such as monoclonal antibodies and other large therapeutic molecules, as well as small molecules and fluids. We refer to the application of rHuPH20 to facilitate the delivery of other drugs or fluids as Enhanze technology. rHuPH20 is also the active ingredient in our first commercially approved product, Hylenex® recombinant.
Our proprietary development pipeline consists primarily of pre-clinical and clinical stage product candidates in oncology and diabetes.oncology. Our lead oncology program is PEGPH20 (PEGylated recombinant human hyaluronidase), a new molecular entity under developmentwe are developing in combination with currently approved cancer therapies as a candidate for the systemic treatment of tumors that accumulate HA. WhenWe have demonstrated that when HA accumulates in a tumor, it can cause higher pressure in the tumor, reducing blood flow into the tumor and with that, reduced access of cancer therapies to the tumor. PEGPH20 works by temporarily degrading HA surrounding some cancer cellsThrough our efforts and results in reduced pressureefforts of our partners and increased blood flow to the cancer with increased amounts of anticancer treatments administered concomitantly gaining access to the tumor. Wecollaborators, we are currently in Phase 3 clinical testing for PEGPH20 with ABRAXANE® (nab-paclitaxel) and gemcitabine in stage IV pancreatic ductal adenocarcinoma (“PDA”) (HALO 109-301), in Phase 1b clinical testing for PEGPH20 with KEYTRUDA® (pembrolizumab) in non-small cell lung cancer and gastric cancer (HALO 107-101), in Phase 1b/2 clinical testing for PEGPH20 with HALAVEN® (eribulin) in patients treated with up to two lines of prior therapy for HER2-negative metastatic pancreaticbreast cancer, (Study 109-202) , and we have recently initiated ain Phase 1b/2 clinical trial in non-small cell lung cancer (Study 107-201). We have also been investigating testing for PEGPH20 with TecentriqHylene®x recombinant for use as pre-treatment (atezolizumab) in patients with Type 1 diabetes using pumps and recombinant human cathepsin L (HTI-501) for the treatment of cellulite.
Our recent receipt of Fast Track and Orphan Drug designationspreviously treated metastatic PDA, in Phase 1b/2 clinical testing for PEGPH20 new pre-clinical data further supporting the pan-tumor potentialwith Tecentriq in patients with gastric cancer and in Phase 1b/2 clinical testing for PEGPH20 with Tecentriq in patients with cholangiocarcinoma and investigator interest in both pancreatic and lunggall bladder cancer trials have confirmed PEGPH20(HALO 110-101/MATRIX).
We refer to the application of rHuPH20 to facilitate the delivery of other drugs or fluids as our priority product candidate for investment. As a result of ongoing evaluationsENHANZE® Technology. We license the ENHANZE Technology to confirm and focus on the highest value opportunities, we have made the decision to seekform collaborations with third partiesbiopharmaceutical companies that develop or explore other strategic alternatives in order to exploit our diabetes and dermatology programs.
Regarding Enhanze, wemarket drugs requiring or benefiting from injection via the subcutaneous route of administration. We currently have ENHANZE collaborations with F. Hoffmann-La Roche, Ltd. and Hoffmann-La Roche, Inc. (Roche), Baxter Healthcare Corporation (Baxter)Baxalta US Inc. and Baxalta GmbH (Baxalta Incorporated was acquired by Shire plc in June 2016) (Baxalta), Pfizer Inc. (Pfizer) and, Janssen Biotech, Inc. (Janssen), withAbbVie, Inc. (AbbVie), Eli Lilly and Company (Lilly), Bristol-Myers Squibb Company (BMS) and Alexion Pharma Holding (Alexion).We receive royalties from two of these collaborations, including royalties from sales of one product approved in the U.S. and three products approved for marketing in Europe from which we are receiving royalties and several others at various stages of development.


31



Our operations to date have involved: (i) building infrastructure for and staffing our operations; (ii) acquiring, developing and securing proprietary protection for our technology; (iii) developing our proprietary product pipeline; (iv) entering into and supporting our collaborations with other companies to advance licensed product candidates; and (v) selling our own approved commercial product, Hylenex recombinant. Currently, we have received only limited revenue from the sales of Hylenex recombinant, in addition to other revenuesBaxalta collaboration and two products from our collaborations.
the Roche collaboration. Future potential revenues from the sales and/or royalties of our approved products, product candidates, which have not been approved or have recently been approvedand ENHANZE collaborations will depend on the ability of Halozyme and our collaborators to develop, manufacture, secure and maintain regulatory approvals for approved products and product candidates and commercialize the product candidates. We have incurred net operating losses each year since inception, with an accumulated deficit of approximately $450.4 million as of December 31, 2014.


Our 20142017 and recent key accomplishments and events are as follows:
ENHANZE collaborations
In January 2015, we disclosed initial efficacy and safety data from an interim assessment of Stage 1 of Study 109-202,2018, Roche initiated a Phase 2 multicenter, randomized clinical trial evaluating PEGPH20 as1 study for an unnamed target with the ENHANZE Technology, triggering a first-line therapy for patients with stage IV metastatic pancreatic cancer. Refer to PEGPH20 section under Part I, Item 1, Business, for further discussion of Study 109-202 data. We also presented the final results from Study 109-201, a multi-center, international open label dose escalation Phase 1b clinical study of PEGPH20 in combination with gemcitabine for the treatment of patients with stage IV metastatic pancreatic cancer at the 2015 Gastrointestinal Cancers Symposium (also known as ASCO-GI meeting). Refer to PEGPH20 section under Part I, Item 1, Business, for further discussion of Study 109-202 data.
$1.0 million milestone payment.
In December 2014,2017, we and JanssenAlexion entered into a collaboration and license agreement, under which Alexion has the worldwide license to develop and commercialize product combining our rHuPH20 enzyme with up to four targets from Alexion’s portfolio of products for an upfront payment of $40.0 million. Targets may be selected on an exclusive basis. Alexion elected two targets on an exclusive basis, including a C5 complement inhibitor, and has an option to select two additional targets within five years from the effective date of the agreement.
During the fourth quarter of 2017, Baxalta and Roche achieved sales milestones for commercial products using the ENHANZE Technology, triggering $5.0 million and $7.0 million in milestone payments, respectively.
During the fourth quarter of 2017, Janssen initiated the first of four currently active Phase 3 studies of daratumumab combined with the ENHANZE Technology in amyloidosis patients, multiple myeloma patients and smoldering myeloma patients, triggering a $15.0 million milestone payment.
In September 2017, we entered into a collaboration and license agreement with BMS, under which BMS has the worldwide license to develop and commercialize products combining our Enhanze technologyENHANZE Technology with Janssen proprietary biologicsBMS immuno-oncology targets directed toat up to five targets. Targets may be selected oneleven targets for an exclusive or non-exclusive basis. We received $15 million for the licenseupfront payment of one specified exclusive target$105.0 million. BMS has designated multiple immuno-oncology targets including programmed death 1 (PD-1) and fourhas an option to select additional targets which Janssen haswithin five years from the effective date.
In September 2017, we entered into an agreement with Roche for the right to elect upondevelop and commercialize one additional exclusive target using our ENHANZE Technology for an upfront payment of additional fees. We are also eligible to receive additional payments upon Janssen's achievement of specified development, regulatory and sales-based events, totaling up to $566$30.0 million.
In November 2014, we announced we had completed a corporate strategy review, including a portfolio review, which resulted in a decision to focus our resources on advancing PEGPH20 and to expand utilization of our Enhanze platform. The recent Fast Track and Orphan Drug designations for PEGPH20, new pre-clinical data further supporting the pan-tumor potential for PEGPH20, and investigator interest in both pancreatic and lung cancer trials have confirmed PEGPH20 as our priority product for investment. With respect to our program investigating Hylenex recombinant for use with insulin pumps, we believe that additional clinical data will be required and are continuing to seek clarity with the U.S. Food and Drug Administration (FDA) on what data will be required, if any. Once we receive clarity, we intend to seek to enter into collaborations with third parties or explore other strategic alternatives in order to exploit this opportunity. We completed a reduction in our workforce of approximately 13% to align with these strategic priorities.


32



In October 2014, Baxter announced the launch and first shipments of HYQVIA [Immune Globulin Infusion 10% (Human) with rHuPH20], Baxter's subcutaneous immunoglobulin treatment for adult patients with primary immunodeficiency in the U.S. HYQVIA was approved by the FDA in September 2014, and is the first subcutaneous immune globulin (IG) treatment approved for primary immunodeficiency patients with a dosing regimen requiring only one infusion up to once per month (every three to four weeks) and one injection site per infusion to deliver a full monthly therapeutic dose of IG. The majority of primary immunodeficiency patients today receive intravenous infusions in a doctor’s office or infusion center, and current subcutaneous IG treatments require weekly or bi-weekly treatment with multiple infusion sites per treatment. The FDA's approval of HYQVIA was a significant milestone for us as it represented the first U.S. approved Biologics License Application (BLA) which utilized our rHuPH20 platform.
In July 2014, we resumed enrollment and dosingAugust 2017, Lilly initiated a Phase 1 study of patientsan investigational new therapy in our ongoing Study 109-202 evaluating PEGPH20 in patientscombination with pancreatic cancer under a revised clinical protocol. The study had previously been placed on clinical hold in April 2014 pending review by the independent Data Monitoring Committee (DMC) for the study and by the FDA of a possible difference in the thromboembolic event rate between the patients treated with PEGPH20 versus the patients treated without PEGPH20 in the trial.rHuPH20.
In June 2014, Roche launched2017, the subcutaneous (SC) formulationFDA approved Genentech’s RITUXAN HYCELA, a combination of MabThera® (rituximab) usingrituximab and rHuPH20, (MabThera SC) in Europe for the treatmentCLL and two types of patients with common forms of non-HodgkinNHL, follicular lymphoma (NHL). Roche received European marketing approval in March 2014. The first commercial launch of MabThera SC triggered a $5 million milestone payment under the License and Collaboration Agreement between Halozyme and Roche. Following the launch of Herceptin® SC in September 2013, MabThera SC is the second novel subcutaneous formulation of one of Roche's oncology products using our patented Enhanze technology to be launched in Europe.diffuse large B-cell lymphoma.
In March 2014, Roche received European marketing approval for Roche's SC formation of MabThera(rituximab) for the treatment of patients with common forms of non-Hodgkin lymphoma (NHL). This is the second European approval for a novel subcutaneous formulation of one of Roche's oncology products using our rHuPH20 technology.
Clinical trials
In January 2018, the Phase 1b portion of the study of HALAVEN (eribulin) with PEGPH20 in HER2-negative metastatic breast cancer closed enrollment. As a result of an Eisai portfolio decision, no further clinical development is planned on the Phase 2 portion of the study. Data analysis is ongoing and a submission of the results of this study to a scientific forum is expected in the second half of 2018.
In October 2017, Genentech initiated a Phase 1b/2 clinical trial evaluating PEGPH20 in combination with Tecentriq in patients with gastric cancer.
In October 2017, we initiated the second study in our clinical agreement with Genentech, a Phase 1b/2 open-label randomized study to assess Tecentriq in combination with PEGPH20 and chemotherapy in patients with cholangiocarcinoma and gall bladder cancer.
In July 2017, Genentech initiated a Phase 1b/2 clinical trial evaluating PEGPH20 in combination with Tecentriq in patients with previously treated metastaticPDA.
In June 2017, results from Study HALO-202 were presented at the European Society for Medical Oncology (ESMO) World Congress of Gastrointestinal Cancer and the Annual Meeting of the American Society of Clinical Oncology (ASCO). The presentations expanded on the topline results announced in January 2017 with additional data from the study as of December 2016.


In April 2017, we presented at the annual meeting of the American Association of Cancer Research (AACR) that, in preclinical models, PEGPH20 increases the number of cancer-fighting white blood cells accumulating in the tumor and the effectiveness of immunotherapies, which builds upon prior preclinical findings and continues to support the potential benefits of remodeling the tumor microenvironment.
In March 2017, SWOG, an independent network of researchers that design and conduct cancer clinical trials, stopped enrollment in a Phase 1b/2 trial evaluating PEGPH20 plus modified FOLFIRINOX chemotherapy versus modified FOLFIRINOX alone in patients with previously untreated metastatic pancreas cancer. SWOG’s independent Data Monitoring Committee found, based on preliminary data, that the addition of PEGPH20 given every two weeks to modified FOLFIRINOX would be unlikely to demonstrate a statistically significant improvement in the primary endpoint of overall survival. SWOG further reported that a higher rate of death was observed in the PEGPH20 arm versus modified FOLFIRINOX alone. In January 2018, SWOG presented final overall survival (OS) and progression-free survival (PFS) data from the study at the ASCO-GI conference, which was consistent with the preliminary data findings. Our PEGPH20 studies and clinical collaborations in combination with agents other than modified FOLFIRINOX continue unchanged.
In January 2017, we announced topline results from the combined analysis of Stage 1 and Stage 2, and Stage 2 alone, of the Study 109-202, based on a December 2016 data cutoff. Among the findings, the overall study population showed a statistically significant increase in PFS in the 84 total HA-High patients treated with PEGPH20 plus ABRAXANE and gemcitabine when compared to HA-High patients receiving ABRAXANE and gemcitabine alone. Stage 2 of the study, which completed enrollment in February 2016, showed a 91 percent improvement in median PFS for HA-High patients in the PEGPH20 arm, 8.6 months compared to 4.5 months in the control arm, and achieved its primary endpoint to evaluate and demonstrate a reduction in the rate of TE events in the PEGPH20 arm.
Financing
In May 2017, we completed an underwritten public offering pursuant to which we sold 11.5 million shares of common stock, including 1.5 million shares sold pursuant to the full exercise of an option to purchase additional shares granted to the underwriters. All of the shares were offered at a public offering price of $12.50 per share, generating approximately $134.9 million in net proceeds, after deducting underwriting discounts and commissions and other offering expenses. We intend to use the net proceeds from this offering to fund continued development of our PEGPH20 oncology program and for other general corporate purposes.
Results of Operations
Comparison of Years Ended December 31, 2014, 20132017, 2016 and 20122015
Product Sales, Net Product sales, increasednet were as follows (in thousands):
  2017 Change 2016 Change 2015
Sales of bulk rHuPH20:          
Roche $22,325
 (10)% $24,786
 9% $22,773
Baxalta 11,717
 5 % 11,117
 73% 6,410
Other 1,204
 (10)% 1,332
 73% 772
Sales of Hylenex
 15,150
 (6)% 16,157
 
 16,127
Total product sales, net $50,396
 (6)% $53,392
 16% $46,082


Product sales, net decreased in 20142017 compared to 2013, by $13.4 million, or 55%, primarily2016, mainly due to a $9.8 million increasedecreases in productthe sales of bulk rHuPH20 to Roche and Hylenex, offset by an increase in sales of bulk rHuPH20 to Baxalta. Product sales, net increased in 2016 compared to 2015 due to an increase in the sales of bulk rHuPH20 to Baxalta and Roche. In 2017 and 2016, we performed services for Roche collaboration products andto bring on-line a $4.1 million increasesecond contract manufacturing facility for bulk rHuPH20. This new facility will become the primary source for Roche of bulk rHuPH20 once it receives regulatory approval. As a result, we anticipate Roche will deplete their existing inventory of rHuPH20 ahead of the transition to the new facility, which will result in lower bulk product sales in 2018. We anticipate Baxalta will deplete their existing inventory of rHuPH20 as part of a planned change to a more efficient manufacturing process, which will also result in lower bulk rHuPH20 product sales in 2018. We expect that future product sales of Hylenex recombinant. Hylenex recombinant product sales increased to $13.2 million in 2014 from $9.0 million in 2013. Product sales increased in 2013 compared to 2012 by $21.6 million,be flat or 746%, primarily due to $14.8 million in product sales of bulk rHuPH20experience modest growth, although there may be periods with declining revenue as we experience competition for Roche and Baxter collaboration products, Herceptin SC and HyQvia. The increase was also due to a $6.8 million increase in product sales of Hylenex recombinant. Subsequent to the receipt of the European marketing approval of Roche's Herceptin SC product in August 2013 and MabThera SC product in March 2014 and Baxter's HYQVIA product in May 2013, revenue from bulk rHuPH20 supply for these collaboration products was recorded as product sales revenue, instead of revenues under collaborative agreements.market share.
Royalties Royalty revenue was $9.4$63.5 million in 20142017 compared to $33,000$51.0 million in 2013. Royalties relate primarily to sales of Roche's Herceptin SC.2016 and $31.0 million in 2015. The increase was mainly due to the launchdriven by higher sales of Herceptin SC and MabThera SC (RITUXAN HYCELA™ in September 2013.the U.S.) by Roche and of HYQVIA by Baxalta. We recognize royalties on sales of the collaboration products by the collaborators in the quarter following the quarter in which the corresponding sales occur.occurred. In general, we expect royalty revenue to increase in future periods reflecting expected increases in sales of collaboration products, although there may be periods with flat or declining royalty revenue as sales of products under collaborations vary.


33



Revenues Under Collaborative Agreements Revenues under collaborative agreements for the years ended December 31, 2014, 2013 and 2012 were as follows (in thousands):
Upfront license fees, license fees for the election of additional targets, event-based payments, license maintenance fees and amortization of deferred upfront and other license fees: 2017 Change 2016 Change 2015
         
BMS $101,400
 n/a
 $
 n/a
 $
Alexion 40,000
 n/a
 
 n/a
 
Roche 33,330
 902 % 3,328
 2% 3,269
Janssen 15,000
 5,900 % 250
 n/a
 
Baxalta 810
 6 % 765
 
 765
Lilly 
 (100)% 8,000
 (68)% 25,000
AbbVie 
 (100)% 6,000
 (74)% 23,000
Pfizer 
 (100)% 2,500
 25 % 2,000
 2014 Change 2013 Change 2012 190,540
 814 % 20,843
 (61%) 54,034
Upfront payments, license maintenance fees and amortization of deferred upfront and license fees:          
Reimbursements for research and development services:          
Roche 6,900
 (63)% 18,700
 632% 2,556
Janssen $15,000
 n/a
 $
 
 $
 3,302
 61 % 2,051
 146 % 834
Roche 3,028
 31% 2,308
 14% 2,016
Pfizer 1,000
 (33%) 1,500
 (84%) 9,500
Baxter 765
 27% 604
 25% 483
Other 
 (100%) 2,000
 (18%) 2,429
 19,793
 209% 6,412
 (56%) 14,428
Milestone payments:          
Roche 
 
 
 (100%) 8,000
          
Reimbursements for research and development services and supply of bulk rHuPH20:          
Roche(1)
 6,923
 (64%) 19,086
 115% 8,897
Baxter(1)
 1,209
 (70%) 4,059
 (40%) 6,742
Pfizer 121
 (79%) 589
 n/a
 
Baxalta 1,585
 311 % 386
 32% 292
Other 40
 (78%) 181
 (87%) 1,371
 383
 14 % 335
 18% 284
 8,293
 (65%) 23,915
 41% 17,010
 12,170
 (43)% 21,472
 441% 3,966
Total revenues under collaborative agreements $28,086
 (7%) $30,327
 (23%) $39,438
 $202,710
 379 % $42,315
 (27%) $58,000
_______________
(1)Subsequent to the European approvals of Roche's Herceptin SC product in August 2013 and MabThera SC product in March 2014 and Baxter's HYQVIA product in May 2013, revenue from supply of bulk rHuPH20 for those products to the collaborators was recorded as product sales.
Revenue from license fees increased in 2017, compared to 2016 due to $171.4 million upfront license revenue for the 2017 Roche, BMS and Alexion agreements, and $15.0 million for clinical milestones under the Janssen Collaboration. In 2014,2016, we recognized $15.0$15.5 million in license fee and milestone revenue in connection with the Lilly, AbbVie and Pfizer collaborations. In 2015, we recognized $48.0 million in license fee revenue in connection with the Janssen Collaboration. In 2012, we recognized $9.5 millionLilly and AbbVie collaborations related to upfront payments. Revenue from upfront licenses fees, license fees for the election of additional targets, event-based payments, license maintenance fees and amortization of deferred upfront and other license fees vary from period to period based on our ENHANZE collaboration activity. We expect these revenues to continue to fluctuate in license fee revenue in connection with the Pfizer Collaboration. In 2012, we also recognized $8.0 million upon achievement of certainfuture periods based on our collaborators’ abilities to meet various clinical and regulatory milestones under the Roche Collaboration. set forth in such agreements and our abilities to obtain new collaborative agreements.


Revenue from reimbursements for research and development services, and bulk rHuPh20including clinical supply of rHuPH20, decreased in 20142017 compared to 20132016 mainly due to revenue from supply of bulk rHuPH20 for Roche collaboration products being recognized as product sales revenue in the current period, as opposed to revenue from reimbursements for research and development services in the same period in 2013. The decrease was also due to a decrease in reimbursements forservices provided to Roche related to the validation of a new manufacturing facility, partially offset by an increase in services provided to supportBaxalta and an increase in clinical supply of rHuPH20 provided to Janssen. The validation of the launches bynew Roche facility was completed in the second quarter of 2017 and, Baxter.therefore, we expect to continue to see a decrease in research and development service revenue associated with this project going forward. Revenue from reimbursements for research and development services and bulk rHuPh20 supply increased in 20132016 compared to 20122015 mainly due to services provided for the increase in reimbursementsnew manufacturing facility for manufacturing services to support the launches by Roche. Research and development services rendered by us on behalf of our collaborators are at the request of the collaborators; therefore, the amount of future revenues related to reimbursable research and development services and supply of bulk rHuPH20 is uncertain. We expect the non-reimbursement revenues under our collaborative agreements to continue to fluctuate in future periods based on our collaborators’ abilities to meet various clinical and regulatory milestones set forth in such agreements and our abilities to obtain new collaborative agreements.
Cost of Product Sales Cost of product sales increasedwere $31.2 million in 20142017 compared to 2013, by $16.5$33.2 million or 264%, primarilyin December 31, 2016 and $29.2 million in 2015. The decrease of $2.0 million in cost of product sales in 2017 compared to 2016 was mainly due to the increased producta decrease in sales of bulk rHuPH20 for Herceptin SC. Costto Roche. The increase of $4.0 million in cost of product sales increased in 20132016 compared to 2012, by $5.2 million, or 471%, primarily2015 was due to a $2.8$5.8 million increase in cost of product sales relatedof bulk rHuPH20 due to the increasedan increase in sales to collaboration partners, partially offset by a $1.8 million decrease in Hylenex recombinant product sales and $2.3 million in cost of product sales, related to the product sales of bulk rHuPH20 for Herceptin SC.


34



Prior to European marketing approvals of Roche's collaboration products, Herceptin SC in August 2013 and MabThera SC in March 2014, and Baxter's collaboration HYQVIA product in May 2013, all costs related to the manufacturing of bulk rHuPH20 for these collaboration products were charged to research and development expenses in the periods such costs were incurred. Therefore, cost of product sales of bulk rHuPH20 for these collaboration products in 2013 was materially reduced due to the exclusion of the manufacturing costs that were charged to research and development expenses in the periods prior to receiving marketing approvals.
Cost of product sales of bulk rHuPH20 for collaboration products in 2014 excluded $1.0 milliona decrease in manufacturing costs, of which $0.9 million and $0.1 million were charged to research and development expenses for 2013 and 2012, respectively. Cost of product sales of bulk rHuPH20 for collaboration products in 2013 excluded $10.0 million in manufacturing costs, of which $9.0 million and $1.0 million were charged to research and development expenses in 2013 and 2012, respectively. The estimated selling price of the zero-cost inventory of bulk rHuPH20 for Herceptin SC on hand as of December 31, 2013, was approximately $1.3 million. We sold all of this inventory in 2014. Going forward, the cost of product sales is expected to be approximately 83% of bulk rHuPH20 product sales revenue. There was no HyQvia API inventory on hand as of December 31, 2014 and 2013.costs.
Research and Development Research and development expenses incurred for the years ended December 31, 2014, 2013consist of external costs, salaries and 2012benefits and allocation of facilities and other overhead expenses related to research manufacturing, clinical trials, preclinical and regulatory activities. Research and development expenses incurred were as follows (in thousands):
  2014 Change 2013 Change 2012
Programs          
Product Candidates:          
PEGPH20 $34,857
 86 % $18,742
 50 % $12,479
Ultrafast insulin program 22,424
 (9)% 24,723
 371 % 5,251
Hylenex recombinant
 5,318
 (50)% 10,734
 (8)% 11,682
HTI-501 1,447
 (47)% 2,712
 38 % 1,962
Enhanze collaborations(1)
 6,799
 (78)% 31,104
 19 % 26,152
rHuPH20 platform(2)
 5,807
 (1)% 5,895
 (23)% 7,705
Other 3,044
 12 % 2,730
 (43)% 4,813
Total research and development expenses $79,696
 (18)% $96,640
 38 % $70,044
  2017 Change 2016 Change 2015
Programs          
PEGPH20 $123,932
 15 % $108,102
 43% $75,616
ENHANZE collaborations and rHuPH20 platform 19,197
 (37)% 30,398
 189% 10,514
Other 7,514
 (39)% 12,342
 74% 7,106
Total research and development expenses $150,643
 
 $150,842
 62% $93,236
_______________
(1)Subsequent to the European approvals of Roche's Herceptin SC product in August 2013 and MabThera SC product in March 2014 and Baxter's HYQVIA product in May 2013, the manufacturing costs of bulk rHuPH20 for these collaboration products were capitalized as inventory.
(2)Includes research, development and manufacturing expenses related to our proprietary rHuPH20 enzyme. These expenses were not designated to a specific program at the time the expenses were incurred.
Research and development expenses relating to our PEGPH20 program increased in 2014 increased2017 by 86%,15% compared to 20132016, and increased in 2016 by 43% compared to 2015, primarily due to the increased clinical trial activities mostly relatingactivities. We expect these expenses to Study 109-202. Research andcontinue to increase in future periods reflecting expected increases in our PEGPH20 development expenses relating to Hylenex recombinant program decreased in 2014 by 50% compared to 2013 mainly due to the completion of the technology transfer and validation campaign with a second manufacturer for Hylenex recombinant at the end of 2013. activities.
Research and development expenses relating to our EnhanzeENHANZE collaborations and our rHuPH20 platform in 20142017 decreased by 78%37%, compared to 2016, primarily due to a $12.0 million decrease resulting from capitalizingin manufacturing costs for approved collaboration productsexpenses related to Roche, due to work associated with bringing on-line a second contract manufacturing facility. As we completed the validation of the new manufacturing facility in the current period, an $8.1 millionsecond quarter of 2017, we expect these expenses to continue to decrease in other outsourced regulatorygoing forward. The rHuPH20 platform includes research, development and manufacturing activitiesexpenses related to support Roche andour proprietary rHuPH20 enzyme. These expenses were not designated to a $2.5 million decrease in preclinical activities to support Baxter. Subsequent tospecific program at the European approvals of Roche's Herceptin SC product in August 2013 and MabThera SC product in March 2014 and Baxter's HYQVIA product in May 2013,time the manufacturing costs of bulk rHuPH20 for these collaboration productsexpenses were capitalized as inventory. We expect research and development costs to increase in future periods as we continue with our clinical trial programs and continue to develop and manufacture our product candidates.


35



Research and development expenses increased in 2013 compared to 2012 by $26.6 million, or 38%.incurred. Research and development expenses relating to our ultrafast insulinENHANZE collaborations and PEGPH20 programsour rHuPH20 platform in 2016 increased in 2013by 189% compared to 2012 by $19.5 million, or 371%, and $6.3 million, or 50%, respectively,2015, primarily due to a $17.0 million increase in manufacturing expenses related to bringing on-line the increased clinical trial activities relating to the CONSISTENT 1 and on-going Phase 2 PEGPH20 clinical trials. new Roche manufacturing facility.
Research and development expenses relatingrelated to our Enhanze collaborations increasedother programs in 20132017 decreased by 39% compared to 2012 by $5.0 million, or 19%,2016 primarily due to a $9.8 million increase in manufacturing activities to support Roche's preparation for the launches of its collaboration product and product candidates; offset in part by a $4.6 million decrease in manufacturingpreclinical development of HTI-1511 and PEG-ADA2. Research and development expenses related to support Baxter's launch of its collaboration product.other programs in 2016 increased by 74% compared to 2015, primarily due to expenses incurred in our preclinical product programs.
Selling, General and Administrative Selling, general and administrative (SG&A) expenses increased in 20142017 compared to 20132016 by $3.6$8.0 million, or 11%17%, due to the increase in compensation costs, including a $2.3 million increase in stock-based compensation.
SG&A expensesand increased in 20132016 compared to 20122015 by $7.5$5.8 million, or 30%15%, primarily due to a $3.9 million increaseincreases in compensation costs,expense including a $0.9 millionstock compensation. We expect SG&A expenses to increase moderately in stock-based compensation, mainly resulting from an increase in headcountfuture periods as our operations expand and higher bonus accruals, and a $1.8 million increase in marketing activitieswe prepare for commercial launch.Hylenex recombinant product.


Interest Expense Interest expense included interest expense and amortization of the debt discount related to the long-term debt acquired in December 2012. The increase of $2.3debt. Interest expense increased by $2.0 million in 20142017 compared to 2016, and increased by $14.8 million in 2016 as compared to 2013 was2015, primarily due to interest expense incurred on the $20.0Royalty-backed Loan we received in January 2016.
Income Taxes Income tax benefit was $1.4 million increasein 2017 compared to income tax expense of $1.2 million in 2016 and was primarily comprised of U.S. federal alternative minimum tax expense in the principal balanceamount of $4.1 million offset by a U.S federal alternative minimum tax credit of $5.5 million. The U.S. federal AMT was eliminated via the Tax Cuts and Jobs Act that was enacted on December 22, 2017. The AMT credit carryovers will be used to offset regular tax liability for any taxable year beginning after 2017. If not utilized before 2022, any remaining AMT credit carryforward amount is fully refundable. The AMT credit carryforward of $5.5 million was recognized as a deferred tax asset at December 31, 2017 as realization is certain. For the years ended December 31, 2017 and 2016, we generated taxable income in the U.S., which was partially offset by utilizing net operating losses carried forward from earlier years. No income tax expense was recognized during the year ended December 2013.31, 2015.
Liquidity and Capital Resources
Our principal sources of liquidity are our existing cash, cash equivalents and available-for-sale marketable securities. As of December 31, 2014,2017, we had cash, cash equivalents and marketable securities of approximately $135.6$469.2 million. We will continue to have significant cash requirements to support product development activities. The amount and timing of cash requirements and cash on hand will depend on the progress and success of our clinical development programs, regulatory and market acceptance, and the resources we devote to research and other commercialization activities.activities and the achievement of various milestones and royalties under our existing collaborative agreements.
We believe that our current cash, cash equivalents and marketable securities will be sufficient to fund our operations for at least the next twelve months. We currently anticipate total net cash burn of approximately $35 million to $45 million for the year ending December 31, 2015, depending on the progress of various preclinical and clinical programs, the timing of our manufacturing scale up and the achievement of various milestones and royalties under our existing collaborative agreements. We expect to fund our operations going forward with existing cash resources, anticipated revenues from our existing collaborations and cash that we may raise through future transactions. We may finance futureraise cash needs through any one of the following financing vehicles: (i) the public offering of securities; (ii) new collaborative agreements; (iii) expansions or revisions to existing collaborative relationships; (iv) private financings; and/or (v) other equity or debt financings.financings; and/or (vi) monetizing assets.
In February 2012,2017, we filed an automatic shelf registration statement on Form S-3 (Registration No. 333-179444)333-216315) with the SEC, which allowsallow us, from time to time, to offer and sell equity, debt securities and warrants to purchase any of such securities, either individually or in units. In May 2017, we completed an underwritten public offering pursuant to which we sold 11.5 million shares of common stock, generating $134.9 million in net proceeds, after deducting underwriting discounts and commissions and other offering expenses. We may, in the future, offer and sell additional equity, debt securities and warrants to purchase any of such securities, either individually or in units to raise capital to fund the continued development of our product candidates, the commercialization of our products or for other general corporate purposes.
Our existing cash, cash equivalents and marketable securities may not be adequate to fund our operations until we become cash flow positive,profitable, if ever. We cannot be certain that additional financing will be available when needed or, if available, financing will be obtained on favorable terms. If we are unable to raise sufficient funds, we may need to delay, scale back or eliminate some or all of our research and development programs, delay the launch of our product candidates, if approved, and/or restructure our operations. If we raise additional funds by issuing equity securities, substantial dilution to existing stockholders could result. If we raise additional funds by incurring debt financing, the terms of the debt may involve significant cash payment obligations, the issuance of warrants that may ultimately dilute existing stockholders when exercised and covenants that may restrict our ability to operate our business.


36



Cash Flows
Operating Activities
Net cash provided by operations was $134.1 million in 2017 compared to net cash used in operations of $50.4 million in 2016. The increase in cash provided by operations was mainly due to an increase in operating income driven by license payments and milestones achieved and changes in working capital for the year ended December 31, 2017 compared to the corresponding period in the prior year.


Net cash used in operations was $47.5$50.4 million in 20142016 compared to $49.3$37.1 million of net cash used in operations in 2013.2015. The $1.8$13.3 million decreaseincrease in utilization of cash in operations was mainly due to an increase in operating losses compared to the receipt of a $15.0 million license fee payment from the Janssen Collaboration;prior year, partially offset in part by the timing of the collection of accounts receivable and the payment of accounts payable.
Net cash used in operations was $49.3 million in 2013 compared to $64.3 million of net cash used in 2012. The $15.0 million decrease in utilization of cash in operations was mainly due to receipts of the first commercial sale milestone payments totaling $14.0 million in 2013 from Roche and Baxter and timing of the collection of accounts receivable and the payment of accounts payable.receivable.
Investing Activities
Net cash used in investing activities was $33.0$163.7 million in 20142017 compared to $47.9 millionnet cash used in 2013 and $1.4investing activities of $76.8 million in 2012. This decrease in 2014 compared to 2013 was primarily due to a $53.9 million increase in proceeds from maturities of marketable securities; offset in part by a $39.9 million increase in purchases of marketable securities in 2014.2016. The increase in net cash used in investing activities in 2013 as compared to 2012 was primarily due to thenet purchases of marketable securities of $48.9using cash provided by operating and financing activities.
Net cash used in investing activities was $76.8 million in 2013.2016 compared to net cash provided of $5.9 million in 2015. The change was primarily due to net purchases of marketable securities using the proceeds from the Royalty-backed Loan.
Financing Activities
Net cash provided by financing activities was $114.5 million in 2014 compared to $25.1 million in 2013 and $112.8$131.7 million in 2012. Net cash provided by financing activities in 2014 consisted of $107.72017, primarily due to $134.9 million in net proceeds from the sale of our common stock in February 2014 and $6.8May 2017, compared to cash provided by financing activities of $150.6 million in 2016, when we drew net proceeds from option exercises.of $148.0 million on the Royalty-backed Loan.
Net cash provided by financing activities was $150.6 million in 2016 compared to $13.1 million in 2015. Net cash provided by financing activities in 20132015 consisted of net proceeds of $20.0 million from the amended long-term debt and $5.1$13.1 million in net proceeds from option exercises. Net cash provided by financing activities in 2012 consisted of net proceeds of $81.5 million from the sale of our common stock in February 2012, net proceeds of $29.7 million from long-term debt, and $1.7 million in net proceeds from option exercises.options exercised.
Long-Term Debt
Royalty-backed Loan
In January 2016, through our wholly-owned subsidiary Halozyme Royalty LLC (Halozyme Royalty), we received a $150 million loan (the Royalty-backed Loan) pursuant to a credit agreement (the Credit Agreement) with BioPharma Credit Investments IV Sub, LP and Athyrium Opportunities II Acquisition LP (the Royalty-backed Lenders). Under the terms of the Credit Agreement, Halozyme Therapeutics, Inc. transferred to Halozyme Royalty the right to receive royalty payments from the commercial sales of ENHANZE products owed under the Roche Collaboration and Baxalta Collaboration (Collaboration Agreements). The royalty payments from the Collaboration Agreements will be used to repay the principal and interest on the loan (the Royalty Payments).  The Royalty-backed Loan bears interest at a per annum rate of 8.75% plus the three-month LIBOR rate. The three-month LIBOR rate is subject to a floor of 0.7% and a cap of 1.5%. The interest rate as of December 2013,31, 2017 was 10.3%. The outstanding balance of the Royalty-backed Loan as of December 31, 2017 was $146.5 million.
The Credit Agreement provides that none of the Royalty Payments were required to be applied to the Royalty-backed Loan prior to January 1, 2017, 50% of the Royalty Payments were required to be applied to the Royalty-backed Loan between January 1, 2017 and January 1, 2018 and thereafter all Royalty Payments must be applied to the Royalty-backed Loan. However, the amounts available to repay the Royalty-backed Loan are subject to caps of $13.75 million per quarter in 2017, $18.75 million per quarter in 2018, $21.25 million per quarter in 2019 and $22.5 million per quarter in 2020 and thereafter. Amounts available to repay the Royalty-backed Loan will be applied first, to pay interest and second, to repay principal on the Royalty-backed Loan. Any accrued interest that is not paid on any applicable quarterly payment date, as defined, will be capitalized and added to the principal balance of the Royalty-backed Loan on such date. Halozyme Royalty will be entitled to receive and distribute to Halozyme any Royalty Payments that are not required to be applied to the Royalty-backed Loan or which are in excess of the foregoing caps.
The final maturity date of the Royalty-backed Loan will be the earlier of (i) the date when principal and interest is paid in full, (ii) the termination of Halozyme Royalty’s right to receive royalties under the Collaboration Agreements, and (iii) December 31, 2050. Currently, we estimate that the loan will be repaid in the first quarter of 2020. This estimate could be adversely affected and the repayment period could be extended if future royalty amounts are less than currently expected. Under the terms of the Credit Agreement, at any time after January 1, 2019, Halozyme Royalty may, subject to certain limitations, prepay the outstanding principal of the Royalty-backed Loan in whole or in part, at a price equal to 105% of the outstanding principal on the Royalty-backed Loan, plus accrued but unpaid interest. The Royalty-backed Loan constitutes an obligation of Halozyme Royalty, and is non-recourse to Halozyme. Halozyme Royalty retains its right to the Royalty Payments following repayment of the loan.


Oxford and SVB Loan and Security Agreement
In June 2016, we entered into an Amended and Restateda Loan and Security Agreement (the Loan Agreement) with Oxford Finance LLC (Oxford) and Silicon Valley Bank (SVB) (collectively, the Lenders), amendingproviding a senior secured loan facility of up to an aggregate principal amount of $70 million, comprising a $55.0 million draw in June 2016 and restating in its entirety our originalan additional $15.0 million tranche, which we had the option to draw during the second quarter of 2017 and did not exercise. The proceeds were partially used to pay the outstanding principal and final payment owed on a previous loan agreement with the Lenders, dated December 2012.Lenders. The Loan Agreement provided for an additional $20 million principal amount of new term loan, bringing the total term loan balance to $50 million. Theremaining proceeds are to bebeing used for working capital and general business requirements. The amended term loan facility matures on January 1, 2018. The outstanding term loan was $49.9 million as of December 31, 2014, net of unamortized debt discount of $0.1 million.
In January 2015, we and the Lenders entered into a second amendment to the Loan Agreement (the Amendment) amending and restating the loan repayment schedule of the Loan Agreement. The amended and restated loan repayment schedule provides for interest only payments in arrears through January 2016,for the first 18 months, followed by consecutive equal monthly payments of principal and interest in arrears starting in February 2016 and continuing through the previously established maturity date. Consistent with the original loan, thedate of January 1, 2021. The Loan Agreement provides for a 7.55% interest rate on the term loan and a final interest payment equal to 8.5%5.50% of the originalinitial $55 million principal amount, or $4.25 million, whichamount. The final payment is due when the term loanLoan Agreement becomes due or upon the prepayment of the facility. We have the option to prepay the outstanding balance of the term loanLoan Agreement in full, subject to a prepayment fee of 2% in the first year and 1% to 3% depending upon whenin the prepayment occurs.second year of the term loan. The outstanding term loan balance was $55.9 million as of December 31, 2017.
The amended and restated term loan facilityLoan Agreement is secured by substantially all of the assets of the Company and its subsidiary, Halozyme, Inc., except that the collateral does not include any equity interests in Halozyme, Inc., and any intellectual property (including all licensing, collaboration and similar agreements relating thereto), and certain other excluded assets. The Loan Agreement contains customary representations, warranties and covenants by us, which covenants limit our ability to convey, sell, lease, transfer, assign or otherwise dispose of certain of our assets; engage in any business other than the businesses currently engaged in by us or reasonably related thereto; liquidate or dissolve; make certain management changes; undergo certain change of control events; create, incur, assume, or be liable with respect to certain indebtedness; grant certain liens; pay dividends and make certain other restricted payments;


37



make certain investments; make payments on any subordinated debt; and enter into transactions with any of our affiliates outside of the ordinary course of business or permit our subsidiaries to do the same.same; and make any voluntary prepayment of or modify certain terms of the Royalty-backed Loan. In addition, subject to certain exceptions, we are required to maintain with SVB our primary deposit accounts, securities accounts and commodities, and to do the same for our domestic subsidiary.
The Loan Agreement also contains customary indemnification obligations and customary events of default, including, among other things, our failure to fulfill certain of our obligations under the Loan Agreement and the occurrence of a material adverse change which is defined as a material adverse change in our business, operations or condition (financial or otherwise), a material impairment of the prospect of repayment of any portion of the loan, or a material impairment in the perfection or priority of lender’sthe Lender’s lien in the collateral or in the value of such collateral.collateral or the occurrence of an event of default under the Royalty-backed Loan. In the event of default by us under the Loan Agreement, the Lenders would be entitled to exercise their remedies thereunder, including the right to accelerate the debt, upon which we may be required to repay all amounts then outstanding under the Loan Agreement, which could harm our financial condition.
Off-Balance Sheet Arrangements
As of December 31, 2014,2017, we did not have any relationships with unconsolidated entities or financial partnerships, such as entities often referred to as structured finance or special purpose entities, which would have been established for the purpose of facilitating off-balance sheet arrangements or other contractually narrow or limited purposes. In addition, we did not engage in trading activities involving non-exchange traded contracts. As such, we are not materially exposed to any financing, liquidity, market or credit risk that could arise if we had engaged in thesesuch relationships.


Contractual Obligations
As of December 31, 2014,2017, future minimum payments due under our contractual obligations are as follows (in thousands):
  Payments Due by Period
Contractual Obligations(1,5)
 Total 
Less than
1 Year
 1-3 Years 4-5 Years 
More than
5 Years
Long-term debt, including interest(2)
 $62,366
 $3,775
 $58,591
 $
 $
Operating leases(3)
 6,519
 2,133
 4,386
 
 
License payments 250
 250
 
 
 
Third-party manufacturing obligations(4)
 13,104
 13,104
 
 
 
Purchase obligations 456
 230
 226
 
 
Total $82,695
 $19,492
 $63,203
 $
 $
  Payments Due by Period
Contractual Obligations(1)
 Total 
Less than
1 Year
 1-3 Years 4-5 Years 
More than
5 Years
Long-term debt, including current portion(2)
 $205,157
 $77,211
 $127,946
 $
 $
Interest on long-term debt(3)
 26,792
 16,914
 9,878
 
 
Operating leases(4)
 13,113
 2,415
 8,079
 2,619
 
Third-party manufacturing obligations(5)
 12,257
 7,507
 4,750
 
 
Purchase obligations 445
 386
 59
 
 
Total $257,764
 $104,433
 $150,712
 $2,619
 $
_______________
(1)Does not include milestone or contractual payment obligations contingent upon the achievement of certain milestones or events if the amount and timing of such obligations are unknown or uncertain. Our in-license agreement is cancelable by us with written notice within 90 days. We may be required to pay up to approximately $9.3$8.0 million in milestone payments, plus sales royalties, in the event that all scientific researchregulatory and commercial milestones under these agreements is successful.the in-license agreement are achieved. Also excludes contractual obligations already recorded on our consolidated balance sheet as current liabilities.
(2)Long-term debt obligationsconsists of the Royalty-backed Loan and the Loan Agreement. Obligations include future quarterly principal payments for the Royalty-backed Loan based on an estimate of future royalty amounts. This estimate could be adversely affected and the repayment period could be extended if future royalty amounts are less than currently expected. Obligations also include future quarterly principal payments and a final payment of $3.03 million for the Loan Agreement due in January 2021.
(3)Interest on long-term debt includes future monthly interest payments for the Loan Agreement based on a fixed rate of 7.55%8.25%. Interest on long-term debt also includes quarterly interest payments on the Royalty-backed Loan, which bears interest at a per annum rate of 8.75% plus the three-month LIBOR rate. The three-month LIBOR rate is subject to a floor of 0.7% and a final paymentcap of $4.25 million1.5%. Future interest obligations for our long-term debt duethe Royalty-backed Loan were estimated using rates in January 2018.effect as of December 31, 2017.
(3)(4)Includes minimum lease payments related to leases of our office and research facilities and certain autos under non-cancelable operating leases.
(4)(5)
We have contracted with third-party manufacturers for the supply of bulk rHuPH20 and fill/finish of Hylenex recombinant. Under these agreements, we are required to purchase certain quantities each year during the terms of the agreements. The amounts presented represent our estimates of the minimum required payments under these agreements.
(5)Excludes contractual obligations already recorded on our consolidated balance sheet as current liabilities.


38



Contractual obligations for purchases of goods or services include agreements that are enforceable and legally binding onto us and that specify all significant terms, including fixed or minimum quantities to be purchased; fixed, minimum or variable price provisions; and the approximate timing of the transaction. For obligations with cancellation provisions, the amounts included in the preceding table were limited to the non-cancelable portion of the agreement terms or the minimum cancellation fee.
For thecertain restricted stock units and performance stock units granted, the number of shares issued on the date the restricted stock units vest is net of the minimum statutory withholding requirements that we pay in cash to the appropriate taxing authorities on behalf of our employees. The obligation to pay the relevant taxing authority is not included in the preceding table, as the amount is contingent upon continued employment. In addition, the amount of the obligation is unknown, as it is based in part on the market price of our common stock when the awards vest.


The expected timing of payments of the obligations above is estimated based on current information. Timing of payments and actual amounts paid may be different, depending on the time of receipt of goods or services, or changes to agreed-upon amounts for some obligations.
Our future capital uses and requirements depend on numerous forward-looking factors. These factors may include, but are not limited to, the following:
the rate of progress and cost of research and development activities;
the number and scope of our research activities;
the costs of filing, prosecuting, defending and enforcing any patent claims and other intellectual property rights;
our ability to establish and maintain product discovery and development collaborations, including scale-up manufacturing costs for our collaborators’ product candidates;
the amount of royalties from our collaborators;
the amount of product sales for Hylenex recombinant;
the costs of obtaining and validating additional manufacturers of Hylenex recombinant;
the effect of competing technological and market developments;
the costs of preparing for and launching a new commercial product;
the terms and timing of any collaborative, licensing and other arrangements that we may establish; and
the extent to which we acquire or in-license new products, technologies or businesses.
Critical Accounting Policies and Estimates
ThisThe discussion and analysis of our financial condition and results of operations are based on our consolidated financial statements, which have been prepared in accordance with U.S. generally accepted accounting principles, or U.S. GAAP. The preparation of our consolidated financial statements requires us to make estimates and judgments that affect the reported amounts of assets, liabilities, revenues and expenses and related disclosure of contingent assets and liabilities. We review our estimates on an ongoing basis. We base our estimates on historical experience and on various other assumptions 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. Actual results may differ from these estimates under different assumptions or conditions. Our significant accounting policies are outlined in Note 2 to the Consolidated Financial Statements included in the Form 10-K. We believe the following accounting policies to be critical to the judgments and estimates used in the preparation of our consolidated financial statements.
Revenue Recognition
We generate revenues from product sales and collaborative agreements. Payments received under collaborative agreements may include nonrefundable fees at the inception of the agreements, license fees, milestone payments for specific achievements designated in the collaborative agreements, reimbursements of research and development services and supply of bulk rHuPH20 and/or royalties on sales of products resulting from collaborative arrangements.
Revenue Recognition
MethodologyJudgment and UncertaintiesEffect if Actual Results Differ From Assumptions
For collaborative arrangements, we generally recognize revenues and income for each identified unit of accounting within the multiple element arrangements based on the nature and timing of the delivery process.

Management performs detailed reviews of all of our significant contracts. At the inception of the arrangement, consideration is allocated to all identified units of accounting based on their relative selling price. On an ongoing basis, the units of accounting are evaluated against revenue recognition criteria.
 The relative selling price for each deliverable is determined using vendor specific objective evidence (“VSOE”) of selling price or third-party evidence of selling price if VSOE does not exist. If neither VSOE nor third-party evidence of selling price exists, we use our best estimate of the selling price for the deliverable.Changes in the allocation of the sales price between delivered and undelivered elements can impact the timing of revenue recognition but do not change the total revenue recognized under any agreement.
We recognize revenue in accordance with the authoritative guidance on revenue recognition. Revenue is recognized when all of the following criteria are met: (1) persuasive evidence of an arrangement exists; (2) delivery has occurred or services have been rendered; (3) the seller’s price to the buyer is fixed or determinable; and (4) collectibility is reasonably assured.


39

Debt Classification
MethodologyJudgment and UncertaintiesEffect if Actual Results Differ From Assumptions
The short-term and long-term classification of outstanding debt represents management's best estimate of the timing of the amounts to be repaid. These estimates are based on contractual obligations, anticipated timing of royalty payments received and changes in LIBOR interest rates.Royalty payments are estimated using partner insight to the marketplace, historical trends and our knowledge of the therapeutic space.
The short-term and long-term portion of the debts may change and the repayment term may be shortened or extended depending on the actual level of royalty payments received. The actual repayment period could vary materially from our estimate to the extent that royalty payments from our partners are lower than our current estimates, which could arise due to factors beyond our control, such as competitive factors, decreased market acceptance or a failure by our partners to successfully commercialize in territories where regulatory approval has been received.

Currently, we do not believe that we have significant amount of risk relative to the repayment of the debt. A 10% reduction in the amount of anticipated royalties would not change our expected repayment period at maximum contractual interest rates.
Share-Based Payments
MethodologyJudgment and UncertaintiesEffect if Actual Results Differ From Assumptions
The Company maintains a Stock Incentive Plan, which provides for share-based payment awards, including stock options, restricted stock and performance awards. We determine the fair value of our stock option awards and performance awards at the date of grant using a Black-Scholes model. We determine the fair value of our restricted stock awards at the date of grant using the closing market value of our common stock on the date of grant.
Option-pricing models and generally accepted valuation techniques require management to make assumptions and to apply judgment to determine the fair value of our awards. These assumptions and judgments include estimating the future volatility of our stock price, expected dividend yield and future employee stock option exercise behaviors. Changes in these assumptions can materially affect the fair value estimate.

Our performance awards require management to make assumptions regarding the likelihood of achieving long-term Company goals.

We do not currently believe there is a reasonable likelihood that there will be a material change in estimates or assumptions we use to determine stock-based compensation expense. However, if actual results are not consistent with our estimates or assumptions, we may be exposed to changes in share-based compensation expense that could be material.

If actual results are not consistent with the assumptions used, the share-based compensation expense reported in our financial statements may not be representative of the actual economic cost of the share-based compensation. A 10% change in our share-based compensation expense for the year ended December 31, 2017, would have affected pre-tax earnings by approximately $3.1 million in 2017.


Refer to Note 2 for a further discussion of our revenue recognition policies for product sales and revenues under our collaborative agreements and Note 4 for a further discussion of our collaborative agreements.
Share-Based Payments
We use the fair value method to account for share-based payments in accordance with the authoritative guidance for share-based compensation. The fair value of each option award is estimated on the date of grant using a Black-Scholes-Merton option pricing model (Black-Scholes model) that uses assumptions regarding a number of complex and subjective variables. Changes in these assumptions may lead to variability with respect to the amount of expense we recognize in connection with share-based payments. Refer to Note 2 for a further discussion of share-based payments.
Research and Development Expenses
Research and development expenses include salaries and benefits, facilities and other overhead expenses, external clinical trial expenses, research related manufacturing services, contract services and other outside expenses. Research and development expenses are charged to operations as incurred when these expenditures relate to our research and development efforts and have no alternative future uses.After receiving marketing approval from the FDA or comparable regulatory agencies in foreign countries for a product, costs related to purchases or manufacturing of bulk rHuPH20 for such product are capitalized as inventory. The manufacturing costs of bulk rHuPH20 for the collaboration products, Herceptin SC, MabThera SC and HYQVIA, which were incurred after the receipt of the European marketing approvals are capitalized as inventory. Refer to Note 2 for a further discussion of research and development expenses.
Due to the uncertainty in obtaining the FDA and other regulatory approvals, our reliance on third parties and competitive pressures, we are unable to estimate with any certainty the additional costs we will incur in the continued development of our proprietary product candidates for commercialization. However, we expect our research and development expenses to increase this year as we continue with our clinical trial programs and continue to develop and manufacture our product candidates.
Clinical development timelines, likelihood of success and total costs vary widely. We anticipate that we will make ongoing determinations as to which research and development projects to pursue and how much funding to direct to each project on an ongoing basis in response to existing resource levels, the scientific and clinical progress of each product candidate, and other market and regulatory developments. We plan on focusing our resources on those proprietary and collaboration product candidates that represent the most valuable economic and strategic opportunities.
Product candidate completion dates and costs vary significantly for each product candidate and are difficult to estimate. The lengthy process of seeking regulatory approvals and the subsequent compliance with applicable regulations require the expenditure of substantial resources. Any failure by us to obtain, or any delay in obtaining, regulatory approvals could cause our research and development expenditures to increase and, in turn, have a material adverse effect on our results of operations. We cannot be certain when, or if, our product candidates will receive regulatory approval or whether any net cash inflow from our other product candidates, or development projects, will commence.
Research and Development Expenses - Clinical Trial Accruals
MethodologyJudgment and UncertaintiesEffect if Actual Results Differ From Assumptions
All of our clinical trials have been executed with support from contract research organizations, (CROs), and other vendors. We accrue costs for clinical trial activities performed by CROs based upon the estimated amount of work completed on each trial.
For clinical trial expenses, the significant factors used in estimating accruals include the number of patients enrolled, the activities to be performed for each patient, the number of active clinical sites, and the duration for which the patients will be enrolled in the trial. We monitor patient enrollment levels and related activities to the extent possible through internal reviews, correspondence with CROs and review of contractual terms.

We base our estimates on the best information available at the time. However, additional information may become available to us, which may allow us to make a more accurate estimate in future periods. If we do not identify costs that we have begun to incur or if we underestimate or overestimate the level of services performed or the costs of these services, our actual expenses could differ from our estimates. There were no such significant changes during the years ended December 31, 2017, 2016 or 2015.
Recent Accounting Pronouncements
Refer to Note 2, Summary of Significant Accounting Policies - Pending Adoption, of Recent Accounting Pronouncements,our consolidated financial statements for a discussion of recent accounting pronouncements and their effect, if any, on us.


40



Item 7A.Quantitative and Qualitative Disclosures About Market Risk
As of December 31, 2014,2017, our cash equivalents and marketable securities consisted of investments in money market funds, andU.S. Treasury securities, corporate debt obligations.obligations and commercial paper. These investments were made in accordance with our investment policy which specifies the categories, allocations, and ratings of securities we may consider for investment. The primary objective of our investment activities is to preserve principal while at the same time maximizing the income we receive without significantly increasing risk. Some of the financial instruments that we invest in could be subject to market risk. This means that a change in prevailing interest rates may cause the value of the instruments to fluctuate. For example, if we purchase a security that was issued with a fixed interest rate and the prevailing interest rate later rises, the value of that security will probably decline. As of December 31, 2014 basedBased on our current investment portfolio as of December 31, 2017, we do not believe that our results of operations would be materially impacted by an immediate change of 10% in interest rates.
We do not hold or issue derivatives, derivative commodity instruments or other financial instruments for speculative trading purposes. Further, we do not believe our cash, cash equivalents and marketable securities have significant risk of default or illiquidity. We made this determination based on discussions with our investment advisors and a review of our holdings. While we believe our cash, cash equivalents and marketable securities do not contain excessive risk, we cannot provide absolute assurance that in the future our investments will not be subject to adverse changes in market value. All of our cash equivalents and marketable securities are recorded at fair market value.
Item 8.Financial Statements and Supplementary Data
Our financial statements are annexed to this report beginning on page F-1.
Item 9.Changes In and Disagreements with Accountants on Accounting and Financial Disclosure
None.


Item 9A.ControlControls and Procedures
Evaluation of Disclosure Controls and Procedures
We maintain disclosure controls and procedures that are designed to ensure that information required to be disclosed in our Exchange Act reports is recorded, processed, summarized and reported within the timelines specified in the Securities and Exchange Commission’s rules and forms, and that such information is accumulated and communicated to our management, including our Chief Executive Officer and Chief Financial Officer, as appropriate, to allow timely decision regarding required disclosure. In designing and evaluating the disclosure controls and procedures, management recognized that any controls and procedures, no matter how well designed and operated, can only provide reasonable assurance of achieving the desired control objectives, and in reaching a reasonable level of assurance, management necessarily was required to apply its judgment in evaluating the cost-benefit relationship of possible controls and procedures.
Under the supervision and with the participation of our management, including our principal executive officer and principal financial officer, we conducted an evaluation of our disclosure controls and procedures, as such term is defined under Rule 13a-15(e) promulgated under the Securities Exchange Act of 1934, as amended. Based on this evaluation, our principal executive officer and our principal financial officer concluded that our disclosure controls and procedures were effective as of the end of the period covered by this Annual Report on Form 10-K.
Changes in Internal Control Over Financial Reporting
There have been no significant changes in our internal control over financial reporting that occurred during the quarter ended December 31, 20142017 that have materially affected, or are reasonably likely to materially affect, our internal control over financial reporting.


41



Management’s Report on Internal Control Over Financial Reporting
Our management is responsible for establishing and maintaining adequate internal control over financial reporting. Internal control over financial reporting is defined in Rule 13a-15(f) and Rule 15d-15(f) promulgated under the Securities Exchange Act of 1934 as a process designed by, or under the supervision of, our principal executive and principal financial officers and effected by our board of directors, management and other personnel, to provide reasonable assurance regarding the reliability of financial reporting and the preparation of financial statements for external purposes in accordance with generally accepted accounting principles and includes those policies and procedures that:
Pertain to the maintenance of records that in reasonable detail accurately and fairly reflect the transactions and dispositions of our assets;
Provide reasonable assurance that transactions are recorded as necessary to permit preparation of financial statements in accordance with generally accepted accounting principles, and that receipts and expenditures are being made only in accordance with authorizations of our management and directors; and
Provide reasonable assurance regarding prevention or timely detection of unauthorized acquisition, use or disposition of our assets that could have a material effect on our financial statements.
Because of its inherent limitations, internal control over financial reporting may not prevent or detect misstatements. Projections of any evaluation of effectiveness to future periods are subject to the risk that controls may become inadequate because of changes in conditions, or that the degree of compliance with the policies or procedures may deteriorate.
Our management assessed the effectiveness of our internal control over financial reporting as of December 31, 2014.2017. In making this assessment, our management used the criteria set forth by the Committee of Sponsoring Organizations of the Treadway Commission (COSO) in Internal Control-Integrated Framework (2013 framework) (the COSO criteria). Based on our assessment, management concluded that, as of December 31, 2014,2017, our internal control over financial reporting is effective based on the COSO criteria.


42



The independent registered public accounting firm that audited the consolidated financial statements that are included in this Annual Report on Form 10-K has issued an audit report on the effectiveness of our internal control over financial reporting as of December 31, 2014.2017. The report appears below.


Report of Independent Registered Public Accounting Firm
The Board of Directors and Stockholders
Halozyme Therapeutics, Inc.
Opinion on Internal Control over Financial Reporting
We have audited Halozyme Therapeutics, Inc.’s internal control over financial reporting as of December 31, 2014,2017, based on criteria established in Internal Control - Integrated Framework issued by the Committee of Sponsoring Organizations of the Treadway Commission (2013 framework) (the COSO criteria). In our opinion, Halozyme Therapeutics, Inc.’s (the Company) maintained, in all material respects, effective internal control over financial reporting as of December 31, 2017, based on the COSO criteria.
We also have audited, in accordance with the standards of the Public Company Accounting Oversight Board (United States) (PCAOB), the consolidated balance sheets of the Company as of December 31, 2017 and 2016, and the related consolidated statements of operations, comprehensive loss, cash flows, and stockholders’ equity (deficit) for each of the three years in the period ended December 31, 2017 , and the related notes and the financial statement schedule listed in the Index at Item 15(a) and our report dated February 20, 2018 expressed an unqualified opinion thereon.
Basis for Opinion
The Company’s management is responsible for maintaining effective internal control over financial reporting and for its assessment of the effectiveness of internal control over financial reporting included in the accompanying Management’s Report on Internal Control Over Financial Reporting. Our responsibility is to express an opinion on the company’sCompany’s internal control over financial reporting based on our audit. We are a public accounting firm registered with the PCAOB and are required to be independent with respect to the Company in accordance with the U.S. federal securities laws and the applicable rules and regulations of the Securities and Exchange Commission and the PCAOB.
We conducted our audit in accordance with the standards of the Public Company Accounting Oversight Board (United States).PCAOB. Those standards require that we plan and perform the audit to obtain reasonable assurance about whether effective internal control over financial reporting was maintained in all material respects.
Our audit included obtaining an understanding of internal control over financial reporting, assessing the risk that a material weakness exists, testing and evaluating the design and operating effectiveness of internal control based on the assessed risk, and performing such other procedures as we considered necessary in the circumstances. We believe that our audit provides a reasonable basis for our opinion.
Definition and Limitations of Internal Control Over Financial Reporting
A company’s internal control over financial reporting is a process designed to provide reasonable assurance regarding the reliability of financial reporting and the preparation of financial statements for external purposes in accordance with generally accepted accounting principles. A company’s internal control over financial reporting includes those policies and procedures that (1) pertain to the maintenance of records that, in reasonable detail, accurately and fairly reflect the transactions and dispositions of the assets of the company; (2) provide reasonable assurance that transactions are recorded as necessary to permit preparation of financial statements in accordance with generally accepted accounting principles, and that receipts and expenditures of the company are being made only in accordance with authorizations of management and directors of the company; and (3) provide reasonable assurance regarding prevention or timely detection of unauthorized acquisition, use, or disposition of the company’s assets that could have a material effect on the financial statements.
Because of its inherent limitations, internal control over financial reporting may not prevent or detect misstatements. Also, projections of any evaluation of effectiveness to future periods are subject to the risk that controls may become inadequate because of changes in conditions, or that the degree of compliance with the policies or procedures may deteriorate.
In our opinion, Halozyme Therapeutics, Inc. maintained, in all material respects, effective internal control over financial reporting as of December 31, 2014, based on the COSO criteria.
We also have audited, in accordance with the standards of the Public Company Accounting Oversight Board (United States), the consolidated balance sheets of Halozyme Therapeutics, Inc. as of December 31, 2014 and 2013, and the related consolidated statements of operations, comprehensive loss, cash flows, and stockholders’ equity (deficit) for each of the three years in the period ended December 31, 2014 of Halozyme Therapeutics, Inc. and our report dated March 2, 2015 expressed an unqualified opinion thereon.
                                                                                                   /s/    Ernst & Young LLP
San Diego, California
March 2, 2015February 20, 2018


43



Item 9B.Other Information
None.
PART III
Item 10.Directors, Executive Officers and Corporate Governance
The information required by this item regarding directors is incorporated by reference to our definitive Proxy Statement (the Proxy Statement) to be filed with the Securities and Exchange Commission in connection with our 20152018 Annual Meeting of Stockholders under the heading “Election of Directors.” The information required by this item regarding compliance with Section 16(a) of the Securities Exchange Act of 1934, as amended, is incorporated by reference to the information under the caption “Compliance with Section 16(a) of the Exchange Act” to be contained in the Proxy Statement. The information required by this item regarding our code of ethics is incorporated by reference to the information under the caption “Code of Conduct and Ethics”Ethics and Corporate Governance Guidelines” to be contained in the Proxy Statement. The information required by this item regarding our audit committee is incorporated by reference to the information under the caption “Board Meetings and Committees—Audit Committee” to be contained in the Proxy Statement. The information required by this item regarding material changes, if any, to the process by which stockholders may recommend nominees to our board of directors is incorporated by reference to the information under the caption “Board Meetings and Committees—Nominating and Governance Committee” to be contained in the Proxy Statement.
Executive Officers
Helen I. Torley, M.B. Ch. B., M.R.C.P. (52)(55), President, Chief Executive Officer and Director. Dr. Torley joined Halozyme in January 2014 as President and Chief Executive Officer and isas a member of Halozyme’s Board of Directors. Throughout her career, Dr. Torley has led several successful product launches, including Kyprolis®Kyprolis®, Prolia®Prolia®, Sensipar®Sensipar®, and Miacalcin®Miacalcin®. Prior to joining Halozyme, Dr. Torley previously served as Executive Vice President and Chief Commercial Officer for Onyx Pharmaceuticals (Onyx) from August 2011 to December 2013 overseeing the collaboration with Bayer on Nexavar®Nexavar® and Stivarga®Stivarga® and the U.S. launch of Kyprolis. She was responsible for the development of Onyx's commercial capabilities in ex-US markets and in particular, in Europe. Prior to Onyx, Dr. Torley spent 1410 years in management positions at Amgen Inc., most recently serving as Vice President and General Manager of both the US Nephrology Business Unit from 2003 to 2009 and the U.S. Bone Health Business Unit.Unit from 2009 to 2011. From 1997 to 2002, she held various senior management positions at Bristol-Myers Squibb, including Regional Vice President of Cardiovascular and Metabolic Sales and Head of Cardiovascular Global Marketing. She began her career at Sandoz/Novartis, where she ultimately served as Vice President of Medical Affairs, developing and conducting post-marketing clinical studies across all therapeutic areas, including oncology. Within the past five years, Dr. Torley served on the board of directors of Relypsa, Inc., a biopharmaceutical company. Before joining the industry, Dr. Torley was in medical practice as a senior registrar in rheumatology at the Royal Infirmary in Glasgow, Scotland. Dr. Torley received her Bachelor of Medicine and Bachelor of Surgery degrees (M.B. Ch.B.) from the University of Glasgow and is a Member of the Royal College of Physicians (M.R.C.P).
David A. RamsayLaurie D. Stelzer (50), Senior Vice President, Chief Financial Officer. Mr. RamsayMs. Stelzer joined Halozyme in 2003June 2015 as Chief Financial Officer and served in that capacity until 2009 when he was appointedSenior Vice President, Corporate Development. After spending four years in various commercial and operational roles, Mr. Ramsay was appointed Chief Financial Officer. Prior to joining Halozyme, heMs. Stelzer served in various financial roles including Vice President, Chief Financial Officer of Lathian Systems. Priorfrom April 2014 to Lathian, Mr. Ramsay was Vice President, Treasurer of ICN Pharmaceuticals, now called Valeant Pharmaceuticals International, a multinational, specialty pharmaceutical company. Mr. Ramsay joined Valeant from ARCO, where he spent four years in various financial roles, most recently servingJanuary 2015 as Manager, Financial Planning & Analysis for the company’s Retail Marketing division. Prior to ARCO, he served as Vice President, Controller for Security Pacific Asian Bank, a subsidiary of Security Pacific Corporation. He began his career as an Auditor at Deloitte & Touche, where he obtained his CPA license. Mr. Ramsay received his B.S in Business Administration from the University of California, Berkeley, and his MBA in Finance and Strategic Management from The Wharton School at the University of Pennsylvania.


44



Athena Countouriotis, M.D. (43), Senior Vice President Chief Medical Officer. Dr. Countouriotis joined Halozyme in 2015. From Februaryof Finance supporting R&D, Technical Operations and M&A at Shire, Inc., a biopharmaceutical company. Prior to that she was the Division CFO for the Regenerative Medicine Division and the Head of Investor Relations at Shire from March 2012 to January 2015, Dr. Countouriotis served as chief medical officer at Ambit Biosciences Corporation which was acquired by Daiichi Sankyo in NovemberApril 2014. From August 2007Prior to February 2012, Dr. Countouriotis was a clinical leader within the Pfizer Inc. Oncology Business Unit. From October 2005 to August 2007, she was directorShire, Ms. Stelzer held positions of oncology global clinical research at Bristol-Myers Squibb Company, a publicly-traded global pharmaceutical company, withincreasing responsibility for leading clinical development15 years at Amgen, Inc., a biopharmaceutical company, including Interim Treasurer, Head of Sprycel® in acute lymphoblastic leukemiaEmerging Markets Expansion, Executive Director of Global Commercial Finance and chronic myeloid leukemia. EarlierHead of Global Accounting. Early in her career, she held various finance and accounting positions in the position as associate medical director at Cell Therapeutics, Inc., a biopharmaceutical company. Dr. Countouriotisreal estate and banking industries. Ms. Stelzer received a B.S.her MBA from the University of California, Los Angeles, and an M.D. at Tufts UniversityAnderson School of Medicine. She received her initial training in pediatrics at the University of California, Los Angeles, and additional training at the Fred Hutchinson Cancer Research Centera Bachelor of Science in Accounting from Arizona State University.


Harry J. Leonhardt, Esq. (61), Senior Vice President, General Counsel, Chief Compliance Officer and Corporate Secretary. Mr. Leonhardt joined Halozyme in April 2015 as Senior Vice President, General Counsel, Chief Compliance Officer and Corporate Secretary. Mr. Leonhardt brings more than 30 years of executive management, corporate legal, intellectual property, compliance, business development and mergers and acquisitions experience to Halozyme, with an extensive background in the Pediatric Hematology/Oncology Program.biotechnology industry. Prior to joining Halozyme, Mr. Leonhardt was an arbitrator before the International Centre for Dispute Resolution and a consultant in the biotechnology industry from January 2013 to April 2015. He served as Senior Vice President, Legal and Compliance, and Corporate Secretary at Amylin Pharmaceuticals, Inc., a biotechnology company, from September 2011 to January 2013 and previously served in other senior management legal positions at Amylin since September 2007. Prior to Amylin, he served as Senior Vice President, General Counsel and Corporate Secretary at Senomyx, Inc. from September 2003 to September 2007. From February 2001 to September 2003, Mr. Leonhardt was Executive Vice President, General Counsel and Corporate Secretary at Genoptix, Inc. and from July 1996 to November 2000, he served as Vice President and then Senior Vice President, General Counsel and Corporate Secretary at Nanogen, Inc. Prior to Nanogen, Mr. Leonhardt held positions of increasing responsibility at Allergan, Inc. including Chief Litigation Counsel and General Counsel for European Operations. Early in his career, he was an attorney at Lyon & Lyon LLP where he represented a number of prominent clients in the biotech, pharmaceutical and consumer products industries. Mr. Leonhardt received a B.S. in Pharmacy from the University of the Sciences and a Juris Doctorate from the University of Southern California School of Law.
Item 11.Executive Compensation
The information required by this item is incorporated by reference to the information under the caption “Executive Compensation” to be contained in the Proxy Statement.
Item 12.Security Ownership of Certain Beneficial Owners and Management and Related Stockholder Matters
Other than as set forth below, the information required by this item is incorporated by reference to the information under the caption “Security Ownership of Certain Beneficial Owners and Management and Related Stockholder Matters” to be contained in the Proxy Statement.
Equity Compensation Plan Information
The following table summarizes our compensation plans under which our equity securities are authorized for issuance as of December 31, 2014:2017:
Plan Category 
Number of
Shares to Be
Issued upon
Exercise of
Outstanding
Options and
Restricted Stock
Units
(a)
 
Weighted-Average
Exercise Price
of Outstanding
Options and
Restricted Stock
Units(2)
(b) 
 
Number of
Shares
Remaining
Available for
Future Issuance
under Equity
Compensation
Plans (Excluding
Shares Reflected
in Column (a))
(c)
 
Number of Shares 
to be Issued upon
Exercise of
Outstanding Options
and Restricted Stock
Units
(a)
 
Weighted Average
Exercise Price
of Outstanding
Options(2)
(b) 
 
Number of Shares
Remaining Available
for Future Issuance
under Equity
Compensation
Plans (Excluding
Shares Reflected
in Column (a))
(c)
Equity compensation plans approved by stockholders (1)
 7,247,452
 $9.18 4,681,212
Equity compensation plans approved by stockholders (1)
 13,023,641
 $12.24 6,552,249
Equity compensation plans not approved by stockholders 
  
Equity compensation plans not approved by stockholders 
  
 7,247,452
 $9.18 4,681,212
 13,023,641
 $12.24 6,552,249
_____________________
(1)Represents stock options, restricted stock units, and performance restricted stock units under the Amended and Restated 2011 Stock Plan, 2008 Stock Plan 2006 Stock Plan, 2005 Outside Directors’ Stock Plan, and 20042006 Stock Plan.
(2)This amount does not include restricted stock units and performance restricted stock units as there is no exercise price for restricted stocksuch units.


Item 13.Certain Relationships and Related Transactions, and Director Independence
The information required by this item is incorporated by reference to the information under the caption “Certain Relationships and Related Transactionsand “Corporate Governance - Director Independence” to be contained in the Proxy Statement.
Item 14.Principal Accounting Fees and Services
The information required by this item is incorporated by reference to the information under the caption “Principal Accounting Fees and Services” to be contained in the Proxy Statement.


45



PART IV
Item 15.Exhibits and Financial Statement Schedules
(a)Documents filed as part of this report.
1.   Financial Statements 
  
  Page
Report of Independent Registered Public Accounting Firm  
Consolidated Balance Sheets at December 31, 20142017 and 20132016  
Consolidated Statements of Operations for Each of the Years Ended December 31, 2014, 2013
2017, 2016 and 2012
2015
  
Consolidated Statements of Comprehensive LossIncome (Loss) for Each of the Years Ended December 31, 2014, 2013
2017,
     2016 and 20122015
 
Consolidated Statements of Cash Flows for Each of the Years Ended December 31, 2014, 20132017, 2016 and 20122015  
Consolidated Statements of Stockholders’ Equity (Deficit) for Each of the Years Ended December  31, 2017,
     2014, 20132016 and 20122015
  F-6
Notes to the Consolidated Financial Statements  F-7
2.   List of all Financial Statement schedules.
The following financial statement schedule of Halozyme Therapeutics, Inc. is filed as part of this Annual Report on Form 10-K on page F-33 and should be read in conjunction with the consolidated financial statements of Halozyme Therapeutics, Inc.
Page
Schedule II: Valuation and Qualifying Accounts
All other schedules are omitted because they are not applicable or the required information is shown in the Financial Statements or notes thereto.
3.   List of Exhibits required by Item 601 of Regulation S-K. See part (b) below.


(b)Exhibits.
The exhibits listed in the accompanying “Exhibit Index” are incorporated herein by reference.
Incorporated by Reference
ExhibitFiled
NumberExhibit TitleHerewithFormFile No.Date Filed
3.110-Q001-323358/7/2013
3.28-K001-3233512/19/2016
10.1SB-2333-1147764/23/2004
10.28-K001-323351/12/2006
10.3#8-K001-323353/24/2006
10.4#10-Q001-323358/8/2006
10.5#10-Q001-323358/8/2006
10.6#8-K001-323353/19/2008
10.7#10-Q001-323358/7/2009
10.8#10-Q001-323358/7/2009
10.9#DEF-14A001-323353/23/2016
10.10#8-K001-323355/6/2011
10.11#8-K001-323355/6/2011
10.12#10-Q001-323358/10/2015
10.13#10-Q001-323358/10/2015
10.14#8-K001-323355/6/2011
10.15#8-K001-323355/6/2011
10.16#10-Q001-3233511/9/2015
10.17#10-Q001-3233511/9/2015
10.18#10-Q001-3233511/9/2015
10.19#10-K001-323352/28/2017
10.20#8-K001-3233512/20/2007


Incorporated by Reference
ExhibitFiled
NumberExhibit TitleHerewithFormFile No.Date Filed
10.21#10-Q001-323355/9/2008
10.22#10-Q001-3233511/9/2015
10.238-K001-323356/16/2011
10.248-K001-323357/5/2017
10.258-K001-323356/16/2011
10.268-K001-323357/5/2017
10.2710-K001-323353/1/2013
10.2810-Q001-323355/8/2013
10.298-K001-323357/5/2017
10.30*10-K001-323352/29/2016
10.31#DEF-14A001-323353/23/2016
10.3210-Q001-323358/9/2016
10.3310-K001-323352/28/2017
10.34X
10.35#8-K001-323355/9/2017
10.36#8-K001-323359/29/2017
21.1X
23.1X
31.1X
31.2X


Incorporated by Reference
ExhibitFiled
NumberExhibit TitleHerewithFormFile No.Date Filed
32X
101.INSXBRL Instance DocumentX
101.SCHXBRL Taxonomy Extension SchemaX
101.CALXBRL Taxonomy Extension Calculation LinkbaseX
101.DEFXBRL Taxonomy Extension Definition LinkbaseX
101.LABXBRL Taxonomy Extension Label LinkbaseX
101.PREXBRL Taxonomy Presentation LinkbaseX
_______________
*Confidential treatment has been granted (or requested) for certain portions of this exhibit. These portions have been omitted from this agreement and have been filed separately with the Securities and Exchange Commission.
#Indicates management contract or compensatory plan or arrangement.
(c)
Financial Statement Schedules.  See Item 15(a) 2 above.

Item 16.Form 10-K Summary
None.

46



SIGNATURES
Pursuant to the requirements of Section 13 or 15(d) of the Securities Exchange Act of 1934, the Registrant has duly caused this report to be signed on its behalf by the undersigned, thereunto duly authorized.
 
    
Halozyme Therapeutics, Inc.,
a Delaware corporation
    
Date: March 2, 2015February 20, 2018   By: /s/    Helen I. Torley, M.B. Ch.B., M.R.C.P.
        Helen I. Torley, M.B. Ch.B., M.R.C.P.
        President and Chief Executive Officer


47



POWER OF ATTORNEY
Know all persons by these presents, that each person whose signature appears below constitutes and appoints Helen I. Torley and David A. Ramsay,Laurie D. Stelzer, and each of them, as his true and lawful attorneys-in-fact and agents, with full power of substitution and resubstitution, for him and in his name, place, and stead, in any and all capacities, to sign any and all amendments to this Annual Report, and to file the same, with all exhibits thereto, and other documents in connection therewith, with the Securities and Exchange Commission, granting unto said attorneys-in-fact and agents, and each of them, full power and authority to do and perform each and every act and thing requisite and necessary to be done in connection therewith, as fully to all intents and purposes as he might or could do in person, hereby ratifying and confirming that all said attorneys-in-fact and agents, or any of them or their or his substitute or substituted, may lawfully do or cause to be done by virtue thereof.
Pursuant to the requirements of the Securities Exchange Act of 1934, this Annual Report has been signed below by the following persons on behalf of the Registrant and in the capacities and on the dates indicated.
Signature  Title Date
   
/s/    Helen I. Torley, M.B. Ch.B., M.R.C.P.  President and Chief Executive Officer March 2, 2015February 20, 2018
       Helen I. Torley, M.B. Ch.B., M.R.C.P.  (Principal Executive Officer), Director 
     
/s/   David A. RamsayLaurie D. Stelzer  Senior Vice President and Chief Financial Officer March 2, 2015February 20, 2018
       David A. RamsayLaurie D. Stelzer (Principal Financial and Accounting Officer) 
     
/s/    Kathryn E. FalbergConnie L. Matsui  Chair of the Board of Directors March 2, 2015February 20, 2018
       Kathryn E. FalbergConnie L. Matsui   
     
/s/ Jean-Pierre Bizzari Director March 2, 2015February 20, 2018
     Jean-Pierre Bizzari
/s/    James M. DalyDirectorFebruary 20, 2018
       James M. Daly
/s/    Jeffrey W. HendersonDirectorFebruary 20, 2018
       Jeffrey W. Henderson    
     
/s/    Kenneth J. Kelley  Director March 2, 2015February 20, 2018
       Kenneth J. Kelley   
     
/s/    Randal J. Kirk  Director March 2, 2015February 20, 2018
       Randal J. Kirk
/s/    Connie L. MatsuiDirectorMarch 2, 2015
       Connie L. Matsui
/s/    John S. Patton, Ph.D.DirectorMarch 2, 2015
       John S. Patton, Ph.D.   
     
/s/    Matthew L. Posard Director March 2, 2015February 20, 2018
       Matthew L. Posard    


48



Report of Independent Registered Public Accounting Firm
The Board of Directors and Stockholders
Halozyme Therapeutics, Inc.

Opinion on the Financial Statements
We have audited the accompanying consolidated balance sheets of Halozyme Therapeutics, Inc. (the Company) as of December 31, 20142017 and 2013,2016, and the related consolidated statements of operations, comprehensive loss, cash flows, and stockholders’ equity (deficit) for each of the three years in the period ended December 31, 2014.  Our audits also included2017, and the related notes and the financial statement schedule listed in the Index at Item 15(a) (collectively referred to as the “consolidated financial statements”). These financial statements and schedule are the responsibility of the Company's management. Our responsibility is to express an opinion on these financial statements and schedule based on our audits.
We conducted our audits in accordance with the standards of the Public Company Accounting Oversight Board (United States). Those standards require that we plan and perform the audit to obtain reasonable assurance about whether the financial statements are free of material misstatement. An audit includes examining, on a test basis, evidence supporting the amounts and disclosures in the financial statements. An audit also includes assessing the accounting principles used and significant estimates made by management, as well as evaluating the overall financial statement presentation. We believe that our audits provide a reasonable basis for our opinion.
In our opinion, the consolidated financial statements referred to above, present fairly, in all material respects, the consolidated financial position of Halozyme Therapeutics, Inc.the Company at December 31, 20142017 and 2013,2016, and the consolidated results of its operations and its cash flows for each of the three years in the period ended December 31, 2014,2017, in conformity with U.S. generally accepted accounting principles. Also, in our opinion, the related financial statement schedule, when considered in relation to the basic financial statements taken as a whole, presents fairly in all material respects the information set forth therein. 
We also have audited, in accordance with the standards of the Public Company Accounting Oversight Board (United States) (PCAOB)Halozyme Therapeutics, Inc.’sthe Company’s internal control over financial reporting as of December 31, 2014,2017, based on criteria established in Internal Control - Integrated Framework issued by the Committee of Sponsoring Organizations of the Treadway Commission (2013 framework) and our report dated March 2, 2015February 20, 2018 expressed an unqualified opinion thereon.
Basis for Opinion
These financial statements are the responsibility of the Company’s management. Our responsibility is to express an opinion on the Company’s financial statements based on our audits. We are a public accounting firm registered with the PCAOB and are required to be independent with respect to the Company in accordance with the U.S. federal securities laws and the applicable rules and regulations of the Securities and Exchange Commission and the PCAOB.
We conducted our audits in accordance with the standards of the PCAOB. Those standards require that we plan and perform the audit to obtain reasonable assurance about whether the financial statements are free of material misstatement, whether due to error or fraud. Our audits included performing procedures to assess the risks of material misstatement of the financial statements, whether due to error or fraud, and performing procedures that respond to those risks. Such procedures included examining, on a test basis, evidence regarding the amounts and disclosures in the financial statements. Our audits also included evaluating the accounting principles used and significant estimates made by management, as well as evaluating the overall presentation of the financial statements. We believe that our audits provide a reasonable basis for our opinion.
/s/    Ernst & Young LLP
We have served as the Company’s auditor since 2006.

San Diego, California
March 2, 2015February 20, 2018




F-1



HALOZYME THERAPEUTICS, INC.
CONSOLIDATED BALANCE SHEETS
(inIn thousands, except per share data)amounts)
 December 31,
2014
 December 31,
2013
 December 31,
2017
 December 31,
2016
ASSETSASSETS    
Current assets:        
Cash and cash equivalents $61,389
 $27,357
 $168,740
 $66,764
Marketable securities, available-for-sale 74,234
 44,146
 300,474
 138,217
Accounts receivable, net 9,149
 9,097
 22,133
 15,680
Inventories 6,406
 6,170
 5,146
 14,623
Prepaid expenses and other assets 10,143
 8,425
 13,879
 21,248
Total current assets 161,321
 95,195
 510,372
 256,532
Property and equipment, net 2,951
 3,422
 3,520
 4,264
Prepaid expenses and other assets 1,205
 2,676
 5,553
 219
Restricted cash 500
 500
 500
 500
Total assets $165,977
 $101,793
 $519,945
 $261,515
        
LIABILITIES AND STOCKHOLDERS’ EQUITY (DEFICIT)LIABILITIES AND STOCKHOLDERS’ EQUITY (DEFICIT)    
Current liabilities:        
Accounts payable $3,003
 $3,135
 $7,948
 $3,578
Accrued expenses 13,961
 14,369
 39,601
 28,821
Deferred revenue, current portion 7,367
 7,398
 6,568
 4,793
Current portion of long-term debt, net 77,211
 17,393
Total current liabilities 24,331
 24,902
 131,328
 54,585
Deferred revenue, net of current portion 47,267
 45,745
 54,297
 39,825
Long-term debt, net 49,860
 49,772
 125,140
 199,228
Other long-term liabilities 3,167
 1,364
 814
 358
Commitments and contingencies (Note 9) 
 
 
 
Stockholders’ equity (deficit):        
Preferred stock — $0.001 par value; 20,000 shares authorized; no shares issued and outstanding 
 
Common stock — $0.001 par value; 200,000 shares authorized; 125,721 and 114,534 shares issued and outstanding at December 31, 2014 and 2013, respectively 126
 115
Preferred stock - $0.001 par value; 20,000 shares authorized; no shares
issued and outstanding
 
 
Common stock - $0.001 par value; 200,000 shares authorized; 142,789 and
129,502 shares issued and outstanding at December 31, 2017 and 2016,
respectively
 143
 130
Additional paid-in capital 491,694
 361,930
 731,044
 552,737
Accumulated other comprehensive (loss) income (41) 17
Accumulated other comprehensive loss (450) (6)
Accumulated deficit (450,427) (382,052) (522,371) (585,342)
Total stockholders’ equity (deficit) 41,352
 (19,990) 208,366
 (32,481)
Total liabilities and stockholders’ equity (deficit) $165,977
 $101,793
 $519,945
 $261,515
See accompanying notes to consolidated financial statements.


F-2



HALOZYME THERAPEUTICS, INC.
CONSOLIDATED STATEMENTS OF OPERATIONS
(inIn thousands, except per share data)amounts)

 Year Ended December 31, Year Ended December 31,
 2014 2013 2012 2017 2016 2015
Revenues:            
Product sales, net $37,823
 $24,439
 $2,887
 $50,396
 $53,392
 $46,082
Royalties 63,507
 50,984
 30,975
Revenues under collaborative agreements 28,086
 30,327
 39,438
 202,710
 42,315
 58,000
Royalties 9,425
 33
 
Total revenues 75,334
 54,799
 42,325
 316,613
 146,691
 135,057
Operating expenses:            
Cost of product sales 22,732
 6,246
 1,094
 31,152
 33,206
 29,245
Research and development 79,696
 96,640
 70,044
 150,643
 150,842
 93,236
Selling, general and administrative 35,942
 32,347
 24,812
 53,816
 45,853
 40,028
Total operating expenses 138,370
 135,233
 95,950
 235,611
 229,901
 162,509
Operating loss (63,036) (80,434) (53,625)
Operating income (loss) 81,002
 (83,210) (27,452)
Other income (expense):            
Investment and other income, net 242
 229
 73
 2,592
 1,326
 422
Interest expense (5,581) (3,274) 
 (21,984) (19,977) (5,201)
Net loss $(68,375) $(83,479) $(53,552)
Income (loss) before income taxes 61,610
 (101,861) (32,231)
Income tax (benefit) expense (1,361) 1,162
 
Net income (loss) $62,971
 $(103,023) $(32,231)
            
Basic and diluted net loss per share $(0.56) $(0.74) $(0.48)
Net income (loss) per share:      
Basic $0.46
 $(0.81) $(0.25)
Diluted $0.45
 $(0.81) $(0.25)
            
Shares used in computing basic and diluted net loss per share 122,690
 112,805
 111,077
Shares used in computing net income (loss) per share:      
Basic 136,419
 127,964
 126,704
Diluted 139,068
 127,964
 126,704
See accompanying notes to consolidated financial statements.


F-3



HALOZYME THERAPEUTICS, INC.
CONSOLIDATED STATEMENTS OF COMPREHENSIVE LOSSINCOME (LOSS)
(inIn thousands)
  Year Ended December 31,
  2014 2013 2012
Net loss $(68,375) $(83,479) $(53,552)
Other comprehensive (loss) income:      
Unrealized (loss) gain on marketable securities (58) 17
 
Total comprehensive loss $(68,433) $(83,462) $(53,552)
  Year Ended December 31,
  2017 2016 2015
Net income (loss) $62,971
 $(103,023) $(32,231)
Other comprehensive income (loss):      
Unrealized (loss) gain on marketable securities (430) 93
 (58)
Foreign currency translation adjustment (14) 
 
Total comprehensive income (loss) $62,527
 $(102,930) $(32,289)
See accompanying notes to consolidated financial statements.


F-4



HALOZYME THERAPEUTICS, INC.
CONSOLIDATED STATEMENTS OF CASH FLOWS
(inIn thousands)
 Year Ended December 31, Year Ended December 31,
 2014 2013 2012 2017 2016 2015
Operating activities:            
Net loss $(68,375) $(83,479) $(53,552)
Adjustments to reconcile net loss to net cash used in operating activities:      
Net income (loss) $62,971
 $(103,023) $(32,231)
Adjustments to reconcile net income (loss) to net cash provided by (used in) operating activities:      
Share-based compensation 15,274
 9,538
 8,349
 30,670
 25,585
 20,838
Depreciation and amortization 1,762
 1,227
 1,079
 2,161
 2,410
 1,677
Non-cash interest expense 2,025
 156
 9
 1,761
 2,896
 1,243
Amortization of premiums on marketable securities, net 1,457
 1,116
 
Payment-in-kind interest expense on long-term debt 
 13,184
 879
(Accretion of discounts) amortization of premiums on marketable securities, net (303) 552
 
Loss on disposal of equipment 233
 
 7
 46
 8
 8
Deferral of unearned revenue 22,759
 701
 4,379
Recognition of deferred revenue (6,512) (9,304) (5,789)
Deferral of rent expense 13
 
 441
Recognition of deferred rent (185) (370) (276)
Other (16) 
 
Changes in operating assets and liabilities:            
Accounts receivable, net (52) 6,606
 (13,441) (6,453) 16,730
 (23,261)
Inventories (236) (3,499) (2,103) 9,477
 (5,134) (3,083)
Prepaid expenses and other assets (265) 1,959
 (4,421) 2,035
 5,626
 (15,774)
Restricted cash 
 (100) 50
Accounts payable and accrued expenses (816) 7,888
 (3,263) 15,629
 (244) 13,866
Deferred revenue 1,490
 9,297
 2,962
Other liabilities (15) (48) 45
Net cash used in operating activities (47,518) (49,339) (64,279)
Net cash provided by (used in) operating activities 134,053
 (50,383) (37,083)
Investing activities:            
Purchases of marketable securities (88,884) (48,947) 
 (398,187) (155,412) (71,482)
Proceeds from maturities of marketable securities 57,301
 3,375
 
 235,805
 81,783
 79,730
Purchases of property and equipment (1,368) (2,297) (1,413) (1,350) (3,137) (2,360)
Net cash used in investing activities (32,951) (47,869) (1,413)
Net cash (used in) provided by investing activities (163,732) (76,766) 5,888
Financing activities:            
Proceeds from issuance of common stock, net 107,713
 
 81,477
 134,874
 
 
Proceeds from issuance of common stock under equity incentive
plans, net
 6,788
 5,079
 1,680
Proceeds from issuance of long-term debt, net 
 19,985
 29,661
 
 203,006
 
Repayment of long-term debt (15,995) (54,250) 
Proceeds from issuance of common stock under equity incentive plans, net of taxes paid related to net share settlement 12,776
 1,865
 13,098
Net cash provided by financing activities 114,501
 25,064
 112,818
 131,655
 150,621
 $13,098
Net increase (decrease) in cash and cash equivalents 34,032
 (72,144) 47,126
Cash and cash equivalents at beginning of period 27,357
 99,501
 52,375
Cash and cash equivalents at end of period $61,389
 $27,357
 $99,501
Net increase (decrease) in cash, cash equivalents and restricted cash 101,976
 23,472
 (18,097)
Cash, cash equivalents and restricted cash at beginning of period 67,264
 43,792
 61,889
Cash, cash equivalents and restricted cash at end of period $169,240
 $67,264
 $43,792
            
Supplemental disclosure of cash flow information:      
Interest paid $3,460
 $3,099
 $19
Supplemental disclosure of non-cash investing and financing activities:      
Amounts accrued for purchases of property and equipment $156
 $100
 $154
Capitalized property and liability associated with a build-to-suit lease
arrangement
 $
 $(1,450) $1,450


  Year Ended December 31,
  2017 2016 2015
Supplemental disclosure of cash flow information:      
Interest paid $20,295
 $3,886
 $3,775
Income taxes paid $3,015
 $1,441
 $
Supplemental disclosure of non-cash investing and financing activities:      
Amounts accrued for purchases of property and equipment $189
 $75
 $473
See accompanying notes to consolidated financial statements.


F-5



HALOZYME THERAPEUTICS, INC.
CONSOLIDATED STATEMENTS OF STOCKHOLDERS’ EQUITY (DEFICIT)
(in thousands)
 Common Stock 
Additional
Paid-In
Capital
 
Accumulated
Other
Comprehensive
Income (Loss)
 
Accumulated
Deficit
 
Total
Stockholders’
Equity (Deficit)
 Common Stock 
Additional
Paid-In
Capital
 
Accumulated
Other
Comprehensive
Income (Loss)
 
Accumulated
Deficit
 
Total
Stockholders’
Equity (Deficit)
 Shares Amount  Shares Amount 
BALANCE AT JANUARY 1, 2012 103,989
 $104
 $255,818
 $
 $(245,021) $10,901
BALANCE AT JANUARY 1, 2015 125,721
 $126
 $491,694
 $(41) $(450,427) $41,352
Share-based compensation expense 
 
 20,838
 
 
 20,838
Issuance of common stock pursuant to exercise of stock options and vesting of restricted stock units, net 2,056
 2
 13,096
 
 
 13,098
Issuance of restricted stock awards, net 375
 
 
 
 
 
Other comprehensive loss 
 
 
 (58) 
 (58)
Net loss 
 
 
 
 (32,231) (32,231)
BALANCE AT DECEMBER 31, 2015 128,152
 128
 525,628
 (99) (482,658) 42,999
Adjustment to beginning retained earnings 
 
 (339) 
 339
 
Share-based compensation expense 
 
 25,585
 
 
 25,585
Issuance of common stock pursuant to exercise of stock options and vesting of restricted stock units and performance restricted stock units, net 570
 1
 1,947
 
 
 1,948
Issuance of restricted stock awards, net 780
 1
 (84) 
 
 (83)
Other comprehensive income 
 
 
 93
 
 93
Net loss 
 
 
 
 (103,023) (103,023)
BALANCE AT DECEMBER 31, 2016 129,502
 130
 552,737
 (6) (585,342) (32,481)
Share-based compensation expense 
 
 8,349
 
 
 8,349
 
 
 30,670
 
 
 30,670
Issuance of common stock for cash, net 7,820
 8
 81,469
 
 
 81,477
 11,500
 11
 134,863
 
 
 134,874
Issuance of common stock pursuant to exercise of stock options and vesting of restricted stock units, net 526
 1
 1,679
 
 
 1,680
Issuance of restricted stock awards 374
 
 
 
 
 
Net loss 
 
 
 
 (53,552) (53,552)
BALANCE AT DECEMBER 31, 2012 112,709
 113
 347,315
 
 (298,573) 48,855
Share-based compensation expense 
 
 9,538
 
 
 9,538
Issuance of common stock pursuant to exercise of stock options and vesting of restricted stock units, net 1,363
 1
 5,078
 
 
 5,079
Issuance of restricted stock awards 462
 1
 (1) 
 
 
Other comprehensive income 
 
 
 17
 
 17
Net loss 
 
 
 
 (83,479) (83,479)
BALANCE AT DECEMBER 31, 2013 114,534
 115
 361,930
 17
 (382,052) (19,990)
Share-based compensation expense 
 
 15,274
 
 
 15,274
Issuance of common stock for cash, net 8,846
 9
 107,704
 
 
 107,713
Issuance of common stock pursuant to exercise of stock options and vesting of restricted stock units, net 1,552
 1
 6,787
 
 
 6,788
Issuance of restricted stock awards 789
 1
 (1) 
 
 
Issuance of common stock pursuant to exercise of stock options and vesting of restricted stock units and performance restricted stock units, net 1,796
 2
 12,774
 
 
 12,776
Cancellation of restricted stock awards, net (9) 
 
 
 
 
Other comprehensive loss 
 
 
 (58) 
 (58) 
 
 
 (444) 
 (444)
Net loss 
 
 
 
 (68,375) (68,375)
BALANCE AT DECEMBER 31, 2014 125,721
 $126
 $491,694
 $(41) $(450,427) $41,352
Net income 
 
 
 
 62,971
 62,971
BALANCE AT DECEMBER 31, 2017 142,789
 $143
 $731,044
 $(450) $(522,371) $208,366
See accompanying notes to consolidated financial statements.


F-6



Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements
1.Organization and Business
1. Organization and Business
Halozyme Therapeutics, Inc. is a biotechnology company focused on developing and commercializing novel oncology therapies. We are seeking to translate our unique knowledge of the tumor microenvironment to create novel therapies that canhave the potential to improve cancer patient survival. Our research primarily focuses on human enzymes that alter the extracellular matrix and tumor microenvironment. The extracellular matrix is a complex matrix of proteins and carbohydrates surrounding the cell that provides structural support in tissues and orchestrates many important biological activities, including cell migration, signaling and survival. Over many years, we have developed unique technology and scientific expertise enabling us to pursue this target-rich environment for the development of therapies.
Our proprietary enzymes can beare used to facilitate the delivery of injected drugs and fluids, potentially enhancing the efficacy and the convenience of other drugs or can be used to alter abnormal tissue structures for potential clinical benefit. We have chosen to exploit our technology and expertise inusing a balanced waytwo pillar strategy that we believe enables us to modulate bothmanage risk and spendcost by: (1) developing our own proprietary products in therapeutic areas with significant unmet medical needs, with a focus on oncology, and (2) licensing our technology to biopharmaceutical companies to collaboratively develop products whichthat combine our technology with the collaborators'collaborators’ proprietary compounds.
The majority of our approved product and product candidates are based on rHuPH20, our patented recombinant human hyaluronidase enzyme. rHuPH20 is the active ingredient in our first commercially approved product, Hylenex® recombinant, and it works by temporarily breaksbreaking down hyaluronic acid (HA)hyaluronan (or “HA”), a naturally occurring substancecomplex carbohydrate that is a major component of the extracellular matrix in tissues throughout the body such as skin and cartilage. We believe this temporary degradation creates an opportunistic window for the improved subcutaneous delivery of injectable biologics, such as monoclonal antibodies and other large therapeutic molecules, as well as small molecules and fluids. We refer to the application of rHuPH20 to facilitate the delivery of other drugs or fluids as Enhanze technology. rHuPH20 is also the active ingredient in our first commercially approved product, HylenexENHANZE® recombinant.Technology. We license the ENHANZE Technology to form collaborations with biopharmaceutical companies that develop or market drugs requiring or benefiting from injection via the subcutaneous route of administration.
We currently have ENHANZE collaborations with F. Hoffmann-La Roche, Ltd. and Hoffmann-La Roche, Inc. (“Roche”), Baxalta US Inc. and Baxalta GmbH (Baxalta Incorporated was acquired by Shire plc in June 2016) (“Baxalta”), Pfizer Inc. (“Pfizer”), Janssen Biotech, Inc. (“Janssen”), AbbVie, Inc. (“AbbVie”), Eli Lilly and Company (“Lilly”), Bristol-Myers Squibb Company (“BMS”) and Alexion Pharma Holding (“Alexion”).We receive royalties from two of these collaborations, including royalties from sales of one product from the Baxalta collaboration and two products from the Roche collaboration. Future potential revenues from the sales and/or royalties of our approved products, product candidates, and ENHANZE collaborations will depend on the ability of Halozyme and our collaborators to develop, manufacture, secure and maintain regulatory approvals for approved products and product candidates and commercialize product candidates.
Our proprietary development pipeline consists primarily of pre-clinical and clinical stage product candidates in oncology and diabetes.oncology. Our lead oncology program is PEGPH20 (PEGylatedPegvorhyaluronidase alfa (“PEGPH20”, PEGylated recombinant human hyaluronidase), a new molecular entity under developmentwe are developing in combination with currently approved cancer therapies as a candidate for the systemic treatment of tumors that accumulate HA. WhenWe have demonstrated that when HA accumulates in a tumor, it can cause higher pressure in the tumor, reducing blood flow into the tumor and with that, reduced access of cancer therapies to the tumor. PEGPH20 workshas been demonstrated in animal models to work by temporarily degrading HA surrounding some cancer cells and resultsresulting in reduced pressure and increased blood flow to the cancer withtumor thereby enabling increased amounts of anticancer treatments administered concomitantly gaining access to the tumor. WeThrough our efforts and efforts of our partners and collaborators, we are currently in Phase 3 clinical testing for PEGPH20 with ABRAXANE® (nab-paclitaxel) and gemcitabine in stage IV pancreatic ductal adenocarcinoma (“PDA”) (HALO 109-301), in Phase 1b clinical testing forPEGPH20 with KEYTRUDA® (pembrolizumab) in non-small cell lung cancer and gastric cancer (HALO 107-101), in Phase 1b/2 clinical testing for PEGPH20 with HALAVEN® (eribulin) in patients treated with up to two lines of prior therapy for HER2-negative metastatic pancreaticbreast cancer, (Study 109-202),in Phase 1b/2 clinical testing for PEGPH20 with Tecentriq® (atezolizumab) in patients with previously treated metastatic PDA, in Phase 1b/2 clinical testing for PEGPH20 with Tecentriq in

F-8

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


patients with gastric cancer and we have recently initiated ain Phase 1b/2 clinical trialtesting for PEGPH20 with Tecentriq in non-small cell lungpatients with cholangiocarcinoma and gall bladder cancer (Study 107-201)(HALO 110-101/MATRIX).
Regarding Enhanze, we currently have collaborations with F. Hoffmann-La Roche, Ltd. and Hoffmann-La Roche, Inc. (Roche), Baxter Healthcare Corporation (Baxter), Pfizer Inc. (Pfizer) and Janssen Biotech, Inc. (Janssen), with one product approved in the U.S. and three products approved for marketing in Europe from which we are receiving royalties and several others at various stages of development.
Except where specifically noted or the context otherwise requires, references to “Halozyme,” “the Company,” “we,” “our,” and “us” in these Notesnotes to Consolidated Financial Statementscondensed consolidated financial statements refer to Halozyme Therapeutics, Inc. and its wholly owned subsidiary, Halozyme, Inc., and Halozyme, Inc.'s’s wholly owned subsidiary,subsidiaries, Halozyme Holdings Ltd.


, Halozyme Royalty LLC, Halozyme Switzerland GmbH and Halozyme Switzerland Holdings GmbH.
F-7


2. Summary of Significant Accounting Policies

2.Summary of Significant Accounting Policies
Basis of Presentation
The consolidated financial statements include the accounts of Halozyme Therapeutics, Inc. and our wholly owned subsidiary, Halozyme, Inc., and Halozyme, Inc.'s’s wholly owned subsidiary,subsidiaries, Halozyme Holdings Ltd., Halozyme Royalty LLC, Halozyme Switzerland GmbH and Halozyme Switzerland Holdings GmbH. All intercompany accounts and transactions have been eliminated.
Reclassifications
Certain prior period amounts have been reclassified to conform to the current period presentation. Specifically, we have reclassified $0.6 million from accrued expenses to other long-term liabilities in the consolidated balance sheet at December 31, 2013.
Use of Estimates
The preparation of consolidated financial statements in conformity with U.S. generally accepted accounting principles (“U.S. GAAP”) requires management to make estimates and assumptions that affect the amounts reported in our consolidated financial statements and accompanying notes. On an ongoing basis, we evaluate our estimates and judgments, which are based on historical and anticipated results and trends and on various other assumptions that management believes to be reasonable under the circumstances. By their nature, estimates are subject to an inherent degree of uncertainty and, as such, actual results may differ from management’s estimates.
Cash Equivalents and Marketable Securities
Cash equivalents consist of highly liquid investments, readily convertible to cash, that mature within ninety days or less from the date of purchase. OurAs of December 31, 2017, our cash equivalents consistconsisted of money market funds.funds and commercial paper.
Marketable securities are investments with original maturities of more than ninety days from the date of purchase that are specifically identified to fund current operations. Marketable securities are considered available-for-sale. These investments are classified as current assets, even though the stated maturity date may be one year or more beyond the current balance sheet date which reflects management'smanagement’s intention to use the proceeds from the sale of these investments to fund our operations, as necessary. Such available-for-sale investments are carried at fair value with unrealized gains and losses recorded in other comprehensive gain (loss) and included as a separate component of stockholders'stockholders’ equity (deficit). The cost of marketable securities is adjusted for amortization of premiums or accretion of discounts to maturity, and such amortization or accretion is included in investment and other income, net in the condensed consolidated statements of operations. We use the specific identification method for calculating realized gains and losses on marketable securities sold. Realized gains and losses and declines in value judged to be other-than-temporary on marketable securities, if any, are included in investment and other income, net in the consolidated statements of operations.
Restricted Cash
Under the terms of the leases onof our facilities, we are required to maintain letters of credit as security deposits during the terms of such leases. At December 31, 20142017 and 2013,2016, restricted cash of $0.5 million was pledged as collateral for the letters of credit.
Fair Value of Financial Instruments
The authoritative guidance for fair value measurements establishes a three tier fair value hierarchy, which prioritizes the inputs used in measuring fair value. These tiers include: Level 1, defined as observable inputs such as quoted prices in active markets; Level 2, defined as inputs other than quoted prices in active markets that are either directly or indirectly observable; and Level 3, defined as unobservable inputs in which little or no market data exists, therefore requiring an entity to develop its own assumptions.

F-9

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Our financial instruments include cash equivalents, available-for-sale marketable securities, accounts receivable, prepaid expenses and other assets, accounts payable, accrued expenses and long-term debt. Fair value estimates of these instruments are made at a specific point in time, based on relevant market information. These estimates may be subjective in nature and involve uncertainties and matters of significant judgment and therefore cannot be determined with precision. The carrying amount of cash equivalents,


F-8



accounts receivable, prepaid expenses and other assets, accounts payable and accrued expenses are generally considered to be representative of their respective fair values because of the short-term nature of those instruments. Further, basedBased on Level 3 inputs and the borrowing rates currently available for loans with similar terms, we believe the fair value of long-term debt approximates its carrying value.
Available-for-sale marketable securities consist of corporate debt securities, U.S. Treasury securities and commercial paper, and certificates of deposit and wereare measured at fair value using Level 1 and Level 2 inputs. Level 2 financial instruments are valued using market prices on less active markets and proprietary pricing valuation models with observable inputs, including interest rates, yield curves, maturity dates, issue dates, settlement dates, reported trades, broker-dealer quotes, issue spreads, benchmark securities or other market related data. We obtain the fair value of Level 2 investments from our investment manager, who obtains these fair values from a third-party pricing service.source. We validate the fair values of Level 2 financial instruments provided by our investment manager by comparing these fair values to a third-party pricing source.
The following table summarizes, by major security type, our cash equivalents and marketable securities that are measured at fair value on a recurring basis and are categorized using the fair value hierarchy (in thousands):
  December 31, 2014 December 31, 2013
  Level 1 Level 2 Total estimated fair value Level 1 Level 2 Total estimated fair value
Cash equivalents:            
Money market funds $42,685
 $
 $42,685
 $5,711
 $
 $5,711
             
Available-for-sale marketable
   securities:
            
Corporate debt securities 
 74,234
 74,234
 
 35,147
 35,147
Commercial paper 
 
 
 
 5,999
 5,999
Certificate of deposit 
 
 
 
 3,000
 3,000
  $42,685
 $74,234
 $116,919
 $5,711
 $44,146
 $49,857
There were no transfers between Level 1 and Level 2 of the fair value hierarchy for the years ended December 31, 2014 and 2013. We have no instruments that are classified within Level 3 as of December 31, 2014 and 2013.
Concentrations of Credit Risk, Sources of Supply and Significant Customers
We are subject to credit risk from our portfolio of cash equivalents and marketable securities. These investments were made in accordance with our investment policy which specifies the categories, allocations, and ratings of securities we may consider for investment. The primary objective of our investment activities is to preserve principal while at the same time maximizing the income we receive without significantly increasing risk. We maintain our cash and cash equivalent balances with one major commercial bank and marketable securities with another financial institution. Deposits held with the financial institutions exceed the amount of insurance provided on such deposits. We are exposed to credit risk in the event of a default by the financial institutions holding our cash, cash equivalents and marketable securities to the extent recorded on the consolidated balance sheet.sheets.
We are also subject to credit risk from our accounts receivable related to our product sales and revenues under our license and collaborative agreements. We have license and collaborative agreements with pharmaceutical companies under which we receive payments for license fees, milestone payments for specific achievements designated in the collaborative agreements, reimbursements of research and development services and supply of bulk formulation of rHuPH20. In addition, we sell Hylenex® recombinant in the United States to a limited number of established wholesale distributors in the pharmaceutical industry. Credit is extended based on an evaluation of the customer’s financial condition, and collateral is not required. Management monitors our exposure to accounts receivable by periodically evaluating the collectibility of the accounts receivable based on a variety of factors including the length of time the receivables are past due, the financial health of the customer and historical experience. Based upon the review of these factors, we recorded no allowance for doubtful accounts at December 31, 20142017 and 2013.2016. Approximately 76% and 81%86% of the accounts receivable balance at December 31, 2014 and 2013, respectively,2017 represents amounts due from Roche and Pfizer.Baxalta. Approximately 81% of the accounts receivable balance at December 31, 2016 represents amounts due from Roche and Baxalta.


F-9



The following table indicates the percentage of total revenues in excess of 10% with any single customer:
 Year Ended December 31,
 2014 2013 2012
Roche57% 64% 45%
Janssen20%  
Baxter3% 10% 17%
Pfizer1% 4% 22%
  Year Ended December 31,
  2017 2016 2015
Roche 38% 63% 42%
BMS 32%  
Alexion 13%  
Baxalta 7% 12% 7%
Lilly  6% 19%
AbbVie  4% 17%

F-10

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


We attribute revenues under collaborative agreements, including royalties, to the individual countries where the collaborator is headquartered. We attribute revenues from product sales to the individual countries to which the product is shipped. Worldwide revenues from external customers are summarized by geographic location in the following table (in thousands):
Year Ended December 31, Year Ended December 31,
2014 2013 2012 2017 2016 2015
United States$31,397
 $19,019
 $22,724
 $196,274
 $52,292
 $77,149
Switzerland42,791
 35,157
 18,913
 119,136
 93,067
 57,136
All other foreign1,146
 623
 688
 1,203
 1,332
 772
Total revenues$75,334
 $54,799
 $42,325
 $316,613
 $146,691
 $135,057
For the years ended December 31, 2014, 2013 and 2012, we had no foreign based operations. As of December 31, 20142017 and 2013,2016, we had $0.4 million and $0.8less than $0.1 million of research equipment in Germany.
We rely on two third-party manufacturers for the supply of bulk rHuPH20 for use in the manufacture of Hylenex recombinant and our other collaboration products and product candidates. Payments due to these suppliers represented 0%4% and 9%13% of the accounts payable balance at December 31, 20142017 and 2013,2016, respectively. We also rely on a third-party manufacturer for the fill and finish of Hylenex recombinant product under a contract. Payments due to this supplier represented 6%1% and 2% of the accounts payable balance at December 31, 20142017 and 2013,2016, respectively.
Accounts Receivable, Net
Accounts receivable is recorded at the invoiced amount and is non-interest bearing. Accounts receivable is recorded net of allowances for doubtful accounts, cash discounts for prompt payment, distribution fees and chargebacks. We recorded no allowance for doubtful accounts at December 31, 20142017 and 20132016 as the collectibility of accounts receivable was reasonably assured.
Inventories
Inventories are stated at lower of cost or market.net realizable value. Cost is determined on a first-in, first-out basis. Net realizable value is the estimated selling price in the ordinary course of business, less reasonably predictable costs of completion, disposal, and transportation. Inventories are reviewed periodically for potential excess, dated or obsolete status. Management evaluatesWe evaluate the carrying value of inventories on a regular basis, taking into account such factors as historical and anticipated future sales compared to quantities on hand, the price we expect to obtain for products in their respective markets compared with historical cost and the remaining shelf life of goods on hand.
Prior to receiving marketing approval from the U.S. Food and Drug Administration (“FDA”) or comparable regulatory agencies in foreign countries, costs related to purchases of bulk rHuPH20 and raw materials and the manufacturing of the product candidates are recorded as research and development expense. All direct manufacturing costs incurred after receiving marketing approval are capitalized as inventory. Inventories used in clinical trials are expensed at the time the inventories are packaged for the clinical trials.
As of December 31, 20142017 and 2013,2016, inventories consisted of $3.0$2.9 million and $2.6$2.3 million, respectively, of Hylenex recombinant inventory, respectively,net, and $3.4$2.2 million and $3.5$12.3 million, respectively, of bulk rHuPH20 respectively, for use in the manufacture of Roche'sBaxalta’s and Roche’s collaboration products. Roche received European marketing approval for Herceptin SC® and MabThera® SC in August 2013 and March 2014, respectively. As such, direct manufacturing costs of bulk rHuPH20 for these collaboration products incurred after the receipt of the European marketing approvals are capitalized as inventory.


F-10



Property and Equipment, Net
Property and equipment are recorded at cost, less accumulated depreciation and amortization. Equipment is depreciated using the straight-line method over theirits estimated useful liveslife of three years and leasehold improvements are amortized using the straight-line method over the estimated useful life of the asset or the lease term, whichever is shorter. Leased buildings under build-to-suit lease arrangements are capitalized and included in property and equipment when we are involved in the construction of the structural improvements or take construction risk prior

F-11

Halozyme Therapeutics, Inc.
Notes to the commencement of the lease. Upon completion of the construction under the build-to-suit leases, we assess whether those arrangements qualify for sales recognition under the sale-leaseback accounting guidance. If we continue to be the deemed owner, the facilities would be accounted for as financing leases.Consolidated Financial Statements — (Continued)


Impairment of Long-Lived Assets
We account for long-lived assets in accordance with authoritative guidance for impairment or disposal of long-lived assets. Long-lived assets are reviewed for events or changes in circumstances, which indicate that their carrying value may not be recoverable. For the yearyears ended December 31, 2014, we recorded an impairment of $0.2 million relating to manufacturing equipment. For the year ended December 31, 2013,2017 and 2016, there was no impairment of the value of long-lived assets.
Deferred Rent
Rent expense is recorded on a straight-line basis over the initial term of the lease. The difference between rent expense accrued and amounts paid under lease agreements is recorded as deferred rent and is included in accrued expenses and other long-term liabilities, as applicable, in the accompanying consolidated balance sheets.
Comprehensive Income (Loss)
Comprehensive income (loss) is defined as the change in equity during the period from transactions and other events and circumstances from non-owner sources.
Revenue Recognition
We generate revenues from product sales and collaborative agreements. Paymentspayments received under collaborative agreementsagreements. Collaborative agreement payments may include nonrefundable fees at the inception of the agreements, license fees, milestone and event-based payments for specific achievements designated in the collaborative agreements, reimbursements of research and development services and supply of bulk rHuPH20, and/or royalties on sales of products resulting from collaborative arrangements.
We recognize revenues in accordance with the authoritative guidance for revenue recognition. We recognize revenue when all of the following criteria are met: (1) persuasive evidence of an arrangement exists; (2) delivery has occurred or services have been rendered; (3) the seller’s price to the buyer is fixed or determinable; and (4) collectibility is reasonably assured.
Product Sales, Net
Hylenex Recombinant
In December 2011, we reintroduced Hylenex recombinant to the market. We sell Hylenex recombinant in the U.S. to wholesale pharmaceutical distributors, who sell the product to hospitals and other end-user customers. Sales to wholesalers provide for selling prices that are fixed on the date of sale, although we offer discounts to certain group purchasing organizations (“GPOs”), hospitals and government programs. The wholesalers take the title to the product, bear the risk of loss of ownership and have economic substance to the inventory. Further, we have no significant obligations for future performance to generate pull-through sales.
Prior to December 31, 2013, Hylenex recombinant had a limited sales history, and we could not reliably estimate expected returns and chargebacks of the product at the time the product was sold to the wholesalers. Accordingly, we deferred the recognition of revenue on sales of Hylenex recombinant to wholesalers, and instead, recognized revenue at the time when evidence existed to confirm that pull-through sales from wholesalers to the hospitals or other end-user customers had occurred or the right of return no longer existed, whichever occurred earlier. At the time product sales revenue was recognized, we recorded allowances for product returns and chargebacks based on our best estimates at the time. Shipments of product that were not recognized as revenue were treated as deferred revenue.


F-11



At December 31, 2013, we hadWe have developed sufficient historical experience and data to reasonably estimate future returns and chargebacks of Hylenex recombinant. As a result, effective December 31, 2013, we began recognizingrecognize Hylenex recombinant product sales and related cost of product sales at the time title transfers to the wholesalers.
Upon recognition of revenue from product sales of Hylenex recombinant, we record certain sales reserves and allowances as a reduction to gross revenue. These reserves and allowances include:
Product Returns. We allow the wholesalers to returninclude amounts for product that is damaged or received in error. In addition, we accept unused product to be returned beginning six months prior to and ending twelve months following product expiration. Our estimates for expected returns of expired products are based(based primarily on an ongoing analysis of historical return patterns.
Distribution Fees. Thepatterns), distribution fees, based on contractually determined rates, arise from contractual agreements we have with certain wholesalers for distribution services they provide with respect to Hylenex recombinant. These fees are generally a fixed percentage of the price of the product purchased by the wholesalers.
Prompt Payment Discounts. We offer cash discounts to certain wholesalers as an incentive to meet certain payment terms. We estimate prompt payment discounts, based on contractual terms, historical utilization rates, as available, and our expectations regarding future utilization rates.    
Other DiscountsGPO fees and Fees. We provideother discounts to end-user members of certain GPOs under collective purchasing contracts between us and the GPOs. We also provide discounts to certain hospitals, who are members of the GPOs, with which we do not have contracts. The end-user members purchase products from the wholesalers at a contracted discounted price, and the wholesalers then charge back to us the difference between the current retail price and the price the end-users paid for the product. We also incur GPO administrative service fees for these transactions. In addition, we provide predetermined discounts under certain government programs. Our estimate for these chargebacks and fees take into consideration contractual terms, historical utilization rates, as available, and our expectations regarding future utilization rates.
Allowances for product returns and chargebacks are based on amounts owed or to be claimed on the related sales. We believe that our estimated product returns for Hylenex recombinant requires a high degree of judgment and is subject to change based on our experience and certain quantitative and qualitative factors. In order to develop a methodology to reliably estimate future returns and provide a basis for recognizing revenue on sales to wholesale distributors, we analyzed many factors, including, without limitation: (1) actual Hylenex recombinant product return history, taking into account product expiration dating at the time of shipment, (2) re-order activities of the wholesalers as well as their customers and (3) levels of inventory at the wholesale channel. We have monitored actual return history on an individual product lot basis since product launch. We considered the dating of product at the time of shipment into the distribution channel and changes in the estimated levels of inventory within the distribution channel to estimate our exposure for returned product. We considered historical chargebacks activity and current contract prices to estimate our exposure for returned product. Based on such data, we believe we have the information needed to reasonably estimate product returns and chargebacks.fees.
We recognize product sales reserves and allowances as a reduction of product sales in the same period the related revenue is recognized. Because of the shelf life of Hylenex recombinant and our lengthy return period, there may be a significant period of time between when the product is shipped and when we issue credits on returned product. If actual product return results differ from our estimates, we will be required to make adjustments to these allowances in the future, which could have an effect on product sales revenue and earnings in the period of adjustments.

F-12

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Bulk rHuPH20
Subsequent to receiving marketing approval from the FDA or comparable regulatory agencies in foreign countries, sales of bulk rHuPH20 for use in collaboration commercial products are recognized as product sales when the materials have met all the specifications required for the customer'scustomer’s acceptance and title and risk of loss have transferred to the customer. Following the receiptsreceipt of European marketing approvals of Roche'sRoche’s Herceptin SC product in August 2013 and MabThera® SC product in March 2014 and Baxter'sBaxalta’s HYQVIA® product in May 2013, revenue from the sales of bulk rHuPH20 for these collaboration products arehas been recognized as product sales. For the years ended December 31, 2014 and 2013, we recognized $23.5 million and $13.7 million in product sales of bulk rHuPH20 for Roche's collaboration products, respectively. For the years ended December 31, 2014 and 2013, we recognized zero and $1.1 million in product sales of bulk rHuPH20 for Baxter's collaboration product, respectively.


F-12



Revenues under Collaborative Agreements
We have entered into license and collaboration agreements under which theour collaborators obtained worldwide rights for the use of our proprietary rHuPH20 enzyme in the development and commercialization of their biologic compounds identified as targets. These agreements may also contain other elements. Pursuant to the collaborators’ biologic compounds. The collaborativeterms of these agreements, contain multiple elementscollaborators could be required to make various payments to us for each target, including nonrefundable payments at the inception of the arrangement,upfront license fees, exclusivity fees, payments based on achievement of specified milestones designated in the collaborative agreements, annual maintenance fees, reimbursements of research and development services, payments for supply of bulk rHuPH20 forused by the collaborator and/or royalties on sales of products resulting from collaborative agreements. We analyze each element of our collaborative agreements and consider a variety of factors in determining the appropriate method of revenue recognition of each element.
In order to account for the multiple-element arrangements, we identify the deliverables included within the collaborative agreement and evaluate which deliverables represent units of accounting. We then determine the appropriate method of revenue recognition for each unit based on the nature and timing of the delivery process. Analyzing the arrangement to identify deliverables requires the use of judgment, and each deliverable may be an obligation to deliver services, a right or license to use an asset, or another performance obligation. The deliverables under our collaborative agreements include (i) the license to our rHuPH20 technology, (ii) at the collaborator’s request, research and development services which are reimbursed at contractually determined rates, and (iii) at the collaborator’s request, supply of bulk rHuPH20 which is reimbursed at our cost plus a margin. A delivered item is considered a separate unit of accounting when the delivered item has value to the collaborator on a standalone basis based on the consideration of the relevant facts and circumstances for each arrangement. Factors considered inWe base this determination includeon the collaborators’ ability to use the delivered items on their own without us supplying undelivered items, which we determine taking into consideration factors such as the research capabilities of the collaborator, and the availability of research expertise in this field in the general marketplace, and the ability to procure the supply of bulk rHuPH20 from the marketplace.
Arrangement consideration is allocated at the inception of the agreement to all identified units of accounting based on their relative selling price. The relative selling price for each deliverable is determined using vendor specific objective evidence (“VSOE”) of selling price or third-party evidence of selling price if VSOE does not exist. If neither VSOE nor third-party evidence of selling price exists, we use our best estimate of the selling price for the deliverable. The amount of allocable arrangement consideration is limited to amounts that are not contingent upon the delivery of additional items or meeting other specified performance conditions. The consideration received is allocated among the separate units of accounting and the applicable revenue recognition criteria are applied to each of the separate units. Changes in the allocation of the sales price between delivered and undelivered elements can impact revenue recognition but do not change the total revenue recognized under any agreement.
Nonrefundable upfront license fee paymentsfees are recognized upon delivery of the license if facts and circumstances dictate that the license has standalone value from the undelivered items, which generally include research and development services and the manufacture of bulk rHuPH20, the relative selling price allocation of the license is equal to or exceeds the upfront license fee, persuasive evidence of an arrangement exists, our price to the collaborator is fixed or determinable and collectibility is reasonably assured. Upfront license fee paymentsfees are deferred if facts and circumstances dictate that the license does not have standalone value. The determination of the length of the period over which to defer revenue is subject to judgment and estimation and can have an impact on the amount of revenue recognized in a given period.

F-13

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


When collaborators have rights to elect additional targets, the rights are assessed as to whether they represent deliverables at the inception of the arrangement. In assessing these contingent deliverables, we consider whether the right is a substantive option. We consider a right to be a substantive option if the election of the additional targets is not essential to the functionality of the other elements in the arrangement and if we are truly at risk of the right being exercised. If the right is determined to be a substantive option, we further consider whether the right is priced at a significant and incremental discount that should be accounted for as an element of the arrangement. If a right is determined to be a substantive option and is not priced at a significant and incremental discount, it is not treated as a deliverable in the arrangement and receives no allocation at the inception of the arrangement of the original arrangement consideration. The termsright is then accounted for when and if it is exercised. When collaborators have a right to return targets, the right is assessed as to whether the target may be returned for a refund of the purchase price, for a credit applied to amounts owed, or in exchange for other dissimilar products. We also assess if we have sufficient history to estimate the likelihood of return. If a right of return is considered to exist and we determine there is not sufficient history to estimate the likelihood of return, the consideration allocated to returnable targets is recorded as deferred revenue on the consolidated balance sheet until the right of return is exercised or expires.
Certain of our collaborative agreements provide for milestone payments upon achievement of certain development and regulatory events and/or specified sales volumes of commercialized products by the collaborator. We account for milestone payments in accordance with the provisions of ASU No. 2010-17, Revenue Recognition - Milestone Method(“Milestone Method of Accounting”). We recognize consideration that is contingent upon the achievement of a milestone in its entirety as revenue in the period in which the milestone is achieved only if the milestone is substantive in its entirety. A milestone is considered substantive when it meets all of the following criteria:
1.The consideration is commensurate with either the entity’s performance to achieve the milestone or the enhancement of the value of the delivered item(s) as a result of a specific outcome resulting from the entity’s performance to achieve the milestone,milestone;
2.The consideration relates solely to past performance,performance; and
3.The consideration is reasonable relative to all of the deliverables and payment terms within the arrangement.
A milestone is defined as an event (i) that can only be achieved based in whole or in part on either the entity’s performance or on the occurrence of a specific outcome resulting from the entity’s performance, (ii) for which there is substantive uncertainty at the date the arrangement is entered into that the event will be achieved, and (iii) that would result in additional payments being due to the vendor.


F-13



Reimbursements of research and development services are recognized as revenue during the period in which the services are performed as long as there is persuasive evidence of an arrangement, the fee is fixed or determinable and collection of the related receivable is reasonably assured. Revenue from the manufacture of bulk rHuPH20 is recognized when the materials have met all specifications required for the collaborator'scollaborator’s acceptance and title and risk of loss have transferred to the collaborator. We do not directly control when any collaborator will request research and development services or supply of bulk rHuPH20; therefore, we cannot predict when we will recognize revenues in connection with research and development services and supply of bulk rHuPH20.
Since we receive royalty reports 60 days after quarter end, royalty revenue from sales of collaboration products by our collaborators will beis recognized in the quarter following the quarter in which the corresponding sales occurred.
The collaborative agreements typically provide the collaborators the right to terminate such agreement in whole or on a product-by-product or target-by-target basis at any time upon 30 to 90 days prior written notice to us. There are no performance, cancellation, termination or refund provisions in any of our collaborative agreements that contain material financial consequences to us.
Refer to Note 4, Collaborative Agreements, for further discussion on our collaborative arrangements.

F-14

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Cost of Product Sales
Cost of product sales consists primarily of raw materials, third-party manufacturing costs, fill and finish costs, freight costs, internal costs and manufacturing overhead associated with the production of Hylenex recombinant and bulk rHuPH20 for use in approved collaboration products. Cost of product sales also consists of the write-down of excess, dated and obsolete inventories and the write-off of any inventories that do not meet certain product specifications, if any.
Prior to European marketing approvals of Roche's collaboration products Herceptin SC in August 2013 and MabThera SC in March 2014, and Baxter's collaboration product HYQVIA in May 2013, all costs related to the manufacturing of bulk rHuPH20 for these collaboration products were charged to research and development expenses in the periods such costs were incurred. Therefore, cost of product sales of these bulk rHuPH20 for the year ended December 31, 2013 was materially reduced due to the exclusion of the manufacturing costs that were charged to research and development expenses in the periods prior to receiving marketing approvals.
For the year ended December 31, 2014, cost of product sales of bulk rHuPH20 excluded $1.0 million in manufacturing costs, of which $0.9 million and $0.1 million were charged to research and development expenses in the years ended December 31, 2013 and 2012, respectively. For the year ended December 31, 2013, cost of product sales of bulk rHuPH20 excluded $10.0 million in manufacturing costs, of which $9.0 million and $1.0 million were charged to research and development expenses in the years ended December 31, 2013 and 2012, respectively. As of December 31, 2013, bulk rHuPH20 inventory for collaboration products which were sold in 2014 excluded $1.0 million in manufacturing costs. There was no bulk rHuPH20 for HyQvia on hand as of December 31, 2014 and 2013.
Research and Development Expenses
Research and development expenses include salaries and benefits, facilities and other overhead expenses, external clinical trial expenses, research related manufacturing services, contract services and other outside expenses. Research and development expenses are charged to operationsoperating expenses as incurred when these expenditures relate to our research and development efforts and have no alternative future uses. After receiving approval from the FDA or comparable regulatory agencies in foreign countries for a product, costs related to purchases and manufacturing of bulk rHuPH20 for such product are capitalized as inventory. The manufacturing costs of bulk rHuPH20 for the collaboration products, Herceptin SC, MabThera SC (RITUXAN HYCELA™ in the U.S.) and HYQVIA, incurred after the receipt of the European marketing approvals are capitalized as inventory.
In accordance with certain research and development agreements, weWe are obligated to make certain upfront payments upon execution of the agreement.certain research and development agreements. Advance payments, including nonrefundable amounts, for goods or services that will be used or rendered for future research and development activities are deferred and capitalized.deferred. Such amounts will beare recognized as an expense as the related goods are delivered or the related services are performed or such time when we do not expect the goods to be delivered or services to be performed.
Milestone payments that we make in connection with in-licensed technology for a particular research and development project that have no alternative future uses (in other research and development projects or otherwise) and therefore no separate economic valuesvalue are expensed as research and development costs at the time the costs are incurred. We currently have no in-licensed technologies that have alternative future uses in research and development projects or otherwise.


F-14



Clinical Trial Expenses
PaymentsWe make payments in connection with our clinical trials are often made under contracts with multiple contract research organizations that conductsupport conducting and managemanaging clinical trials on our behalf.trials. The financial terms of these agreements are subject to negotiation and vary from contract to contract and may result in uneven payment flows. Generally, these agreements set forth the scope of work to be performed at a fixed fee, unit price or on a time and materials basis. PaymentsA portion of our obligation to make payments under these contracts dependdepends on factors such as the successful enrollment or treatment of patients or the completion of other clinical trial milestones.
Expenses related to clinical trials are accrued based on our estimates and/or representations from service providers regarding work performed, including actual level of patient enrollment, completion of patient studies and progress of the clinical trials. Other incidental costs related to patient enrollment or treatment are accrued when reasonably certain. If the contracted amounts we are obligated to pay under our clinical trial agreements are modified (for instance, as a result of changes in the clinical trial protocol or scope of work to be performed), we modifyadjust our accruals accordingly on a prospective basis. Revisions in the scope of a contractto our contractual payment obligations are charged to expense in the period in which the facts that give rise to the revision become reasonably certain. Historically, we have had no material changes in clinical trial expense accruals that had a material impact on our consolidated results of operations or financial position.
Share-Based Compensation
We record compensation expense associated with stock options, restricted stock awards (“RSAs”), restricted stock units (“RSUs”), and other share-based awardsRSUs with performance conditions (“PRSUs”) in accordance with the authoritative guidance for stock-based compensation. The cost of employee services received in exchange for an award of an equity instrument is measured at the grant date, based on the estimated fair value of the award, and is recognized as expense on a straight-line basis net of estimated forfeitures, over the requisite service period of the award. Share-based compensation expense recognized during the period is based on the value of the portion of share-based payment awards that is ultimately expected to vest during the period. Share-based compensation expense for an award with a performance condition is recognized when the achievement of such performance condition is determined to be probable. If the outcome of such performance condition is not determined to be probable or is not met, no compensation expense is recognized and any previously recognized compensation expense is reversed. AsForfeitures are recognized as a reduction of share-based compensation expense recognized is based on awards ultimately expectedas they occur.

F-15

Halozyme Therapeutics, Inc.
Notes to vest, it has been reduced for estimated forfeitures. The guidance requires forfeitures to be estimated at the time of grant and revised, if necessary, in subsequent periods if actual forfeitures differ from those estimates. Pre-vesting forfeitures were estimated to be approximately 10% for employees for the years ended December 31, 2014, 2013 and 2012 based on our historical experience for the years then ended.Consolidated Financial Statements — (Continued)


Income Taxes
We provide for income taxes using the liability method. Under this method, deferred income tax assets and liabilities are determined based on the differences between the financial statement carrying amounts of existing assets and liabilities at each year end and their respective tax bases and are measured using enacted tax rates in effect for the year in which the differences are expected to affect taxable income. Significant judgment is required by management to determine our provision for income taxes, our deferred tax assets and liabilities, and the valuation allowance to record against our net deferred tax assets, which are based on complex and evolving tax regulations throughout the world. Deferred tax assets and other tax benefits are recorded when it is more likely than not that the position will be sustained upon audit. ValuationWhile we have begun to utilize certain of our net operating losses, we have not yet established a track record of profitability. Accordingly, valuation allowances have been establishedrecorded to reduce our net deferred tax assets to zero, with the exception of the alternative minimum tax ("AMT") credit carryover. Under the Tax Cuts and Jobs Act enacted in December 2017, the AMT credit carryover will either be utilized, or if unutilized fully refunded in 2022. For all other deferred tax assets the valuation allowance will reduce the net value to zero until such time as we believe that it is more likely than not that such assets will not be realized.can demonstrate an ability to realize them.
Net LossIncome (Loss) Per Share
Basic net lossincome (loss) per common share is computed by dividing lossnet income (loss) for the period by the weighted average number of common shares outstanding during the period, without consideration for common stock equivalents. StockOutstanding stock options, unvested restricted stock awards (“RSAs”)RSAs, unvested RSUs and unvested restricted stock units (“RSUs”)PRSUs are considered common stock equivalents and are only included in the calculation of diluted earnings per common share when net income is reported and their effect is dilutive. BecauseFor the years ended December 31, 2017, 2016 and 2015, approximately 7.1 million, 13.8 million, and 9.8 million shares, respectively, of our net loss, outstanding stock options, outstandingunvested RSAs, unvested RSUs and unvested RSAs totaling approximately 8,405,903, 8,070,141 and 7,444,333PRSUs were excluded from the calculation of diluted net lossincome (loss) per common share for the years ended December 31, 2014, 2013 and 2012, respectively, because their effect was anti-dilutive. SinceA reconciliation of the performance conditions for performance restricted stock units (“PRSUs”) were not satisfied at December 31, 2014, such securities are excluded from potentially dilutive securities.numerators and the denominators of the basic and diluted net income (loss) per common share computations is as follows (in thousands, except per share amounts):


F-15
  Year Ended December 31,
  2017 2016 2015
Numerator:      
Net income (loss) $62,971
 $(103,023) $(32,231)
Denominator:      
Weighted average common shares outstanding for basic
net income (loss) per share
 136,419
 127,964
 126,704
Net effect of dilutive common stock equivalents 2,649
 
 
Weighted average common shares outstanding for diluted
net income (loss) per share
 139,068
 127,964
 126,704
Net income (loss) per share:      
Basic $0.46
 $(0.81) $(0.25)
Diluted $0.45
 $(0.81) $(0.25)



Segment Information
We operate our business in one segment, which includes all activities related to the research, development and commercialization of our proprietary enzymes. This segment also includes revenues and expenses related to (i) research and development and APIbulk rHuPH20 manufacturing activities conducted under our collaborative agreements with third parties and (ii) product sales of Hylenex recombinant. The chief operating decision-maker reviews the operating results on an aggregate basis and manages the operations as a single operating segment. Our long-lived assets located in foreign countries had minimal book value as of December 31, 2017 and 2016.

F-16

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Adoption and Pending Adoption of Recent Accounting Pronouncements
In August 2014,The following table provides a brief description of recently issued accounting standards, those adopted in the Financial Accounting Standards Board (“FASB”) issued Accounting Standards Update No. 2014-15, Presentationcurrent period and those not yet adopted:
StandardDescriptionEffective Date
Effect on the Financial
Statements or Other Significant Matters
In July 2015, the FASB issued ASU 2015-11, Inventory: Simplifying the Measurement of Inventory.The new guidance requires that for entities that measure inventory using the first-in, first-out method, inventory should be measured at the lower of cost or net realizable value. Topic 330, Inventory, currently requires an entity to measure inventory at the lower of cost or market. Market could be replacement cost, net realizable value, or net realizable value less an approximate normal profit margin. Net realizable value is the estimated selling price in the ordinary course of business, less reasonably predictable costs of completion, disposal, and transportation.January 1, 2017.The adoption did not have a material impact on our consolidated financial position or results of operations.
In November 2015, the FASB issued ASU 2015-17, Income Taxes (Topic 740), Balance Sheet Classification of Deferred Tax Assets
The amendments in this update simplify the presentation of deferred income taxes by requiring that deferred tax liabilities and assets be classified as noncurrent in a classified statement of financial position.

January 1, 2017
The adoption of this guidance did not have a significant impact on the Company’s financial statements.

In August 2016, the FASB issued ASU 2016-15, Statement of Cash Flows: Classification of Certain Cash Receipts and Cash Payments.

In November 2016, the FASB issued ASU 2016-18, Statement of Cash Flows: Restricted Cash.
Current U.S. GAAP either is unclear or does not include specific guidance on the eight cash flow classification issues included in ASU 2016-15. The new guidance is an improvement to U.S. GAAP and is intended to reduce the current and potential future diversity in practice. ASU 2016-18 provides additional classification guidance for restricted cash, which requires that restricted cash be included with cash and cash equivalents when reconciling the beginning-of-period and end-of-period total amounts shown on the statement of cash flows.January 1, 2018. We have elected to early adopt as of January 1, 2017.Cash and cash equivalents at the beginning-of-period and end-of-period total amounts in the Consolidated Statements of Cash Flows have been adjusted to include $0.5 million of restricted cash for each of the periods presented.

F-17

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — Going Concern (“ASU 2014-15”). The provisions of ASU 2014-15 provide that in connection with preparing financial statements for each annual and interim reporting period, an entity's management should evaluate whether there are conditions or events, considered in the aggregate, that raise substantial doubt about the entity's ability to continue as a going concern within one year after the date that the financial statements are issued (or within one year after the date that the financial statements are available to be issued when applicable). ASU 2014-15 is effective for the annual reporting period ending after December 15, 2016, and for annual and interim periods thereafter. Early adoption is permitted. The adoption of ASU 2014-15 will not have a material impact on our consolidated financial position or results of operations.(Continued)
In May 2014, the FASB issued Accounting Standards Update No. 2014-09, Revenue from Contracts with Customers (“ASU 2014-09”). ASU 2014-09 will eliminate transaction-specific and industry-specific revenue recognition guidance under current GAAP and replace it with a principle-based approach for determining revenue recognition. ASU 2014-09 will require that companies recognize revenue based on the value of transferred goods or services as they occur in the contract. ASU 2014-09 also will require additional disclosure about the nature, amount, timing and uncertainty of revenue and cash flows arising from customer contracts, including significant judgments and changes in judgments and assets recognized from costs incurred to obtain or fulfill a contract. ASU 2014-09 is effective for annual reporting periods beginning after December 15, 2016. Early application is not permitted. Entities can transition to the standard either retrospectively or as a cumulative-effect adjustment as of the date of adoption. We have not yet selected a transition method and we are currently evaluating the effect that the updated standard will have on our consolidated financial statements and related disclosures.
In July 2013, the FASB issued Accounting Standards Update No. 2013-11, Income Taxes (Topic 740), Presentation of an Unrecognized Tax Benefit When a Net Operating Loss Carryforward, a Similar Tax Loss, or a Tax Credit Carryforward Exists (“ASU 2013-11”).The provisions of ASU 2013-11 require entities to present unrecognized tax benefits as a decrease in a net operating loss, similar tax loss or tax credit carryforward if certain criteria are met. The determination of whether a deferred tax asset is available is based on the unrecognized tax benefit and the deferred tax asset that exists at the reporting date and presumes disallowance of the tax position at the reporting date. The guidance will eliminate the diversity in practice in the presentation of unrecognized tax benefits but will not alter the way in which entities assess deferred tax assets for realizability. The amendments are effective for fiscal years, and interim periods within those fiscal years, beginning after December 15, 2014. The amendments should be applied prospectively to unrecognized tax benefits that exist at the effective date. Early adoption is permitted. The adoption of ASU 2013-11 will not have a material impact on our consolidated financial position or results of operations.


F-16
StandardDescriptionEffective Date
Effect on the Financial
Statements or Other Significant Matters
In January 2016, the FASB issued ASU 2016-01, Financial Instruments - Overall; Recognition and Measurement of Financial Assets and Financial Liabilities.The new guidance supersedes the guidance to classify equity securities with readily determinable fair values into different categories (that is, trading or available-for-sale) and requires equity securities to be measured at fair value with changes in the fair value recognized through net income. The new guidance requires public business entities that are required to disclose fair value of financial instruments measured at amortized cost on the balance sheet to measure that fair value using the exit price notion consistent with Topic 820, Fair Value Measurement.January 1, 2018.We currently do not hold equity securities, and we are evaluating the effect the updated standard will have on our consolidated financial statements and related disclosures.
In May 2014, the FASB issued ASU 2014-09, Revenue from Contracts with Customers (Topic 606). In March, April, May and December 2016, the FASB issued additional guidance related to Topic 606.
The new standard will supersede nearly all existing revenue recognition guidance. Under Topic 606, an entity is required to recognize revenue upon transfer of promised goods or services to customers in an amount that reflects the expected consideration to be received in exchange for those goods or services. Topic 606 defines a five-step process in order to achieve this core principle, which may require the use of judgment and estimates, and also requires expanded qualitative and quantitative disclosures relating to the nature, amount, timing and uncertainty of revenue and cash flows arising from contracts with customers, including significant judgments and estimates used. The new standard also defines accounting for certain costs related to origination and fulfillment of contracts with customers, including whether such costs should be capitalized. The new standard permits adoption either by using (i) a full retrospective approach for all periods presented in the period of adoption or (ii) a modified retrospective approach where the new standard is applied in the financial statements starting with the year of adoption. Under both approaches, cumulative impact of the adoption is reflected as an adjustment to retained earnings (accumulated equity (deficit)) as of the earliest date presented in accordance with the new standard.


January 1, 2018. Early adoption is permitted.
We plan to implement the new guidance on January 1, 2018 using the modified retrospective approach. We have substantially completed our evaluation of the effect that the updated standard will have on our consolidated financial statements and related disclosures. Adoption of the new guidance will impact the timing of recognition of payments related to certain of our license and collaboration agreements (1) and the timing of recognition of our sales-based royalties.(2) This standard will have a material impact on our consolidated financial statements.


F-18

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


StandardDescriptionEffective Date
Effect on the Financial
Statements or Other Significant Matters
In February 2016, the FASB issued ASU 2016-02, Leases.The new guidance requires lessees to recognize assets and liabilities for most leases and provides enhanced disclosures.January 1, 2019. Early adoption is permitted.We are currently evaluating the effect the updated standard will have on our consolidated financial statements and related disclosures and do not intend to early adopt. We anticipate recognition of additional assets and corresponding liabilities related to our leases on our consolidated balance sheet.
In June 2016, the FASB issued ASU 2016-13, Financial Instruments - Credit Losses (Topic 326), Measurement of Credit Losses on Financial Instruments
The standard amends the impairment model by requiring entities to use a forward-looking approach based on expected losses to estimate credit losses for most financial assets and certain other instruments that aren’t measured at fair value through net income.

January 1, 2020
The Company does not believe the adoption will have a material impact on our consolidated financial position or results of operations.

_______________
3.(1)Marketable SecuritiesUnder the new standard, we are required to assess whether licenses granted under our collaboration and license agreements are distinct in the context of the agreement from other performance obligations and functional when granted. We expect that license-related amounts, including upfront payments, exclusive designation fees, annual license maintenance fees, additional target fees, development, regulatory and sales-based milestones will be recognized, generally, at a point in time when earned. Currently, these amounts related to certain of our license and collaboration agreements are being amortized over the term of the collaboration agreement. For example, during the year ended December 31, 2017, we recognized revenue from amortization of license payments of $4.1 million. Total deferred revenue related to license payments under collaboration agreements as of December 31, 2017 was $51.8 million, which will be eliminated and recorded as a reduction to our accumulated deficit upon adoption of Topic 606. Under the new standard, license revenues would have totaled $198.4 million for the year ended December 31, 2017.
(2)Under the new standard, we expect sales-based royalties will be recognized in the quarter they are earned based on estimates, with a true-up to actual results following in the subsequent quarter. Sales-based royalty revenue earned under our collaboration and license agreements is presently recognized when the royalty reports are made available. Upon adoption of Topic 606, we will reduce our accumulated deficit and increase our accounts receivable, net, by the amount earned but not yet reported in our consolidated balance sheet of approximately $19.4 million. In 2017, we recognized royalty revenues of $63.5 million. Under the new standard, royalty revenues would have totaled $68.9 million for the year ended December 31, 2017. We have established a process to estimate sales-based royalty revenues in the quarter in which the sales occur going forward.

F-19

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


3. Fair Value Measurement
Available-for-sale marketable securities consisted of the following (in thousands):
 December 31, 2014 December 31, 2017
Description Amortized Cost Gross Unrealized Gains Gross Unrealized Losses Estimated Fair Value
 Amortized Cost Gross Unrealized Gains Gross Unrealized Losses Estimated Fair Value
Corporate debt securitiesCorporate debt securities $74,275
 $2
 $(43) $74,234
 $117,427
 $
 $(235) $117,192
U.S. Treasury securities 66,601
 
 (201) 66,400
Commercial paper 116,882
 
 
 116,882
 $300,910
 $
 $(436) $300,474
 December 31, 2013 December 31, 2016
Description Amortized Cost Gross Unrealized Gains Gross Unrealized Losses Estimated Fair Value
 Amortized Cost Gross Unrealized Gains Gross Unrealized Losses Estimated Fair Value
Corporate debt securitiesCorporate debt securities $35,130
 $20
 $(3) $35,147
 $40,221
 $1
 $(15) $40,207
U.S. Treasury securities 94,002
 24
 (16) 94,010
Commercial paperCommercial paper 5,999
 
 
 5,999
 4,000
 
 
 4,000
Certificate of deposit 3,000
 
 
 3,000
 $44,129
 $20
 $(3) $44,146
 $138,223
 $25
 $(31) $138,217
As of December 31, 2014, all of our2017, 28 available-for-sale marketable securities were scheduled to mature within the next twelve months. There were $57.3with a fair market value of $183.6 million of available-for-sale securities that matured during the year ended December 31, 2014. There were no realized gains or losses for the year ended December 31, 2014 and 2013. As of December 31, 2014, we had 14 available-for-sale marketable securities in a gross unrealized loss position of $0.4 million, all of which had been in such position for less than twelve12 months. Based on our review of these marketable securities, we believe we had no other-than-temporary impairments on these securities as of December 31, 20142017, because we do not intend to sell these securities and it is not more likely than notmore-likely-than-not that we will be required to sell these securities before the recovery of their amortized cost basis.
Contractual maturities of available-for-sale debt securities are as follows (in thousands):
  December 31, 2017 December 31, 2016
  Estimated Fair Value
Due within one year $213,426
 $132,221
After one but within five years 87,048
 5,996
  $300,474
 $138,217

F-20

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


The following table summarizes, by major security type, our cash equivalents and available-for-sale marketable securities that are measured at fair value on a recurring basis and are categorized using the fair value hierarchy (in thousands):
  December 31, 2017 December 31, 2016
  Level 1 Level 2 Total estimated fair value Level 1 Level 2 Total estimated fair value
Cash equivalents:            
Money market funds $142,091
 $
 $142,091
 $60,916
 $
 $60,916
Commercial paper 
 15,700
 15,700
 
 
 
Available-for-sale marketable
   securities:
            
Corporate debt securities 
 117,192
 117,192
 
 40,207
 40,207
U.S. Treasury securities 66,400
 
 66,400
 94,010
 
 94,010
Commercial paper 
 116,882
 116,882
 
 4,000
 4,000
  $208,491
 $249,774
 $458,265
 $154,926
 $44,207
 $199,133
There were no transfers between Level 1 and Level 2 of the fair value hierarchy during the year ended December 31, 2017. We had no instruments that were classified within Level 3 as of December 31, 2017 and 2016.
4.Collaborative Agreements
4. Collaborative Agreements
Roche Collaboration
In December 2006, we and Roche entered into a licensecollaboration and collaborativelicense agreement, under which Roche obtained a worldwide exclusive license to develop and commercialize product combinations of our patented rHuPH20 enzyme and up to thirteen Roche target compounds (the “Roche Collaboration”). As of December 31, 2014, Roche initially had elected a total of fivethe exclusive right to apply rHuPH20 to three pre-defined Roche biologic targets and retainswith the option to develop and commercialize rHuPH20 with threeten additional targets. Roche had the right to exercise this option to identify additional targets provided thatfor ten years. As of the ten year anniversary in December 2016, Roche continues to pay annual maintenance fees to us.had elected a total of eight targets, two of which are exclusive.
In August 2013, Roche received European marketing approval in August 2013 for its collaboration product, Herceptin SC, for the treatment of patients with HER2-positive breast cancer and launched Herceptin SC in the European Union (“EU”) in September 2013.
In March 2014, Roche received European marketing approval for its collaboration product, MabThera SC, for the treatment of patients with common forms of non-Hodgkin lymphoma (“NHL”). In June 2014, Roche launched MabThera SC in the EU which triggered a $5.0 million sales-based paymentEU. In May 2016, Roche announced that the EMA approved Mabthera SC to us fortreat patients with chronic lymphocytic leukemia (“CLL”). In June 2017, the achievement of the first commercial sale pursuant to the termsFDA approved Genentech’s (a member of the Roche Collaboration.Group) RITUXAN HYCELA™, a combination of rituximab and rHuPH20 (approved and marketed under the MabThera SC brand in countries outside the U.S.), for CLL and two types of NHL, follicular lymphoma and diffuse large B-cell lymphoma. Following FDA approval, Genentech launched RITUXAN HYCELA in the U.S.
Roche assumes all development, manufacturing, clinical, regulatory, sales and marketing costs under the Roche Collaboration, while we are responsible for the supply of bulk rHuPH20. We are entitled to receive reimbursements for providing research and development services and supplying bulk rHuPH20 to Roche at its request.


F-17



Under the terms of the Roche Collaboration, Roche pays us a royalty on each product commercialized under the agreement consisting of a mid-single digit percent of the net sales of such product. Unless terminated earlier in accordance with its terms, the Roche Collaboration continues in effect until the expiration of Roche'sRoche’s obligation to pay royalties. Roche has the obligation to pay royalties to us with respect to each product commercialized in each country, during the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the Roche Collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country. In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term.

F-21

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Payments received from Roche, excluding royalties and reimbursements for providing research and development services and supplying bulk rHuPH20, since inception of the Roche Collaboration are as follows (in thousands):
 As of December 31, 2017
Upfront license fee payment for the application of rHuPH20 to the initial exclusive targets$20,000
Election of additional exclusive targets and annual license maintenance fees for the
   right to designate the remaining targets as exclusive targets
23,000
Clinical development milestone payments13,000
Regulatory milestone payments8,000
Sales-based milestone payments22,000
Total payments received$86,000
Due to our continuing involvement obligations (for example, support activities associated with rHuPH20) under the Roche Collaboration, revenues from the upfront payment, exclusive designation fees, annual license maintenance fees and sales-based milestone payments were deferred and are being amortized over the remaining term of the Roche Collaboration.
For each of the years ended December 31, 2017, 2016 and 2015, we recognized $3.3 million of Roche deferred revenues, excluding reimbursements for providing research and development services and supplying bulk rHuPH20, as revenues under collaborative agreements. Total Roche deferred revenues, excluding deferred revenues related to reimbursements for providing research and development services and supplying bulk rHuPH20, were $39.4 million and $35.7 million as of December 31, 2017 and 2016, respectively.
In September 2017, we and Roche entered into an agreement providing Roche the right to develop and commercialize products with one additional exclusive target using our ENHANZE Technology for an upfront payment of $30.0 million (the “2017 Roche Collaboration”). The upfront license payment may be followed by event-based payments subject to Roche’s achievement of specified development, regulatory and sales-based milestones. In addition, Roche will pay royalties to us if products under the collaboration are commercialized consisting of a mid-single digit percent of the net sales of such product. Unless terminated earlier in accordance with its terms, the 2017 Roche Collaboration continues in effect until the expiration of Roche’s obligation to pay royalties. Roche has the obligation to pay royalties to us with respect to each product commercialized in each country, during the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the 2017 Roche Collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country. In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term. Roche may terminate the agreement prior to expiration for any reason in its entirety upon 90 days prior written notice to us. Upon any such termination, the license granted to Roche (in total or with respect to the terminated target, as applicable) will terminate provided, however, that in the event of expiration of the agreement, the licenses granted will become perpetual, non-exclusive and fully paid.
At the inception of the 2017 Roche Collaboration, we identified the deliverables in the arrangement to include the license, research and development services and supply of bulk rHuPH20 and determined that each individually represent separate units of accounting, because each deliverable has standalone value. The estimated selling prices for the units of accounting we identified were determined based on market conditions, the terms of comparable collaborative arrangements for similar technology in the pharmaceutical and biotech industry and entity-specific factors such as the terms of our previous collaborative agreements, our pricing practices and pricing objectives. The arrangement consideration was allocated to the deliverables based on the relative selling price method and the nature of the research and development services to be performed for the collaborator. The amount allocable to the delivered unit or units of accounting is limited to the amount that is not contingent upon the delivery of additional items or meeting other specified performance conditions (non-contingent amount). As such, we excluded from the allocable arrangement consideration the event-based payments, milestone payments and royalties regardless of the probability of receipt. Based on the results of our analysis, we allocated the $30.0 million to the license fee deliverable. We determined that the upfront payment was earned upon the granting of the worldwide, exclusive right to our technology to Roche. As a result, we recognized the $30.0 million license fee under the 2017 Roche Collaboration as revenues under collaborative agreements for the year ended December 31, 2017.

F-22

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Baxalta Collaboration
In September 2007, we and Baxalta entered into a collaboration and license agreement, under which Baxalta obtained a worldwide, exclusive license to develop and commercialize HYQVIA, a combination of Baxalta’s current product GAMMAGARD LIQUID and our patented rHuPH20 enzyme (the “Baxalta Collaboration”). In 2013, the European Commission granted Baxalta marketing authorization in all EU Member States for the use of HYQVIA (solution for subcutaneous use), a combination of GAMMAGARD LIQUID and rHuPH20 in dual vial units, as replacement therapy for adult patients with primary and secondary immunodeficiencies, and Baxalta launched HYQVIA in the EU. In 2014, Baxalta launched HYQVIA in the U.S following the FDA’s approval of HYQVIA for treatment of adult patients with primary immunodeficiency. In May 2016, Baxalta announced that HYQVIA received a marketing authorization from the European Commission for a pediatric indication, which is being launched in Europe to treat primary and certain secondary immunodeficiencies.
The Baxalta Collaboration is applicable to both kit and formulation combinations. Baxalta assumes all development, manufacturing, clinical, regulatory, sales and marketing costs under the Baxalta Collaboration, while we are responsible for the supply of bulk rHuPH20. We perform research and development activities and supply bulk rHuPH20 at the request of Baxalta, and are reimbursed by Baxalta under the terms of the Baxalta Collaboration. In addition, Baxalta has certain product development and commercialization obligations in major markets identified in the Baxalta Collaboration.
Under the terms of the Baxalta Collaboration, Baxalta pays us a royalty consisting of a mid-single digit percent of the net sales of HYQVIA. Unless terminated earlier in accordance with its terms, the Baxalta Collaboration continues in effect until the expiration of Baxalta’s obligation to pay royalties to us. Baxalta has the obligation to pay royalties to us, with respect to each product commercialized in each country, during the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the Baxalta Collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country. In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term.
Payments received from Baxalta, excluding royalties and reimbursements for providing research and development services and supplying bulk rHuPH20, since inception of the collaboration agreement are as follows (in thousands):
 As of December 31, 2017
Upfront license fee payment for the application of rHuPH20 to the initial exclusive target$10,000
Regulatory milestone payments3,000
Sales-based milestone payments9,000
Total payments received$22,000
Due to our continuing involvement obligations (for example, support activities associated with rHuPH20 enzyme), the upfront license fee and sales-based milestone payments were deferred and are being recognized over the term of the Baxalta Collaboration.
For each of the years ended December 31, 2017, 2016 and 2015, we recognized $0.8 million of Baxalta deferred revenues, excluding reimbursements for providing research and development services and supplying bulk rHuPH20, as revenues under collaborative agreements. Total Baxalta deferred revenues, excluding reimbursements for providing research and development services and supplying bulk rHuPH20, were $12.4 million and $8.2 million as of December 31, 2017 and 2016, respectively.
Other Collaborations
In December 2017, we and Alexion entered into a collaboration and license agreement, under which Alexion has the worldwide license to develop and commercialize products combining our patented rHuPH20 enzyme with Alexion proprietary biologics directed at up to four targets (the “Alexion Collaboration”). Targets, once selected, will be on an exclusive, global basis. As of December 31, 2017, Alexion has elected two specific exclusive targets. Alexion has the right to elect up to two additional targets for additional fees. The upfront license payment may be followed by event-based payments subject to Alexion’s achievement of specified development, regulatory and sales-based milestones. In addition, Alexion will pay royalties to us if products under the collaboration are commercialized. Unless terminated earlier in accordance with its terms, the Alexion Collaboration continues in effect until the later of: (i) expiration of the last to expire of the valid claims of or patents covering rHuPH20 or other specified

F-23

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


patents developed under the collaboration which valid claim covers a product developed under the collaboration, and (ii) expiration of the last to expire royalty term for a product developed under the collaboration. The royalty term of a product developed under the Alexion Collaboration, with respect to each country, consists of the period equal to the longer of (a) duration of any valid claim of our patent covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country. In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term. Alexion may terminate the agreement prior to the expiration for any reason in its entirety upon 90 days prior written notice to us. Upon such termination, the license granted to Alexion (in total or with respect to the terminated target, as applicable) will terminate provided, however, that in the event of expiration of the agreement, the licenses granted will become perpetual, non-exclusive and fully paid.
In September 2017, we and BMS entered into a collaboration and license agreement, effective November 2017, under which BMS has the worldwide license to develop and commercialize products combining our rHuPH20 enzyme with BMS proprietary biologics directed at up to eleven targets (the “BMS Collaboration”). Targets, once selected, will be on an exclusive, global basis, with the exception of one co-exclusive target. As of December 31, 2017, BMS has elected several specified exclusive targets, including programmed death 1 (PD-1), and has the right to elect additional targets, some of which are subject to additional fees. The upfront license payment may be followed by event-based payments subject to BMS’s achievement of specified development, regulatory and sales-based milestones. In addition, BMS will pay royalties to us if products developed under the collaboration are commercialized. Unless terminated earlier in accordance with its terms, the BMS Collaboration continues in effect until the later of: (i) expiration of the last to expire of the valid claims of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers a product developed under the collaboration, and (ii) expiration of the last to expire royalty term for a product developed under the collaboration. The royalty term of a product developed under the BMS Collaboration, with respect to each country, consists of the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country.
As In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term. BMS may terminate the agreement prior to expiration for any reason in its entirety upon prior written notice to us. Upon any such termination, the license granted to BMS (in total or with respect to the terminated target, as applicable) will terminate provided, however, that in the event of December 31, 2014, we have received $78.3 million, excluding royalties and reimbursements for providing research and development services and supplying bulk rHuPH20, from Roche. The amounts received consistedexpiration of the $20.0 million upfront license fee payment foragreement, the application of rHuPH20 to the initial three Roche exclusive targets, $22.3 million in connection with Roche's election of two additional exclusive targetslicenses granted will become perpetual, non-exclusive and annual license maintenance fees for the right to designate the remaining targets as exclusive targets, fully paid.$13.0 million in clinical development milestone payments, $8.0 million in regulatory milestone payments
In December 2015, we and $15.0 million in sales-based payments. Due to our continuing involvement obligations (for example, support activities associated with rHuPH20), revenues from the upfront payment, exclusive designation fees, annual license maintenance fees and sales-based payment were deferred and are being amortized over the remaining term of the Roche Collaboration.
For the years ended December 31, 2014, 2013 and 2012, we recognized approximately $8.1 million, $4.6 million, and $2.0 million, respectively, of Roche deferred revenues as revenues under collaborative agreements. In addition, for the years ended December 31, 2014, 2013 and 2012, we recognized approximately $2.0 million, $1.3 million and $1.4 million, respectively, of deferred bulk rHuPH20 sales revenue as product sales revenue. Total Roche deferred revenues was approximately $42.7 million and $41.6 million as of December 31, 2014 and 2013, respectively. For the year ended December 31, 2012, we recognized $8.0 million as revenues under collaborative agreements in accordance with the Milestone Method related to the achievement of certain regulatory milestones pursuant to the terms of the Roche Collaboration. No such revenues were recognized for the years ended December 31, 2014 and 2013.
Gammagard Collaboration
In September 2007, weLilly entered into a collaboration and license and collaborative agreement, with Baxter, under which Baxter obtained aLilly has the worldwide exclusive license to develop and commercialize HYQVIA, a combination of Baxter's current product GAMMAGARD LIQUID andproducts combining our patented rHuPH20 enzyme with Lilly proprietary biologics directed at up to five targets (the “Baxter“Lilly Collaboration”). Targets, once selected, will be on an exclusive, global basis, with the exception of one semi-exclusive target. As of December 31, 2017, Lilly has elected two specified exclusive targets and one specified semi-exclusive target. Lilly has the right to elect up to two additional targets for additional fees. The upfront license payment may be followed by event-based payments subject to Lilly’s achievement of specified development, regulatory and sales-based milestones. In May 2013, the European Commission granted Baxter marketing authorization in all EU Member States for the use of HYQVIA (solution for subcutaneous use), a combination of GAMMAGARD LIQUID and rHuPH20 in dual vial units, as replacement therapy for adult patients with primary and secondary immunodeficiencies. Baxter launched HYQVIA in the first EU country in July 2013 and in additional EU countries in the second half of 2013 and in 2014. The FDA approved HYQVIA for treatment of adult patients with primary immunodeficiency in September 2014. In October 2014, Baxter announced the launch and first shipments of HYQVIA in the U.S.
The Baxter Collaboration is applicableaddition, Lilly will pay royalties to both kit and formulation combinations. Baxter assumes all development, manufacturing, clinical, regulatory, sales and marketing costsus if products under the Baxter Collaboration, while wecollaboration are responsible for the supply of bulk rHuPH20. We perform research and development activities and supply bulk rHuPH20 at the request of Baxter, and are reimbursed by Baxter under the terms of the Baxter Collaboration. In addition, Baxter has certain product development and commercialization obligations in major markets identified in the Baxter Collaboration.
commercialized. Unless terminated earlier in accordance with its terms, the BaxterLilly Collaboration continues in effect until the later of: (i) expiration of Baxter's obligationthe last to pay royalties. Baxter hasexpire of the obligationvalid claims of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers a product developed under the collaboration, and (ii) expiration of the last to pay royalties,expire royalty term for a product developed under the collaboration. The royalty term of a product developed under the Lilly Collaboration, with respect to each product in each country, duringconsists of the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country.


F-18



As In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term. Lilly may terminate the agreement prior to expiration for any reason in its entirety upon 60 days prior written notice to us. Upon any such termination, the license granted to Lilly (in total or with respect to the terminated target, as applicable) will terminate provided, however, that in the event of December 31, 2014, we have received $17.0 million under the Gammagard Collaboration, excluding royalties. The amounts received consistedexpiration of the $10.0 million upfrontagreement, the licenses granted will become perpetual, non-exclusive and fully paid.
In June 2015, we and AbbVie entered into a collaboration and license fee payment, a $3.0 million regulatory milestone paymentagreement, under which AbbVie has the worldwide license to develop and a $4.0 million sales-based payment. Baxter pays us a royaltycommercialize products combining our patented rHuPH20 enzyme with AbbVie proprietary biologics directed at up to nine targets (the “AbbVie Collaboration”). Targets, once selected, will be on HYQVIA consisting of a mid-single digit percent of the net sales of such product. Due to our continuing involvement obligations (for example, support activities associated with rHuPH20 enzyme), the upfront and sales-based payments were deferred and are being recognized over the term of the Baxter Collaboration. We recognized revenue from the upfront and sales-based payments in the amount of approximately $0.8 million, $0.6 million and $0.5 million for the years ended December 31, 2014, 2013 and 2012, respectively. Deferred revenue relating to the upfront and sales-based payments under the Gammagard Collaboration was approximately $10.9 million and $10.5 million asan exclusive, global basis. As of December 31, 20142017, AbbVie has elected one specified exclusive target, and 2013, respectively.subsequently returned the target. AbbVie has the right
Other Collaborations
F-24

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


to elect up to eight additional targets for additional fees. The upfront license payment may be followed by event-based payments subject to AbbVie’s achievement of specified development, regulatory and sales-based milestones. In addition, AbbVie will pay tiered royalties to us if products under the collaboration are commercialized. Unless terminated earlier in accordance with its terms, the AbbVie Collaboration continues in effect until the later of: (i) expiration of the last to expire of the valid claims of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers a product developed under the collaboration, and (ii) expiration of the last to expire royalty term for a product developed under the collaboration. The royalty term of a product developed under the AbbVie Collaboration, with respect to each country, consists of the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country. In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term. AbbVie may terminate the agreement prior to expiration for any reason in its entirety or on a target-by-target basis upon 90 days prior written notice to us. Upon any such termination, the license granted to AbbVie (in total or with respect to the terminated target, as applicable) will terminate provided, however, that in the event of expiration of the agreement, the licenses granted will become perpetual, non-exclusive and fully paid.
In December 2014, we and Janssen entered into a collaboration and license agreement, under which Janssen has the worldwide license to develop and commercialize products combining our patented rHuPH20 enzyme with Janssen proprietary biologics directed at up to five targets (the “Janssen Collaboration”). Targets, mayonce selected, will be selected on an exclusive, or non-exclusiveglobal basis. As of December 31, 2014, we have received a $15.0 million payment for the license fee of2017, Janssen has elected one specified exclusive target andtarget. Janssen has the right to elect four additional targets in the future upon payment of additional fees. In addition, Janssen will pay royalties to us if products under the collaboration are commercialized. Unless terminated earlier in accordance with its terms, the Janssen Collaboration continues in effect until the later of (i) expiration of the last to expire of the valid claims of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers a product developed under the collaboration, and (ii) expiration of the last to expire royalty term for a product developed under the collaboration. The royalty term of a product developed under the Janssen Collaboration, with respect to each country, consists of the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country. In the event such valid claims expire, the royalty rate is reduced for the remaining royalty term. Janssen may terminate the agreement prior to expiration for any reason in its entirety or on a target-by-targetproduct-by-product basis upon 90 days prior written notice to us. Upon any such termination, the license granted to Janssen (in total or with respect to the terminated target, as applicable) will terminate provided, however, that in the event of expiration of the agreement, the licenses granted will become perpetual, non-exclusive and fully paid-up.paid.
In December 2012, we and Pfizer entered into a collaboration and license agreement, under which Pfizer has the worldwide license to develop and commercialize products combining our patented rHuPH20 enzyme with Pfizer proprietary biologics directed at up to six targets (the “Pfizer Collaboration”). Targets may be selected on an exclusive or non-exclusive basis. As of December 31, 2014, we have received $11.0 million in upfront and license fee payments for the licenses to four2017, Pfizer has elected five specified exclusive targets and has returned two additional targets which of its elected targets. Pfizer has the right to elect three additional targets in the future, upontwo of which are contingent on payment of additional fees. In addition, Pfizer will pay royalties to us if products under the collaboration are commercialized. Unless terminated earlier in accordance with its terms, the Pfizer Collaboration continues in effect until the later of (i) expiration of the last to expire of the valid claims of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers a product developed under the collaboration, and (ii) expiration of the last to expire royalty term for a product developed under the collaboration. The royalty term of a product developed under the Pfizer Collaboration, with respect to each country, consists of the period equal to the longer of: (a) the duration of any valid claim of our patents covering rHuPH20 or other specified patents developed under the collaboration which valid claim covers the product in such country or (b) ten years following the date of the first commercial sale of such product in such country. Royalties are subject to adjustment as set forth in the agreement. Pfizer may terminate the agreement prior to expiration for any reason in its entirety or on a target-by-target basis upon 30 days prior written notice to us. Upon any such termination, the license granted to Pfizer (in total or with respect to the terminated target, as applicable) will terminate, provided, however, that in the event of expiration of the agreement, the licenses granted will become perpetual, non-exclusive and fully paid-up.paid.


F-19
F-25

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Payments received under collaboration agreements for upfront license fees, license fees for the election of additional targets, maintenance fees and event-based payments since inception of the collaboration agreements are as follows (in thousands):
 As of December 31, 2017
Alexion Collaboration$40,000
BMS Collaboration105,000
Lilly Collaboration33,000
AbbVie Collaboration29,000
Janssen Collaboration30,250
Pfizer Collaboration16,500
Total payments received$253,750
At the inception of the Pfizer, Janssen, AbbVie, Lilly, BMS and JanssenAlexion arrangements, we identified the deliverables in each arrangement to include the license, research and development services and supply of bulk rHuPH20. We have determined that the license, research and development services and supply of bulk rHuPH20 individually represent separate units of accounting, because each deliverable has standalone value. We determined that the rights to elect additional targets in the future upon the payment of additional license fees are substantive options that are not priced at a significant and incremental discount. Therefore, we determined for each collaboration that the rights to elect additional targets are not deliverables at the inception of the arrangement. The estimated selling prices for thesethe units of accounting we identified were determined based on market conditions, the terms of comparable collaborative arrangements for similar technology in the pharmaceutical and biotech industry and entity-specific factors such as the terms of our previous collaborative agreements, our pricing practices and pricing objectives. The arrangement consideration was allocated to the deliverables based on the relative selling price method and the nature of the research and development services to be performed for the collaborator.
The amount allocable to the delivered unit or units of accounting is limited to the amount that is not contingent upon the delivery of additional items or meeting other specified performance conditions (the noncontingent(non-contingent amount). As such, we excluded from the allocable arrangement consideration the event-based payments, milestone payments, annual exclusivity fees and royalties regardless of the probability of receipt. Based on the results of our analysis, we allocated the $11.0 million license fees from Pfizer and the $15.0 million upfront license fee from Janssenfollowing amounts to the license fee deliverable under each of the arrangements. arrangement (in thousands):
 
Consideration allocated to
license fees:
Alexion Collaboration$40,000
BMS Collaboration101,400
Lilly Collaboration33,000
AbbVie Collaboration23,000
Janssen Collaboration15,250
Pfizer Collaboration12,500
Total consideration allocated to license fees$225,150
We determined that the upfront paymentsconsideration allocated to the license fees were earned upon the granting of the worldwide, exclusive right to our technology to the collaborators in these arrangements. As a result, we recognized the $11.0 millionconsideration allocated to license fees under the Pfizer Collaboration and the $15.0 million upfront license fee under the Janssen Collaboration as revenues under collaborative agreements in the period when such license fees were earned. There were no revenuesWe recognized revenue related to event-based payments or milestone payments under these collaborations of $15.0 million, $6.0 million, and $1.0 million for the years ended December 31, 2014, 20132017, 2016 and 2012.2015, respectively.
The collaborators are each solely responsible for the development, manufacturing and marketing of any products resulting from their respective collaborations. We are entitled to receive payments for research and development services and supply of bulk rHuPH20 API to these collaborators if requested by suchany collaborator. We recognize amounts allocated to research and development services as revenues

F-26

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


under collaborative agreements as the related services are performed. We recognize amounts allocated to the sales of APIbulk rHuPH20 as revenues under collaborative agreements or product sales, as appropriate, when such APIbulk rHuPH20 has met all required specifications by the collaborators and the related title and risk of loss and damages have passed to the collaborators. We cannot predict the timing of delivery of research and development services and APIbulk rHuPH20 as they are at the collaborators'collaborators’ requests.
In May 2011 and June 2011, we entered into collaboration and license agreements with ViroPharma Incorporated and Intrexon Corporation, respectively. These collaboration agreements were terminated effective May 2014.
Pursuant to the terms of our collaboration agreements withthe Roche Collaboration and the Pfizer Collaboration, certain future payments meet the definition of a milestone in accordance with the Milestone Method of Accounting. We are entitled to receive additional milestone payments under our collaboration agreements with Roche and Pfizer for the successful development of the elected targets in the aggregate of up to approximately $55.0$62.5 million upon achievement of specified clinical development milestone events and up to approximately $12.0$12.0 million upon achievement of specified regulatory milestone events in connection with specified regulatory filings and receipt of marketing approvals.
5.Certain Balance Sheet Items
5. Certain Balance Sheet Items
Accounts receivable, net consisted of the following (in thousands):
 December 31,
2014
 December 31,
2013
 December 31,
2017
 December 31,
2016
Accounts receivable from product sales to collaborators $6,361
 $4,495
 $18,475
 $7,854
Accounts receivable from revenues under collaborative agreements 2,142
 6,151
Accounts receivable from other product sales 2,133
 1,505
 2,075
 2,234
Accounts receivable from revenues under collaborative agreements 1,266
 3,707
Subtotal 9,760
 9,707
 22,692
 16,239
Allowance for distribution fees and discounts (611) (610) (559) (559)
Total accounts receivable, net $9,149
 $9,097
 $22,133
 $15,680


F-20



Inventories consisted of the following (in thousands):
 December 31,
2014
 December 31,
2013
 December 31,
2017
 December 31,
2016
Raw materials $553
 $1,137
 $377
 $761
Work-in-process 5,207
 4,280
 2,131
 12,850
Finished goods 646
 753
 2,638
 1,012
Total inventories $6,406
 $6,170
 $5,146
 $14,623
Prepaid expenses and other assets consisted of the following (in thousands):
 December 31,
2014
 December 31,
2013
 December 31,
2017
 December 31,
2016
Prepaid manufacturing expenses $6,339
 $5,884
 $2,337
 $9,663
Prepaid research and development expenses 2,380
 3,522
 7,793
 8,613
Other prepaid expenses 1,094
 1,339
 2,585
 1,661
Other assets 1,535
 356
 6,717
 1,530
Total prepaid expenses and other assets 11,348
 11,101
 19,432
 21,467
Less long-term portion 1,205
 2,676
 5,553
 219
Total prepaid expenses and other assets, current $10,143
 $8,425
 $13,879
 $21,248
Prepaid manufacturing expenses include slot reservation fees and other amounts paid to contract manufacturing organizations. Such amounts are reclassified to work-in-process inventory once the manufacturing process has commenced.

F-27

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Property and equipment, net consisted of the following (in thousands):
 December 31,
2014
 December 31,
2013
 December 31,
2017
 December 31,
2016
Research equipment $8,474
 $7,714
 $10,970
 $10,479
Computer and office equipment 2,178
 1,949
 3,725
 3,373
Leasehold improvements 1,518
 1,408
 2,715
 2,331
Subtotal 12,170
 11,071
 17,410
 16,183
Accumulated depreciation and amortization (9,219) (7,649) (13,890) (11,919)
Property and equipment, net $2,951
 $3,422
 $3,520
 $4,264
Depreciation and amortization expense was approximately $1.8$2.2 million, $1.2$2.4 million, and $1.1$1.7 million for the years ended December 31, 2014, 20132017, 2016 and 2012,2015, respectively.
Accrued expenses consisted of the following (in thousands):
  December 31,
2014
 December 31,
2013
Accrued compensation and payroll taxes $5,923
 $7,075
Accrued outsourced research and development expenses 4,383
 3,377
Accrued outsourced manufacturing expenses 2,112
 3,233
Other accrued expenses 2,023
 1,235
Total accrued expenses 14,441
 14,920
Less long-term accrued outsourced research and development expenses 480
 551
     Total accrued expenses, current $13,961
 $14,369


F-21



Long-term accrued outsourced research and development is included in other long-term liabilities in the consolidated balance sheets.
  December 31,
2017
 December 31,
2016
Accrued outsourced research and development expenses $18,757
 $9,522
Accrued compensation and payroll taxes 13,384
 11,539
Accrued outsourced manufacturing expenses 2,504
 3,225
Other accrued expenses 5,396
 4,552
     Total accrued expenses 40,041
 28,838
Less long-term portion 440
 17
     Total accrued expenses, current $39,601
 $28,821
Deferred revenue consisted of the following (in thousands):
 December 31,
2014
 December 31,
2013
 December 31,
2017
 December 31,
2016
Collaborative agreements $53,479
 $51,185
    
License fees and event-based payments:    
Roche $39,379
 $35,709
Other 15,999
 8,209
 55,378
 43,918
Reimbursement for research and development services 
 700
Product sales 1,155
 1,958
 5,487
 
Total deferred revenue 54,634
 53,143
 60,865
 44,618
Less current portion 7,367
 7,398
 6,568
 4,793
Deferred revenue, net of current portion $47,267
 $45,745
 $54,297
 $39,825

F-28

6.Long-Term Debt, Net
Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


6. Long-Term Debt, Net
Royalty-backed Loan
In January 2016, through our wholly-owned subsidiary Halozyme Royalty LLC (“Halozyme Royalty”), we received a $150 million loan (the “Royalty-backed Loan”) pursuant to a credit agreement (the “Credit Agreement”) with BioPharma Credit Investments IV Sub, LP and Athyrium Opportunities II Acquisition LP (the “Royalty-backed Lenders”). Under the terms of the Credit Agreement, Halozyme Therapeutics, Inc. transferred to Halozyme Royalty the right to receive royalty payments from the commercial sales of ENHANZE products owed under the Roche Collaboration and Baxalta Collaboration (“Collaboration Agreements”). The royalty payments from the Collaboration Agreements will be used to repay the principal and interest on the loan (the “Royalty Payments”).  The Royalty-backed Loan bears interest at a per annum rate of 8.75% plus the three-month LIBOR rate. The three-month LIBOR rate is subject to a floor of 0.7% and a cap of 1.5%. The interest rate as of December 31, 2017 and 2016 was 10.25% and 9.71%, respectively.
The Credit Agreement provides that none of the Royalty Payments were required to be applied to the Royalty-backed Loan prior to January 1, 2017, 50% of the Royalty Payments are required to be applied to the Royalty-backed Loan between January 1, 2017 and January 1, 2018 and thereafter all Royalty Payments must be applied to the Royalty-backed Loan. However, the amounts available to repay the Royalty-backed Loan are subject to caps of $13.75 million per quarter in 2017, $18.75 million per quarter in 2018, $21.25 million per quarter in 2019 and $22.5 million per quarter in 2020 and thereafter. Amounts available to repay the Royalty-backed Loan will be applied first to pay interest and second to repay principal on the Royalty-backed Loan. Any accrued interest that is not paid on any applicable quarterly payment date, as defined, will be capitalized and added to the principal balance of the Royalty-backed Loan on such date. Halozyme Royalty will be entitled to receive and distribute to Halozyme any Royalty Payments that are not required to be applied to the Royalty-backed Loan or which are in excess of the foregoing caps.
Because the repayment of the term loan is contingent upon the level of Royalty Payments received, the repayment term may be shortened or extended depending on the actual level of Royalty Payments. The final maturity date of the Royalty-backed Loan will be the earlier of (i) the date when principal and interest is paid in full, (ii) the termination of Halozyme Royalty’s right to receive royalties under the Collaboration Agreements, and (iii) December 31, 2050.  Currently, we estimate that the loan will be repaid in the first quarter of 2020. This estimate could be adversely affected and the repayment period could be extended if future royalty amounts are less than currently expected. Under the terms of the Credit Agreement, at any time after January 1, 2019, Halozyme Royalty may, subject to certain limitations, prepay the outstanding principal of the Royalty-backed Loan in whole or in part, at a price equal to 105% of the outstanding principal on the Royalty-backed Loan, plus accrued but unpaid interest. The Royalty-backed Loan constitutes an obligation of Halozyme Royalty, and is non-recourse to Halozyme. Halozyme Royalty retains its right to the Royalty Payments following repayment of the loan.
As of December 31, 2017, we were in compliance with all covenants under the Royalty-backed Loan and there was no material adverse change in our business, operations or financial condition.
During the year ended December 31, 2016, accrued interest in the amount of $13.2 million was capitalized and added to the principal balance of the Royalty-backed Loan. We began making principal and interest payments against the Royalty-backed Loan in the first quarter of 2017 and therefore had no capitalized interest in the year ended December 31, 2017. In addition, we recorded accrued interest, which is included in accrued expenses, of $0.7 million as of December 31, 2017 and 2016, respectively.
In connection with the Royalty-backed Loan, we paid the Royalty-backed Lenders a fee of $1.5 million and incurred additional debt issuance costs totaling $0.4 million, which includes expenses that we paid on behalf of the Royalty-backed Lenders and expenses incurred directly by us. Debt issuance costs and the lender fee have been netted against the debt as of December 31, 2017, and are being amortized over the estimated term of the debt using the effective interest method. For the years ended December 31, 2017 and 2016, the Company recognized interest expense, including amortization of the debt discount, related to the Royalty-backed Loan of $16.4 million and $14.5 million, respectively. The assumptions used in determining the expected repayment term of the debt and amortization period of the issuance costs requires that we make estimates that could impact the short- and long-term classification of these costs, as well as the period over which these costs will be amortized. The outstanding balance of the Royalty-backed Loan as of December 31, 2017 was $146.5 million, net of unamortized debt discount of $0.8 million.

F-29

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Oxford and SVB Loan and Security Agreement
In December 2013, we entered into an Amended and Restated Loan and Security Agreement (the “Loan“Original Loan Agreement”) with Oxford Finance LLC (“Oxford”) and Silicon Valley Bank (“SVB”) (collectively, the “Lenders”), amending and restating in its entirety our originalprevious loan agreement with the Lenders, dated December 2012. The Original Loan Agreement provided for an additional $20 million principal amount of new term loan, bringing the total term loan balance to $50 million. The proceeds are to be used for working capital and general business requirements. The amended term loan facility matureswas scheduled to mature on January 1, 2018.
In January 2015, we entered into the second amendment to the Original Loan Agreement with the Lenders, amending and restating the term loan repayment schedules of the Original Loan Agreement. The amended and restated term loan repayment schedule providesprovided for interest only payments through January 2016, followed by consecutive equal monthly payments of principal and interest in arrears starting in February 2016 and continuing through the previously established maturity date of January 1, 2018. Consistent with the original loan, the amended Original Loan Agreement providesprovided for a 7.55% interest rate on the term loan and a final interest payment equal to 8.5% of the original principal amount, or $4.25 million, which iswas due when the term loan became due or upon the prepayment of the facility.
In June 2016, we entered into a Loan and Security Agreement (the “Loan Agreement”) with the Lenders, providing a senior secured loan facility of up to an aggregate principal amount of $70.0 million, comprising a $55.0 million draw in June 2016 and an additional $15.0 million tranche, which we had the option to draw during the second quarter of 2017 and did not exercise. The initial proceeds carry an interest rate of 8.25% and were partially used to pay the outstanding principal and final payment of $4.25 million owed on the Original Loan Agreement with the Lenders. The remaining proceeds are being used for working capital and general business requirements. The repayment schedule provides for interest only payments for the first 18 months, followed by consecutive equal monthly payments of principal and interest in arrears through the maturity date of January 1, 2021. The Loan Agreement provides for a final payment equal to 5.50% of the initial $55.0 million principal amount. The final payment is due when the Loan Agreement becomes due or upon the prepayment of the facility. We have the option to prepay the outstanding balance of the term loanLoan Agreement in full, subject to a prepayment fee of 2% in the first year and 1% to 3% depending upon whenin the prepayment occurs.second year of the Loan Agreement.
In connection with the term loan,Loan Agreement, the debt offering costs have been recorded as a debt discount in our condensed consolidated balance sheets which, together with the final payment and fixed interest rate payments, are being amortized and recorded as interest expense throughout the life of the term loan using the effective interest rate method.
The amended term loanLoan Agreement is secured by substantially all of the assets of the Company and our subsidiary, Halozyme, Inc., except that the collateral does not include any equity interests in Halozyme, Inc., any of our intellectual property (including all licensing, collaboration and similar agreements relating thereto), and certain other excluded assets. The Loan Agreement contains customary representations, warranties and covenants by us, which covenants limit our ability to convey, sell, lease, transfer, assign or otherwise dispose of certain of our assets; engage in any business other than the businesses currently engaged in by us or reasonably related thereto; liquidate or dissolve; make certain management changes; undergo certain change of control events; create, incur, assume, or be liable with respect to certain indebtedness; grant certain liens; pay dividends and make certain other restricted payments; make certain investments; make payments on any subordinated debt; and enter into transactions with any of our affiliates outside of the ordinary course of business or permit our subsidiaries to do the same.same; and make any voluntary prepayment of or modify certain terms of the Royalty-backed Loan. In addition, subject to certain exceptions, we are required to maintain with SVB our primary deposit accounts, securities accounts and commodities, and to do the same for our domestic subsidiary.subsidiary, Halozyme, Inc.
The Loan Agreement also contains customary indemnification obligations and customary events of default, including, among other things, our failure to fulfill certain of our obligations under the Loan Agreement and the occurrence of a material adverse change which is defined as a material adverse change in our business, operations, or condition (financial or otherwise), a material impairment of the prospect of repayment of any portion of the loan, or a material impairment in the perfection or priority of lender'sthe Lender’s lien in the collateral or in the value of such collateral.collateral or the occurrence of an event of default under the Royalty-backed Loan. In the event of default by us under the Loan Agreement, the Lenders would


F-22



be entitled to exercise their remedies thereunder, including the right to accelerate the debt, upon which we may be required to repay all amounts then outstanding under the Loan Agreement, which could harm our financial condition.

F-30

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


 As of December 31, 2014,2017, we were in compliance with all material covenants under the Loan Agreement and there was no material adverse change in our business, operations or financial condition.
Interest expense, including amortization of the debt discount, related to the Loan Agreement totaled $5.5 million, $20.0 million and $5.2 million for the years ended December 31, 2017, 2016 and 2015, respectively. Accrued interest, which is included in accrued expenses, was $0.4 million and $1.1 million as of December 31, 2017 and 2016, respectively. The outstanding term loan balance was $55.9 million as of December 31, 2017, inclusive of $1.3 million of accretion of the final payment and net of unamortized debt discount related to offering costs of $0.4 million.
Future maturities and interest payments under the term loanof long-term debt as of December 31, 2014,2017, are as follows (in thousands):
2015 $3,775
2016 25,077
2017 27,013
2018 6,501
 $94,125
2019 
 105,758
2020 27,311
2021��4,755
2022 
Total minimum payments 62,366
 231,949
Less amount representing interest (12,366) (26,792)
Gross balance of long-term debt 50,000
 205,157
Less unamortized debt discount (140) (2,806)
Present value of long-term debt 49,860
 202,351
Less current portion of long-term debt 
 (77,211)
Long-term debt, less current portion and unamortized debt discount $49,860
 $125,140
Interest expense, including amortization of debt discount, related to the long-term debt for the years ended December 31, 2014 and 2013 was approximately $5.6 million and $3.3 million, respectively.
7.Stockholders’ Equity (Deficit)
During 2014, we issued an aggregate of 1,432,206 shares of common stock, in connection with the exercises of stock options for cash in the aggregate amount of approximately $7.8 million. In addition, we issued 789,345 shares of common stock, net of RSAs canceled, in connection with the grants of RSAs and 120,043 shares of common stock upon vesting of certain RSUs. The RSU holders surrendered 74,325 RSUs to pay for minimum withholding taxes totaling approximately $1.0 million.
In February 2014, we completed an underwritten public offering and issued 8,846,153 shares of common stock, including 1,153,846 shares sold pursuant to the full exercise of an over-allotment option granted to the underwriters. All of the shares were offered at a public offering price of $13.00 per share, generating approximately $107.7 million in net proceeds.7. Share-based Compensation
During 2013, we issued an aggregate of 1,270,362 shares of common stock in connection with the exercises of stock options for cash in the aggregate amount of approximately $5.5 million. In addition, we issued 461,729 shares of common stock, net of RSAs canceled, in connection with the grants of RSAs and 92,201 shares of common stock upon vesting of certain RSUs. The RSU holders surrendered 61,923 RSUs to pay for minimum withholding taxes totaling approximately $0.4 million.
In February 2012, we completed an underwritten public offering and issued 7,820,000 shares of common stock, including 1,020,000 shares sold pursuant to the full exercise of an over-allotment option granted to the underwriter. All of the shares were offered at a public offering price of $10.61 per share, generating approximately $81.5 million in net proceeds. Of the 7,820,000 shares of common stock sold, Randal J. Kirk, a member of our board of directors, through his affiliates, purchased 1,360,000 shares of common stock in this offering at the public offering price of $10.61 per share for a total of approximately $14.4 million.


F-23



8.Equity Incentive Plans
We currently grant stock options, restricted stock awards and restricted stock units under the Amended and Restated 2011 Stock Plan ("(“2011 Stock Plan"Plan”). In, which was approved by the stockholders on May 2013, our stockholders approved the Amended and Restated 2011 Stock Plan, which6, 2016 and provides for the grant of up to 12.544.2 million shares of common stock (subject to certain limitations as described in the Amended and Restated 2011 Stock Plan) to selected employees, consultants and non-employee members of our Board of Directors (“Outside Directors”) as stock options, stock appreciation rights, restricted stock awards, restricted stock unit awards and performance awards. The 2011 Stock Plan was approved by the stockholders. Awards are subject to terms and conditions established by the Compensation Committee of our Board of Directors. During the year ended December 31, 2014,2017, we granted share-based awards under the 2011 Stock Plan. At December 31, 2014, 7,247,4522017, 13,023,641 shares were subject to outstanding awards and 4,681,2126,552,249 shares were available for future grants of share-based awards. At the present time, management intends to issue new common shares upon the exercise of stock options, issuance of restricted stock awards and settlement of restricted stock units.
Total share-based compensation expense related to share-based awards was comprised of the following (in thousands):
 Year Ended December 31, Year Ended December 31,
 2014 2013 2012 2017 2016 2015
Research and development $7,939
 $4,476
 $4,191
 $13,080
 $11,470
 $9,795
Selling, general and administrative 7,335
 5,062
 4,158
 17,590
 14,115
 11,043
Share-based compensation expense $15,274
 $9,538
 $8,349
 $30,670
 $25,585
 $20,838

F-31

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Share-based compensation expense by type of share-based award (in thousands):
  Year Ended December 31,
  2017 2016 2015
Stock options $19,583
 $16,544
 $11,145
RSAs, RSUs and PRSUs 11,087
 9,041
 9,693
  $30,670
 $25,585
 $20,838
Total unrecognized estimated compensation expensecost by type of award and the weighted-average remaining requisite service period over which such expense is expected to be recognized (in thousands, unless otherwise noted):
 December 31, 2014 December 31, 2017
 Unrecognized
Expense
 Remaining
Weighted-Average
Recognition Period
(years)
 
Unrecognized
Expense
 
Remaining
Weighted-Average
Recognition Period
(years)
Stock options $15,675
 2.7 $36,914
 2.4
RSAs $7,153
 1.6 $4,610
 1.6
RSUs $3,991
 2.6 $15,965
 2.6
Cash flows resulting from tax deductions in excess of the cumulative compensation cost recognized for options exercised (excess tax benefits) are classified as cash inflows provided by financing activities and cash outflows used in operating activities. Due to our net loss position, no tax benefits have been recognized in the consolidated statements of cash flows.
Stock Options. Options granted under the Plans must have an exercise price equal to at least 100% of the fair market value of our common stock on the date of grant. The options will generally have a maximum contractual term of ten years and vest at the rate of one-fourth of the shares on the first anniversary of the date of grant and 1/48 of the shares monthly thereafter. Certain option awards provide for accelerated vesting if there is a change in control (as defined in the Plans).


F-24
F-32

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


A summary of our stock option award activity as of and for the years ended December 31, 2014, 20132017, 2016 and 20122015 is as follows: 

 
Shares
Underlying
Stock Options

Weighted
Average Exercise
Price per Share

Weighted Average
Remaining
Contractual Term (years)
 
Aggregate
Intrinsic
Value
 Shares
Underlying
Stock Options
 Weighted
Average Exercise
Price per Share
 Weighted
Average
Remaining
Contractual
Term (years)
 Aggregate
Intrinsic
Value
Outstanding at January 1, 2012 5,869,784
 $5.82    
Outstanding at January 1, 2015 6,353,892
 $9.18    
Granted 1,215,442
 $9.90   3,973,604
 $16.26  
Exercised (444,637) $4.56   (1,926,368) $7.49  
Canceled/forfeited (260,722) $8.34   (407,936) $10.64  
Outstanding at December 31, 2012 6,379,867
 $6.59  
Outstanding at December 31, 2015 7,993,192
 $13.03  
Granted 1,806,392
 $7.14   4,466,306
 $9.03  
Exercised (1,270,362) $4.34   (413,248) $6.88  
Canceled/forfeited (214,982) $8.18   (955,054) $12.42  
Outstanding at December 31, 2013 6,700,915
 $7.11  
Outstanding at December 31, 2016 11,091,196
 $11.70  
Granted 2,271,143
 $13.02   2,717,614
 $12.60  
Exercised (1,432,206) $5.43   (1,514,826) $9.24  
Canceled/forfeited (1,185,960) $9.39   (1,185,518) $11.89  
Outstanding at December 31, 2014 6,353,892
 $9.18 6.5 
$11.2 million
Vested and expected to vest at December 31, 2014 6,025,883
 $9.05 6.4 
$10.9 million
Exercisable at December 31, 2014 3,944,408
 $7.36 4.6 
$8.9 million
Outstanding at December 31, 2017 11,108,466
 $12.24 7.0 
$90.8 million
Vested and expected to vest at December 31, 2017 11,108,466
 $12.24 7.0 
$90.8 million
Exercisable at December 31, 2017 5,493,802
 $12.31 6.1 
$44.7 million
The weighted average grant-dategrant date fair values of options granted during the years ended December 31, 2014, 20132017, 2016 and 20122015 were $8.13$7.86 per share, $4.40$5.36 per share and $5.63$9.60 per share, respectively. The total intrinsic value of options exercised during the years ended December 31, 2014, 20132017, 2016 and 20122015 was approximately $8.1$10.0 million,, $8.3 $1.4 million and $2.9$16.2 million,, respectively. Cash received from stock option exercises for the years ended December 31, 2014, 20132017, 2016 and 20122015 was approximately $7.8$14.0 million,, $5.5 $2.8 million and $2.0$14.4 million,, respectively.
The exercise price of stock options granted is equal to the closing price of the common stock on the date of grant. The fair value of each option award is estimated on the date of grant using the Black-Scholes-Merton option pricing model (“Black-Scholes model”) that uses the assumptions noted in the following table.. Expected volatility is based on historical volatility of our common stock. The expected term of options granted is based on analyses of historical employee termination rates and option exercises. The risk-free interest rate is based on the U.S. Treasury yield for a period consistent with the expected term of the option in effect at the time of the grant. The dividend yield assumption is based on the expectation of no future dividend payments by us. Assumptionspayments. The assumptions used in the Black-Scholes model were as follows:
 Year Ended December 31, Year Ended December 31,
 2014
2013
2012 2017 2016 2015
Expected volatility 66.6-71.8%

70.1-72.5%

64.0-69.2%
 69.8-71.7% 67.5-71.9% 66.2-67.4%
Average expected term (in years) 5.7

5.7

5.6
 5.6 5.4 5.6
Risk-free interest rate 1.73-2.04%

0.86-2.00%

0.80-1.15%
 1.73-2.13% 1.00-1.90% 1.34-1.92%
Expected dividend yield 0%
0%
0%   


F-25
F-33

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Restricted Stock AwardsRestricted stock awardsRSAs are grants that entitle the holder to acquire shares of our common stock at zero or a fixed price, which is typically nominal.cost. The shares covered by a restricted stock awardRSA cannot be sold, pledged, or otherwise disposed of until the award vests and any unvested shares may be reacquired by us for the original purchase price following the awardee’s termination of service. The restricted stock awardsRSAs will generally vest at the rate of one-fourth of the shares on each anniversary of the date of grant. Annual grants of restricted stock awardsRSAs to Outsidethe Board of Directors typically vest in full the first day the awardee may trade our stock in compliance with our insider trading policy following the date immediately preceding the first annual meeting of stockholders following the grant date.approximately one year.
The following table summarizes our restricted stock awardRSA activity during the years ended December 31, 2014, 20132017, 2016 and 2012:2015:

 
Number of
Shares

Weighted  Average
Grant Date
Fair Value
 Number of
Shares
 Weighted
Average
Grant Date
Fair Value
Unvested at January 1, 2012 347,883
 $6.51
Unvested at January 1, 2015 1,158,451
 $10.26
Granted 380,158
 $10.29 515,695
 $15.00
Vested (339,758) $6.51 (721,990) $10.11
Forfeited (5,963) $10.81 (140,676) $11.84
Unvested at December 31, 2012 382,320
 $10.21
Unvested at December 31, 2015 811,480
 $13.13
Granted 476,096
 $6.88 968,652
 $8.41
Vested (211,178) $8.78 (296,831) $12.76
Forfeited (14,367) $8.17 (180,198) $10.33
Unvested at December 31, 2013 632,871
 $8.23
Unvested at December 31, 2016 1,303,103
 $10.09
Granted 1,055,122
 $11.15 98,945
 $14.15
Vested (263,765) $8.33 (514,613) $10.23
Forfeited (265,777) $10.86 (108,485) $9.62
Unvested at December 31, 2014 1,158,451
 $10.26
Unvested at December 31, 2017 778,950
 $10.59
The estimated fair value of the restricted stock awards isRSAs was based on the closing market value of our common stock on the date of grant. The total grant-dategrant date fair value of restricted stock awardsRSAs vested during the years ended December 31, 2014, 20132017, 2016 and 20122015 was approximately $2.2$5.3 million,, $1.9 $3.8 million and $2.2$7.3 million,, respectively. We recognized approximately $5.4 million, $2.2 million and $2.1 millionThe fair value of share-based compensation expense related to restricted stock awards forRSAs vested during the years ended December 31, 2014, 20132017, 2016 and 2012,2015, was approximately $6.6 million, $2.5 million and $13.9 million, respectively.


F-26
F-34

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Restricted Stock Units. A restricted stock unitRSU is a promise by us to issue a share of our common stock upon vesting of the unit. The restricted stock unitsRSUs will generally vest at the rate of one-fourth of the shares on each anniversary of the date of grant.
The following table summarizes our restricted stock unitRSU activity during the years ended December 31, 2014, 20132017, 2016 and 2012:2015:

 
Number of
Shares

Weighted
Average
Grant Date
Fair Value

Weighted
Average  Remaining
Contractual Term (yrs)
 
Aggregate
Intrinsic
Value
 Number of
Shares
 Weighted
Average
Grant Date
Fair Value
 Weighted
Average
Remaining
Contractual
Term (yrs)
 Aggregate
Intrinsic
Value
Unvested at January 1, 2012 148,000
 
    
Unvested at January 1, 2015 462,322
 $11.12    
Granted 682,146
 $10.61   422,492
 $14.75  
Vested (128,000) 
   (134,088) $10.93  
Forfeited (20,000) 
   (84,512) $10.86  
Outstanding at December 31, 2012 682,146
 
  
Outstanding at December 31, 2015 666,214
 $13.49  
Granted 323,700
 $6.69   796,582
 $8.17  
Vested (154,124) 
   (218,279) $12.74  
Forfeited (115,367) 
   (77,948) $10.99  
Outstanding at December 31, 2013 736,355
 $9.06  
Outstanding at December 31, 2016 1,166,569
 $10.16  
Granted 305,535
 $13.71   1,378,273
 $12.13  
Vested (194,368) $9.12   (378,406) $10.48  
Forfeited (385,200) $8.84   (251,261) $11.11  
Outstanding at December 31, 2014 462,322
 $11.12 2.6 
$4.5 million
Outstanding at December 31, 2017 1,915,175
 $11.39 1.4 
$38.8 million
The estimated fair value of the restricted stock unitsRSUs was based on the closing market value of our common stock on the date of grant. The total intrinsicgrant date fair value of restricted stock unitsRSUs vested during the years ended December 31, 2014, 20132017, 2016 and 20122015 was approximately $2.6$4.0 million, $1.1$2.8 million and $0.9$1.5 million,, respectively. We recognized approximately $2.0 million, $1.8 million and $1.5 millionThe fair value of share-based compensation expense related to the restricted stock units forRSUs vested during the years ended December 31, 2014, 20132017, 2016 and 2012,2015 was approximately $4.7 million, $2.1 million and $1.8 million, respectively.

F-35

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Performance Restricted Stock Units. A PRSU is a promise by us to issue a share of our common stock upon achievement of a specific performance condition.
In July 2014, we granted 540,742 PRSUs toThe following table summarizes our executive officers, which are not included inPRSU activity during the table above. For the yearyears ended December 31, 2014, there were 109,5042017, 2016 and 2015:
  Number of
Shares
 
Weighted
Average
Grant Date
Fair Value
Outstanding at January 1, 2015 431,238
 
$8.91
Granted 118,209
 
$11.19
Vested (83,380) 
$9.48
Forfeited (156,360) 
$9.21
Outstanding at December 31, 2015 309,707
 
$9.48
Granted 
 
Vested (30,037) 
$9.49
Forfeited (79,415) 
$9.44
Outstanding at December 31, 2016 200,255
 
$9.49
Granted 
 
Vested 
 
Forfeited (200,255) 
$9.49
Outstanding at December 31, 2017 
 
The estimated fair value of the PRSUs forfeited. Aswas based on the closing market value of December 31, 2014,our common stock on the aggregatedate of grant. The total grant date fair value and intrinsic value of PRSUs vested during the 431,238 PRSUs outstanding was $4.2 million. For the yearyears ended December 31, 2014, there2017, 2016 and 2015 was no expense recognized relatedapproximately zero, $0.3 million and $0.8 million, respectively.
8. Stockholders’ Equity (Deficit)
In May 2017, we completed an underwritten public offering pursuant to which we sold 11.5 million shares of common stock, including 1.5 million shares sold pursuant to the PRSUs, asfull exercise of an option to purchase additional shares granted to the achievementunderwriters. All of the performance conditions was not determinedshares were offered at a public offering price of $12.50 per share, generating $134.9 million in net proceeds, after deducting underwriting discounts and commissions and other offering expenses. We intend to be probable.use the net proceeds from this offering to fund continued development of our PEGPH20 oncology program and for other general corporate purposes.
During the years ended December 31, 2017, 2016 and 2015, we issued an aggregate of 1,514,826, 413,248 and 1,926,368 shares of common stock, respectively, in connection with the exercises of stock options, for net proceeds of approximately $14.0 million, $2.8 million and $14.4 million, respectively. For the years ended December 31, 2017, 2016 and 2015, we issued 281,398, 134,944 and 82,069 shares of common stock, respectively, upon vesting of certain RSUs for which the RSU holders surrendered 97,008, 83,335 and 52,019 RSUs, respectively, to pay for minimum withholding taxes totaling approximately $1.9 million, $0.8 million and $0.7 million, respectively. In addition, we canceled 9,540 shares of common stock, net of issuances and issued 780,066 and 375,019 shares of common stock, net of cancellations in connection with grants of RSAs during the years ended December 31, 2017, 2016 and 2015, respectively.

F-36

9.Commitments and Contingencies
Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


9. Commitments and Contingencies
Operating Leases
Our administrative offices and research facilities are located in San Diego, California. We lease an aggregate of approximately 76,000 square feet of office and research space in four buildings. The leases commenced in June 2011 and November 2013 and continue through January 2018.2023. The leases are subject to approximately 2.5% to 3.0% annual increases throughout the terms of the leases. We also pay a pro rata share of operating costs, insurance costs, utilities and real property taxes.
We received incentives under the leases, including tenant improvement allowances and reduced or free rent, for which the unamortized deferred rent balances associated with these incentives was $1.0$0.3 million and $0.4 million as of December 31, 20142017 and 2013.2016, respectively.
In November 2015, we opened a satellite office in South San Francisco, California. We lease approximately 10,000 square feet of office space. The lease commenced in November 2015 and continues through January 2021. The lease is subject to approximately 3.0% annual increases throughout the term of the lease. We also pay a pro rata share of operating costs, insurance costs, utilities and real property taxes. We received incentives under the lease, including tenant improvement allowances and reduced or free rent, for which the unamortized deferred rent balances associated with these incentives was $0.3 million and $0.4 million as of December 31, 2017 and 2016, respectively.
Additionally, we lease certain office equipment under operating leases. Total rent expense was approximately $1.9$2.3 million,, $1.7 $2.2 million and $1.6$1.9 million for the years ended December 31, 2014, 20132017, 2016 and 2012,2015, respectively.


F-27



Approximate annual future minimum operating lease payments as of December 31, 20142017 are as follows (in thousands): 
Year: 
Operating
Leases
 Operating
Leases
2015 $2,133
2016 2,142
2017 2,162
2018 82
 $2,415
2019 2,785
2020 2,824
2021 2,470
2022 2,506
Total minimum lease payments $6,519
 $13,000
Other Commitments
In order to scale up the production of bulk rHuPH20 and to identify another manufacturer that would help meet anticipated production obligations arising from our proprietary programs and our collaborations, we entered into a Technology Transfer Agreement and a Clinical Supply Agreement with Cook Pharmica LLC (“Cook”). The technology transfer was completed in 2008. In 2009, multiple batches of bulk rHuPH20 were produced to support planned future clinical studies.
In March 2010, we entered into a Commercial Supply Agreement with Cook (the “Cook Commercial Supply Agreement”). Under the terms of the Cook Commercial Supply Agreement, Cook will manufacture certain batches of bulk rHuPH20 that will be used for commercial supply of certain products and product candidates. Under the terms of the Cook Commercial Supply Agreement, we are committed to certain minimum annual purchases of bulk rHuPH20 equal to four quarters of forecasted supply. At December 31, 2014, we had no minimum purchase obligation in connection with the Cook Commercial Supply Agreement.
In March 2010, we amended our Commercial Supply Agreement (the “March 2010 Avid Amendment”) with Avid Bioservices, Inc. (“Avid”) which was originally entered into in February 2005 and amended in December 2006. Under the terms of the March 2010 Avid Amendment, we are committed to certain minimum annual purchases of bulk rHuPH20 equal to three quarters of forecasted supply. In addition, Avid has the right to manufacture and supply a certain percentage of bulk rHuPH20 that will be used in Hylenex recombinant. At December 31, 2014, we had no minimum purchase obligations with this agreement.
In March 2010, we entered into a second Commercial Supply Agreement with Avid (the “Avid Commercial Supply Agreement”). Under the terms of the Avid Commercial Supply Agreement, we are committed to certain minimum annual purchases of bulk rHuPH20 equal to three quarters of forecasted supply. In addition, Avid has the right to manufacture and supply a certain percentage of bulk rHuPH20 that will be used in the collaboration products. At December 31, 2014,2017, we had a $9.6$7.2 million minimum purchase obligation in connection with this agreement.
In June 2011, we entered into a commercial manufacturing and supply agreement with Baxter, under which Baxter provides the final fill and finishing steps in the production process of Hylenex recombinant for a limited period of time. The initial term of the agreement with Baxter was extended to December 31, 2015, subject to further extensions in accordance with the terms and conditions of the agreement. At December 31, 2014, we had a minimum purchase obligation in connection with this agreement of approximately $1.7 million.
In June 2011, we entered into a services agreement with another third party manufacturerPatheon for the technology transfer and manufacture of Hylenex recombinant. At December 31, 2014,2017, we had a $1.2$0.3 million minimum purchase obligation in connection with this agreement.
Contingencies
We have entered into an in-licensing agreement with a research organization, which is cancelable at our option with 90 days written notice. Under the terms of this agreement, we have received a license to the know-how and technology claimed, in certain patents or patent applications. We are required to pay fees, milestones and/or royalties on future sales of products employing the technology or falling under claims of a patent, and some of the agreements require minimum royalty payments. We continually reassess the value of the license agreement. If the in-licensed and research candidate is successfully developed, we may be required to pay milestone payments of approximately $9.3$8.0 million over the life of this agreement in addition to royalties on sales of the affected products. Due to the uncertainties of the development process, the timing and probability of the remaining milestone and royalty payments cannot be accurately estimated.


F-28
F-37

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


Legal Contingencies
From time to time, we may be involved in disputes, including litigation, relating to claims arising out of operations in the normal course of our business. Any of these claims could subject us to costly legal expenses and, while we generally believe that we have adequate insurance to cover many different types of liabilities, our insurance carriers may deny coverage or our policy limits may be inadequate to fully satisfy any damage awards or settlements. If this were to happen, the payment of any such awards could have a material adverse effect on our consolidated results of operations and financial position. Additionally, any such claims, whether or not successful, could damage our reputation and business. We currently are not a party to any legal proceedings, the adverse outcome of which, in management’s opinion, individually or in the aggregate, would have a material adverse effect on our consolidated results of operations or financial position.
10.Income Taxes
The Tax Cuts and Jobs Act (the “Act”) was enacted in December 2017. The Act reduces the U.S. federal corporate tax rate from 35% to 21%. As of December 31, 2017, we have not completed our accounting for the tax effects of the Act; however, we have made a reasonable estimate of the effects on our existing deferred tax balances. We remeasured certain deferred tax assets and liabilities based on the rates at which they are expected to reverse in the future. However, we are still analyzing certain aspects of the Act and refining our calculations, which could potentially affect the measurement of these balances or potentially give rise to new deferred tax amounts. The provisional amount recorded related to the remeasurement of our deferred tax balance was $17.1 million. The provisional amount of $17.1 million was fully offset by a change in the valuation allowance.
Total income (loss) before income taxes summarized by region were as follows (in thousands):
  Year Ended December 31,
  2017 2016 2015
United States $160,938
 $6,384
 $11,724
Foreign (99,328) (108,245) (43,955)
Net income (loss) before income taxes $61,610
 $(101,861) $(32,231)
Significant components of our net deferred tax assets at December 31, 2014 and 2013 are shown belowassets/(liabilities) were as follows (in thousands).
  December 31,
  2017 2016
Deferred tax assets:    
Net operating loss carryforwards $32,630
 $103,296
Deferred revenue 8,815
 15,354
Research and development and orphan drug credits 75,224
 73,701
Share-based compensation 7,423
 8,844
Alternative minimum tax credit 5,532
 1,494
Other, net 2,270
 1,021
  131,894
 203,710
Valuation allowance for deferred tax assets (126,189) (203,370)
Deferred tax assets, net of valuation 5,705
 340
Deferred tax liabilities:    
Depreciation (173) (340)
Total deferred tax liabilities (173) (340)
Net deferred tax asset (liability) $5,532
 $

F-38

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


A valuation allowance of $179.0$126.2 million and $162.0$203.4 million has been established to offset the net deferred tax assets as of December 31, 20142017 and 2013,2016, respectively, as realization of such assets is uncertain. Under the Act, the AMT credit carryovers can be used to offset regular tax liability for taxable years beginning after 2017. If not utilized before 2022, any remaining AMT credit carryover will be fully refundable. Accordingly, the recognized deferred tax asset, on a provisional basis, as of December 31, 2017 is the AMT credit carryover that will either be utilized or refunded.
Income tax expense was comprised of the following components (in thousands):
  December 31,

 2014
2013
Deferred tax assets: 


Net operating loss carryforwards $120,707
 $116,572
Deferred revenue 18,034
 13,324
Research and development credits 34,146
 28,867
Share-based compensation 5,381
 2,495
Other, net 891
 853

 179,159
 162,111
Valuation allowance for deferred tax assets (178,965) (161,968)
Deferred tax assets, net of valuation 194
 143
Deferred tax liabilities:    
Depreciation (194) (143)
Net deferred tax liabilities (194) (143)
Net deferred tax assets $
 $
  Year Ended December 31,
  2017 2016 2015
Current - federal $4,051
 $1,145
 $
Current - state 120
 17
 
Deferred - federal (5,532) 
 
Deferred - state 
 
 
  $(1,361) $1,162
 $
The provision for income taxes on earnings subject to income taxes differs from the statutory federal income tax rate due to the following (in thousands):
  December 31,

 2014
2013
2012
Federal income tax at 34% $(23,247) $(28,383) $(18,208)
State income tax, net of federal benefit (1,761) (1,745) (3,023)
Increase in valuation allowance 16,998
 33,525
 20,954
Foreign income subject to tax at other than federal statutory rate 12,747
 
 
Tax effect on non-deductible expenses and other 540
 5,219
 1,293
Research and development credits (5,277) (8,616) (1,016)

 $
 $
 $
  Year Ended December 31,
  2017 2016 2015
Federal income tax expense (benefit) at 34% $20,947
 $(34,633) $(10,959)
State income tax benefit, net of federal income tax impact 930
 (653) 5,524
(Decrease) increase in valuation allowance (77,181) 11,252
 4,045
Enactment of the Tax Cuts and Jobs Act 17,132
 
 
Foreign income subject to tax at other than federal statutory rate 33,674
 36,803
 14,945
Shared-based compensation 525
 3,735
 (4,990)
Non-deductible expenses and other 5,779
 698
 6,457
Research and development credits, net 4,162
 (1,084) (3,861)
Orphan drug credits, net of federal add back (7,329) (14,956) (11,161)
  $(1,361) $1,162
 $

At December 31, 2017, our unrecognized tax benefit and uncertain tax positions were $14.4 million. None of this amount would affect the effective tax rate and $14.4 million would affect the effective tax rate in the event the valuation allowance was removed. Of the unrecognized tax benefits, we do not expect any significant changes to occur in the next 12 months. Interest and/or penalties related to uncertain income tax positions are recognized by us as a component of income tax expense. For the years ended December 31, 2017, 2016 and 2015, we recognized no interest or penalties.
The following table summarizes the activity related to our unrecognized tax benefits (in thousands):
  Year Ended December 31,
  2017 2016 2015
Gross unrecognized tax benefits at beginning of period $12,799
 $4,898
 $
Increases in tax positions for prior years 
 5,615
 
Decreases in tax positions for prior years (2,518) (4,898) 
Increases in tax positions for current year 4,147
 7,184
 4,898
Gross unrecognized tax benefits at end of period $14,428
 $12,799
 $4,898

F-29
F-39

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


At December 31, 2014,2017, we had federal, California and Californiaother state tax net operating loss carryforwards of approximately $341.9$88.5 million, $243.1 million and $307.9$13.9 million,, respectively. Included in these amounts are
The following table shows key expiration dates of the federal and California net operating losses of approximately $34.3 million and $32.8 million, respectively, attributable to stock option, RSA and RSU deductions for which the tax benefit will be credited to equity when realized. The federal and California tax loss carryforwards will begin to expire in 2018 and 2015, respectively, unless previously utilized.(in thousands):
    Expires in:
  Net Operating Loss 2018 2021 and beyond 2028 and beyond
Federal $88,516
 
 $88,516
 
California $243,080
 
 
 $243,080
At December 31, 2014,2017, we also had federal and California research and development tax credit carryforwards of approximately $26.1$19.1 million and $12.2$11.7 million,, respectively. The federal research and development tax credits will begin to expire in 2024 unless previously utilized. The California research and development tax credits will carryforward indefinitely until utilized. Additionally, we had Orphan Drug Credit carryforwards of $57.3 million which will begin to expire in 2035.
Pursuant to Internal Revenue Code Section 382, the annual use of the net operating loss carryforwards and research and development tax credits could be limited by any greater than 50% ownership change during any three-yearthree year testing period. As a result of any such ownership change, portions of our net operating loss carryforwards and research and development tax credits are subject to annual limitations. We recently completed an updatedA Section 382 analysis regarding the limitation of the net operating losses and research and development credits was completed as of June 30, 2014.November 1, 2017. Based upon the analysis, we do not believe an ownership change occurred during 2017. Based upon previous analysis it was determined that ownership changes occurred in prior years. However,years; however, the annual limitations on net operating loss and research and development tax credit carryforwards will not have a material impact on the future utilization of such carryforwards.
At December 31, 2014 and 2013, our unrecognized income tax benefits and uncertain tax positions were not material and would not, if recognized, affect the effective tax rate. Interest and/or penalties related to uncertain income tax positions are recognized by us as a component of income tax expense. For the years ended December 31, 2014, 2013 and 2012, we recognized no interest or penalties.
We do not provide for U.S. income taxes on the undistributed earnings of our foreign subsidiarysubsidiaries as it is our intention to utilize those earnings in the foreign operations for an indefinite period of time. At December 31, 2014,2017 and 2016, there were no undistributed earnings in the foreign subsidiary.subsidiaries.
We are subject to taxation in the U.S. and in various state and foreign jurisdictions. Our tax years for 1998 and forward are subject to examination by the U.S. and California tax authorities due to the carryforward of unutilized net operating losses and research and development credits.
A new Swiss subsidiary, Halozyme Switzerland GmbH, was formed during the fourth quarter of 2016 and obtained a tax ruling from Canton of Basel Stadt for its operations in Switzerland. The tax ruling is dated December 21, 2016, and will continue for a period of ten years, not to extend beyond December 31, 2026. The combined income tax burden at the federal, cantonal and communal level will not exceed 10% during the period covered by the ruling. As a result of foreign losses and a full valuation allowance, no net tax benefit was derived for the year ended December 31, 2017 as a result of the tax ruling.
11.Employee Savings Plan
We have an employee savings plan pursuant to Section 401(k) of the Internal Revenue Code. All employees are eligible to participate, provided they meet the requirements of the plan. We are not required to make matching contributions under the plan. However, we voluntarily contributed to the plan approximately $0.7$1.2 million,, $0.6 $1.0 million and $0.5$0.7 million for the years ended December 31, 2014, 20132017, 2016 and 2012,2015, respectively.

F-40

Halozyme Therapeutics, Inc.
Notes to Consolidated Financial Statements — (Continued)


12.Related Party Transactions
In June 2011, we and Intrexon entered into the Intrexon Collaboration, under which Intrexon obtained a worldwide exclusive license for the use of rHuPH20 enzyme in the development of a subcutaneous injectable formulation of Intrexon’s recombinant human alpha 1-antitrypsin (rHuA1AT). The Intrexon Collaboration was terminated in May 2014. Intrexon’s chief executive officer and chairman of its board of directors, Randal J. Kirk, is also a member of our Board of Directors. The collaborative arrangement with Intrexon was reviewed and approved by our Board of Directors in accordance with our related party transaction policy. For the years ended December 31, 2014, 2013 and 2012, we recognized zero, $1.0 million and $1.0 million, respectively, in revenue under collaborative agreements pursuant to the terms of the Intrexon Collaboration. See Note 4, Collaborative Agreements, for a further discussion of the Intrexon Collaboration.


F-30



13.Restructuring Expense
In November 2014, we completed a corporate reorganization to focus our resources on advancing our PEGPH20 oncology proprietary program and Enhanze collaborations. This reorganization resulted in a reduction in the workforce of approximately 13%, primarily in research and development.
We recorded approximately $1.2 million of severance pay and benefits expense in connection with the reorganization, of which $1.1 million and $0.1 million was included in research and development expense and selling, general and administrative expense, respectively, in the consolidated statement of operations for the year ended December 31, 2014. No other restructuring charges were incurred. We made cash payments of $0.7 million related to the restructuring expense for the year ended December 31, 2014. As of December 31, 2014, the restructuring liability was approximately $0.5 million and was included in current accrued expenses. The balance is expected to be paid in 2015.


F-31



14.Summary of Unaudited Quarterly Financial Information
The following is a summary of our unaudited quarterly results for the years ended December 31, 20142017 and 20132016 (in thousands):
 Quarter Ended Quarter Ended
2014 (Unaudited): March 31, June 30, September 30, December 31,
Total revenues(1)
 $11,966
 $18,385
 $14,606
 $30,377
2017 (Unaudited): March 31, June 30, September 30, December 31,
Total revenues (1) (2)
 $29,568
 $33,750
 $63,731
 $189,564
Gross profit on product sales $3,890
 $4,992
 $5,257
 $5,105
Total operating expenses $57,094
 $59,228
 $55,654
 $63,635
Net income (loss) $(32,897) $(30,763) $2,749
 $123,882
Net income (loss) per share:        
Basic $(0.26) $(0.23) $0.02
 $0.87
Diluted $(0.26) $(0.23) $0.02
 $0.85
Shares used in computing net income (loss) per share:        
Basic 128,615
 134,013
 141,190
 141,718
Diluted 128,615
 134,013
 143,236
 145,633
 Quarter Ended
2016 (Unaudited): March 31, June 30, September 30, December 31,
Total revenues $42,499
 $33,336
 $31,853
 $39,003
Gross profit on product sales $3,048
 $3,570
 $4,476
 $3,997
 $5,178
 $5,391
 $4,197
 $5,420
Total operating expenses $37,185
 $33,325
 $33,632
 $34,228
 $58,668
 $55,059
 $54,596
 $61,578
Net loss $(26,548) $(16,273) $(20,280) $(5,274) $(19,816) $(26,875) $(28,946) $(27,386)
Net loss per share, basic and diluted $(0.22) $(0.13) $(0.16) $(0.04) $(0.16) $(0.21) $(0.23) $(0.21)
Shares used in computing basic and diluted net loss
per share
 118,943
 123,710
 124,041
 124,272
 127,615
 127,958
 128,154
 128,185
        
 Quarter Ended
2013 (Unaudited): March 31, June 30, September 30, December 31,
Total revenues $11,833
 $14,454
 $16,013
 $12,499
Gross profit on product sales(2)
 $769
 $1,816
 $9,342
 $6,266
Total operating expenses $30,330
 $36,574
 $34,507
 $33,822
Net loss $(19,289) $(22,912) $(19,292) $(21,986)
Net loss per share, basic and diluted $(0.17) $(0.20) $(0.17) $(0.19)
Shares used in computing basic and diluted net loss
per share
 112,417
 112,486
 112,765
 113,550
_______________
(1)Revenues for the quarter ended December 31, 20142017 included $101.4 million, $40.0 million and $15.0 million in revenue under collaborative agreementsarrangements from theBMS, Alexion and Janssen, Collaboration.respectively.
(2)Gross profit on product salesRevenues for the quartersquarter ended June 30, 2013, September 30, 2013 and December 31, 2013 excluded manufacturing costs related to2017 included $30.0 million in revenue under collaborative arrangements from the product sales of bulk rHuPH20 for Herceptin SC and HyQvia in the amounts of $0.9 million, $6.5 million and $2.6 million, respectively. Such costs were incurred prior to European marketing approvals for Herceptin SC and HyQvia, and therefore, they were charged to research and development expenses in the periods the costs were incurred.2017 Roche Collaboration.


F-32



HALOZYME THERAPEUTICS, INC.

Halozyme Therapeutics, Inc.
Schedule II
Valuation and Qualifying Accounts
(in thousands)
 Balance at Beginning of Period Additions Deductions Balance at End of Period Balance at Beginning of Period Additions Deductions Balance at End of Period
For the year ended December 31, 2014        
For the year ended December 31, 2017        
Accounts receivable allowances (1)
 $610
 $4,518
 $(4,519) $609
 $559
 $4,645
 $(4,645) $559
For the year ended December 31, 2013        
For the year ended December 31, 2016        
Accounts receivable allowances (1)
 $178
 $2,979
 $(2,547) $610
 $967
 $4,795
 $(5,203) $559
For the year ended December 31, 2012        
For the year ended December 31, 2015        
Accounts receivable allowances (1)
 $15
 $771
 $(608) $178
 $611
 $4,150
 $(3,794) $967
_______________
(1)
Allowances are for chargebacks, prompt payment discounts and distribution fees related to Hylenex recombinant product sales.


F-33
F-42



Exhibit Index
Incorporated by Reference
ExhibitFiled
NumberExhibit TitleHerewithFormFile No.Date Filed
2.1Agreement and Plan of Merger, dated November 14, 2007, by and between the Registrant and the Registrant’s predecessor Nevada corporation8-K001-3233511/20/2007
3.1Composite Certification of Incorporation10-Q001-323358/7/2013
3.2Certificate of Designation, Preferences and Rights of the terms of the Series A Preferred Stock8-K001-3233511/20/2007
3.3Bylaws, as amended8-K001-3233512/12/2011
4.1Amended Rights Agreement between Corporate Stock Transfer, as rights agent, and Registrant, dated November 12, 200710-K001-323353/14/2008
10.1License Agreement between University of Connecticut and Registrant, dated November 15, 2002SB-2333-1147764/23/2004
10.2First Amendment to the License Agreement between University of Connecticut and Registrant, dated January 9, 20068-K001-323351/12/2006
10.3*Commercial Supply Agreement with Avid Bioservices, Inc. and Registrant, dated February 16, 20058-K001-323352/22/2005
10.4*First Amendment to the Commercial Supply Agreement between Avid Bioservices, Inc. and Registrant, dated December 15, 20068-K001-3233512/21/2006
10.5*Clinical Supply Agreement between Cook Pharmica, LLC and Registrant, dated August 15, 200810-Q001-3233511/7/2008
10.6#2004 Stock Plan and Form of Option Agreement thereunderSB-2333-1147767/23/2004
10.7#Halozyme Therapeutics, Inc. 2005 Outside Directors’ Stock Plan8-K001-323357/6/2005
10.8#Form of Stock Option Agreement (2005 Outside Directors’ Stock Plan)10-Q001-323358/8/2006
10.9#Form of Restricted Stock Agreement (2005 Outside Directors’ Stock Plan)10-Q001-323358/8/2006
10.10#Halozyme Therapeutics, Inc. 2006 Stock Plan8-K001-323353/24/2006
10.11#Form of Stock Option Agreement (2006 Stock Plan)10-Q001-323358/8/2006
10.12#Form of Restricted Stock Agreement (2006 Stock Plan)10-Q001-323358/8/2006
10.13#Halozyme Therapeutics, Inc. 2008 Stock Plan8-K001-323353/19/2008





Incorporated by Reference
ExhibitFiled
NumberExhibit TitleHerewithFormFile No.Date Filed
10.14#Form of Stock Option Agreement (2008 Stock Plan)10-Q001-323358/7/2009
10.15#Form of Restricted Stock Agreement (2008 Stock Plan)10-Q001-323358/7/2009
10.16#Halozyme Therapeutics, Inc. 2008 Outside Directors’ Stock Plan8-K001-323353/19/2008
10.17#Form of Restricted Stock Agreement (2008 Outside Directors’ Stock Plan)10-Q001-323358/7/2009
10.18#Halozyme Therapeutics, Inc. Amended and Restated 2011 Stock PlanDEF14A001-323354/11/2013
10.19#Form of Stock Option Agreement (2011 Stock Plan)8-K001-323356/16/2011
10.20#Form of Stock Option Agreement for Executive Officers (2011 Stock Plan)8-K001-323356/16/2011
10.21#Form of Restricted Stock Units Agreement (2011 Stock Plan)8-K001-323356/16/2011
10.22#Form of Restricted Stock Award Agreement (2011 Stock Plan)8-K001-323356/16/2011
10.23#Form of Indemnity Agreement for Directors and Executive Officers8-K001-3233512/20/2007
10.24#Severance Policy10-Q001-323355/9/2008
10.25#Form of Change In Control Agreement with CEO10-K001-323352/28/2013
10.26#Form of Change In Control Agreement with CEO10-K001-323352/28/2014
10.27#Form of Amended and Restated Change In Control Agreement with Officer10-K001-323352/28/2013
10.28Separation Agreement and General Release of All Claims between Halozyme, Inc. and James Shaffer10-Q001-323355/12/2014
10.29*Enhanze Technology License and Collaboration Agreement between Baxter Healthcare Corporation, Baxter Healthcare S.A. and Registrant, dated September 7, 20078-K001-323359/12/2007
10.30*License and Collaboration Agreement between F. Hoffmann-La Roche Ltd, Hoffmann-La Roche Inc. and Registrant dated December 5, 20068-K/A001-3233512/15/2006
10.31Standard Industrial Net Lease (11388 Sorrento Valley Road), effective as of July 26, 20078-K001-323357/31/2007
10.32Amended and Restated Lease (11388 Sorrento Valley Road), effective as of June 10, 20118-K001-323356/16/2011
10.33Lease (11436 Sorrento Valley Road), effective as of April 201310-K001-323352/28/2013





Incorporated by Reference
ExhibitFiled
NumberExhibit TitleHerewithFormFile No.Date Filed
10.34First modification to Lease (11436 Sorrento Valley Road)10-Q001-323355/8/2013
10.35Loan and Security Agreement and Disbursement Letter, dated December 28, 201210-K001-323352/28/2013
10.36First Amendment to Loan and Security Agreement and Disbursement Letter, dated February 5, 201310-K001-323352/28/2013
10.37Amended and Restated Loan and Security Agreement, dated December 27, 201310-K001-323352/28/2014
10.38Consent and First Amendment to Amended and Restated Loan and Security Agreement, dated June 10, 201410-Q001-323358/11/2014
10.39Second Amendment to Amended and Restated Loan and Security Agreement, dated January 23, 2015X
21.1Subsidiaries of RegistrantX
23.1Consent of Independent Registered Public Accounting FirmX
31.1Certification of Chief Executive Officer pursuant to Rule 13a-14(a) and 15d-14(a) of the Securities Exchange Act of 1934, as amendedX
31.2Certification of Chief Financial Officer pursuant to Rule 13a-14(a) and 15d-14(a) of the Securities Exchange Act of 1934, as amendedX
32Certification of Chief Executive Officer and Chief Financial Officer pursuant to 18 U.S.C. 1350, as adopted pursuant to Section 906 of the Sarbanes-Oxley Act of 2002X
101.INSXBRL Instance DocumentX
101.SCHXBRL Taxonomy Extension SchemaX
101.CALXBRL Taxonomy Extension Calculation LinkbaseX
101.DEFXBRL Taxonomy Extension Definition LinkbaseX
101.LABXBRL Taxonomy Extension Label LinkbaseX
101.PREXBRL Taxonomy Presentation LinkbaseX
_______________
*Confidential treatment has been granted for certain portions of this exhibit. These portions have been omitted from this agreement and have been filed separately with the Securities and Exchange Commission.
#Indicates management contract or compensatory plan or arrangement.