You should read the following discussion and analysis of our financial condition and results of operations together with our financial statements and related notes included in this Quarterly Report on Form 10-Q. The following discussion contains forward-looking statements that involve risks and uncertainties. Our actual results and the timing of certain events could differ materially from those anticipated in these forward-looking statements as a result of certain factors, including those discussed below and elsewhere in this Quarterly Report. This discussion should be read in conjunction with the accompanying unaudited consolidated financial statements and the audited consolidated financial statements and notes thereto included in our Annual Report on Form 10-K for the year ended December 31, 2020 filed with the Securities and Exchange Commission on March 25, 2021 (the "2020 Annual Report"). This discussion may contain forward-looking statements that involve risks and uncertainties. See "Forward-Looking Statements." You should review the disclosure under the heading "Risk Factors" in this Quarterly Report on Form 10-Q and the 2020 Annual Report for a discussion of important factors that could cause our actual results to differ materially from those anticipated in these forward-looking statements.

OVERVIEW

We are a biopharmaceutical company primarily engaged in the development of immune therapies and vaccines. Our allogeneic vaccine platform is based on secreted gp96 and designed to activate the immune system. This platform has broad applications in cancer and infectious disease. Our platform leverages a natural molecular warning system that presents antigens to the immune system. HS-110 (viagenpumatucel-L) is our first allogeneic ("off-the-shelf") cell line biologic product candidate in a series of proprietary immunotherapies designed to stimulate a patient's T cells to fight cancer. HS-130 is another allogeneic cell line engineered to express the extracellular domain of OX40 ligand as a fusion protein (OX40L-Fc). This biologic is a key costimulator of T cells, with potential to augment antigen-specific CD4+ and CD8+ T cell responses. We have initiated development of a new COVID-19 vaccine program under our Zolovax, Inc. subsidiary that utilizes our gp96 platform to chaperone SARS-CoV-2 antigens, eliciting an immune response against the spike glycoprotein. Our subsidiary Pelican Therapeutics, Inc. ("Pelican"), is developing PTX-35, a novel T cell co-stimulator agonist antibody targeting TNFRSF25.

These programs are designed to harness innate and/or antigen specific immune activation and tolerance mechanisms to reprogram immunity and provide a long-term, durable clinical effect. We have completed the following clinical milestones, completed patient enrollment for our HS-110 Phase 2 non-small cell lung cancer (NSCLC) clinical trial, completed enrollment and dosed fifteen patients in our HS-130 Phase 1 clinical trial and dosed fifteen patients in our PTX-35 Phase 1a clinical trial.

We are also providing pre-clinical, CMC development, and administrative support for these programs; while constantly focusing on protecting and expanding our intellectual property in areas of strategic interest. As we advance our clinical programs, we are in close contact with our CROs and clinical sites to monitor the impact of COVID-19 on our studies, current timelines, and costs.

In an effort to decrease our dependence upon third party manufacturers and enhance efficiency, we are designing and building a cGMP facility in San Antonio, Texas for the development of bioanalytics, process development and manufacturing activities. We also expect to offer fee-for-service contracting to external customers after completion of the build out.

We are also expanding our infectious disease/biothreat programs and have formed a biothreat advisory board to aid in advancing our biothreat initiatives. In addition, our Skunkworx Bio, Inc. subsidiary is utilizing advanced computational methods and bioinformatics to further enhance target precision and is developing proprietary "pocket biologics" to identify miniature proteins which bind to critical domains of druggable targets.



                                       23

  Table of Contents

Our Clinical Programs

We completed patient enrollment in our HS-110 Phase 2 non-small cell lung cancer (NSCLC) clinical trial and completed enrollment of patients in our HS-130 Phase 1 clinical trial that combines HS-110 with HS-130 to drive immune surveillance. Our PTX-35 safety and dose escalation Phase 1 clinical trial also continues to enroll patients. We are also providing pre-clinical, CMC development, and administrative support for these efforts; while constantly focusing on protecting and expanding our intellectual property in areas of strategic interest. We currently do not have any FDA approved products or sales and we have not generated any significant revenue since our inception nor revenue from product sales. We expect to continue to incur significant expenses and increased operating losses over the next several years. We anticipate that our expenses will increase substantially in the following areas:

? ongoing clinical trials of our product candidates;

? maintaining, expanding, and protecting our intellectual property portfolio;

? gaining regulatory approvals for our product pipeline;

? continued research and development efforts;

? the building and operation of our facility for the development of bioanalytics,

process development and manufacturing activities;

increased operational, financial, management information systems, and personnel

? - including personnel to support our product development and commercialization

efforts; and

? expenses incurred with operating as a public company.






About our gp96 Platform

Our gp96 platform is designed to activate and expand tumor antigen specific "killer" T cells to destroy a patient's cancer. By turning immunologically "COLD tumors HOT," we believe our platform can become an essential component of the immuno-oncology regimen to enhance the effectiveness and durability of checkpoint inhibitors and other cancer therapies, thereby improving outcomes for those patients less likely to benefit from checkpoint inhibitors alone.

We believe this is a highly differentiated approach as our platform delivers a broad range of tumor antigens that are previously unrecognized by the patient's immune system. Tumor antigens are processed and chaperoned by a powerful and naturally occurring immune adjuvant, gp96, to activate an anti-cancer immune response. Our cancer vaccine therapeutics are replication incompetent, "off-the-shelf", allogenic cell-based therapies that are locally administered into the skin. The treatment primes innate immune recognition and activates T cells to seek and destroy the cancer cells throughout the body. These gp96 agents can be administered with a variety of immuno-modulators to enhance a patient's immune response through T cell activation.

Unlike many other "patient specific" or autologous immunotherapy approaches, our drug biotherapeutics are fully allogeneic, "off-the-shelf" products. This provides a means to quickly administer the biotherapeutic without the need to extract and expand blood or tumor tissue from individual patients or create individualized treatment based on the patient's haplotype. Our gp96 product candidates are produced from allogeneic cell lines expressing tumor-specific proteins common among cancers. Because each patient receives the same treatment, we believe that our immunotherapy approach offers superior speed to initiation, logistics, manufacturing efficiencies, and importantly, cost benefits, compared to "personalized" precision medicine approaches.

Our gp96 platform also delivers antigen-driven T cell activation and specific co-stimulation in a single product. The vaccine delivers both the gp96 heat shock protein and a T cell co-stimulatory fusion protein (OX40L) as a single therapeutic, without the need for multiple, independent biologic products. This dual approach has several potential advantages including: (a) enhanced activation of antigen-specific CD8+ T cells; (b) boosting the number of antigen-specific CD8+ and CD4+ T cells compared to OX40L alone; (c) stimulation of T cell memory function to remain effective after treatment, even in the event of cancer remission; (d) demonstration of less toxicity, as the source of cancer associated antigens and co-stimulator are supplied at the same time locally in the draining lymph nodes, which drives targeted, cancer specific immunity towards the tumor rather than throughout the body; and (e) simplification of combination cancer immunotherapy versus systemic co-stimulation with conventional monoclonal antibodies (mAbs).





                                       24

  Table of Contents

An Allogenic Cell-Based Approach to Activating the Immune System

Our gp96 platform is an allogenic cell-based, T cell-stimulating platform that functions as an immune activator to stimulate and expand T effector cells. The key component of this innovative immunotherapy platform is the dual functionality of the gp96 heat shock protein.

As a molecular chaperone, gp96 is typically found within the cell's endoplasmic reticulum and facilitates the folding of newly synthesized proteins for functionalized tasks. When a cell abnormally dies through necrosis or infection, gp96 is naturally released into the surrounding microenvironment. This event becomes a Danger Associated Molecular Protein, or "DAMP", a molecular warning signal for localized innate activation of the immune system. In this context, gp96 serves as a potent adjuvant, or immune stimulator, via Toll-Like Receptor 4/2 (TLR4 and TLR2) signaling which serves to activate professional antigen presenting cells (APCs), such as dendritic cells that upregulate T cell costimulatory ligands, major histocompatibility (MHC) molecules and immune activating cytokines. It is among the most powerful adjuvants found in the body and uniquely shows exclusive specificity to CD8+ "killer" T cells through cross-presentation of the gp96-chaperoned tumor associated peptide antigens directly to MHC class I molecules for direct activation and expansion of CD8+ T cells. Thus, gp96 plays a central role driving mechanism of action for our T cell activating platform immuno-therapies; mimicking necrotic cell death and activating a powerful, tumor antigen-specific T cell immune response to attack the patient's cancer cells.

About COVID-19 Program

Besides its utility in oncology, our gp96 platform has been shown to activate the human immune system to combat infectious diseases. Our collaborators have laid a solid foundation by engineering different pathogenic antigens into our platform. Previous preclinical studies using our gp96 platform includes SIV/HIV, Malaria and Zika. We initiated a COVID-19 vaccine program in collaboration with the University of Miami in March 2020.

During the first quarter of 2020, preclinical studies using a cell-based COVID-19 vaccine expressing gp96-Ig, OX40L-Ig, and the SARS-CoV-2 spike glycoprotein (ZVX-60) were started under a sponsored research agreement with the University of Miami Miller School of Medicine. In July 2020, we announced proof-of-concept data demonstrating effective vaccine immunogenicity in relevant preclinical models, including expansion of human-HLA-restricted T cells against immunodominant epitopes of SARS-CoV-2 spike glycoprotein. Data showed robust T cell mediated immune response directed against the spike protein of SARS-CoV-2. In August 2020, we announced publication of positive preclinical COVID-19 vaccine results, which included supporting data that our gp96-based COVID-19 vaccine induces systemic and tissue-specific (e.g. lung) memory CD8+ T cells and tissue-resident memory CD8+ T cells. These results were published in the peer-reviewed journal Frontiers in Immunology in January 2021. In January 2021, we communicated that Waisman Biomanufacturing was contracted to complete process development and manufacturing for ZVX-60. We believe this COVID-19 vaccine is especially suited for immune compromised patients and is being developed for use as either a standalone, or in combination with other vaccines, to enhance prophylactic protection against COVID-19.

The strategy for this program includes providing prophylactic protection to elderly patients and those with underlying health conditions to drive cellular immune responses via CD8+ T cells, in addition to humoral B cell immune responses. We currently plan to seek grant funding for further development of this program however to date we have not received any grant funding.

About PTX-35

Pelican is focused on developing an agonist PTX-35 mAb, against a T cell costimulatory receptor, TNFRSF25. PTX-35 is designed to harness antigen specific immune activation and tolerance mechanisms to reprogram immunity and provide a long-term, durable clinical effect. TNFRSF25 agonism has been shown to provide highly selective and potent stimulation of antigen experienced 'memory' CD8+ cytotoxic T cells, which are the class of long-lived T cells capable of eliminating tumor cells in patients. Due to the preferential specificity of PTX-35 to antigen experienced CD8+ T cells, this agent represents a promising candidate as a T cell co-stimulator biotherapeutic in cancer patients.





                                       25

  Table of Contents

When combined in preclinical studies with Heat Biologics HS-110 and HS-130 immunotherapies or anti-PD-1 checkpoint inhibitor, PTX-35 has been shown to enhance antigen specific T cell activation to eliminate tumor cells. Pelican is also developing other immune biologics that target TNFRSF25 modulators for various immunotherapy approaches.

Our Bioanalytics, Process Development and Manufacturing Activities

To promote efficiency and reduce our reliance on third-party CDMO vendors, we are building in-house bioanalytic, process development, and manufacturing capabilities to support our current biotherapeutics and discovery pipeline. Manufacturing will also be offered to third parties as a fee-for-service model. We have entered into a lease for a 20,441 square foot facility in San Antonio, Texas where we plan to conduct such services and are currently in the process of building the manufacturing facility. Our expansion in Texas is part of a company-wide-growth strategy to enhance efficiency and decrease our dependence on third-party CDMO vendors as we advance our clinical trials and translate our research and development pipeline. We estimate that the investment to build out the facility with labs, equipment, and staff will be approximately $26 million.

Equipment purchases of $8.2 million through third quarter of 2021 and

approximately $6.4 million for lab equipment expected to be spent in the fourth quarter of 2021 and first quarter of 2022 are included in the $26 million. This excludes federal new market tax credits, historical designation federal and state tax credits, as well as city and county tax abatement incentives with the City of San Antonio and Bexar County, respectively. We intend to fund this initiative with current working capital. The potential value of tax credits and tax incentives to Scorpion are estimated at approximately $4.5 million based on the total cost of the build out, employees hired, real property, and other factors. Operations at the facility are projected to commence by second quarter of 2022, and we expect to fill production capacity by immediately transitioning our outsourced manufacturing and development to in-house. Fee-for-service contracting with be offered to external customers after completion of the build out. However, there can be no assurance that we will be successful in these new operations. As of November 10, 2021, $10.4 million has been spent on laboratory related manufacturing equipment for the San Antonio Facility.





Recent Developments

On August 19, 2021, we announced the formation of a biothreat advisory board comprised Andrew C. Weber, Former Assistant Secretary of Defense for Nuclear, Chemical & Biological Defense Programs; Dr. Gregory Koblentz, Associate Professor at George Mason University and leading expert on chemical and biological weapons; David Lasseter, Former Deputy Assistant Secretary of Defense for Countering Weapons of Mass Destruction, Former US Representative Jack Kingston, who also serves as the Secretariat of the Alliance for Biosecurity; and former two-term U.S. senator and Arkansas Attorney General Mark Pryor who currently serves with the law firm of Brownstein Hyatt Farber Schrek.

On October 5, 2021, Scorpion Biological Services, Inc. entered into a lease with Merchants Ice II, LLC (the "Landlord"), pursuant to which the Company will lease approximately 20,144 square feet of office and lab space located at 1305 E. Houston Street, San Antonio, Texas 78205 for general office, laboratory, research, analytical, and/or biomanufacturing purposes.

CRITICAL ACCOUNTING POLICIES AND SIGNIFICANT JUDGMENTS AND ESTIMATES

We believe that several accounting policies are important to understanding our historical and future performance. We refer to these policies as "critical" because these specific areas generally require us to make judgments and estimates about matters that are uncertain at the time we make the estimate, and different estimates-which also would have been reasonable-could have been used, which would have resulted in different financial results.

Our management's discussion and analysis of financial condition and results of operations is based on our consolidated financial statements, which have been prepared in accordance with 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, revenue and expenses and related disclosure of contingent assets and liabilities. On an ongoing basis, we evaluate our estimates based on historical experience and make various assumptions, which management believes to be reasonable under the circumstances, which form the basis for judgments about the carrying values of assets and liabilities that are not readily apparent from other sources. Actual results may differ from these estimates under different assumptions or conditions.



                                       26

Table of Contents

The notes to our consolidated financial statements contained herein and to our audited consolidated financial statements contained in our 2020 Annual Report contain a summary of our significant accounting policies. We consider the following accounting policies critical to the understanding of the results of our operations:



 ? Revenue;


 ? In-process R&D;


 ? Goodwill impairment;


 ? Income tax;


? Contingent consideration;

? Stock-based compensation;

? Research and development costs, including clinical and regulatory costs; and

? Recent accounting pronouncements.

RESULTS OF OPERATIONS

Comparison of the Three Months Ended September 30, 2021 and 2020

Revenues. For the three months ended September 30, 2021 we recognized $0.5 million of grant revenue for qualified expenditures under the CPRIT grant. For the three months ended September 30, 2020, we recognized $0.8 million of grant revenue for qualified expenditures under the CPRIT grant. The decrease in grant revenue in the current-year period primarily reflects the expected timing of completion of deliveries under the current phase of the contracts. As of September 30, 2021, we had a grant receivable balance of $0.9 million for CPRIT proceeds not yet received, but for which the costs had been incurred or the conditions of the award had been met. We continue our efforts to secure future non-dilutive grant funding to subsidize ongoing research and development costs.

Research and development expense. Research and development expenses increased approximately 37.5% to $4.4 million for the three months ended September 30, 2021 compared to $3.2 million for the three months ended September 30, 2020. The components of R&D expense are as follows, in millions:






                                                    For the Three Months Ended
                                                          September 30,
                                                     2021                2020
Programs
HS-110                                           $        0.2        $        0.1
HS-130                                                    0.3                 0.2
PTX-35                                                    1.1                 0.5
COVID-19                                                  0.7                 0.1
Other programs                                              -                 0.1
Unallocated research and development expenses             2.1                 2.2
                                                 $        4.4        $        3.2

HS-110 expense was $0.2 million, reflecting the current-period mix of

? development activities, primarily due to increased costs associated with our

Phase 2 trial.

? HS-130 expense was $0.3 million and included regulatory consulting and

investigator site payments for the ongoing Phase 1 clinical trial.

PTX-35 expense increased $0.6 million primarily due to dosing of patients,

? third-party regulatory consulting and investigator site payments for the

ongoing Phase 1 clinical trial.

? COVID-19 program was $0.7 million and primarily represents sponsored research

agreement costs and manufacturing costs.

? Other programs include preclinical costs associated with our Zika program, T

cell costimulatory programs, and laboratory supplies.




                                       27

  Table of Contents

? Unallocated research expenses primarily reflects personnel costs, including

stock-based compensation from stock awards.

General and administrative expense. General and administrative expense was $3.4 million and $6.6 million for the three months ended September 30, 2021 and 2020. The decrease was due to a decrease in stock-based compensation expense of $3.9 million due to our CEO being granted immediately vesting stock options in the third quarter of 2020 that did not recur in 2021, off-set by increased personnel costs of $0.2 million, increase in employee travel of $0.1 million, increase in D&O insurance of $0.1 million, and an increase of $0.3 million for consulting and other professional expenses to manage the business.

Change in fair value of contingent consideration. The change in fair value of contingent consideration was $0.3 million for the three months ended September 30, 2021, compared to $0.2 million for the three months ended September 30, 2020. The change in the 2021 period primarily reflects the increased timeline of the Phase 1a trial and the remeasurement of discounted cash flows for the passage of time and milestone achievement.

Total non-operating income (loss). Total non-operating income was $0.02 million for the three months ended September 30, 2021 which primarily consisted of $0.2 million of interest income, and ($0.2) million of unrealized losses on short-term investment balances. Total non-operating income was $0.2 million for the three months ended September 30, 2020 which primarily consisted of interest income on cash and short-term investments.

Net loss attributable to Heat Biologics, Inc. We had a net loss attributable to Heat Biologics, Inc. of $7.4 million, or ($0.30) per basic and diluted share for the three months ended September 30, 2021 compared to a net loss of $8.9 million, or ($0.43) per basic and diluted share for the three months ended September 30, 2020.

Comparison of the Nine Months Ended September 30, 2021 and 2020

Revenues. For the nine months ended September 30, 2021, we recognized $1.5 million of grant revenue for qualified expenditures under the CPRIT grant and NIH grant. For the nine months ended September 30, 2020, we recognized $2.3 million of grant revenue for qualified expenditures under the CPRIT grant. The decrease in grant revenue in the current-year period primarily reflects the expected timing of completion of deliveries under the current phase of the contracts. As of September 30, 2021, we had a grant receivable balance of $0.9 million for CPRIT proceeds not yet received but for which the costs had been incurred or the conditions of the award had been met. We continue our efforts to secure future non-dilutive grant funding to subsidize ongoing research and development costs.

Research and development expense. Research and development expenses increased approximately 36.4% to $12.0 million for the nine months ended September 30, 2021 compared to $8.8 million for the nine months ended September 30, 2020. The components of R&D expense are as follows, in millions:




                                                   For the Nine Months Ended
                                                         September 30,
                                                     2021               2020
Programs
HS-110                                          $          1.4       $       0.5
HS-130                                                     0.6               0.6
PTX 35                                                     2.2               1.6
COVID-19                                                   1.6               0.2
Other programs                                             0.2               0.2
Unallocated research and development expenses              6.0               5.7
                                                $         12.0       $       8.8

HS-110 expense increased $0.9 million, reflecting the current-period mix of

? development activities, primarily due to increased costs associated with the

transition of patients from active treatment into long-term follow-up, and

increased manufacturing costs.




                                       28

  Table of Contents

HS-130 expense remained steady at $0.6 million due to the completion of

? enrollment of patients, third-party regulatory consulting and investigator site

payments for the ongoing Phase 1 clinical trial.

? PTX-35 expense was $2.2 million primarily consisting of manufacturing

development and patient dosing.

? COVID-19 program was $1.6 million and primarily represents sponsored research

agreement costs and manufacturing costs.

? Other programs include preclinical costs associated with our Zika program, T

cell costimulatory programs, and laboratory supplies.

? Unallocated research expenses primarily reflects personnel costs, including

stock-based compensation from stock awards.

General and administrative expense. General and administrative expense was $11.0 million and $11.7 million for the nine months ended September 30, 2021 and 2020. The decrease was primarily due to a decrease in stock-based compensation expense of $1.7 million due to our CEO being granted immediately vesting stock options in the third quarter of 2020 that did not recur in 2021, an increase in D&O insurance premiums of $0.3 million, an increase in consulting expenses of $0.6 million, an increase in personnel costs of $0.2 million, an increase in franchise taxes of $0.2 million, and a decrease in public company expenses of $0.3 million.

Change in fair value of contingent consideration. The change in fair value of contingent consideration was $0.4 million for the nine months ended September 30, 2021, compared to $1.0 million in the nine months ended September 30, 2020. The change in the 2021 period primarily reflects the increased timeline of the Phase 1a trial and the remeasurement of discounted cash flows for the passage of time and milestone achievement.

Total non-operating income (loss). Other income was $0.1 million for the nine months ended September 30, 2021, compared to a loss of $(0.7) million for the nine months ended September 30, 2020. The change of $0.8 million primarily consists of a decrease in extinguishment expense and changes in fair value related to warrants of ($1.1) million, and a increase in interest income on cash and short-term investment balances of $0.2 million and a decrease of $0.5 million of unrealized losses on short-term investment balances.

Net loss attributable to Heat Biologics, Inc. We had a net loss attributable to Heat Biologics, Inc. of $21.5 million, or ($0.87) per basic and diluted share for the nine months ended September 30, 2021 compared to a net loss of $19.6 million, or ($1.42) per basic and diluted share for the nine months ended September 30, 2020.

LIQUIDITY AND CAPITAL RESOURCES

Sources of Liquidity

Since our inception in June 2008, we have incurred significant losses and we have financed our operations with net proceeds from the private placement of our preferred stock, common stock and debt. Since our initial public offering, we have primarily financed our operations with net proceeds from the public offering of our securities and to a lesser extent, the proceeds from the exercise of warrants. On January 21, 2020, we closed an underwritten public offering of shares of our common stock and warrants to purchase shares of our common stock pursuant to which we received net proceeds of approximately $6.4 million. For the year ended December 31, 2020 we received net proceeds of approximately $114.4 million from sales of our common stock in at-the-market offerings. For the nine months ended September 30, 2021 we received net proceeds of $25.6 million from the sale of 2,106,027 shares of our common stock in at-the-market offerings. We had no sales of our common stock in at-the-market offerings for the three months ended September 30, 2021. As of September 30, 2021, we had an accumulated deficit of $152.2 million. We had net losses of $26.4 million and $20.4 million for the years ended December 31, 2020 and 2019, respectively. We had net losses of $21.5 million and $19.6 million for the nine months ended September 30, 2021 and 2020, respectively.

We expect to incur significant expenses and continued losses from operations for the foreseeable future. We expect our expenses to increase in connection with our ongoing activities, particularly as we continue the research and development and advance our clinical trials of, and seek marketing approval for, our product candidates and as we add to our product candidate pipeline including expansion of our infectious disease/biothreat programs. Our expenses will also increase due



                                       29

  Table of Contents

to our recent new lease obligations for the manufacturing facility in San Antonio and related equipment expenses. In addition, if we obtain marketing approval for any of our product candidates, we expect to incur significant commercialization expenses related to product sales, marketing, manufacturing, and distribution. Although we currently have sufficient funds to complete our Phase 2 clinical trial, as currently planned, and expect that we will have sufficient funds to fund our operations into 2024, we will need to obtain substantial additional future funding in connection with our future planned clinical trials, the manufacturing facility that we are building out in San Antonio, Texas and any new programs or ventures we pursue. While we are currently funding vaccine development and preclinical studies, we do not expect to use significant corporate resources to advance our COVID-19 program. We are applying for several large grants to support clinical development of this program and are engaged in collaboration discussions, which we believe may provide attractive and non-dilutive pathways to help accelerate development of our COVID-19 program; however, to date we have not received any grant funding for such program and there can be no assurance that we will receive such grant funding or if received, the amount of such grant funding. Adequate additional financing may not be available to us on acceptable terms, or at all. If we are unable to raise capital when needed or on attractive terms, we would be forced to delay, reduce or eliminate our research and development programs or any future commercialization efforts. To meet our capital needs, we are considering multiple alternatives, including, but not limited to, additional equity financings, which include sales of our common stock under at-the-market offerings, if available, debt financings, partnerships, collaborations and other funding transactions. This is based on our current estimates, and we could use our available capital resources sooner than we currently expect. We will need to generate significant revenues to achieve profitability, and we may never do so. As of September 30, 2021, we had approximately $108.9 million in cash and cash equivalents and short-term investments.

Cash Flows

Operating activities. The use of cash during the nine months ended September 30, 2021 and 2020 resulted primarily from our net losses adjusted for non-cash charges and changes in components of working capital. Net cash used in operating activities during the nine months ended September 30, 2021 was $26.7 million compared to $15.6 million during the same period in 2020. The increase was primarily due to an increased net loss of $2.0 million, a decrease in stock-based compensation of $1.7 million, a decrease in the change in fair value of common stock warrants of $1.0 million, a decrease in the change in fair value of contingent consideration of $0.6 million, a decrease in deferred revenue of $1.6 million, an increase in other assets of $8.2 million and an increase in accounts payable and accrued expenses of $1.2 million. The increase in other assets relates to reimbursements made to Merchants Ice, LLC for equipment in our new San Antonio lease that will be classified as a right of use asset upon lease commencement.

Investing activities. Net cash used in investing activities was $1.9 million during the nine months ended September 30, 2021 compared to $87.1 million during the same period in 2020. The decrease is from the change in net purchases of short-term investments of $86.3 million from 2021 to 2020 and the increase in purchases of property and equipment of $1.1 million.

Financing activities. Net cash provided by financing activities was $25.6 million during the nine months ended September 30, 2021 compared to $118.4 million during the nine months ended September 30, 2020. The decrease of $92.8 million was primarily due to a $90.0 million net decrease of sales of our common stock through an at-the-market Common Stock Sales Agreement with B. Riley FBR, Inc. and Cantor Fitzgerald & Co., net of the decrease in related stock issuance costs of $2.4 million, partially offset by a public offering of shares that occurred in 2020 of $6.6 million.

Current and Future Financing Needs

We have incurred an accumulated deficit of $152.2 million through September 30, 2021. We have incurred negative cash flows from operations since we started our business. We have spent, and expect to continue to spend, substantial amounts in connection with implementing our business strategy, including our planned product development efforts, our clinical trials, and our research and discovery efforts.

In order to promote efficiency and reduce our reliance on third-party vendors, we plan to enhance our in-house development of bioanalytic, process development and manufacturing capabilities and offer such services to third parties for fees. We have entered into a lease for a 20,441 square foot facility in San Antonio, TX to conduct such services and



                                       30

Table of Contents

are currently building the facility. Our proposed expansion in Texas is part of a company-wide-growth strategy to enhance efficiency and decrease our dependence on third-party vendors as we advance our clinical trials and general research and development. We estimate that the investment to build out the facility with labs, equipment, and staff will be approximately $26 million, without taking into account federal new market tax credits based on the location in San Antonio, federal and state historical tax credits based on the historical designation of the facility, as well as city and county tax abatement incentives with the City of San Antonio and Bexar County, respectively. Equipment purchases of $8.2 million through third quarter 2021 and approximately $6.4 million for lab equipment expected to be spent in the fourth quarter of 2021 and first quarter of 2022 are included in the $26 million. We intend to fund this initiative with current working capital. The potential value of tax credits and tax incentives to Scorpion are estimated to be up to approximately $4.5 million based on the total cost of the build out, employees hired, real property, and other factors. Operations at the facility are projected to commence by second quarter of 2022, and we expect to fill production capacity by transitioning our outsourced manufacturing and development to in-house immediately and followed by contracting with external customers. However, there can be no assurance that we will be successful in these new operations. As of November 10, 2021 we have spent $10.4 million on laboratory related manufacturing equipment for the San Antonio facility.

We intend to meet our financing needs through multiple alternatives, including, but not limited to, cash on hand, additional equity financings, debt financings and/or funding from partnerships or collaborations and potential revenue, if any, from our planned development and manufacturing facility.

However, the actual amount of funds we will need to operate is subject to many factors, some of which are beyond our control. These factors include the following:

? the progress of our research activities;

? the number and scope of our research programs;

? the progress of our preclinical and clinical development activities;

? the progress of the development efforts of parties with whom we have entered

into research and development agreements;

? our expansion plans and cash needs of any new projects such as our planned

development and manufacturing facility described above;

the cost we incur to build the facility for the development of bioanalytics,

? process development and manufacturing activities, the cost of the equipment

required for such facility and the revenue derived from such facility

? our ability to maintain current research and development licensing arrangements

and to establish new research and development and licensing arrangements;

? our ability to achieve our milestones under licensing arrangements;

? the costs involved in prosecuting and enforcing patent claims and other

intellectual property rights;

? the costs and timing of regulatory approvals;

? the receipt of grant funding if any; and

? clinical laboratory development and testing.

We have based our estimate on assumptions that may prove to be wrong. We may need to obtain additional funds sooner or in greater amounts than we currently anticipate. Potential sources of financing include strategic relationships, public or private sales of our equity or debt and other sources. We may seek to access the public or private equity markets when conditions are favorable due to our long-term capital requirements. We do not have any committed sources of financing at this time, and it is uncertain whether additional funding will be available when we need it on terms that will be acceptable to us, or at all. If we raise funds by selling additional shares of common stock, such as through the Amended and Restated Common Stock Sales Agreement with B. Riley FBR, Inc. and Cantor Fitzgerald & Co., or other securities convertible into common stock, the ownership interest of our existing stockholders will be diluted. If we are not able to obtain financing when needed, we may be unable to carry out our business plan. As a result, we may have to significantly limit our operations and our business, financial condition and results of operations would be materially harmed. While we are experiencing limited financial impacts at this time, given the global economic slowdown, the overall disruption of global healthcare systems and the other risks and uncertainties associated with the pandemic, our business, financial condition, results of operations and growth prospects could be materially adversely affected.



                                       31

Table of Contents

OFF-BALANCE SHEET ARRANGEMENTS

We did not have during the periods presented, and we do not currently have, any off-balance sheet arrangements, as defined under Securities and Exchange Commission rules.

© Edgar Online, source Glimpses