The following discussions should be read in conjunction with our financial statements and related notes thereto included in this Annual Report. The following discussion contains forward-looking statements made pursuant to the safe harbor provisions of Section 27A of the Securities Act and Section 21E of the Securities Exchange Act of 1934 and the Private Securities Litigation Reform Act of 1995. These statements are based on our beliefs and expectations about future outcomes and are subject to risks and uncertainties that could cause actual results to differ materially from anticipated results. Factors that could cause or contribute to such differences include those described under "Part I, Item 1A - Risk Factors" appearing in this Annual Report and factors described in other cautionary statements, cautionary language and risk factors set forth in other documents that we file with the Securities and Exchange Commission. We undertake no obligation to publicly update forward-looking statements, whether as a result of new information, future events or otherwise.





Overview


Celsion Corporation ("Celsion" and the "Company") is a fully integrated, clinical stage biotechnology company focused on advancing a portfolio of innovative treatments including DNA-based immunotherapies, next generation vaccines and directed chemotherapies through clinical trials and eventual commercialization. The Company's product pipeline includes GEN-1, a DNA-based immunotherapy for the localized treatment of ovarian cancer and ThermoDox®, a proprietary heat-activated liposomal encapsulation of doxorubicin, currently under investigator-sponsored development for several cancer indications. Celsion has two feasibility stage platform technologies for the development of novel nucleic acid-based immunotherapies and next generation vaccines and other anti-cancer DNA or RNA therapies. Both are novel synthetic, non-viral vectors with demonstrated capability in nucleic acid cellular transfection.





IMMUNO-ONCOLOGY Program


On June 20, 2014, the Company completed the acquisition of substantially all of the assets of EGEN, a private company located in Huntsville, Alabama. Pursuant to the Asset Purchase Agreement, CLSN Laboratories acquired all of EGEN's right, title and interest in substantially all of the assets of EGEN, including cash and cash equivalents, patents, trademarks and other intellectual property rights, clinical data, certain contracts, licenses and permits, equipment, furniture, office equipment, furnishings, supplies and other tangible personal property. A key asset acquired from EGEN was the TheraPlas technology platform. The first drug candidate developed from this technology platform is GEN-1.





THERAPLAS Technology Platform


TheraPlas is a technology platform for the delivery of DNA and mRNA therapeutics via synthetic non-viral carriers and is capable of providing cell transfection for double-stranded DNA plasmids and large therapeutic RNA segments such as mRNA. There are two components of the TheraPlas system, a plasmid DNA or mRNA payload encoding a therapeutic protein, and a delivery system. The delivery system is designed to protect the DNA/mRNA from degradation and promote trafficking into cells and through intracellular compartments. We designed the delivery system of TheraPlas by chemically modifying the low molecular weight polymer to improve its gene transfer activity without increasing toxicity. We believe that TheraPlas may be a viable alternative to current approaches to gene delivery due to several distinguishing characteristics, including enhanced molecular versatility that allows for complex modifications to potentially improve activity and safety.

The design of the TheraPlas delivery system is based on molecular functionalization of polyethyleneimine (PEI), a cationic delivery polymer with a distinct ability to escape from the endosomes due to heavy protonation. The transfection activity and toxicity of PEI is tightly coupled to its molecular weight; therefore the clinical application of PEI is limited. We have used molecular functionalization strategies to improve the activity of low molecular weight PEIs without augmenting their cytotoxicity. In one instance, chemical conjugation of a low molecular weight branched BPEI1800 with cholesterol and polyethylene glycol (PEG) to form PEG-PEI-Cholesterol (PPC) dramatically improved the transfection activity of BPEI1800 following in vivo delivery. Together, the cholesterol and PEG modifications produced approximately 20-fold enhancement in transfection activity. Biodistribution studies following intraperitoneal or subcutaneous administration of DNA/PPC nanocomplexes showed DNA delivery localized primarily at the injection site with only small amount escaping into the systemic circulation. PPC is the delivery component of our lead TheraPlas product, GEN-1, which is in clinical development for the treatment of ovarian cancer. The PPC manufacturing process has been scaled up from bench scale (1-2 g) to 0.6Kg, and several current Good Manufacturing Practice ("cGMP") lots have been produced with reproducible quality.





57






We believe that TheraPlas has emerged as a viable alternative to current approaches due to several distinguishing characteristics such as strong molecular versatility that may allow for complex modifications to potentially improve activity and safety with little difficulty. The biocompatibility of these polymers reduces the risk of adverse immune response, thus allowing for repeated administration. Compared to naked DNA or cationic lipids, TheraPlas is generally safer, more efficient, and cost effective. We believe that these advantages place Celsion in a strong position to capitalize on this technology platform.

Ovarian Cancer Overview

Ovarian cancer is the most lethal of gynecological malignancies among women with an overall five-year survival rate of 45%. This poor outcome is due in part to the lack of effective prevention and early detection strategies. There were approximately 22,000 new cases of ovarian cancer in the U.S. in 2014 with an estimated 14,000 deaths. Mortality rates for ovarian cancer declined very little in the last forty years due to the unavailability of detection tests and improved treatments. Most women with ovarian cancer are not diagnosed until Stages III or IV, when the disease has spread outside the pelvis to the abdomen and areas beyond causing swelling and pain, where the five-year survival rates are 25 - 41 percent and 11 percent, respectively. First-line chemotherapy regimens are typically platinum-based combination therapies. Although this first line of treatment has an approximate 80 percent response rate, 55 to 75 percent of women will develop recurrent ovarian cancer within two years and ultimately will not respond to platinum therapy. Patients whose cancer recurs or progresses after initially responding to surgery and first-line chemotherapy have been divided into one of the two groups based on the time from completion of platinum therapy to disease recurrence or progression. This time period is referred to as platinum-free interval. The platinum-sensitive group has a platinum-free interval of longer than six months. This group generally responds to additional treatment with platinum-based therapies. The platinum-resistant group has a platinum-free interval of shorter than six months and is resistant to additional platinum-based treatments. Pegylated liposomal doxorubicin, topotecan, and Avastin are the only approved second-line therapies for platinum-resistant ovarian cancer. The overall response rate for these therapies is 10 to 20 percent with median overall survival ("OS") of eleven to twelve months. Immunotherapy is an attractive novel approach for the treatment of ovarian cancer particularly since ovarian cancers are considered immunogenic tumors. IL-12 is one of the most active cytokines for the induction of potent anti-cancer immunity acting through the induction of T-lymphocyte and natural killer cell proliferation. The precedence for a therapeutic role of IL-12 in ovarian cancer is based on epidemiologic and preclinical data.





GEN-1 Immunotherapy


GEN-1 is a DNA-based immunotherapeutic product candidate for the localized treatment of ovarian cancer by intraperitoneally administering an Interleukin-12 ("IL-12") plasmid formulated with our proprietary TheraPlas delivery system. In this DNA-based approach, the immunotherapy is combined with a standard chemotherapy drug, which can potentially achieve better clinical outcomes than with chemotherapy alone. We believe that increases in IL-12 concentrations at tumor sites for several days after a single administration could create a potent immune environment against tumor activity and that a direct killing of the tumor with concomitant use of cytotoxic chemotherapy could result in a more robust and durable antitumor response than chemotherapy alone. We believe the rationale for local therapy with GEN-1 is based on the following:





  ? Loco-regional production of the potent cytokine IL-12 avoids toxicities and
    poor pharmacokinetics associated with systemic delivery of recombinant IL-12;

  ? Persistent local delivery of IL-12 lasts up to one week and dosing can be
    repeated; and

  ? Local therapy is ideal for long-term maintenance therapy.




58

OVATION I Study. In February 2015, we announced that the U.S. Food and Drug Administration ("FDA") accepted, without objection, the Phase I dose-escalation clinical trial of GEN-1 in combination with the standard of care in neoadjuvant ovarian cancer (the "OVATION I Study"). On September 30, 2015, we announced enrollment of the first patient in the OVATION I Study. The OVATION I Study was designed to:





  (i)   identify a safe, tolerable and therapeutically active dose of GEN-1 by
        recruiting and maximizing an immune response;

  (ii)  enroll three to six patients per dose level and evaluate safety and
        efficacy; and

  (iii) attempt to define an optimal dose for a follow-on Phase I/II study.



In addition, the OVATION I Study established a unique opportunity to assess how cytokine-based compounds such as GEN-1, directly affect ovarian cancer cells and the tumor microenvironment in newly diagnosed ovarian cancer patients. The study was designed to characterize the nature of the immune response triggered by GEN-1 at various levels of the patients' immune system, including:





  ? Infiltration of cancer fighting T-cell lymphocytes into primary tumor and
    tumor microenvironment including peritoneal cavity, which is the primary site
    of metastasis of ovarian cancer;

  ? Changes in local and systemic levels of immuno-stimulatory and
    immunosuppressive cytokines associated with tumor suppression and growth,
    respectively; and

  ? Expression profile of a comprehensive panel of immune related genes in
    pre-treatment and GEN-1-treated tumor tissue.



We initiated the OVATION I Study at four clinical sites at the University of Alabama at Birmingham, Oklahoma University Medical Center, Washington University in St. Louis, and the Medical College of Wisconsin. During 2016 and 2017, we announced data from the first fourteen patients in the OVATION I Study. On October 3, 2017, we announced final translational research and clinical data from the OVATION I Study.

Key translational research findings from all evaluable patients are consistent with the earlier reports from partial analysis of the data and are summarized below:





  ? The intraperitoneal treatment of GEN-1 in conjunction with NACT resulted in
    dose dependent increases in IL-12 and Interferon-gamma (IFN-?) levels that
    were predominantly in the peritoneal fluid compartment with little to no
    changes observed in the patients' systemic circulation. These and other
    post-treatment changes including decreases in VEGF levels in peritoneal fluid
    are consistent with an IL-12 based immune mechanism;

  ? Consistent with the previous partial reports, the effects observed in the IHC
    analysis were pronounced decreases in the density of immunosuppressive T-cell
    signals (Foxp3, PD-1, PDL-1, IDO-1) and increases in CD8+ cells in the tumor
    microenvironment;

  ? The ratio of CD8+ cells to immunosuppressive cells was increased in
    approximately 75% of patients suggesting an overall shift in the tumor
    microenvironment from immunosuppressive to pro-immune stimulatory following
    treatment with GEN-1. An increase in CD8+ to immunosuppressive T-cell
    populations is a leading indicator and believed to be a good predictor of
    improved OS; and

  ? Analysis of peritoneal fluid by cell sorting, not reported before, shows a
    treatment-related decrease in the percentage of immunosuppressive T-cell
    (Foxp3+), which is consistent with the reduction of Foxp3+ T-cells in the
    primary tumor tissue, and a shift in tumor naïve CD8+ cell population to more
    efficient tumor killing memory effector CD8+ cells.




59

The Company also reported positive clinical data from the first fourteen patients who completed treatment in the OVATION I Study. GEN-1 plus standard chemotherapy produced no dose limiting toxicities and positive dose dependent efficacy signals which correlate well with positive surgical outcomes as summarized below:





  ? Of the fourteen patients treated in the entire study, two patients
    demonstrated a complete response, ten patients demonstrated a partial response
    and two patients demonstrated stable disease, as measured by RECIST criteria.
    This translates to a 100% disease control rate and an 86% objective response
    rate ("ORR"). Of the five patients treated in the highest dose cohort, there
    was a 100% ORR with one complete response and four partial responses;

  ? Fourteen patients had successful resections of their tumors, with nine
    patients (64%) having a complete tumor resection ("R0"), which indicates a
    microscopically margin-negative resection in which no gross or microscopic
    tumor remains in the tumor bed. Seven out of eight (88%) patients in the
    highest two dose cohorts experienced a R0 surgical resection. All five
    patients treated at the highest dose cohort experienced a R0 surgical
    resection; and

  ? All patients experienced a clinically significant decrease in their CA-125
    protein levels as of their most recent study visit. CA-125 is used to monitor
    certain cancers during and after treatment. CA-125 is present in greater
    concentrations in ovarian cancer cells than in other cells.



On March 2, 2019, the Company announced final progression free survival ("PFS") results from the OVATION I Study. Median PFS in patients treated per protocol (n=14) was 21 months and was 17.1 months for the intent-to-treat ("ITT") population (n=18) for all dose cohorts, including three patients who dropped out of the study after 13 days or less, and two patients who did not receive full NAC and GEN-1 cycles. Under the current standard of care, in women with Stage III/IV ovarian cancer undergoing NAC, their disease progresses within about 12 months on average. The results from the OVATION I Study support continued evaluation of GEN-1 based on promising tumor response, as reported in the PFS data, and the ability for surgeons to completely remove visible tumor at interval debulking surgery. GEN-1 was well tolerated, and no dose-limiting toxicities were detected. Intraperitoneal administration of GEN-1 was feasible with broad patient acceptance.

OVATION 2 Study. The Company held an Advisory Board Meeting on September 27, 2017 with the clinical investigators and scientific experts including those from Roswell Park Cancer Institute, Vanderbilt University Medical School, and M.D. Anderson Cancer Center to review and finalize clinical, translational research and safety data from the OVATION I Study in order to determine the next steps forward for our GEN-1 immunotherapy program.

On November 13, 2017, the Company filed its Phase I/II clinical trial protocol with the FDA for GEN-1 for the localized treatment of ovarian cancer. The protocol is designed with a single dose escalation phase to 100 mg/m² to identify a safe and tolerable dose of GEN-1 while maximizing an immune response. The Phase I portion of the study will be followed by a continuation at the selected dose in approximately 110 patients randomized Phase II study.

In the OVATION 2 Study, patients in the GEN-1 treatment arm will receive GEN-1 plus chemotherapy pre- and post-interval debulking surgery ("IDS"). The OVATION 2 Study will include up to 110 patients with Stage III/IV ovarian cancer, with 12 to 15 patients in the Phase I portion and up to 95 patients in Phase II. The study is powered to show a 33% improvement in the primary endpoint, PFS, when comparing GEN-1 with neoadjuvant + adjuvant chemotherapy versus neoadjuvant + adjuvant chemotherapy alone. The PFS primary analysis will be conducted after at least 80 events have been observed or after all patients have been followed for at least 16 months, whichever is later.

In March 2020, the Company announced encouraging initial clinical data from the first 15 patients enrolled in the Phase I portion of the OVATION 2 Study for patients newly diagnosed with Stage III and IV ovarian cancer. The OVATION 2 Study combines GEN-1, the Company's IL-12 gene-mediated immunotherapy, with standard-of-care neoadjuvant chemotherapy (NACT). Following NACT, patients undergo interval debulking surgery (IDS), followed by three additional cycles of chemotherapy.





60






GEN-1 plus standard NACT produced positive dose-dependent efficacy results, with no dose-limiting toxicities, which correlates well with successful surgical outcomes as summarized below:





  ? Of the 15 patients treated in the Phase I portion of the OVATION 2 Study, nine
    patients were treated with GEN-1 at a dose of 100 mg/m² plus NACT and six
    patients were treated with NACT only. All 15 patients had successful
    resections of their tumors, with eight out of nine patients (88%) in the GEN-1
    treatment arm having an R0 resection, which indicates a microscopically
    margin-negative complete resection in which no gross or microscopic tumor
    remains in the tumor bed. Only three out of six patients (50%) in the NACT
    only treatment arm had a R0 resection.

  ? When combining these results with the surgical resection rates observed in the
    Company's prior Phase Ib dose-escalation trial (the OVATION 1 Study), a
    population of patients with inclusion criteria identical to the OVATION 2
    Study, the data reflect the strong dose-dependent efficacy of adding GEN-1 to
    the current standard of care NACT:




                                                              % of
                                                            Patients
                                                       with R0 Resections
         0, 36, 47 mg/m² of GEN-1 plus NACT     n=12                    42 %
         61, 79, 100 mg/m² of GEN-1 plus NACT   n=17                    82 %




  ? The ORR as measured by Response Evaluation Criteria in Solid Tumors (RECIST)
    criteria for the 0, 36, 47 mg/m² dose GEN-1 patients were comparable, as
    expected, to the higher (61, 79, 100 mg/m²) dose GEN-1 patients, with both
    groups demonstrating an approximate 80% ORR.



On March 23, 2020, the Company announced that the European Medicines Agency (the "EMA") Committee for Orphan Medicinal Products ("COMP") has recommended that GEN-1 be designated as an orphan medicinal product for the treatment of ovarian cancer. GEN-1 is an IL-12 DNA plasmid vector encased in a non-viral nanoparticle delivery system, which enables cell transfection followed by persistent, local secretion of the IL-12 protein. GEN-1 previously received orphan designation from the FDA.

On March 26, 2020, the Company announced with Medidata, a Dassault Systèmes company, that examining matched patient data provided by Medidata in a synthetic control arm ("SCA") with results from the Company's completed Phase Ib dose-escalating OVATION I Study showed positive results in progression-free survival ("PFS"). The hazard ratio ("HR") was 0.53 in the ITT group, showing strong signals of efficacy. Celsion believes these data may warrant consideration of strategies to accelerate the clinical development program for GEN-1 in newly diagnosed, advanced ovarian cancer patients by the FDA. In its March 2019 discussion with Celsion, the FDA noted that preliminary findings from the Phase Ib OVATION I Study were exciting but lacked a control group to evaluate GEN-1's independent impact on impressive tumor response, surgical results and PFS. The FDA encouraged the Company to continue its GEN-1 development program and consult with FDA with new findings that may have a bearing on designations such as Fast Track and Breakthrough Therapy.

SCAs have the potential to revolutionize clinical trials in certain oncology indications and some other diseases where a randomized control is not ethical or practical. SCAs are formed by carefully selecting control patients from historical clinical trials to match the demographic and disease characteristics of the patients treated with the new investigational product. SCAs have been shown to mimic the results of traditional randomized controls so that the treatment effects of an investigational product can be visible by comparison to the SCA. SCAs can help advance the scientific validity of single arm trials, and in certain indications, reduce time and cost, and expose fewer patients to placebos or existing standard-of-care treatments that might not be effective for them.

On July 27, 2020, the Company announced the randomization of the first two patients in the Phase II portion of the OVATION 2 Study with GEN-1 in advanced ovarian cancer. The Company anticipates completing enrollment of up to 110 patients in the second half of 2021. Because this is an open-label study, the Company intends to provide clinical updates throughout the course of treatment including response rates and surgical resection scores.





61






On February 22, 2021, the Company announced that it has received Fast Track designation from the FDA for GEN-1, its DNA-mediated IL-12 immunotherapy currently in Phase II development for the treatment of advanced ovarian cancer.

On February 25, 2021, the Company provided an update on the OVATION 2 Study. The Company reported that approximately one-third, or 34 patients, of the anticipated 110 patients had been enrolled into the OVATION 2 Study, of which 20 are in the treatment arm and 14 are in the control. Currently, 27 patients have had their interval debulking surgery with the following results:





  ? 12 of 15, or 80%, of patients treated with GEN-1 had a R0 resection, which
    indicates a microscopically margin-negative complete resection in which no
    gross or microscopic tumor remains in the tumor bed.
  ? 7 of 12 patients, or 58%, of patients in the control arm had an R0 resection.
  ? This interim data represents a 38% improvement in R0 resection rates for GEN-1
    patients compared with control arm patients and is consistent with the
    reported improvement in resection scores noted in the encouraging Phase I
    OVATION I Study, the manuscript of which has been submitted for peer review
    publication.



The Company further reported that 22 clinical sites in the U.S. and Canada have been initiated, with three more sites expected to be added by the end of the first quarter. Clinical investigators met in early February 2021 in a virtual meeting and expressed excitement about the potential for GEN-1 to treat advanced ovarian cancer and, despite the challenges and earlier delays posed by the COVID-19 pandemic, they remain committed to completing enrollment in the study during the second half of 2021.

PLACCINE DNA VACCINE TECHNOLOGY PLATFORM

In January 2021, the Company announced the filing of a provisional U.S. patent application for a novel DNA-based, investigational vaccine for preventing or treating infections from a broad range of infectious agents including the coronavirus disease using its PLACCINE DNA vaccine technology platform ("PLACCINE"). The provisional patent covers a family of novel composition of multi-cistronic vectors and polymeric nanoparticles that comprise the PLACCINE DNA vaccine platform technology for preventing or treating infectious agents that have the potential for global pandemics, including the SARS-CoV-2 virus and its variations, using the Company's platform technology.

Celsion's PLACCINE DNA vaccine technology platform is characterized by a single multi-cistronic DNA plasmid vector expressing multiple pathogen antigens along with a potent immune modifier and delivered with a synthetic delivery system. It is easily adaptable to creating vaccines for a multitude of pathogens, including emerging pathogens leading to pandemics as well as infectious diseases that have yet to be effectively addressed with current vaccine technologies. This flexible vaccine platform is well supported by an already established supply chain to produce any plasmid vector and its assembly into a respective vaccine formulation.

PLACCINE is an extension of the Company's synthetic, non-viral TheraPlas delivery technology currently in a Phase II trial for the treatment of late-stage ovarian cancer with GEN-1. Celsion's proprietary multifunctional DNA vaccine technology concept is built on the flexible PLACCINE technology platform that is amenable to rapidly responding to the SARS-CoV-2 virus, as well as possible future mutations of SARS-CoV-2, other future pandemics, emerging bioterrorism threats, and novel infectious diseases. Celsion's extensive experience with TheraPlas suggests that the PLACCINE-based nanoparticles are stable at storage temperatures of 4oC to 25oC, making vaccines developed on this platform easily suitable for broad world-wide distribution.

Celsion's vaccine approach is designed to optimize the quality of the immune response dictating the efficiency of pathogen clearance and patient recovery. Celsion has taken a multivalent approach in an effort to generate an even more robust immune response that not only results in a strong neutralizing antibody response, but also a more robust and durable T-cell response. Delivered with Celsion's synthetic polymeric system, the proprietary DNA plasmid is protected from degradation and its cellular uptake is facilitated.





62







COVID-19 Vaccine Overview


Emerging data from the recent literature indicates that the quality of the immune response as opposed to its absolute magnitude is what dictates SARS-CoV-2 viral clearance and recovery and that an ineffective or non-neutralizing enhanced antibody response might actually exacerbate disease. The first-generation COVID-19 vaccines were developed for rapid production and deployment and were not optimized for generating cellular responses that result in effective viral clearance. Though early data has indicated some of these vaccines to be over 95% effective, these first-generation vaccines were primarily designed to generate a strong antibody response and, while they have been shown to provide prophylactic protection against disease, the durability of this protection is currently unclear. The vast majority of these vaccines have been specifically developed to target the SARS-CoV-2 Spike (S) protein (antigen), though it is known that restricting a vaccine to a sole viral antigen creates selection pressure that can serve to facilitate the emergence of viral resistance. Indeed, even prior to full vaccine rollout, it has been observed that the S protein is a locus for rapid evolutionary and functional change as evidenced by the D614G, Y453F, 501Y.V2, and VUI-202012/01 mutations/deletions. This propensity for mutation of the S protein leads to future risk of efficacy reduction over time as these mutations accumulate.

Our Next Generation Vaccine Initiative

Celsion's next generation vaccine initiative stands at the confluence of immunotherapy and immunogenicity and envisions delivery, on a single plasmid, multiple SARS-CoV-2 antigens in conjunction with a potent immune modifier, interleukin-12 (IL-12), which directs a TH-1 immune response, stimulates T-cell immunity, and also promises the promotion of humoral immunity (antibody response). While most COVID-19 vaccines in late-stage clinical development are monovalent (S protein antigen only), Celsion has taken this multivalent approach in an effort to generate an even more robust immune response that not only results in a strong neutralizing antibody response, but also a more robust and durable T-cell response.

Celsion's vaccine candidate approach comprises a single plasmid vector containing the DNA sequence encoding the cytokine IL-12 and multiple SARS-CoV-2 antigens, including S antigen in combination with the membrane (M) or nucleocapsid (N) antigen. Delivery will be evaluated intramuscularly, intradermally, or subcutaneously with a non-viral synthetic DNA delivery carrier that facilitates vector delivery into the cells of the injected tissue and has potential immune adjuvant properties. Unique designs and formulations of Celsion vaccine candidates may offer several potential key advantages.





  ? While the antibodies against S antigen would prevent virus entry into cells,
    the M and N antibodies could help virus clearance through antibody-mediated
    opsonization and phagocytosis. The presentation of multiple antigens on the
    cell surface of vaccine-injected tissue produces a broad variety of killer
    T-cells which could potentially produce more efficient viral clearance than a
    single antigen vaccine.

  ? Since IL-12 is an essential regulator of the differentiation, proliferation,
    and maintenance of T helper 1 (TH-1) cells that generate killer T-cells and
    memory T-cells against virally infected cells, its simultaneous expression
    could boost the viral clearance by the vaccine and improve the immune system's
    memory against any future exposure of the same virus.

  ? Finally, the synthetic polymeric DNA carrier is an important component of the
    vaccine composition as it has the potential to facilitate the vaccine
    immunogenicity by improving vector delivery and, due to potential adjuvant
    properties, attract professional immune cells to the site of vaccine delivery.



Future vaccine technology will need to address viral mutations and the challenges of efficient manufacturing, distribution, and storage. We believe an adaptation of our TheraPlas technology, PLACCINE, has the potential to meet these challenges. Our approach is described in our provisional patent filing and is summarized as a DNA vaccine technology platform characterized by a single plasmid DNA with multiple coding regions. The plasmid vector is designed to express multiple pathogen antigens along with a potent immune modifier. It is delivered via a synthetic delivery system and has the potential to be easily modified to create vaccines against a multitude of infectious diseases, addressing:





  ? Viral Mutations: PLACCINE may offer broad-spectrum and mutational resistance
    (variants) by targeting multiple antigens on a single plasmid vector.




63







  ? Enhanced Efficacy: The potent immune modifier IL-12 may improve humoral and
    cellular responses to viral antigens and can be incorporated in the plasmid.

  ? Durable Efficacy: PLACCINE delivers a DNA plasmid-based antigen that can
    result in durable antigen exposure and a robust vaccine response to viral
    antigens.

  ? Storage & Distribution: PLACCINE allows for stability that is compatible with
    manageable vaccine storage and distribution.

  ? Simple Dosing & Administration: PLACCINE is a synthetic delivery system that
    should require a simple injection that does not require viruses or special
    equipment to deliver its payload.



We are conducting preliminary research associated with our recently announced proprietary DNA vaccine platform provisional patent filing. At the same time, we are redoubling our efforts and R&D resources in our immuno-oncology and next generation vaccine program.

THERMODOX® - DIRECTED CHEMOTHERAPY

Liposomes are manufactured submicroscopic vesicles consisting of a discrete aqueous central compartment surrounded by a membrane bilayer composed of naturally occurring lipids. Conventional liposomes have been designed and manufactured to carry drugs and increase residence time, thus allowing the drugs to remain in the bloodstream for extended periods of time before they are removed from the body. However, the current existing liposomal formulations of cancer drugs and liposomal cancer drugs under development do not provide for the immediate release of the drug and the direct targeting of organ specific tumors, two important characteristics that are required for improving the efficacy of cancer drugs such as doxorubicin. A team of research scientists at Duke University developed a heat-sensitive liposome that rapidly changes its structure when heated to a threshold minimum temperature of 39.5º to 42º Celsius. Heating creates channels in the liposome bilayer that allow an encapsulated drug to rapidly disperse into the surrounding tissue. This novel, heat-activated liposomal technology is differentiated from other liposomes through its unique low heat-activated release of encapsulated chemotherapeutic agents. We are able to use several available focused-heat technologies, such as radiofrequency ablation ("RFA"), microwave energy and high intensity focused ultrasound ("HIFU"), to activate the release of drugs from our novel heat sensitive liposomes.

THERMODOX® for the Treatment of Primary Liver Cancer

Primary Liver Cancer Overview

Hepatocellular carcinoma ("HCC") is one of the most common and deadliest forms of cancer worldwide. It ranks as the third most common solid tumor cancer. It is estimated that up to 90% of liver cancer patients will die within five years of diagnosis. The incidence of primary liver cancer is approximately 35,000 cases per year in the U.S., approximately 65,000 cases per year in Europe and is increasing at approximately 2-3% per year worldwide. Global incidence (per 2017 GLOBALCAN statistics) is reported at 755,000 cases. The World Health Organization (the "WHO") has projected that HCC will be the most prevalent form of cancer by 2030. HCC is commonly diagnosed in patients with longstanding hepatic disease and cirrhosis (primarily due to hepatitis C in the U.S., Japan and Europe and hepatitis B in Asia).

At an early stage, the standard first line treatment for liver cancer is surgical resection of the tumor. Up to 80% of patients are ineligible for surgery or transplantation at time of diagnosis because early-stage liver cancer generally has few symptoms and when finally detected the tumor frequently is too large for surgical resection. There are few alternative treatments since radiation therapy and chemotherapy are largely ineffective in treating liver cancer. For tumors generally up to 5 centimeters in diameter, RFA has emerged as the standard of care treatment which directly destroys the tumor tissue through the application of high temperatures administered by a probe inserted into the core of the tumor. Local recurrence rates after RFA directly correlate to the size of the tumor. For tumors 3 cm or smaller in diameter the recurrence rate has been reported to be 10 - 20%; however, for tumors greater than 3 cm, local recurrence rates of 40% or higher have been observed.





64







Celsion's Approach


While RFA uses extremely high temperatures (greater than 90° Celsius) to ablate the tumor, it may fail to treat micro-metastases in the outer margins of the ablation zone because temperatures in the periphery may not be high enough to destroy cancer cells. Our ThermoDox® treatment approach is designed to utilize the ability of RFA devices to ablate the center of the tumor while simultaneously thermally activating our ThermoDox® liposome to release its encapsulated doxorubicin to kill any remaining viable cancer cells throughout the heated region, including the ablation margins. This novel treatment approach is intended to deliver the drug directly to those cancer cells that survive RFA. This approach is designed to increase the delivery of the doxorubicin at the desired tumor site while potentially reducing drug exposure distant to the tumor site.





OPTIMA Study



The OPTIMA Study represents an evaluation of ThermoDox® in combination with a first line therapy, RFA, for newly diagnosed, intermediate stage HCC patients. The OPTIMA Study was designed to enroll up to 550 patients globally at approximately 65 clinical sites in the U.S., Canada, European Union (EU), China and other countries in the Asia-Pacific region and will evaluate ThermoDox® in combination with standardized RFA, which will require a minimum of 45 minutes across all investigators and clinical sites for treating lesions three to seven centimeters, versus standardized RFA alone. The primary endpoint for the OPTIMA Study is OS, and the secondary endpoints are progression free survival and safety. The statistical plan calls for two interim efficacy analyses by an independent Data Monitoring Committee ("DMC").

On February 24, 2014, we announced that the FDA provided clearance for the OPTIMA Study, which is a pivotal, double-blind, placebo-controlled Phase III trial of ThermoDox®, in combination with standardized RFA, for the treatment of primary liver cancer. The trial design of the OPTIMA Study is based on the comprehensive analysis of data from an earlier Phase III clinical trial called the HEAT Study (the "HEAT Study"). The OPTIMA Study is supported by a hypothesis developed from an OS analysis of a large subgroup of patients from the HEAT Study.

Post-hoc data analysis from our earlier Phase III HEAT Study suggests that ThermoDox® may substantially improve OS, when compared to the control group, in patients if their lesions undergo a 45-minute RFA procedure standardized for a lesion greater than 3 cm in diameter. Data from nine OS sweeps have been conducted since the top line progression free survival PFS data from the HEAT Study were announced in January 2013, with each data set demonstrating substantial improvement in clinical benefit over the control group with statistical significance. On August 15, 2016, we announced updated results from its final retrospective OS analysis of the data from the HEAT Study. These results demonstrated that in a large, well bounded, subgroup of patients with a single lesion (n=285, 41% of the HEAT Study patients), treatment with a combination of ThermoDox® and optimized RFA provided an average 54% risk improvement in OS compared to optimized RFA alone. The HR at this analysis is 0.65 (95% CI 0.45 - 0.94) with a p-value of 0.02. Median OS for the ThermoDox® group has been reached which translates into a two-year survival benefit over the optimized RFA group (projected to be greater than 80 months for the ThermoDox® plus optimized RFA group compared to less than 60 months projection for the optimized RFA only group). This information should be viewed with caution since it is based on a retrospective analysis of a subgroup.

We also conducted additional analyses that further strengthen the evidence for the HEAT Study subgroup.





  ? We commissioned an independent computational model at the University of South
    Carolina Medical School. The results unequivocally indicate that longer RFA
    heating times correlate with significant increases in doxorubicin
    concentration around the RFA treated tissue.

  ? In addition, we conducted a prospective preclinical study in 22 pigs using two
    different manufacturers of RFA and human equivalent doses of ThermoDox® that
    clearly support the relationship between increased heating duration and
    doxorubicin concentrations.



On August 13, 2019, the Company announced that results from an independent analysis of the Company's ThermoDox® HEAT Study conducted by the National Institutes of Health (NIH) were published in the peer-reviewed publication, Journal of Vascular and Interventional Radiology. The analysis was conducted by the intramural research program of the NIH and the NIH Center for Interventional Oncology, with the full data set from the Company's HEAT Study. The analysis evaluated the full data set to determine if there was a correlation between baseline tumor volume and RFA heating time (minutes/tumor volume in milliliters), with or without ThermoDox® treatment, for patients with HCC. The NIH analysis was conducted under the direction of Dr. Bradford Wood, MD, Director, NIH Center for Interventional Oncology and Chief, NIH Clinical Center Interventional Radiology.





65






The article titled, "RFA Duration Per Tumor Volume May Correlate with Overall Survival in Solitary Hepatocellular Carcinoma Patients Treated with RFA Plus Lyso-thermosensitive Liposomal Doxorubicin," discussed the NIH analysis of results from 437 patients in the HEAT Study (all patients with a single lesion representing 62.4% of the study population). The key finding was that increased RFA heating time per tumor volume significantly improved OS in patients with single-lesion HCC who were treated with RFA plus ThermoDox®, compared to patients treated with RFA alone. A one-unit increase in RFA duration per tumor volume was shown to result in about a 20% improvement in OS for patients administered ThermoDox®, compared to RFA alone. The authors conclude that increasing RFA heating time in combination with ThermoDox® significantly improves OS and establishes an improvement of over two years versus the control arm when the heating time per milliliter of tumor is greater than 2.5 minutes. This finding was consistent with the Company's own results, which defined the optimized RFA procedure as a 45-minute treatment for tumors with a diameter of 3 centimeters. Thus, the NIH analysis lent support to the hypothesis underpinning the OPTIMA Study.

In August 2018, the Company announced that the OPTIMA Study was fully enrolled. On August 5, 2019, the Company announced that the prescribed number of OS events had been reached for the first prespecified interim analysis of the OPTIMA Phase III Study. Following preparation of the data, the first interim analysis was conducted by the DMC. The DMC's pre-planned interim efficacy review followed 128 patient events, or deaths, which occurred in August 2019. On November 4, 2019, the Company announced that the DMC unanimously recommended the OPTIMA Study continue according to protocol. The recommendation was based on a review of blinded safety and data integrity from 556 patients enrolled in the OPTIMA Study. Data presented demonstrated that PFS and OS data appeared to be tracking with patient data observed at a similar point in the Company's subgroup of patients followed prospectively in the earlier Phase III HEAT Study, upon which the OPTIMA Study was based.

On April 15, 2020, the Company announced that the prescribed minimum number of events of 158 patient deaths had been reached for the second pre-specified interim analysis of the OPTIMA Phase III Study. The hazard ratio for success at 158 deaths is 0.70, which represents a 30% reduction in the risk of death compared with RFA alone. On July 13, 2020, the Company announced that it has received a recommendation from the DMC to consider stopping the global OPTIMA Study. The recommendation was made following the second pre-planned interim safety and efficacy analysis by the DMC on July 9, 2020. The DMC analysis found that the pre-specified boundary for stopping the trial for futility of 0.900 was crossed with an actual value of 0.903. However, the 2-sided p-value of 0.524 for this analysis provides uncertainty, subsequently, the DMC has left the final decision of whether or not to stop the OPTIMA Study to Celsion. There were no safety concerns noted during the interim analysis. The Company followed the advice of the DMC and considered its options either to stop the study or continue to follow patients after a thorough review of the data, and an evaluation of our probability of success. Timing for this decision is made less urgent by the fact that the OPTIMA Study has been fully enrolled since August 2018 and that the vast majority of the trial expenses have already been incurred.

On August 4, 2020, the Company issued a press release announcing it would continue following patients for OS, noting that the unexpected and marginally crossed futility boundary, suggested by the Kaplan-Meier analysis at the second interim analysis on July 9, 2020, may be associated with a data maturity issue. On October 12, 2020, the Company provided an update on the ongoing data analysis from its Phase III OPTIMA Study with ThermoDox® as well as growing interest among clinical investigators in conducting studies with ThermoDox® as a monotherapy or in combination with other therapies.





  ? Celsion engaged a global biometrics contract research organization, with
    forensic statistical analysis capability that specializes in data management,
    statistical consulting, statistical analysis and data sciences, with
    particular expertise in evaluating unusual data from clinical trials and
    experience with associated regulatory issues. The primary objective of the
    CRO's work was to determine the basis and reasoning behind continuing to
    follow patients for survival, and if there were outside influences that may
    have impacted the forecast of futility.




66







  ? In parallel, the Company submitted all OPTIMA Study clinical trial data to the
    National Institutes of Health (NIH) with the expectation of receivings a report
    on the following:

    ? A Cox Regression Analysis for single solitary lesions including minimum burn
      time per tumor volume, evaluating similarities to the hypothesis generated
      from the NIH paper published in the Journal of Vascular and Interventional
      Radiology, in which the key finding was that increased RFA heating time per
      tumor volume significantly improved OS in patients with single lesion HCC who
      were treated with RFA plus ThermoDox®, compared with patients treated with
      RFA alone.
    ? A site-by-site evaluation for RFA heating time-based anomalies that may have
      contributed to the treatment arm performance.
    ? An image-based evaluation comparing results from the OPTIMA Study to the data
      from the HEAT Study that led to the RFA heating time hypothesis.



On February 11, 2021, the Company provided a final update on the Phase III OPTIMA Study and the decision to stop following patients in the Study. Independent analyses conducted by a global biometrics contract research organization and the NIH, did not find any evidence of significance or factors that would justify continuing to follow patients for OS. Therefore, the Company notified all clinical sites to discontinue following patients. The OPTIMA Study database of 556 patients will now be frozen at 185 patient deaths. While the analyses did identify certain patient subgroups that appear to have had a clinical benefit, the Company concluded that it would not be in its best interest to pursue these retrospective findings as the regulatory hurdles supporting further discussion will be significant.

Investigator-Sponsored Studies with ThermoDox®

Celsion continues working closely and supporting investigations by others throughout the world in breast cancer, pancreatic cancer and in solid tumors in children. Following inquiries from the NIH, we intend to renew our Cooperative Research and Development Agreement (CRADA) with the Institute at a nominal cost, one goal of which is to pursue their interest in a study of ThermoDox® to treat patients with bladder cancer. Importantly, Celsion is developing a business model to support these investigator-sponsored studies in a manner that will not interfere with the Company's focus on our GEN-1 program and vaccine development initiative.

Below are summaries of several investigator-sponsored studies using ThermoDox®:





  ? Oxford University plans to begin enrolling patients in a Phase I pancreatic
    cancer study with ThermoDox® in combination with High Intensity Focused
    Ultrasound (HIFU) in the first half of 2021. The primary objective of this
    trial, the PanDox Study: Targeted Doxorubicin in Pancreatic Tumors, is to
    quantify the enhancement in intratumoral doxorubicin concentration when
    delivered with ThermoDox® and HIFU, versus doxorubicin monotherapy. This study
    is being undertaken pursuant to promising data in a mouse model of pancreatic
    cancer, which was published in the International Journal of Hyperthermia in
    2018. That preclinical study showed a 23x increase in intratumoral doxorubicin
    concentration with ThermoDox® + HIFU, compared with a 2x increase in
    intratumoral doxorubicin concentration with free doxorubicin plus HIFU.

  ? Utrecht University in the Netherlands continues to enroll patients in a Phase
    I breast cancer study to determine the safety, tolerability and feasibility of
    ThermoDox® in combination with Magnetic Resonance Guided High Intensity
    Focused Ultrasound (MR-HIFU) hyperthermia and cyclophosphamide therapy for the
    local treatment of the primary tumor in metastatic breast cancer (mBC). This
    investigator-sponsored study, which is being funded by the Dutch Cancer
    Society, the Center for Translational Molecular Medicine (a public-private
    partnership in the Netherlands), will be conducted at University Medical
    Center Utrecht and will enroll up to 12 newly diagnosed mBC patients. Celsion
    will supply Thermodox® clinical product for the trial.

  ? As evidence of the ongoing support Celsion enjoys from the NIH, they have
    organized a clinical project to evaluate ThermoDox® plus the chemotherapy drug
    mitomycin in bladder cancer. Depending on the NIH timelines, this study may
    commence as early as 2021.




67

Because of the risks and uncertainties discussed in this Annual Report, among others, we are unable to estimate the duration and completion costs of our research and development projects or when, if ever, and to what extent we will receive cash inflows from the commercialization and sale of a product. Our inability to complete any of our research and development activities, preclinical studies or clinical trials in a timely manner or our failure to enter into collaborative agreements when appropriate could significantly increase our capital requirements and could adversely impact our liquidity. While our estimated future capital requirements are uncertain and could increase or decrease as a result of many factors, including the extent to which we choose to advance our research, development activities, preclinical studies and clinical trials, or if we are in a position to pursue manufacturing or commercialization activities, we will need significant additional capital to develop our product candidates through development and clinical trials, obtain regulatory approvals and manufacture and commercialize approved products, if any. We do not know whether we will be able to access additional capital when needed or on terms favorable to us or our stockholders. Our inability to raise additional capital, or to do so on terms reasonably acceptable to us, would jeopardize the future success of our business.

Covenant Not to Compete (CNTC)

Pursuant to the EGEN Purchase Agreement, EGEN provided certain covenants ("Covenant Not to Compete") to the Company whereby EGEN agreed, during the period ending on the seventh anniversary of the closing date of the acquisition on June 20, 2014, not to enter into any business, directly or indirectly, which competes with the business of the Company nor will it contact, solicit or approach any of the employees of the Company for purposes of offering employment.





Business Plan



As a clinical stage biopharmaceutical company, our business and our ability to execute our strategy to achieve our corporate goals are subject to numerous risks and uncertainties. Material risks and uncertainties relating to our business and our industry are described in "Part I, Item 1A. Risk Factors" in this Annual Report on Form 10-K.

Since inception, the Company has incurred substantial operating losses, principally from expenses associated with the Company's research and development programs, clinical trials conducted in connection with the Company's product candidates, and applications and submissions to the U.S. FDA. The Company has not generated significant revenue and has incurred significant net losses in each year since our inception. As of December 31, 2020, the Company has incurred approximately $312 million of cumulative net losses and we had approximately $17.2 million in cash and cash equivalents. We have substantial future capital requirements to continue our research and development activities and advance our product candidates through various development stages. The Company believes these expenditures are essential for the commercialization of its technologies.

The Company expects its operating losses to continue for the foreseeable future as it continues its product development efforts, and when it undertakes marketing and sales activities. The Company's ability to achieve profitability is dependent upon its ability to obtain governmental approvals, manufacture, and market and sell its new product candidates. There can be no assurance that the Company will be able to commercialize its technology successfully or that profitability will ever be achieved. The operating results of the Company have fluctuated significantly in the past.

The actual amount of funds the Company will need to operate is subject to many factors, some of which are beyond the Company's control. These factors include the following:

? the progress of research activities;

? the number and scope of research programs;

? the progress of preclinical and clinical development activities;

? the progress of the development efforts of parties with whom the Company has

entered into research and development agreements;

? the costs associated with additional clinical trials of product candidates;






68






? the ability to maintain current research and development licensing arrangements

and to establish new research and development and licensing arrangements;

? the ability to achieve milestones under licensing arrangements;

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

intellectual property rights; and

? the costs and timing of regulatory approvals.

On July 13, 2020, the Company announced that it has received a recommendation from the independent DMC to consider stopping the global Phase III OPTIMA Study of ThermoDox® in combination with RFA for the treatment of HCC, or primary liver cancer. The recommendation was made following the second pre-planned interim safety and efficacy analysis by the DMC on July 9, 2020. The DMC's analysis found that the pre-specified boundary for stopping the trial for futility of 0.900 was crossed with an actual value of 0.903. The Company followed the advice of the DMC and considered its options to either stop the study or continue to follow patients after a thorough review of the data, and an evaluation of the probability of success. On February 11, 2021, the Company issued a letter to shareholders stating that the Company was notifying all clinical sites to discontinue following patients in the OPTIMA Study.

As more fully discussed below, in June 2020 and as updated in September 2020, the Company filed an application with the New Jersey Economic Development Authority to sell substantially all of its remaining State of New Jersey net operating losses totaling $2.0 million available under the program. On February 12, 2021, the New Jersey Economic Development Authority approved the full amount of the Company's application. In February 2021, the Company entered into an agreement to sell the net operating losses from the 2020 application and expects to receive net proceeds of approximately $1.85 million by the end of the first quarter of 2021.

As more fully discussed in Note 10 to our Consolidated Financial Statements contained in this Form 10-K, during 2021 through the date of the filing of this Annual Report on Form 10-K, the Company has raised $6.9 million in gross proceeds from the use of its JonesTrading Capital on DemandTM financing facility, $35 million in gross proceeds from a registered direct financing completed in January 2021 and approximately $1.5 million in net proceeds through warrant exercises.

With $17.2 million in cash and cash equivalents, coupled with approximately $43 million of gross proceeds received from the sale of equity thus far in 2021 and up to $1.85 million in expected net proceeds from the sale of its State of New Jersey net operating losses it applied for in 2020, the Company believes it has sufficient capital resources to fund its operations through 2023.

The Company has based its estimates on assumptions that may prove to be wrong. The Company may need to obtain additional funds sooner or in greater amounts than it currently anticipates. Potential sources of financing include strategic relationships, public or private sales of the Company's shares or debt, the sale of the Company's State of New Jersey net operating losses and other sources. If the Company raises funds by selling additional shares of common stock or other securities convertible into common stock, the ownership interest of existing stockholders may be diluted.





69







Financing Overview


Equity, Debt and Other Forms of Financing

During 2020, 2019 and 2018, the Company submitted applications to sell a portion of the Company's State of New Jersey net operating losses as part of the Technology Business Tax Certificate Program sponsored by The New Jersey Economic Development Authority. Under the program, emerging biotechnology companies with unused NOLs and unused research and development credits are allowed to sell these benefits to other New Jersey-based companies. In 2018 and 2019, the Company sold NOLs totaling $13 million receiving net proceeds of $12.2 million. In June 2020 and as updated in September 2020, the Company filed an application with the New Jersey Economic Development Authority to sell substantially all of its remaining State of New Jersey net operating losses totaling $2.0 million available under the program. On February 12, 2021, the New Jersey Economic Development Authority approved the full amount of the Company's application. In February of 2021, the Company entered into an agreement to sell the net operating losses from the 2020 application and expects to receive net proceeds of approximately $1.85 million by the end of the first quarter of 2021. Beginning in 2021, the New Jersey State Legislature increased the maximum lifetime benefit per company from $15 million to $20 million, which will allow the Company to participate in this innovative funding program in future years.

In June 2018, the Company entered into a Credit Agreement with Horizon Technology Finance Corporation ("Horizon") that provided $10 million in capital (the "Horizon Credit Agreement"). The obligations under the Horizon Credit Agreement are secured by a first-priority security interest in substantially all assets of Celsion other than intellectual property assets. Payments under the loan agreement are interest only (calculated based on one-month LIBOR plus 7.625%) for the first twenty-four (24) months through July 2020, followed by a 24-month amortization period of principal and interest starting on August 1, 2020 and ending through the scheduled maturity date. On August 28, 2020, in connection with an Amendment to the Horizon Credit Agreement, Celsion repaid $5 million of the $10 million loan and $0.2 million in related end of term charges, and the remaining $5 million in obligations were restructured as more fully discussed in Note 8 to our Consolidated Financial Statements contained in this Form 10-K.

During 2019 and 2020, we issued a total of 21.1 million shares of common stock as discussed below for an aggregate $32.8 million in gross proceeds. During the first quarter of 2021, the Company issued an additional 34.3 million shares of common stock for an aggregate of $43.4 million in gross proceeds as discussed in more detail below.





  ? On October 28, 2019, Company, entered into the 2019 Aspire Purchase Agreement
    with Aspire Capital. The terms and conditions pursuant to the 2019 Aspire
    Purchase Agreement are substantially similar to the 2018 Aspire Purchase
    Agreement. Pursuant to the new 2019 Aspire Purchase Agreement, Aspire Capital
    is committed to purchase up to an aggregate of $10.0 million of shares of the
    Company's common stock over the 24-month term of the 2019 Aspire Purchase
    Agreement. Concurrently with entering into the 2019 Aspire Purchase Agreement,
    the Company also entered into a registration rights agreement with Aspire
    Capital (the "Registration Rights Agreement"), in which the Company agreed to
    file one or more registration statements, as permissible and necessary to
    register under the Securities Act of 1933, as amended (the "Securities Act"),
    registering the sale of the shares of the Company's common stock that have
    been and may be issued to Aspire Capital under the 2019 Aspire Purchase
    Agreement. In consideration for entering into the 2019 Aspire Purchase
    Agreement, the Company issued to Aspire Capital an additional 100,000
    Commitment Shares. On November 8, 2019, the Company filed with the SEC a
    Registration Statement on Form S-1 registering all the shares of common stock
    that may be offered to Aspire Capital from time to time under the 2019 Aspire
    Purchase Agreement. During 2019, the Company sold 0.5 million shares of common
    stock under the 2019 Aspire Purchase Agreement, receiving approximately $0.7
    million in gross proceeds. On March 5, 2020, the Company delivered notice to
    Aspire Capital terminating the 2019 Aspire Purchase Agreement effective as of
    March 6, 2020. During the first quarter of 2020, the Company sold 1.0 million
    shares of common stock under the 2019 Aspire Purchase Agreement and received
    $1.6 million in gross proceeds.




70







  ? On December 4, 2018, the Company entered into a new Capital on DemandTM Sales
    Agreement (the "Capital on Demand Agreement") with JonesTrading Institutional
    Services LLC, as sales agent ("JonesTrading"), pursuant to which the Company
    may offer and sell, from time to time, through JonesTrading shares of common
    stock having an aggregate offering price of up to $16.0 million. The Company
    intends to use the net proceeds from the offering, if any, for general
    corporate purposes, including research and development activities, capital
    expenditures and working capital. The Company is not obligated to sell any
    Common Stock under the Capital on Demand Agreement and, subject to the terms
    and conditions of the Capital on Demand Agreement, JonesTrading will use
    commercially reasonable efforts, consistent with its normal trading and sales
    practices and applicable state and federal law, rules and regulations and the
    rules of The Nasdaq Capital Market, to sell common stock from time to time
    based upon Celsion's instructions, including any price, time or size limits or
    other customary parameters or conditions the Company may impose. Under the
    Capital on Demand Agreement, JonesTrading may sell common stock by any method
    deemed to be an "at the market offering" as defined in Rule 415 promulgated
    under the Securities Act of 1933, as amended. The Capital on Demand Agreement
    will terminate upon the earlier of (i) the sale of all shares of our common
    stock subject to the Sales Agreement, and (ii) the termination of the Capital
    on Demand Agreement by JonesTrading or Celsion. The Capital on Demand
    Agreement may be terminated by JonesTrading or the Company at any time upon 10
    days' notice to the other party, or by JonesTrading at any time in certain
    circumstances, including the occurrence of a material adverse change in the
    Company. The Company did not sell any shares under the Capital on Demand
    Agreement during 2018. During 2019, 2020 and thus far in 2021, the Company
    sold 0.5 million, 5.2 million and 7.2 million shares of common stock under the
    Capital on Demand Agreement, respectively, receiving gross proceeds of
    approximately $1.0 million, $6.2 million and $6.9 million, respectively.

  ? On February 27, 2020, we entered into a Securities Purchase Agreement (the
    "Purchase Agreement") with several institutional investors, pursuant to which
    we agreed to issue and sell, in a registered direct offering (the "February
    2020 Offering"), an aggregate of 4,571,428 shares (the "Shares") of our common
    stock at an offering price of $1.05 per share for gross proceeds of
    approximately $4.8 million before the deduction of the Placement Agent fees
    and offering expenses. The Shares were offered by the Company pursuant to a
    registration statement on Form S-3 (File No. 333-227236). The Purchase
    Agreement contains customary representations, warranties and agreements by the
    Company and customary conditions to closing. In a concurrent private placement
    (the "Private Placement"), the Company agreed to issue to the investors that
    participated in the Offering, for no additional consideration, warrants, to
    purchase up to 2,971,428 shares of Common Stock (the "Original Warrants"). The
    Original Warrants were initially exercisable six months following their
    issuance and were set to expire on the five-year anniversary of such initial
    exercise date. The Warrants had an exercise price of $1.15 per share subject
    to adjustment as provided therein. On March 12, 2020, the Company entered into
    private exchange agreements (the "Exchange Agreements") with holders the
    Warrants. Pursuant to the Exchange Agreements, in return for a higher exercise
    price of $1.24 per share of Common Stock, the Company issued new warrants to
    the Investors to purchase up to 3,200,000 shares of Common Stock (the
    "Exchange Warrants") in exchange for the Original Warrants. The Exchange
    Warrants, like the Original Warrants, are initially exercisable six months
    following their issuance (the "Initial Exercise Date") and expire on the
    five-year anniversary of their Initial Exercise Date. Other than having a
    higher exercise price, different issue date, Initial Exercise Date and
    expiration date, the terms of the Exchange Warrants are identical to those of
    the Original Warrants. On July 31, 2020, the Company filed a Form S-3
    Registration Statement to register the shares of Common Stock issuable under
    the Exchange Warrants; the Registration Statement was declared effective by
    the SEC on August 13, 2020. No Exchange Warrants were exercised during 2020.
    During 2021 thus far, the Company has issued 1.2 million shares pursuant to
    investors exercising Exchange Warrants, receiving approximately $1.5 million
    in gross proceeds.




71







  ? On September 8, 2020, the Company entered into a purchase agreement (the "LPC
    Purchase Agreement") and a Registration Rights Agreement (the "Registration
    Rights Agreement") with Lincoln Park Capital Fund, LLC ("Lincoln Park"),
    pursuant to which, upon the terms and subject to the conditions and
    limitations set forth therein, the Company has the right to sell to Lincoln
    Park up to $26.0 million of shares of the Company's Common Stock at the
    Company's discretion as described below (the " LPC Offering"). Over the
    36-month term of the LPC Purchase Agreement, we have the right, but not the
    obligation, from time to time, in our sole discretion and subject to certain
    conditions, including that the closing price of our Common Stock is not below
    $0.25 per share, to direct Lincoln Park to purchase up to an aggregate amount
    of $26.0 million (subject to certain limitations) of shares of Common Stock.
    Under the Purchase Agreement, on any business day selected by us, we may
    direct Lincoln Park to purchase up to 400,000 shares (the "Regular Purchase
    Share Limit") of our Common Stock (each such purchase, a "Regular Purchase").
    Lincoln Park's maximum obligation under any single Regular Purchase will not
    exceed $1,500,000 unless we mutually agree to increase the maximum amount of
    such Regular Purchase. The purchase price for shares of Common Stock to be
    purchased by Lincoln Park under a Regular Purchase will be the equal to the
    lower of (in each case, subject to the adjustments described in the LPC
    Purchase Agreement): (i) the lowest sale price for our Common Stock on The
    Nasdaq Capital Market on the applicable purchase date, and (ii) the arithmetic
    average of the three lowest sale prices for our Common Stock on The Nasdaq
    Capital Market during the ten trading days prior to the purchase date. If we
    direct Lincoln Park to purchase the maximum number of shares of Common Stock
    we then may sell in a Regular Purchase, then in addition to such Regular
    Purchase, and subject to certain conditions and limitations in the LPC
    Purchase Agreement, we may direct Lincoln Park to make an "accelerated
    purchase" of an additional amount of Common Stock that may not exceed the
    lesser of (i) 300% of the number of shares purchased pursuant to the
    corresponding Regular Purchase and (ii) 30% of the total number of shares of
    our Common Stock traded on The Nasdaq Capital Market during a specified period
    on the applicable purchase date as set forth in the Purchase Agreement. Under
    certain circumstances and in accordance with the Purchase Agreement, the
    Company may direct Lincoln Park to purchase shares in multiple accelerated
    purchases on the same trading day. The Purchase Agreement prohibits us from
    issuing or selling to Lincoln Park under the Purchase Agreement: (i) in excess
    of 6,688,588 shares of our Common Stock (the "Exchange Cap"), unless we obtain
    stockholder approval to issue shares in excess of the Exchange Cap or the
    average price of all applicable sales of our Common Stock to Lincoln Park
    under the LPC Purchase Agreement equal or exceed the lower of (a) the Nasdaq
    Official Closing Price (as defined in the Purchase Agreement) immediately
    preceding the execution of the LPC Purchase Agreement or (b) the average of
    the five Nasdaq Official Closing Prices for the Common Stock immediately
    preceding the execution of the LPC Purchase Agreement, as adjusted in
    accordance with the rules of The Nasdaq Capital Market, and (ii) any shares of
    our Common Stock if those shares, when aggregated with all other shares of our
    Common Stock then beneficially owned by Lincoln Park and its affiliates would
    result in Lincoln Park and its affiliates having beneficial ownership of more
    than 9.99% of the then total outstanding shares of our Common Stock. The LPC
    Purchase Agreement does not limit our ability to raise capital from other
    sources at our sole discretion, except that we may not enter into any equity
    line or similar transaction for 36 months, other than an "at-the-market"
    offering. The LPC Purchase Agreement and the Registration Rights Agreement
    contain customary representations, warranties and agreements of us and Lincoln
    Park, indemnification rights and other obligations of the parties. We have the
    right to terminate the Purchase Agreement at any time on one business days'
    notice to Lincoln Park, at no cost to us. As consideration for entering into
    the Purchase Agreement, we issued 437,828 shares of our Common Stock to
    Lincoln Park (the "LPC Commitment Shares"). We will not receive any cash
    proceeds from the issuance of the LPC Commitment Shares. Also pursuant to the
    Purchase Agreement, Lincoln Park agreed to an initial purchase of 1,000,000
    shares of our Common Stock for an aggregate purchase price of $1,000,000 or
    $1.00 per share. Lincoln Park has covenanted not to cause or engage in any
    manner whatsoever, any direct or indirect short selling or hedging of our
    shares of Common Stock. The Offering is being made pursuant to our effective
    Registration Statement on Form S-3 (File No. 333-227236) (the "Registration
    Statement"), which was previously filed with the SEC on September 7, 2018, and
    declared effective by the SEC on October 12, 2018, and the prospectus
    supplement related to the Offering filed with the SEC on September 8, 2020.
    During 2020 the Company sold and issued an aggregate of 3.3 million shares,
    including the LPC Commitment Shares, under the LPC Purchase Agreement,
    receiving approximately $2.2 million in gross proceeds. During 2020, the
    Company sold and issued an aggregate of 3.3 million shares, including the LPC
    Commitment Shares, under the LPC Purchase Agreement, receiving approximately
    $2.2 million in gross proceeds. During the first quarter of 2021, the Company
    sent a letter to Lincoln Park terminating the LPC Offering effective January
    21, 2021. The Company did not sell any shares under the LPC Purchase Agreement
    during 2021.

  ? On January 22, 2021, the Company entered into a Securities Purchase Agreement
    (the "January 2021 Purchase Agreement") with several institutional investors,
    pursuant to which the Company agreed to issue and sell, in a registered direct
    offering (the "January 2021 Offering"), an aggregate of 25,925,925 shares of
    the Company's common stock at an offering price of $1.35 per share for gross
    proceeds of approximately $35 million before the deduction of Placement Agents
    fees and offering expenses. The shares were offered by the Company pursuant to
    a registration statement on Form S-3 (File No. 333-227236) (the "Registration
    Statement") and a registration statement on Form S-3 (File No. 333-252320)
    filed pursuant to Rule 462 under the Securities Act of 1933, as amended (the
    "Securities Act"). The January 2021 Purchase Agreement contains customary
    representations, warranties and agreements by the Company and customary
    conditions to closing. The closing of the January 2021 Offering occurred on
    January 26, 2021.

    In connection with the January 2021 Offering, the Company entered into a
    placement agent agreement (the "January 2021 Placement Agent Agreement") with
    A.G.P./Alliance Global Partners (together with Brookline Capital Markets, the
    "January 2021 Placement Agents") pursuant to which the Company agreed to pay
    the January 2021 Placement Agents a cash fee equal to 7% of the aggregate
    gross proceeds raised from the sale of the securities sold in the January 2021
    Offering and reimburse the January 2021 Placement Agents for certain of their
    expenses in an amount not to exceed $82,500.




72







    The January 2021 Placement Agent Agreement contains customary representations,
    warranties and agreements by the Company, customary conditions to closing,
    indemnification obligations of the Company and the January 2021 Placement
    Agents, including for liabilities under the Securities Act, other obligations
    of the parties and termination provisions. Under the January 2021 Purchase
    Agreement and January 2021 Placement Agent Agreement, the Company and its
    subsidiary are prohibited, for a period of 90 days after the closing, from
    issuing, entering into any agreement to issue or announcing the issuance or
    proposed issuance of any shares of common stock or any other securities that
    are at any time convertible into, or exercisable or exchangeable for, or
    otherwise entitle the holder thereof to receive common stock, without the
    prior written consent of the placement agents or the investors participating
    in the offering, subject to specific exceptions.



Please refer to Note 2 to our Consolidated Financial Statements contained in this Form 10-K. Also refer to Part II, Item IA, Risk Factors, including, but not limited to, "We will need to raise substantial additional capital to fund our planned future operations, and we may be unable to secure such capital without dilutive financing transactions. If we are not able to raise additional capital, we may not be able to complete the development, testing and commercialization of our product candidates."

Critical Accounting Policies and Estimates

Our financial statements, which appear at Part II, Item 8. Financial Statements and Supplementary Data have been prepared in accordance with accounting principles generally accepted in the U.S., which require that we make certain assumptions and estimates and, in connection therewith, adopt certain accounting policies. Our significant accounting policies are set forth in Note 1 to our Consolidated Financial Statements contained in this Form 10-K. Of those policies, we believe that the policies discussed below may involve a higher degree of judgment and may be more critical to an accurate reflection of our financial condition and results of operations.

In-Process Research and Development, Other Intangible Assets and Goodwill

During 2014, the Company acquired certain assets of EGEN, Inc. As more fully described in Note 5 to our Consolidated Financial Statements contained in this Form 10-K., the acquisition was accounted for under the acquisition method of accounting which required the Company to perform an allocation of the purchase price to the assets acquired and liabilities assumed. Under the acquisition method of accounting, the total purchase price is allocated to net tangible and intangible assets and liabilities based on their estimated fair values as of the acquisition date.





Lease Accounting



In February 2016, the FASB issued Accounting Standards Update ("ASU") No. 2016-02, "Leases" - Topic 842 (ASC Topic 842), which requires that lessees recognize assets and liabilities for leases with lease terms greater than twelve months in the statement of financial position. Leases will be classified as either finance or operating, with classification affecting the pattern of expense recognition in the income statement. This update also requires improved disclosures to help users of financial statements better understand the amount, timing and uncertainty of cash flows arising from leases. The update became effective for fiscal years beginning after December 15, 2018, including interim reporting periods within that reporting period. The FASB subsequently issued the following amendments to ASC Topic 842, which have the same effective date and transition date of January 1, 2019:





  ? ASU No. 2018-10, Codification Improvements to Topic 842, Leases, which amends
    certain narrow aspects of the guidance issued in ASU No. 2016-02; and

  ? ASU No. 2018-11, Leases (Topic 842): Targeted Improvements, which allows for a
    transition approach to initially apply ASU No. 2016-02 at the adoption date
    and recognize a cumulative-effect adjustment to the opening balance of
    retained earnings in the period of adoption as well as an additional practical
    expedient for lessors to not separate non-lease components from the associated
    lease component.




73

We adopted Accounting Standards Codification ("ASC") Topic 842 effective January 1, 2019 and elected to apply the available practical expedients and implement internal controls to enable the preparation of financial information on adoption. We have identified all of our leases which consist of the New Jersey corporate office lease and the Alabama lab facility lease and we estimate the adoption of this standard will result in the recognition of right-of-use assets of approximately $1.4 million, related operating lease liabilities of $1.5 million and reduced other liabilities by approximately $0.1 million on the consolidated balance sheets as of January 1, 2019 of approximately $1.5 million related to our operating lease commitments, with no material impact to the opening balance of retained earnings. See Note 15 to our Consolidated Financial Statements contained in this Form 10-K for further discussions regarding the adoption of ASC Topic 842.

In August 2018, the FASB issued ASU No. 2018-13, Fair Value Measurement: Disclosure Framework - Changes to the Disclosure Requirements for Fair Value Measurement, which adds and modifies certain disclosure requirements for fair value measurements. Under the new guidance, entities will no longer be required to disclose the amount of and reasons for transfers between Level 1 and Level 2 of the fair value hierarchy, or valuation processes for Level 3 fair value measurements. However, public companies will be required to disclose the range and weighted average of significant unobservable inputs used to develop Level 3 fair value measurements, and related changes in unrealized gains and losses included in other comprehensive income. This update is effective for annual periods beginning after December 15, 2019, and interim periods within those periods. The adoption of this standard did not have an impact on the Company's condensed consolidated financial statements.

In December 2019, the FASB issued ASU No. 2019-12, Income Taxes (Topic 740). The standard simplifies the accounting for incomes taxes by removing certain exceptions to the general principles in Topic 740 related to the approach for intra-period tax allocation and the recognition of deferred tax liabilities for outside basis differences. The standard also clarifies the accounting for transactions that result in a step-up in the tax basis of goodwill. The standard also improves consistent application of and simplifies GAAP for other areas of Topic 740 by clarifying and amending existing guidance. The amendment is effective for fiscal years, and interim periods within those fiscal years, beginning after December 15, 2020. Early adoption is permitted. The Company does not believe the adoption of this standard will have a material impact on its condensed consolidated financial statements.

We review our financial reporting and disclosure practices and accounting policies on an ongoing basis to ensure that our financial reporting and disclosure system provides accurate and transparent information relative to the current economic and business environment. As part of the process, the Company reviews the selection, application and communication of critical accounting policies and financial disclosures. The preparation of our financial statements in conformity with accounting principles generally accepted in the U.S. requires that our management make estimates and assumptions that affect the reported amounts of assets and liabilities and disclosure of contingent assets and liabilities at the date of the financial statements and the reported amounts of revenues and expenses during the reporting period. We review our estimates and the methods by which they are determined on an ongoing basis. However, actual results could differ from our estimates.





Results of Operations


Comparison of Fiscal Year Ended December 31, 2020 and Fiscal Year Ended December 31, 2019.

For the year ended December 31, 2020, our net loss was $21.5 million compared to a net loss of $16.9 million for the year ended December 31, 2019. The Company recognized $1.85 million and $1.82 million in tax benefits from the sale of its New Jersey net operating losses under the Technology Business Tax Certificate Program in each of the fourth quarters of 2020 and 2019, respectively. With $17.2 million in cash and cash equivalents, coupled with approximately $43 million of gross proceeds received from the sale of equity in the first quarter of 2021 and up to $1.85 million in expected proceeds from the sale of the State of New Jersey net operating losses it applied for in 2020, the Company believes it has sufficient capital resources to fund its operations through 2023.

Technology Development and Licensing Revenue

In January 2013, we entered into a technology development contract with Hisun, pursuant to which Hisun paid us a non-refundable technology transfer fee of $5.0 million to support our development of ThermoDox® in the China territory. The $5.0 million received as a non-refundable payment from Hisun in the first quarter 2013 has been recorded to deferred revenue and will be amortized over the ten-year term of the agreement; therefore, we recognized revenue of $500,000 in each of the years 2020 and 2019.





74






Research and Development Expenses

Research and development ("R&D") expenses decreased $1.8 million from $13.1 million in 2019 to $11.3 million in 2020. Costs associated with the Phase III OPTIMA Study were $2.2 million in 2020 compared to $4.1 million in 2019. In July 2020, the Company unblinded the OPTIMA Study at the recommendation of the DMC to halt the study due to futility. Costs associated with the OVATION 2 Study were $1.3 million in 2020 compared to $0.6 million in 2019 as the Company initiated enrollment in the Phase 2 portion of the study during the third quarter of 2020. Regulatory costs were $0.6 million in 2020 compared to $1.1 million in 2019. Other clinical costs were $2.0 million in 2020 compared to $2.5 million in 2019. Costs associated with the production of ThermoDox® were $2.1 million during 2020 compared to $1.5 million in 2019. R&D costs associated with the development of GEN-1 to support the OVATION program decreased by $0.2 million to $3.1 million in 2020 compared to $3.3 million in 2019.

General and Administrative Expenses

General and administrative expenses decreased $0.4 million to $7.6 million in 2020 compared to $8.0 million in 2019. This decrease is primarily attributable to lower personnel costs of approximately $0.5 million which included a $0.3 million decrease in non-cash stock compensation expense.

Change in Earn-out Milestone Liability

The total aggregate purchase price for the acquisition of assets from EGEN included potential future earn-out payments contingent upon achievement of certain milestones. The difference between the aggregate $30.4 million in future earn-out payments and the $13.9 million included in the fair value of the acquisition consideration at June 20, 2014 was based on the Company's risk-adjusted assessment of each milestone and utilizing a discount rate based on the estimated time to achieve the milestone. These milestone payments are fair valued at the end of each quarter and any change in their value is recognized in our Consolidated Financial Statements contained in this Form 10-K.

On March 28, 2019, the Company and EGWU, Inc, entered into an amendment to the Asset Purchase Agreement discussed in Note 8 to our Consolidated Financial Statements contained in this Form 10-K. Pursuant to the Amended Asset Purchase Agreement, payment of the earnout milestone liability related to the Ovarian Cancer Indication of $12.4 million has been modified. The Company has the option to make the payment as follows:





  ? $7.0 million in cash within 10 business days of achieving the milestone; or
  ? $12.4 million in cash, common stock of the Company, or a combination of
    either, within one year of achieving the milestone.



The Company provided EGWU, Inc. 200,000 warrants to purchase common stock at a strike price of $0.01 per warrant share as consideration for entering into the amended agreement. These warrants shares have no expiration and were fair valued at $2.00 using the closing price of a share of Celsion stock on the date of issuance offset by the exercise price and recorded $0.4 million as an expense in the income statement and were classified as equity on the balance sheet during 2019. In October of 2020, EGWU, Inc elected to receive 197,260 shares through a non-cash conversion exercised of all 200,000 warrant shares.

At December 31, 2020, the Company fair valued the earn-out milestone liability at $7.0 million and recognized a non-cash charge of $1.3 million during 2020 as a result of the change in the fair value of earn-out milestone liability of $5.7 million at December 31, 2019. In assessing the earnout milestone liability at December 31, 2020, the Company fair valued each of the two payment options per the Amended Asset Purchase Agreement and weighted them at 50% and 50% probability for the $7.0 million and the $12.4 million payments, respectively.

At December 31, 2019, the Company fair valued the earn-out milestone liability at $5.7 million and recognized a non-cash gain of $3.2 million during 2019 as a result of the change in the fair value of earn-out milestone liability of $8.9 million at December 31, 2018. In assessing the earnout milestone liability at December 31, 2019, the Company fair valued each of the two payment options per the Amended Asset Purchase Agreement and weighted them at 80% and 20% probability for the $7.0 million and the $12.4 million payments, respectively.





75







Impairment of IPR&D


IPR&D is reviewed for impairment at least annually as of our third quarter ended September 30 by assessing if any events or changes in circumstances have occurred which indicate that the carrying value of the assets might not be recoverable. At September 30, 2020, after our assessment of the totality of the events that could impair IPR&D, the Company determined certain IPR&D assets related to the development of its GBM product candidate may be impaired. To arrive at this determination, the Company assessed the status of studies in GBM conducted by its competitors and the Company's strategic commitment of resources to its studies in primary liver cancer and ovarian cancer. The Company concluded that the GBM asset, valued at $2.4 million, was fully impaired and wrote off the GBM asset, incurring a non-cash charge of $2.4 million in the third quarter of 2020. During 2019, the Company concluded no IPR&D asset was impaired during that period.

Investment income and interest expense

The Company realized $0.1 million and $0.5 million of investment income from its short-term investments during 2020 and 2019, respectively. In connection with the Horizon Credit Agreement, the Company incurred $1.3 million and $1.4 million in interest expense in 2020 and 2019, respectively.





Income Tax Benefit


Annually, the State of New Jersey enables approved technology and biotechnology businesses with New Jersey net operating tax losses the opportunity to sell these losses through the Technology Business Tax Certificate Program (the "NOL Program"), thereby providing cash to companies to help fund their research and development and business operations. During the fourth quarter of 2018, the Company received eligibility from the New Jersey Economic Development Authority to sell, and did sell, $11.1 million of its unused New Jersey net operating losses under the Technology Business Tax Certificate Program, receiving $10.4 million of non-dilutive funding. The Company received approval from the New Jersey Economic Development Authority to sell $1.9 million of its New Jersey net operating losses recognizing a tax benefit for the year ended December 31, 2019 for the net proceeds (approximately $1.8 million) by reducing the deferred income tax valuation allowance.

In early 2020, the Company entered into an agreement to sell these net operating losses and received net proceeds of approximately $1.82 million in the second quarter of 2020. In June 2020 and as updated in September 2020, the Company filed an application with the New Jersey Economic Development Authority to sell substantially all of its remaining State of New Jersey net operating losses totaling $2.0 million available under the program. On February 12, 2021, the New Jersey Economic Development Authority approved the full amount of the Company's application. In February of 2021, the Company entered into an agreement to sell the net operating losses from the 2020 application and expects to receive net proceeds of approximately $1.85 million by the end of the first quarter of 2021. During 2021, the New Jersey State Legislature increased the maximum lifetime benefit per company from $15 million to $20 million, which will allow the Company to participate in this innovative funding program in future years.





Inflation


We do not believe that inflation has had a material adverse impact on our revenue or operations in any of the past three years.

Financial Condition, Liquidity and Capital Resources

Since inception we have incurred significant losses and negative cash flows from operations. We have financed our operations primarily through the net proceeds from the sales of equity, credit facilities and amounts received under our product licensing agreement with Yakult and our technology development agreement with Hisun. The process of developing ThermoDox®, GEN-1 and other product candidates and technologies requires significant research and development work and clinical trial studies, as well as significant manufacturing and process development efforts. We expect these activities, together with our general and administrative expenses to result in significant operating losses for the foreseeable future. Our expenses have significantly and regularly exceeded our revenue, and we had an accumulated deficit of $312 million at December 31, 2020.





76






At December 31, 2020 we had total current assets of $18.8 million (including cash and cash equivalents of $17.2 million) and current liabilities of $6.8 million, resulting in net working capital of $12.0 million. At December 31, 2019 we had total current assets of $16.2 million (including cash, cash equivalents, short-term investments and interest receivable of $14.9 million) and current liabilities of $7.9 million, resulting in net working capital of $8.3 million. We have substantial future capital requirements to continue our research and development activities and advance our product candidates through various development stages. The Company believes these expenditures are essential for the commercialization of its technologies.

Net cash used in operating activities for 2020 was $15.6 million. Our net loss of $21.5 million for 2020 included the following non-cash transactions: (i) $1.9 million in non-cash stock-based compensation expense, (ii) $2.4 million non-cash charge from the write-off of the IPR&D assets related to the development of its GBM product candidate, (iii) $0.4 million in non-cash interest expense and (iv) $1.3 non-cash charge based on the change in the earn-out milestone liability. The $15.6 million net cash used in operating activities was funded from cash and cash equivalents, short term investments, and cash proceeds received in equity financings during 2020. At December 31, 2020, we had cash and cash equivalents of $17.2 million and coupled with approximately $42 million of gross proceeds received from the sale of equity thus far in 2021 and up to $1.85 million in expected net proceeds from the sale of the State of New Jersey net operating losses it applied for in 2020, the Company believes it has sufficient capital resources to fund its operations through 2023. See Financing Overview as wells as Notes 8, 9 and 10 to our Consolidated Financial Statements contained in this Form 10-K.

The Company may seek additional capital through further public or private equity offerings, debt financing, additional strategic alliance and licensing arrangements, collaborative arrangements, or some combination of these financing alternatives. If we raise additional funds through the issuance of equity securities, the percentage ownership of our stockholders could be significantly diluted, and the newly issued equity securities may have rights, preferences, or privileges senior to those of the holders of our common stock. If we raise funds through the issuance of debt securities, those securities may have rights, preferences, and privileges senior to those of our common stock. If we seek strategic alliances, licenses, or other alternative arrangements, such as arrangements with collaborative partners or others, we may need to relinquish rights to certain of our existing or future technologies, product candidates, or products we would otherwise seek to develop or commercialize on our own, or to license the rights to our technologies, product candidates, or products on terms that are not favorable to us. The overall status of the economic climate could also result in the terms of any equity offering, debt financing, or alliance, license, or other arrangement being even less favorable to us and our stockholders than if the overall economic climate were stronger. We also will continue to look for government sponsored research collaborations and grants to help offset future anticipated losses from operations and, to a lesser extent, interest income.

If adequate funds are not available through either the capital markets, strategic alliances, or collaborators, we may be required to delay or, reduce the scope of, or terminate our research, development, clinical programs, manufacturing, or commercialization efforts, or effect additional changes to our facilities or personnel, or obtain funds through other arrangements that may require us to relinquish some of our assets or rights to certain of our existing or future technologies, product candidates, or products on terms not favorable to us.

Off-Balance Sheet Arrangements

We do not utilize off-balance sheet financing arrangements as a source of liquidity or financing.

© Edgar Online, source Glimpses