The following discussion and analysis of our financial condition and results of operations This discussion contains forward-looking statements that involve risks and uncertainties. Our actual results may differ materially from those discussed in forward-looking statements. Factors that might cause a difference include, but are not limited to, those discussed above under "Cautionary Note Regarding Forward-Looking Statements," and in Item 1A. Risk factors in our Annual Report on Form 10-K for the fiscal year ended December 31, 2021.

Strategic and Clinical Overview

Celsion Corporation ("Celsion" or 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. ThermoDox®, Celsion's proprietary heat-activated liposomal encapsulation of doxorubicin, currently under investigator-sponsored development for several cancer indications, is being managed though Celsion's wholly owned subsidiary, Celsion GmbH. Additionally, 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.



23





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.

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 20,000 new cases of ovarian cancer in the U.S. in 2021 with an estimated 13,000 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. The five-year survival rates for Stages III and IV are 39% and 17%, respectively. First-line chemotherapy regimens are typically platinum-based combination therapies. Although this first line of treatment has an approximate 80% response rate, 55% to 75% 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% 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.



24

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"). 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 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.


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.



25

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 29, 2021, the Company announced final progression free survival ("PFS") results from the OVATION I Study published in the Journal of Clinical Cancer Research. Median PFS in patients treated per protocol (n=14) was 21 months and was 18.4 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 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.



26





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
                                                           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 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 before the end of the third quarter of 2022. Because this is an open-label study, the Company has provided clinical updates throughout the course of treatment including response rates and surgical resection scores.

In February 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 and also 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. Of the 34 patients enrolled in the trial, 27 patients have had their interval debulking surgery with the following results:



  ? 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.

  ? 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.


In June 2022, the Company announced that following a pre-planned interim safety review of 87 as treated patients (46 patients in the experimental arm and 41 patients in the control arm) randomized in the OVATION 2 Study, the Data Safety Monitoring Board (DSMB) unanimously recommended that the OVATION 2 Study continue treating patients with the dose of 100 mg/m2. The DSMB also determined that safety is satisfactory with an acceptable risk/benefit, and that patients tolerate GEN-1 during a course of treatment that lasts up to six months. No dose-limiting toxicities were reported. Interim clinical data from patients who have undergone interval debulking surgery showed that the GEN-1 treatment arm is continuing to show improvement in R0 surgical resection rates and CRS 3 chemotherapy response scores over the control arm. A complete tumor resection (R0) is a microscopically margin-negative resection in which no gross or microscopic tumor remains in the tumor bed. The chemotherapy response score is a three-tier standardized scoring system for histological tumor regression into complete/near complete (CRS 3), partial (CRS 2) and no/minimal (CRS 1) response based on omental examination.



27





Through July 31, 2022, over 90% of the patients have been enrolled in the OVATION 2 study. To date no patient in the treatment arm of the phase 2 portion of the trial has received all 17 doses of the GEN-1 treatment as prescribed in the study protocol. Implications will be assessed in conjunction with the primary end point, PFS, results.

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 TheraPlas platform technology.

Celsion's PLACCINE DNA vaccine technology platform is characterized by a single multi-cistronic DNA plasmid vector expressing multiple pathogen antigens delivered with a synthetic delivery system. We believe it is 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 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.

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 vaccine candidate comprises a single plasmid vector containing the DNA sequence encoding multiple SARS-CoV-2 antigens. 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. 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.



28





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. 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.

  ? Durable Efficacy: PLACCINE delivers a DNA plasmid-based antigen that could
    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.

On September 2, 2021, the Company announced results from preclinical in vivo studies showing production of antibodies and cytotoxic T-cell response specific to the spike antigen of SARS-CoV-2 when immunizing BALB/c mice with the Company's next-generation PLACCINE DNA vaccine platform. Moreover, the antibodies to SARS-CoV-2 spike antigen prevented the infection of cultured cells in a viral neutralization assay. The production of antibodies predicts the ability of PLACCINE to protect against SARS-CoV-2 exposure, and the elicitation of cytotoxic T-cell response shows the vaccine's potential to eradicate cells infected with SARS-CoV-2. These findings demonstrate the potential immunogenicity of Celsion's PLACCINE DNA vaccine, which is intended to provide broad-spectrum protection and resistance against variants by incorporating multiple viral antigens, to improve vaccine stability at storage temperatures of 4o C and above, and to facilitate cheaper and easier manufacturing.

On January 31, 2022, the Company announced it had engaged BIOQUAL, Inc., a preclinical testing contract research organization, to conduct a non-human primate (NHP) challenge study with Celsion's DNA-based approach for a SARS-CoV-2 vaccine. The NHP pilot study follows the generation of encouraging mouse data and will evaluate the Company's lead vaccine formulations for safety, immunogenicity and protection against SARS-CoV-2. In completed preclinical studies, Celsion demonstrated safe and efficient immune responses including IgG response, neutralizing antibodies and T-cell responses that parallel the activity of commercial vaccines following intramuscular (IM) administration of novel vaccine compositions expressing a single viral antigen. In addition, vector development has shown promise of neutralizing activity against a range of SARS-CoV-2 variants. Celsion's novel DNA-based vaccines have been based on a simple intramuscular injection that does not require viral encapsulation or special equipment for administration.

In April 2022, the Company presented its PLACCINE platform technology at the 2022 World Vaccine Congress. In an oral presentation during a Session on Cancer and Immunotherapy, Dr. Khursheed Anwer, Celsion's Chief Science Officer, highlighted the Company's technology platform in his presentation entitled: "Novel DNA Approaches for Cancer Immunotherapies and Multivalent Infectious Disease Vaccines." PLACCINE is demonstrating the potential to be a powerful platform that provides for rapid design capability for targeting two or more different variants of a single virus in one vaccine. There is a clear public health need for vaccines today that address more than one strain of viruses, like COVID-19, which have fast evolving variant capability to offer the widest possible protection. Murine model data has thus far been encouraging and suggests that the Company's approach provides not only flexibility, but also the potential for efficacy comparable to benchmark COVID-19 commercial vaccines with durability to protect for more than 6 months.

In the murine model, our multivalent PLACCINE vaccine targeted against two different variants showed to be immunogenic as determined by the levels of IgG, neutralizing antibodies, and T-cell responses. Additionally, our multivalent vaccine was equally effective against two different variants of the COVID-19 virus while the commercial mRNA vaccine appeared to have lost some activity against the newer variant. The Company continues to evaluate our technology and look forward to the results from our ongoing proof-of-concept non-human primate study evaluating our PLACCINE vaccine against the challenge from live SARS-CoV-2 virus in the second quarter, with durability results available in the second half of this year.



29





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.

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").

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 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 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.

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.



30





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 is 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 to evaluate the use of ThermoDox for the treatment of various cancer. Following inquiries from the NIH, we renewed 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.

Business Plan

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. Food and Drug Administration. The Company has not generated significant revenue and has incurred significant net losses in each year since our inception. As of June 30, 2022, the Company has incurred approximately $xxx million of cumulative net losses. As of June 30, 2022, the Company had $48.1 million in cash and cash equivalents, short-term investments, interest receivable and restricted cash. The Company has substantial future capital requirements to continue its research and development activities and advance its 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.

In January 2020, the World Health Organization declared an outbreak of coronavirus, COVID-19, to be a "Public Health Emergency of International Concern," and the U.S. Department of Health and Human Services declared a public health emergency to aid the U.S. healthcare community in responding to COVID-19. This virus continues to evolve and may have an adverse effect on our operations and product development timelines. Uncertainty with respect to the economic impacts of the pandemic has introduced significant volatility in the financial markets. While the extent to which COVID-19 impacts the Company's future results will depend on future developments, the pandemic and associated economic impacts could result in a material impact to the Company's future financial condition, results of operations and cash flows.

The Company's ability to raise additional capital may be adversely impacted by potential worsening global economic conditions and the recent disruptions to, and volatility in, financial markets in the U.S. and worldwide resulting from the ongoing COVID-19 pandemic and the geopolitical turmoil caused by the war in Ukraine. These disruptions may have a negative impact on the Company's clinical trials process and enrollment of patients. The ongoing geopolitical turmoil caused by the war in Ukraine has contributed to highly volatile financial markets which may impact our ability to access the capital markets and rising levels of inflation which may impact our expenditures and supply chain. The Company continues to monitor its operating activities in light of these events, and it is reasonably possible that these events could have a negative effect on the Company's financial condition and results of operations. The specific impact, if any, is not readily determinable as of the date of the Financial Statements.



31





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;

  ? 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.

Since 2018, the Company has annually 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 New Jersey 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 cumulative New Jersey NOLs from 2011 to 2018 totaling $13 million and received net proceeds of $12.2 million. As part of the Technology Business Tax Certificate Program, the Company sold $1.5 million and $2.0 million of its New Jersey NOLs in 2021 and 2020, respectively. The sale of these net operating losses resulted in net proceeds to the Company of approximately $1.4 million in 2021 and $1.9 million in 2020. 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 funding program in future years for up to an additional $3.5 million in net operating losses under this maximum lifetime benefit. In June 2022, the Company filed an application with The New Jersey Economic Development Authority to sell $1.7 million of its New Jersey NOL's for the tax year 2021 which is expected to generate net proceeds to the Company of approximately $1.6 million.

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 24 months through July 2020, followed by a 21-month amortization period of principal and interest starting on August 1, 2020, and ending through the scheduled maturity date on April 1, 2023. 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 10 to the Financial Statements, in June 2021, the Company entered into a $10 million loan facility with Silicon Valley Bank ("SVB"). The Company immediately used $6 million from this facility to retire all outstanding indebtedness with Horizon and deposited $6 million with SVB as restricted cash as discussed in more detail in Note 4. The remaining $4 million under the SVB loan facility ("SVB Loan Facility") will be available to be drawn down up to 12 months after closing. Payments under the loan agreement are interest only for the first 24 months after loan closing, followed by a 24-month amortization period of principal and interest through the scheduled maturity date.

With $48.1 million in cash and cash equivalents, short-term investments, interest receivable and restricted cash, coupled with approximately $3.5 million of future planned sales of the Company's State of New Jersey net operating losses, the Company believes it has sufficient capital resources to fund its operations into 2025.

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 New Jersey NOLs 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.



32






Financing Overview

Equity, Debt and Other Forms of Financing

On March 19, 2021, the Company filed with the SEC a new $100 million shelf registration statement on Form S-3 (File No. 333-254515) (the "2021 Registration Statement") that allows the Company to issue any combination of common stock, preferred stock or warrants to purchase common stock or preferred stock. This shelf registration was declared effective on March 30, 2021.

During 2021 and 2022 through the date of this Quarterly Report filed on Form 10-Q, we issued a total of 4.4 million shares of common stock as discussed below for an aggregate $65.4 million in gross proceeds.



  ?  On December 4, 2018, the Company entered into the Capital on Demand Agreement
     with 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. During 2021, the Company sold 0.5
     million shares under the Capital on Demand Agreement, receiving approximately
     $6.9 million in gross proceeds under the Capital on Demand Agreement.

  ?  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 1,728,395
     shares of the Company's common stock at an offering price of $20.25 per share
     for gross proceeds of approximately $35 million before the deduction of the
     January 2021 Placement Agents (as defined below) fee and offering expenses.
     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 with A.G.P./Alliance Global Partners ("AGP" and
     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.

  ?  On March 31, 2021, the Company entered into a Securities Purchase Agreement
     (the "March 2021 Purchase Agreement") with several institutional investors,
     pursuant to which the Company agreed to issue and sell, in a registered
     direct offering (the "March 2021 Offering"), an aggregate of 769,230 shares
     of the Company's common stock, at an offering price of $19.50 per share for
     gross proceeds of approximately $15 million before the deduction of the
     placement agents fee and offering expenses. The shares were offered by the
     Company pursuant to the 2021 Registration Statement. The closing of the
     offering occurred on April 5, 2021.

     In connection with the March 2021 Offering, the Company entered into a
     placement agent agreement with AGP, as lead placement agent (together with
     JonesTrading Institutional Services LLC and Brookline Capital Markets, a
     division of Arcadia Securities, LLC, serving as co-placement agents, the
     "March 2021 Placement Agents"), pursuant to which the Company agreed to pay
     the March 2021 Placement Agents an aggregate cash fee equal to 7% of the
     aggregate gross proceeds raised from the sale of the securities sold in the
     offering and reimburse the Placement Agents for certain of their expenses in
     an amount not to exceed $82,500.

  ?  On January 10, 2022, the Company entered into the Preferred Stock Purchase
     Agreement with several institutional investors, pursuant to which the Company
     agreed to issue and sell, in the Preferred Offerings, (i) 50,000 shares of
     Series A Preferred Stock, and (ii) 50,000 shares of Series B Preferred Stock,
     in each case at an offering price of $285 per share, representing a 5%
     original issue discount to the stated value of $300 per share, for gross
     proceeds of each Preferred Offering of $14.25 million, or approximately
     $28.50 million in the aggregate for the Preferred Offerings, before the
     deduction of the Placement Agent's (as defined below) fee and offering
     expenses. The shares of Series A Preferred Stock had a stated value of $300
     per share and were convertible, at a conversion price of $13.65 per share,
     into 1,098,901 shares of common stock (subject in certain circumstances to
     adjustments). The shares of Series B Preferred Stock had a stated value of
     $300 per share and were convertible, at a conversion price of $15.00 per
     share, into 1,000,000 shares of common stock (subject in certain
     circumstances to adjustments). The closing of the Preferred Offerings
     occurred on January 13, 2022.

     The Company held a special meeting of stockholders to consider an amendment
     (the "Amendment") to the Company's Certificate of Incorporation, as amended,
     to effect a reverse stock split of the outstanding shares of common stock
     ("Common Stock") by a ratio to be determined by the Board of Directors of the
     Company (the "Reverse Stock Split"). The investors have agreed in the
     Purchase Agreement to not transfer, offer, sell, contract to sell,
     hypothecate, pledge or otherwise dispose of the shares of the Preferred Stock
     until the Reverse Stock Split, to vote the shares of the Series A Preferred
     Stock purchased in the Preferred Offerings in favor of such Amendment and to
     vote the shares of the Series B Preferred Stock purchased in the Preferred
     Offerings in a manner that "mirrors" the proportions on which the shares of
     Common Stock (excluding any shares of Common Stock that are not voted) and
     Series A Preferred Stock are voted on the Reverse Stock Split and the
     Amendment.



33







    The holders of Preferred Stock were entitled to dividends, on an as-if
    converted basis, equal to dividends actually paid, if any, on shares of Common
    Stock. The Preferred Stock was convertible into shares of Common Stock at a
    rate of $13.65 per share for the Series A Preferred Stock and $15.00 per share
    for the Series B Preferred Stock, subject to adjustment. The Preferred Stock
    was convertible at the option of the holder at any time after the Company had
    received stockholder approval for the Reverse Stock Split and filed the
    requisite Amendment with the Delaware Secretary of State's office to
    effectuate the Reverse Stock Split (the "Reverse Stock Split Date"), subject
    to beneficial ownership limitations set forth in the applicable Certificate of
    Designation. In addition, on or after the Reverse Stock Split Date, and
    subject to the satisfaction of certain conditions, the Company had the right
    to cause the holders of the Preferred Stock to convert their shares of
    Preferred Stock, subject to such beneficial ownership limitations.

    Each holder of the Preferred Stock had the right to cause the Company to
    redeem all or part of their shares of the Preferred Stock from the earlier of
    receipt of stockholder approval of the Reverse Stock Split or of 90 days
    following the original issue date until 120 days following the original issue
    date, the "Redemption Date," in cash at a redemption price equal to 105% of
    the stated value plus an amount equal to accumulated but unpaid dividends, if
    any, on such shares (whether or not earned or declared, but excluding interest
    on such dividends) up to, but excluding, the Redemption Date. In connection
    with the Preferred Offerings, the Company entered into a placement agent
    agreement (the "Placement Agent Agreement") with AGP in which the Company paid
    $1,000,000 as a placement agent fee and $110,000 to reimburse AGP for certain
    expenses related to the Preferred Stock offering.

    On March 3, 2022, the Company redeemed for cash at a price equal to 105% of
    the $300 stated value per share all of its 50,000 outstanding shares of Series
    A Preferred Stock and its 50,000 outstanding Series B Preferred Stock. As a
    result, all shares of the Preferred Stock have been retired and are no longer
    outstanding and Celsion's only class of outstanding stock is its common stock.
    Each share of common stock entitles the holder to one vote.

    The Series A Preferred Stock and Series B Preferred Stock were recorded as a
    liability on the condensed consolidated balance sheet during the first quarter
    of 2022 until the preferred shares were redeemed during the same quarter. The
    Company recognized $4,551,567 as interest expense for the preferred shares
    during the first quarter of 2022, which was composed of: (a) $3,000,000 as the
    difference between the redemption price for the preferred shares and the net
    proceeds received from the issuance of the preferred shares, (b) $1,110,000
    paid to AGP as a placement agent fee and reimbursement for certain expenses,
    and (c) $441,567 in legal fees recognized in the first quarter that were
    attributed to the preferred shares.

  ? On April 6, 2022, the Company entered into a Securities Purchase Agreement
    (the "April 2022 Purchase Agreement") with several institutional investors,
    pursuant to which the Company agreed to issue and sell, in a registered direct
    offering (the "April 2022 Offering"), an aggregate of 1,328,274 shares of the
    Company's common stock at an offering price of $5.27 per share for gross
    proceeds of $7.0 million before the deduction of the April 2022 Placement
    Agent (as defined below) fees and offering expenses. The April 2022 Purchase
    Agreement contains customary representations, warranties and agreements by the
    Company and customary conditions to closing. The closing of the April 2022
    Offering occurred on April 8, 2022.

    In connection with the April 2022 Offering, the Company entered into a
    placement agent agreement with A.G.P./Alliance Global Partners (the "April
    2022 Placement Agent") pursuant to which the Company agreed to pay the April
    2022 Placement Agent a cash fee equal to 6.5% of the aggregate gross proceeds
    raised from the sale of the securities sold in the April 2022 Offering and
    reimburse the April 2022 Placement Agent for certain of their expenses in an
    amount not to exceed $50,000.


Significant Accounting Policies

Our significant accounting policies are more fully described in Note 1 to our consolidated financial statements included in our 2021 Annual Report on Form 10-K for the year ended December 31, 2021 filed with the SEC on March 31, 2022. See Note 3 to the Condensed Consolidated Financial Statements contained in this Quarterly Report on Form 10-Q.

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 "Item 1A. Risk Factors" under "Part II: Other Information" included herein.



34





FINANCIAL REVIEW FOR THE THREE MONTHS ENDED JUNE 30, 2022 AND 2021

Results of Operations

For the three months ended June 30, 2022 our net loss was $6.0 million compared to a net loss of $5.4 million for the same three-month period of 2021.



With $48.1 million in cash and cash equivalents, short-term investments,
interest receivable and restricted cash, coupled with approximately $3.5 million
of future planned sales of the Company's State of New Jersey net operating
losses, the Company believes it has sufficient capital resources to fund its
operations into 2025.

                                                        Three Months Ended June 30,
                                                                            Change Increase
                                                  (In thousands)              (Decrease)
                                                 2022         2021
Licensing Revenue:                            $    125     $    125     $      -            - %

Operating Expenses:
Clinical Research                                1,505        1,210          295         24.4 %

Chemistry, Manufacturing and Controls (CMC) 1,721 1,383 338 24.4 % Research and development expenses

                3,226        2,593          633         24.4 %
General and administrative expenses              2,877        2,603          274         10.5 %
Total operating expenses                         6,103        5,196          907         17.5 %
Loss from operations                          $ (5,978 )   $ (5,071 )   $   (907 )       17.9 %



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 recorded deferred revenue of $125,000 in each of the second quarters of 2022 and 2021.

Research and Development Expenses

Research and development ("R&D") expenses were $3.2 million in the second quarter of 2022 compared $2.6 million in same period of 2021. Costs associated the OVATION 2 Study were $0.4 million in each of the second quarters of 2022 and 2021. Costs associated with the OPTIMA Study were $0.5 million in the second quarter of 2022 compared to $0.2 million in the same period of 2021. In July 2020, the Company unblinded the OPTIMA Study at the recommendation of the DMC to halt the study due to futility. Other clinical and regulatory costs were $0.7 million the second quarter of 2022 compared to $0.6 million in the same period of 2021. R&D costs associated with the development of GEN-1 to support the OVATION 2 Study as well as development of the PLACCINE DNA vaccine technology platform increased to $1.5 million in the second quarter of 2022 compared to $1.0 million in the same period of 2021. CMC costs decreased to $0.3 million in the second quarter of 2022 compared to $0.4 million in the same period of 2021.

General and Administrative Expenses

General and administrative expenses were $2.9 million in the second quarter of 2022 compared to $2.6 million in the same period of 2021. The increase of costs is associated with higher professional fees partially offset by less stock compensation costs.

Impairment of IPR&D Liability

Due to the continuing deterioration of public capital markets in the biotech industry and its impact on market capitalization rates in this section, IPR&D related to the ovarian cancer indication was reviewed for impairment during the second quarter of 2022. Based on the Company's analysis of the IPR&D, the Company has concluded that it is not more than likely that the asset had been impaired as of June 30, 2022. As such, no impairment charges for IPR&D related to the ovarian cancer indication were recorded during the second quarter of 2022.



35





Change in Earn-out Milestone Liability

As of June 30, 2022 and March 31, 2022, the Company fair valued the earn-out milestone liability at $5.4 million with no change recorded to the fair value of the earn-out milestone during the second quarter of 2022.

Investment income and interest expense

The Company recognized interest expense of $0.1 million in the second quarter of 2022 compared to $0.2 million in the same period of 2021. As more fully discussed in Note 10, in June 2021, the Company entered into a $10 million loan facility with Silicon Valley Bank. The Company immediately used $6 million from this facility to retire all outstanding indebtedness with Horizon Technology Finance Corporation. The remaining $4 million will be available to be drawn down up to 12 months after closing and will be used for working capital and to fund the advancement of the Company's product pipelines. In connection with this Horizon Technology Financing Facility, the Company incurred $0.2 million in interest expense in the second quarter of 2021 compared to $0.3 million during the same period of 2020. In connection with the termination of the Horizon Technology Financing Facility in the second quarter of 2021, the Company paid early termination and end of term charges to Horizon and recognized $0.2 million as a loss on debt extinguishment.

Investment income from its short-term investments was insignificant in the second quarter of 2021 and 2022.

FINANCIAL REVIEW FOR THE SIX MONTHS ENDED JUNE 30, 2022 AND 2021

Results of Operations

For the six months ended June 30, 2022 our net loss was $16.5 million compared to a net loss of $11.1 million for the same six-month period of 2021.



                                                     Six Months Ended June 30,
                                                                         Change Increase
                                           (In thousands)                   (Decrease)
                                         2022           2021
Licensing Revenue:                    $      250     $      250     $         -              - %

Operating Expenses:
Clinical Research                          2,826          2,364             462           19.5 %
Chemistry, Manufacturing and
Controls (CMC)                             3,496          2,801             695           24.8 %
Research and development expenses          6,322          5,165           1,157           22.4 %
General and administrative expenses        5,748          5,540             208            3.8 %
Total operating expenses                  12,070         10,705           1,365           12.8 %
Loss from operations                  $  (11,820 )   $  (10,455 )   $    (1,365 )         13.1 %



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 recorded deferred revenue of $250,000 in each of the first two quarters of 2022 and 2021.

Research and Development Expenses

Research and development ("R&D") expenses were $6.3 million in the first half of 2022 compared $5.2 million in same period of 2021. Costs associated the OVATION 2 Study were $0.8 million in the first half of 2022 compared to $0.9 million in the same period of 2021. Costs associated with the OPTIMA Study were $0.5 million in the first half of 2022 compared to $0.4 million in the same period of 2021. In July 2020, the Company unblinded the OPTIMA Study at the recommendation of the DMC to halt the study due to futility. Other clinical and regulatory costs were $1.5 million the first half of 2022 compared to $1.2 million in the same period of 2021. R&D costs associated with the development of GEN-1 to support the OVATION 2 Study as well as development of the PLACCINE DNA vaccine technology platform increased to $2.9 million in the first half of 2022 compared to $2.0 million in the same period of 2021. CMC costs decreased to $0.6 million in the first half of 2022 compared to $0.8 million in the same period of 2021.



36





General and Administrative Expenses

General and administrative expenses were $5.7 million in the first half of 2022 compared to $5.5 million in the same period of 2021. The increase of costs is associated with higher professional fees partially offset by less stock compensation costs.

Impairment of IPR&D Liability

Due to the continuing deterioration of public capital markets in the biotech industry and its impact on market capitalization rates in this section, IPR&D related to the ovarian cancer indication was reviewed for impairment during the first half of 2022. Based on the Company's analysis of the IPR&D, the Company has concluded that it is not more than likely that the asset had been impaired as of June 30, 2022. As such, no impairment charges for IPR&D related to the ovarian cancer indication were recorded during the first half of 2022.

Change in Earn-out Milestone Liability

As of June 30, 2022 and December 31, 2021, the Company fair valued the earn-out milestone liability at $5.4 million with no change recorded to the fair value of the earn-out milestone liability during the first half of 2022.

Investment income and interest expense

The Company recognized interest expense of $4.8 million in the first half of 2022. As more fully discussed in Notes 10 and 11 of the financial statements, the Company expensed $0.2 million in interest due to the Silicon Valley Bank loan facility and $4.6 million in interest attributed to the Series A Preferred Stock and Series B Preferred Stock during the first quarter of 2022.

As more fully discussed in Note 10, in June 2021, the Company entered into a $10 million loan facility with Silicon Valley Bank. The Company immediately used $6 million from this facility to retire all outstanding indebtedness with Horizon Technology Finance Corporation. The remaining $4 million will be available to be drawn down up to 12 months after closing and will be used for working capital and to fund the advancement of the Company's product pipelines. In connection with this Horizon Technology Financing Facility, the Company incurred $0.4 million in interest expense in the first half of 202. In connection with the termination of the Horizon Technology Financing Facility in the second quarter of 2021, the Company paid early termination and end of term charges to Horizon and recognized $0.2 million as a loss on debt extinguishment.

Investment income from its short-term investments was insignificant in the first half of 2021 and 2022.

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 $349 million at June 30, 2022.

At June 30, 2022, we had total current assets of $44.9 million and current liabilities of $6.9 million, resulting in net working capital of $38.0 million. At June 30, 2022, we had cash and cash equivalents, short-term investments, interest receivable on short term investments and money market investments ($6.0 million of which is included in other assets) of $48.1 million. At December 31, 2021 we had total current assets of $51.9 million and current liabilities of $6.8 million, resulting in net working capital of $45.1 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 the first six months of 2022 was $13.4 million. Net cash provided by investing activities was $14.2 million during the first six months of 2022. Cash provided by financing activities during the first six months of 2022 totaled $6.3 million. With $48.1 million in cash and cash equivalents, short-term investments, interest receivable and restricted cash, coupled with approximately $3.5 million of future planned sales of the Company's State of New Jersey net operating losses, the Company believes it has sufficient capital resources to fund its operations into 2025.



37





We expect to seek additional capital through further public or private equity offerings, debt financing, additional strategic alliance and licensing arrangements, collaborative arrangements, potential sales of our net operating losses, 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, collaborators, or sales of our net operating losses, 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 and Contractual Obligations

None.

© Edgar Online, source Glimpses