11/10/2025 | Press release | Distributed by Public on 11/10/2025 15:16
Management's Discussion and Analysis ofFinancial Condition and Results of Operations
The condensed consolidated financial statements and this Management's Discussion and Analysis of Financial Condition and Results of Operations should be read in conjunction with the consolidated financial statements and notes thereto for the year ended December 31, 2024 and the related Management's Discussion and Analysis of Financial Condition and Results of Operations, both of which are contained in our Annual Report on Form 10-K for the fiscal year ended December 31, 2024 filed with the U.S. Securities and Exchange Commission on March 20, 2025 (2024 Annual Report). In addition to historical information, this discussion and analysis contains forward-looking statements that involve risks and uncertainties. Our actual results could differ materially from those expressed or implied in any forward-looking statements as a result of various factors, including those set forth under "Part I. Item 1A. Risk Factors" in our 2024 Annual Report and "Part II. Item 1A. Risk Factors" in this report.
Overview
We are a biotechnology company developing innovative therapeutics targeting serious viral diseases with the potential to improve the lives of patients worldwide. Our pipeline includes multiple clinical-stage investigational therapies, including: (1) two helicase-primase inhibitors (HPIs) targeting herpes simplex virus (HSV) for the treatment of recurrent genital herpes; (2) an orally bioavailable hepatitis delta virus (HDV) entry inhibitor; and (3) a highly potent next-generation capsid assembly modulator (CAM) designed to disrupt the replication cycle of hepatitis B virus (HBV) at several key points. Our pipeline also includes a novel, oral broad-spectrum non-nucleoside polymerase inhibitor (NNPI) targeting transplant-related herpesviruses, which is currently undergoing studies to enable a regulatory filing, and we have additional research programs against multiple antiviral targets. We currently have corporate and administrative offices and research laboratory space in South San Francisco, California.
Our Clinical Programs and Regulatory Filing-Enabling Program
Since the beginning of 2024, we have initiated: (1) both the Phase 1a and Phase 1b portions of a Phase 1a/b study of ABI-5366 (5366), a long-acting HPI for the treatment of recurrent genital herpes; (2) both the Phase 1a and Phase 1b portions of a Phase 1a/b study of ABI-1179 (1179), also a long-acting HPI for recurrent genital herpes; (3) a Phase 1a study of ABI-6250 (6250), an orally bioavailable HDV entry inhibitor; and (4) a Phase 1b study of ABI-4334 (4334), a highly potent, next-generation CAM for chronic HBV infection.
We announced interim results from the Phase 1a portion of the 5366 study in September 2024 and interim results from the Phase 1b portion of the 5366 study in August 2025, interim results from the Phase 1a portion of the 1179 study in February 2025, interim results from the Phase 1a study of 6250 in August 2025 and topline results from the Phase 1b study of 4334 in June 2025.
In addition, in December 2024, we identified a development candidate, ABI-7423 (7423), in our broad-spectrum NNPI program targeting transplant-associated herpesviruses. 7423 is a prodrug, and in October 2025, we transitioned our discovery and development from 7423 to its parent molecule, ABI-7272 (7272), which is currently in regulatory filing-enabling preclinical studies.
Recurrent Genital Herpes/HSV-1 and HSV-2
Genital herpes can be caused by either HSV type 1 (HSV-1) or HSV type 2 (HSV-2). HSV-1 and HSV-2 are acquired by oral or genital contact either during symptomatic or asymptomatic reactivation of the virus. Both viruses replicate in neurons, where they can remain latent for the rest of the individual's life and periodically reactivate, with the virus spreading, replicating and causing disease in epithelial tissues. Initial infection can be asymptomatic or can be marked by serious symptoms, including painful skin lesions, swelling of lymph nodes and urinary problems that can persist for two to three weeks. While genital herpes can be caused by either HSV-1 or HSV-2, recurrences are more likely to be experienced by individuals infected by HSV-2. Genital herpes recurrence can cause painful genital lesions that can lead to increased transmission and debilitate patients, and symptoms may become more serious with additional episodes. Additional complications include increased risk of HIV infection, as 30% of HIV infections acquired through sexual transmission are attributable to HSV-2 infection. In addition, people with recurrent genital herpes often experience associated psychosocial impacts, including anxiety, concerns about transmission, depression and social stigma. Immunocompromised individuals may experience more severe and prolonged symptoms due to increased recurrence rates.
HPIs are antiviral agents in development for the treatment of recurrent genital herpes, with a clinically-validated mechanism of action. HPIs inhibit the HSV helicase-primase complex, which is a unique viral enzyme complex without a human homolog, consisting of helicase, primase and cofactor subunits. These subunits have functions that
are essential for HSV DNA replication and are conserved across HSV-1 and HSV-2. Unlike nucleoside analogs, these compounds do not require phosphorylation by the HSV thymidine kinase (TK) and ongoing viral replication to become active drugs. As a result, HPIs are active immediately upon reactivation of latent HSV-1 and HSV-2. Furthermore, HPIs are active against TK-deficient HSV-1 and HSV-2, which is a major mechanism of resistance to nucleoside analogs.
Most people with initial symptomatic genital herpes who are infected with HSV-2 have frequent recurrences, generally between three and 15 per year. This includes an estimated population of over four million people in the United States and France, Germany, Italy and Spain (collectively, the EU4) and the United Kingdom (UK). In addition, over eight million people have been diagnosed with genital herpes, and it is estimated that there are over 60 million people living with HSV-2 infection. Currently, there are three antiviral drugs (all nucleoside analogs) that have been approved in the United States and the EU4/UK for the treatment of genital herpes. However, no new drugs have been approved in these regions to treat genital herpes for more than 25 years. In addition to the approved nucleoside analogs, agents such as local anesthetics or analgesics may be used to alleviate local symptoms of minor pain and discomfort.
Nucleoside analogs can be administered as episodic therapy as individual outbreaks arise or daily as chronic suppressive therapy for those with high post-exposure recurrences. However, these agents are only partially effective at controlling the infection or reducing transmission risk. With current nucleoside analog therapies, only one out of three people with recurrent genital herpes with six or more recurrences per year are able to make it through a year of treatment without a recurrence. There are still high titer (greater than 104HSV-2 DNA copies/mL) shedding episodes under this current standard of care for recurrent genital herpes, which can lead to recurrent episodes and transmission of genital herpes. In addition, nucleoside analogs also carry a high pill burden as a lifelong daily treatment, with doses ranging from one to three times daily. There is also high treatment variability among those taking nucleoside analogs, as many seeking care may not consistently receive suppressive therapy.
Based on the limitations of current therapies, we see a path to advancing the treatment paradigm for people suffering from recurrent genital herpes. To reach that goal, we discovered and are developing a novel, potent, long-acting HPI for recurrent genital herpes, 5366, which demonstrated low nanomolar potency in vitro against both HSV-1 and HSV-2 clinical isolates and a favorable nonclinical safety profile in the U.S. Food and Drug Administration's (FDA) Good Laboratory Practice (GLP) toxicology studies. In addition, we are developing a second novel, potent, long-acting HPI for genital herpes, 1179, which was exclusively in-licensed to us as part of our collaboration with Gilead, and also has demonstrated low nanomolar potency in vitro against both HSV-1 and HSV-2 clinical isolates and a favorable nonclinical safety profile in the FDA's GLP toxicology studies.
In September 2024, we announced positive interim data for the Phase 1a portion of a Phase 1a/b clinical study of 5366, and in April 2025, we announced additional data for the Phase 1a portion of the study at the 2025 Congress of the European Society of Clinical Microbiology and Infectious Diseases and an encore presentation at the STI & HIV World Congress in July 2025. Interim results exceeded our objectives for this Phase 1a study and supported 5366's progression into the Phase 1b portion of the study, which we initiated during the third quarter of 2024. Across the Part A (Phase 1a) cohorts evaluated, 5366 had a mean half-life of approximately 20 days when dosed orally, supporting once-weekly oral dosing, the target profile for 5366, as well as showing the potential for once-monthly oral dosing. We are exploring both once-weekly and once-monthly oral dosing regimens in the Part B (Phase 1b) portion of the study.
In the Part A cohorts, 5366 has been well-tolerated with a favorable safety profile observed when administered orally up to 350 mg, with exposure up to 98 days. Treatment-emergent adverse events (AEs) were all mild to moderate in intensity and all were considered not related to study treatment by the study investigators; there have been no serious AEs in any dose arm. There were no treatment-related Grade 3 or 4 laboratory abnormalities and no protocol-defined stopping criteria were met. There were no clinically significant electrocardiogram (ECG) abnormalities or patterns of AEs or laboratory abnormalities noted.
We reported interim data from the Phase 1b portion of the 5366 study in August 2025. For the powered antiviral endpoint, HSV-2 shedding rate, highly potent antiviral activity was observed with a 94% reduction compared to placebo (p<0.01) over the 29-day evaluation period in the cohort evaluating a 350 mg weekly dose. This reduction exceeded our target for the study of an 80%-85% reduction in the rate of HSV-2 shedding. For a secondary clinical endpoint of genital lesion rate, a 94% reduction compared to placebo (p<0.01) was observed with the 350 mg weekly dose. The rate of samples with high viral load (i.e., >104copies/mL HSV DNA), a potential surrogate for HSV-2 transmission and a secondary endpoint, was reduced by 98% compared to placebo (p<0.05) in this cohort.
5366 was observed to be well-tolerated at once-a-week oral doses up to 350 mg in participants seropositive for HSV-2 with recurrent genital herpes. Of the treatment-emergent AEs reported, the majority were Grade 1 or Grade 2. One grade 3 AE was reported, hypertriglyceridemia, in a participant with relevant medical history who had Grade 4
elevated triglycerides pre-dose on Day 1. This AE resulted in study discontinuation but was not considered treatment related. There were three participants with treatment-emergent Grade 3 laboratory abnormalities, all considered unrelated to assigned treatment: an exercise-associated elevation in creatine kinase, a decrease in neutrophils and an elevation of cholesterol in the follow-up period in a participant that had a grade 2 elevation at baseline. There did not appear to be a dose-response relationship in either the frequency or severity of treatment-emergent AEs or laboratory abnormalities. There have been no serious AEs reported to date.
Enrollment for the Phase 1b portion of the 5366 study is complete. The observed nonclinical pharmacokinetic PK profile continues to support once-weekly dosing and the potential for once-monthly oral dosing regimens. With these data, we expect to move directly into Phase 2 clinical study preparation for a once-weekly treatment regimen in parallel with completion of this Phase 1b study, which includes an ongoing cohort evaluating a monthly oral dosing regimen. The in-life portions of chronic toxicology studies of 5366 are now complete and these studies are expected to support longer-term dosing in Phase 2.
A late-breaking oral presentation of the interim Phase 1b data reported in August 2025 was presented at the 38thCongress of the International Union Against Sexually Transmitted Infections - Europe, which took place in October 2025 in Athens, Greece.
In addition to 5366, we are also advancing 1179, a structurally-differentiated HPI with single digit nanomolar potency against HSV-1 and HSV-2 and a PK and safety profile to date that is supportive of a potential long-acting treatment by once-weekly oral administration. We submitted the Clinical Trial Application for a Phase 1a/b study of 1179 in September 2024, which was approved in October 2024. We dosed our first participant in the Phase 1a portion of this study during the fourth quarter of 2024.
In February 2025, we announced positive interim data for the Phase 1a portion of the 1179 study, and in July 2025, we announced additional data for the Phase 1a portion of the study at the STI & HIV 2025 World Congress. Interim results exceeded our objectives for this Phase 1a study and support 1179's progression into Phase 1b, and we dosed our first participant in the Phase 1b portion of the study in participants with recurrent genital herpes during the second quarter of 2025. We submitted an Investigational New Drug (IND) application in May 2025 to support expansion of the Phase 1b study to sites in the United States, and we received clearance for the IND in June 2025.
In all three single-dose Phase 1a cohorts completed to date (50 mg, 100 mg and 300 mg), 1179 has been well-tolerated with a favorable safety profile observed. Treatment-emergent AEs were generally Grade 1 in intensity and all were considered not related to study treatment by the study investigators; there have been no serious AEs in any dose cohort. A single, self-limited Grade 2 alanine transaminase (ALT) elevation was observed in a subject receiving the highest dose of 300 mg. There were no clinically significant ECG abnormalities, no significant treatment-related laboratory abnormalities and no protocol-defined stopping criteria were met. Plasma concentrations exceeded necessary protein-adjusted EC50 for inhibition of HSV replication at all dose levels and the observed half-life of approximately four days supports weekly dosing, the target profile for 1179.
The Phase 1b study of 1179 is ongoing concurrently with the ongoing Phase 1b evaluation of 5366, our other long-acting HPI candidate that began dosing in Phase 1b in the fourth quarter of 2024. Enrollment for two cohorts of the Phase 1b portion of the 1179 study is complete. We plan to report interim data on two cohorts of weekly dosing of 1179 and one cohort of monthly dosing of 5366 by the end of 2025, which are expected to be presented together once all virology analyses are completed. The completion of the Phase 1b studies of both 5366 and 1179 constitute an option triggering point under the terms of the Gilead Collaboration Agreement, as defined below under "Collaboration and License Agreement - Gilead Sciences, Inc."
Our HBV and HDV Programs
The World Health Organization (WHO) estimates that 254 million people worldwide are chronically infected with HBV as of 2022, and 1.2 million new infections occur each year. HBV is a leading global cause of chronic liver disease and liver transplants, and the WHO estimates that 1.1 million people died in 2022 from HBV, mostly due to cirrhosis and hepatocellular carcinoma. As of 2022, only approximately 33 million, or 13%, of those chronically infected with HBV, were aware of their infection, and only approximately 7 million, or 3%, of those diagnosed received treatment. HBV is a highly prevalent disease that infects almost three times the number of people infected with hepatitis C virus and HIV infections combined, according to the WHO.
The current standard of care for chronic HBV infection, nucleos(t)ide analog reverse transcriptase inhibitors (NrtIs), are taken life-long and reduce, but do not eliminate, the virus and result in very low cure rates. No new mechanisms of action (MOA) have been approved for the treatment of chronic HBV infection in over 25 years. The focus of our
HBV program is to improve outcomes and increase the number of patients diagnosed and treated through the development of finite and curative therapies targeting an orthogonal MOA.
HDV is a "satellite virus" of HBV because it can only infect people (1) who are already infected with HBV or (2) at the same time as a person is infected with HBV. HDV affects a subset of approximately 12 to 72 million HBV infected people. These individuals infected with HDV, which comprise an estimated 4.5% of hepatitis B surface antigen (HBsAg) positive individuals, experience a substantially increased disease burden, as they account for 18% of cirrhosis and 20% of hepatocellular carcinoma associated with HBV. HDV is considered the most severe form of hepatitis, as 70% of individuals infected with HDV progress to cirrhosis within ten years. While HDV is less prevalent in the United States, it is a significant and serious health problem with inadequate treatment in many parts of Europe, Africa, the Middle East, East Asia and parts of South America. HDV may be significantly underdiagnosed, because there were no HDV-targeted therapies approved until very recently, and the first therapy approved is only approved in Europe. HDV is known to accelerate disease progression and increase the incidence of liver cirrhosis and liver cancer, which results in higher morbidity and mortality rates than HBV alone.
The current standard of care treatment for HDV is off-label pegylated interferon-α (IFN-α) injected weekly or, in Europe, a large, complex peptide inhibitor that requires daily injections, bulevirtide. There are no approved HDV treatments in the United States, and bulevirtide is the only approved HDV treatment in Europe. We believe a safe and effective oral small molecule entry inhibitor would be a significant innovation for people living with HDV, who face a significant and immediate disease burden.
HDV Entry Inhibitor
HDV is a small RNA virus that encodes just two viral proteins and relies on host enzymes as well as the HBsAg from HBV to replicate, which limits the number of HDV-specific antiviral targets. Similar to HBV, HDV utilizes HBsAg to enter hepatocytes by binding the cellular transmembrane protein sodium taurocholate co-transporting peptide (NTCP). NTCP is highly expressed on human hepatocytes, where it serves as one of several proteins involved in the transport of bile acids. The binding of specific small or large molecules to NTCP has been shown to effectively inhibit the interaction of HBsAg with NTCP, which prevents HBV and HDV from infecting hepatocytes.
The inhibition of HBV and HDV infection by molecules that bind NTCP has been demonstrated in vitro, in animal models and clinically. Notably, bulevirtide, a peptide blocker of NTCP, is the only approved therapy for HDV (approved in Europe). Our novel small molecule entry inhibitor, 6250 uses the same clinically-validated MOA as bulevirtide. The binding of NTCP-targeted HBV/HDV entry inhibitors to NTCP has also been shown to inhibit the transport of certain bile acids into cells, which results in plasma elevations of bile acids; this effect has been well-tolerated clinically and may serve as a biomarker of pharmacologically active concentrations of drug in the plasma. In nonclinical studies in non-human primates, clinically-relevant doses of 6250 elevated bile acids to levels similar to those seen in humans with bulevirtide.
We believe a safe and effective oral small molecule entry inhibitor would be a significant innovation for people living with HDV and could significantly improve treatment uptake and diagnosis rates, especially when compared with currently available injectable products.
In September 2023, we nominated 6250, a novel, orally bioavailable small molecule designed to inhibit entry of HBV and HDV by targeting NTCP. In nonclinical studies, 6250 demonstrated low nanomolar potency against all tested HBV/HDV genotypes, favorable selectivity for NTCP versus other bile acid transporters, good oral bioavailability and a PK profile in nonclinical species projected to support once-daily oral dosing. In GLP toxicology studies, 6250 demonstrated a favorable nonclinical safety profile with wide safety margins.
At the European Association for the Study of the Liver's International Liver CongressTM in May 2025 and an encore presentation at the 2025 International HBV meeting in September 2025, 6250 was featured in a poster presentation highlighting nonclinical data supporting the advancement of 6250 into an ongoing Phase 1a clinical study, which was initiated during the fourth quarter of 2024.
In August 2025, we announced interim PK, biomarker and safety data from single-ascending and multiple-ascending doses cohorts in healthy participants. Across the cohorts evaluated to date, a mean half-life of approximately four days was observed for 6250 when dosed orally, supporting the once-daily oral dosing profile target. Given this half-life, accumulation was observed in the multiple-dose cohorts with exposures on the last day of dosing generally reaching six- to seven-fold higher than the exposure seen after the first dose.
Dose-dependent elevations of total serum bile acids (TBAs) were observed for both the 5 mg and 25 mg single-dose cohorts, indicative of NTCP target engagement. In the highest single-dose cohort of 25 mg, coproporphyrin I (CP-1), a biomarker for off-target engagement of the organic anion transporters, OATP1B1 and/or OATP1B3, was also elevated. CP-1 elevation was not noted at the other doses.
Given the predicted 6250 accumulation driven by the long half-life and the observed elevations TBAs for the single-dose cohorts, doses at and below 1 mg daily were selected for the multiple-dose cohorts to characterize the lower end of the dose-response curve. Elevation of TBAs was observed for both the 0.2 mg and 1 mg daily multiple-dose cohorts, consistent with the respective 6250 exposures. Minimal TBA elevation was observed in the 0.05 mg daily multiple-dose cohort.
Treatment-emergent AEs and laboratory abnormalities were all Grade 1 or 2 in severity with the majority being Grade 1. There were no serious AEs in any dose cohort. No protocol defined stopping criteria were met. There were no clinically significant ECG abnormalities or patterns of AEs noted.
One Grade 2 ALT elevation was observed in the cohort evaluating the highest single-dose level of 25 mg. In this cohort, off-target engagement of other liver transporters was also seen as indicated by elevated CP-1 levels. Grade 1 ALT elevations were observed at a low frequency across the other cohorts. All ALT elevations were self-limited, and none were accompanied with elevations in bilirubin or other markers of liver injury.
The elevations resolved in the study period with ongoing drug exposure due to 6250's four-day half-life. We have completed enrollment and the follow-up period in the Phase 1a study, and we are preparing for Phase 2 clinical studies in parallel with the completion of ongoing chronic toxicology studies.
Capsid Assembly Modulator
HBV is a DNA virus that infects hepatocytes and establishes a reservoir of covalently closed circular DNA (cccDNA), a unique viral DNA species that resides in the nucleus of HBV-infected hepatocytes and is associated with viral persistence and chronic infection. No currently approved oral therapies target cccDNA activity directly. As a result, we have worked to discover and develop compounds targeting the core protein, a viral protein involved in numerous aspects of the HBV replication cycle, including the generation of HBV cccDNA.
A benchmark for therapeutic agents aiming to decrease cccDNA levels is the use of several key viral antigens as surrogate biomarkers of active cccDNA. The same biomarkers can be used in both primary human hepatocytes and infected individuals. On this basis, our next-generation CAM, 4334, has shown nonclinical proof of principle. In a variety of cell culture models, 4334 has demonstrated the ability to reduce production of HBV DNA levels as well as the surrogate markers for cccDNA establishment: HBV e antigen (HBeAg), HBV core-related antigen (HBcrAg) and HBV pre-genomic RNA (pgRNA).
As a next-generation CAM, 4334 was optimized to potently disrupt viral replication (MOA #1) and prevent the establishment and replenishment of new cccDNA (MOA #2). In contrast, while they are active against MOA #1, first-generation CAMs have not demonstrated adequate potency to sufficiently block cccDNA formation (MOA #2). Further, the current standard of care, NrtIs, impacts the viral replication cycle after establishment of cccDNA and can only inhibit production of new viral particles, and they do so incompletely. The chemical scaffold of 4334 is novel and distinct from all our prior CAM candidates.
We believe that 4334 has a best-in-class nonclinical profile, with single-digit nanomolar potency against MOA #1 and MOA #2, pan-genotypic activity, an improved resistance profile and a favorable safety profile. Through mechanistic studies presented at multiple conferences, we have demonstrated that 4334 promotes the formation of empty capsids by acceleration of capsid assembly, prevents the formation of cccDNA by disrupting incoming capsids, and prematurely disrupts capsids containing duplex linear DNA, the precursor for integrated HBV DNA.
A Phase 1a study demonstrated that 4334 was well-tolerated when administered orally as single or multiple doses. During the second quarter of 2024, we dosed our first participant in a Phase 1b clinical study of 4334. We reported interim clinical results from the initial 150 mg cohort in December 2024 and topline clinical results including a subsequent 400 mg cohort in June 2025. In both the 150 mg and 400 mg cohorts, 4334 continued to show a half-life supportive of once-daily oral dosing. In addition, results for both cohorts indicated that 4334 maintained clinical exposures multiple folds above those anticipated to be required for potent viral activity and inhibition of cccDNA formation. Mean declines in HBV DNA of 2.9 log10IU/mL and 3.2 log10IU/mL were observed over 28 days in a population of predominately HBeAg negative participants receiving 150 mg and 400 mg, respectively. Among the subset of participants with detectable HBV RNA at baseline, mean declines of 2.5 log10 U/mL and 2.3 log10U/mL were observed over 28 days in the participants receiving 150 mg and 400 mg, respectively. As anticipated, limited
changes in viral antigens were observed for the study population over the 28-day treatment period. These antiviral data are consistent with the high potency seen preclinically for 4334. The safety data also demonstrated that 4334 was well-tolerated with a favorable safety profile observed. The 400 mg cohort was the final cohort for this Phase 1b study and final data will be presented at the American Association for the Study of Liver Disease, The Liver Meeting® in November 2025. The completion of the Phase 1b study of 4334 constitutes an option triggering point under the terms of the Gilead Collaboration Agreement, as defined below under "Collaboration and License Agreement - Gilead Sciences, Inc."
Transplant-Associated Herpesviruses
In a transplant setting, when patients are experiencing immunosuppression, they are at high risk of uncontrolled viral replication and severe disease brought on by one or more herpesviruses, including cytomegalovirus (CMV), HSV-1, HSV-2, varicella zoster virus (VZV) and Epstein-Barr virus (EBV). Each of these herpesviruses are highly prevalent, as approximately (1) 60% of transplant patients are CMV-positive; (2) 60% of transplant patients are HSV-positive; (3) 80% of transplant patients are VZV-positive and (4) 45% of transplant patients are EBV-positive. These viruses establish lifelong latent infections and frequently reactivate in transplant patients due to the use of immunosuppressive drugs following transplantation. These uncontrolled viral infections increase the risk of severe disease and serious complications, including organ rejection, graft loss and death, and impacted approximately 95,000 people receiving transplants in 2021 in the United States and Europe.
While there are approved antivirals that are administered in a transplant setting, currently approved antivirals are only partially efficacious, not active against a broad spectrum of transplant-associated herpesviruses and pose the risk of potentially serious side effects and drug-drug interactions. As a result of these limitations, we identified an opportunity to develop an oral, broad-spectrum NNPI for transplant-associated herpesvirus infections, which could greatly improve efficacy and treatment.
In December 2024, we nominated 7423, a prodrug, as our development candidate to undergo regulatory filing-enabling studies, which are ongoing. In October 2025, we transitioned our discovery and development from 7423 to nominate its parent molecule, 7272, which is currently in regulatory filing-enabling preclinical studies.
Research Programs
In addition to our investigational therapy programs that have nominated development candidates and have advanced into clinical studies or regulatory filing-enabling studies, our research team continues to actively focus on proprietary research to discover and develop novel antivirals to treat serious viral diseases.
Collaboration and License Agreement
Gilead Sciences, Inc.
In October 2023, we entered into an Option, License and Collaboration agreement (the Gilead Collaboration Agreement) with Gilead pursuant to which Gilead (1) exclusively licensed to us its HPI program and its NNPI program, while retaining opt-in rights to these programs and (2) has an option to take an exclusive license, on a program-by-program basis, to all of our other current and future pipeline programs. During the 12-year collaboration term (subject to payment of certain extension fees) and for a specified period thereafter, Gilead may exercise its opt-in rights, on a program-by-program basis, at one of two timepoints-completion of a certain Phase 1 study or completion of a certain Phase 2 study for the first product within the program-upon payment of an opt-in fee ranging from $45.0 million to $125.0 million per program depending on the type of program and when the option is exercised. Pursuant to the Gilead Collaboration Agreement, Gilead made an $84.8 million upfront cash payment to us. In December 2024, we and Gilead entered into the First Amendment to the Gilead Collaboration Agreement, which restructured the timing of specific options exercisable and the fees payable to us under the terms of the Gilead Collaboration Agreement due to an agreed upon development plan for 6250. To facilitate this development plan, (1) we received a payment of $10.0 million from Gilead and (2) the opt-in fee payable by Gilead in connection with 6250 was restructured, though it remains in the range of opt-in fees detailed above. The $10.0 million payment received in connection with the First Amendment to the Gilead Collaboration Agreement is creditable towards future collaboration-related payments payable by Gilead.
If Gilead exercises its opt-in right to any current or future program under the collaboration, we are eligible to receive up to $330.0 million in potential regulatory and commercial milestones on that program, in addition to royalties ranging from the high single-digits to high teens, depending on the clinical stage of the program at the time of the opt-in. Following Gilead's exercise of its option for each program, we may opt in to cover 40% of the research and development costs in the United States and share 40% of the profits and operating loss in the United States for products
within the program in lieu of receiving milestones and royalties for that program in the United States, unless we later opt out of the cost/profit share for the program. Prior to Gilead's potential exercise of its opt-in, we are primarily responsible for all discovery, research and development on both our programs and the two Gilead-contributed programs. Following Gilead's opt-in, Gilead will control the further discovery, research, development, and commercialization on any optioned programs. During the term, Gilead will continue to support the collaboration through extension fees of $75.0 million in each of the third, fifth and seventh anniversaries of the collaboration.
The Gilead Collaboration Agreement is subject to termination by either party for the other party's uncured, material breach or insolvency. Subject to certain limitations, we and Gilead both have certain termination for convenience rights, upon sufficient prior written notice, with respect to programs that one party in-licenses from the other (subject to Gilead's option rights), and with respect to Gilead, for programs it has option rights to (subject to certain time limitations with respect to existing Company programs). Gilead also has a right to terminate the collaborative activities under the Gilead Collaboration Agreement at certain specified points during the collaboration term. Other customary termination rights are further provided in the Gilead Collaboration Agreement.
We and Gilead also entered into a Common Stock Purchase Agreement and an Investor Rights Agreement (together, the Gilead Equity Agreements), which were both amended in June 2024 in connection with a financing transaction, in which a new investor purchased shares of common stock and was issued a warrant (the 2024 Financing Transaction). Pursuant to the Gilead Equity Agreements, Gilead made an upfront equity investment of $15.2 million by purchasing from us 1,089,472 shares of our common stock at a purchase price of $13.92 per share. The terms of the Gilead Equity Agreements provided Gilead the right to elect to purchase additional shares of common stock from us at a premium in an amount that results in Gilead owning 29.9% of our then-outstanding voting common stock. This right was exercised in December 2024, at a purchase price of $21.37 per share, which represents a 35% premium to the 30-trading day volume weighted average price immediately prior to the date of purchase. The Gilead Equity Agreements also include a three-year standstill provision and a two-year lockup provision, each with customary exceptions, and provide Gilead with certain other stock purchase rights and registration rights, as well as the right to designate two directors (or, alternatively, board observers at Gilead's election) to our board of directors. In December 2023, Gilead designated Tomas Cihlar, Ph.D. to serve on our board of directors, and in March 2024, Gilead designated Robert D. Cook II to serve on our board of directors.
Gilead participated in the 2024 Financing Transaction on the same terms as the new investor pursuant to the anti-dilution provision in the Investor Rights Agreement. We and Gilead entered into a Securities Purchase Agreement for the issuance and sale, in a private placement, of 179,500 shares of our common stock and a warrant to purchase up to 179,500 shares of our common stock. The warrant sold to Gilead has an exercise price equal to $17.00 per share, became immediately exercisable on the date of issuance and will expire on June 18, 2029. Subject to certain exceptions, neither Gilead nor its affiliates may exercise any portion of the warrant to the extent that Gilead would own more than 19.9% of the number of our shares of common stock outstanding immediately after giving effect to such exercise.
Gilead also participated in a financing transaction in August 2025 on the same as the new investors. We and Gilead entered into a Securities Purchase Agreement for the issuance and sale, in a private placement, of 2,295,920 shares of our common stock at a combined purchase price of $19.60 per shares and accompanying one-half of one Class A Warrant and one-half of one Class B Warrant (collectively, the August 2025 Private Placement). The common stock, the Class A Warrant and the Class B Warrant were sold to Gilead pursuant to the terms of the Investor Rights Agreement. The Class A Warrant and the Class B Warrant each provide for the right to purchase up to 1,147,960 shares of our common stock. The Class A Warrant has an exercise price of $21.60 per share, became immediately exercisable on the date of issuance and will expire on or prior to the earlier of (a) August 11, 2030 (five years from the date of issuance) and (b) the date that is 30 days after the public announcement that we have completed enrollment of at least 200 patients total for our Phase 2 clinical study evaluating 5366 versus valacyclovir. The Class B Warrant has an exercise price of $21.60 per share and is exercisable between November 15, 2026 and December 31, 2026. Notwithstanding the foregoing, if, prior to November 15, 2026, we publicly announce that we have received at least $75.0 million in the aggregate of non-dilutive capital in connection with a collaboration agreement, then the Class B Warrant automatically terminates in full.
In October 2025, as required under the registration rights terms of the Investor Rights Agreement, we filed a Registration Statement on Form S-3 with the Securities and Exchange Commission (SEC) to register all of the shares of our common stock that have been issued and sold to Gilead, as well as all of the shares of common stock that can be acquired by exercising the warrants that have been issued and sold to Gilead. The SEC is neither reviewing nor accelerating the effectiveness of registration statements during the ongoing shutdown of the U.S. government.
Results of Operations
Comparison of the Three Months Ended September 30, 2025 and 2024
Collaboration Revenue
The following table summarizes the period-over-period changes in our collaboration revenue (in thousands, except for percentages):
|
Three Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
Collaboration revenue from a related party |
$ |
10,789 |
$ |
6,845 |
$ |
3,944 |
58 |
% |
||||||||
Collaboration revenue was $10.8 million for the three months ended September 30, 2025 compared to $6.8 million for the same period in 2024. The $3.9 million increase was primarily due to more costs incurred under the Gilead Collaboration Agreement during the three months ended September 30, 2025.
Research and Development Expenses
Research and development expenses consist primarily of employee-related expenses, fees paid to contract research organizations and contract manufacturing organizations, lab supplies and other third-party expenses that support our research and discovery, nonclinical and clinical activities. External costs represent a significant portion of our research and development expenses, which we track on a program-by-program basis following the nomination of a development candidate. We use our employee and infrastructure resources, as well as certain third-party costs, across multiple research and development programs, and we do not specifically allocate these costs to our programs.
The following table summarizes the period-over-period changes in our research and development expenses (in thousands, except for percentages):
|
Three Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
External program expenses: |
||||||||||||||||
|
5366 |
$ |
2,430 |
$ |
1,797 |
$ |
633 |
35 |
% |
||||||||
|
1179 |
2,982 |
654 |
2,328 |
356 |
% |
|||||||||||
|
6250 |
714 |
1,298 |
(584 |
) |
(45 |
%) |
||||||||||
|
4334 |
113 |
842 |
(729 |
) |
(87 |
%) |
||||||||||
|
7272 |
(1) |
354 |
- |
354 |
100 |
% |
||||||||||
|
Research and discovery |
2,190 |
2,251 |
(61 |
) |
(3 |
%) |
||||||||||
|
Vebicorvir (VBR) |
- |
(67 |
) |
(2) |
67 |
(100 |
%) |
|||||||||
|
Total external program expenses |
8,783 |
6,775 |
2,008 |
30 |
% |
|||||||||||
|
Employee and contractor-related expenses |
6,934 |
6,072 |
862 |
14 |
% |
|||||||||||
|
Facility and other expenses |
870 |
668 |
202 |
30 |
% |
|||||||||||
|
Total research and development expenses |
$ |
16,587 |
$ |
13,515 |
$ |
3,072 |
23 |
% |
||||||||
Research and development expenses were $16.6 million for the three months ended September 30, 2025 compared to $13.5 million for the same period in 2024. The $3.1 million increase was primarily driven by higher external program expenses as we advance our pipeline. Most notably, our 1179 and 5366 programs incurred additional costs due to significant patient enrollment in their respective Phase 1b studies. Employee and contractor-related expenses also increased due to the recognition of stock-based compensation expense on previously granted performance-based
restricted stock units now deemed probable of vesting. These increases were partially offset by lower expenses for our 4334 program following the completion of its Phase 1b study.
General and Administrative Expenses
General and administrative expenses consist primarily of salaries and other related costs for personnel in executive, finance, accounting, business development, information technology, legal and human resources functions. Other significant costs include facility costs not otherwise included in research and development expenses, insurance costs, legal fees relating to patents and corporate matters and fees for accounting and consulting services.
The following table summarizes the period-over-period changes in our general and administrative expenses (in thousands, except for percentages):
|
Three Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
General and administrative expenses |
$ |
5,085 |
$ |
4,286 |
$ |
799 |
19 |
% |
||||||||
General and administrative expenses were $5.1 million for the three months ended September 30, 2025 compared to $4.3 million for the same period in 2024. The $0.8 million increase was primarily driven by higher professional fees related to patent filings, commercial planning and market research, as well as an increase in stock-based compensation expense from the recognition of expense on previously granted performance-based restricted stock units now deemed probable of vesting.
Interest and Other Income, Net
Interest income consists of interest earned on our cash and cash equivalents and available-for-sale marketable securities. The following table summarizes the period-over-period changes in our interest and other income, net (in thousands, except for percentages):
|
Three Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
Interest and other income, net |
$ |
1,687 |
$ |
1,343 |
$ |
344 |
26 |
% |
||||||||
Interest and other income, net was $1.7 million for the three months ended September 30, 2025, compared to $1.3 million for the same period in 2024. The $0.3 million increase was primarily due to more interest earned on marketable securities from having a larger portfolio balance after our financing in August 2025.
Comparison of the Nine Months Ended September 30, 2025 and 2024
Collaboration Revenue
The following table summarizes the period-over-period changes in our collaboration revenue (in thousands, except for percentages):
|
Nine Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
Collaboration revenue from a related party |
$ |
29,834 |
$ |
21,163 |
$ |
8,671 |
41 |
% |
||||||||
Collaboration revenue was $29.8 million for the nine months ended September 30, 2025 compared to $21.2 million for the same period in 2024. The $8.7 million increase was due to more costs incurred under the Gilead Collaboration Agreement during the nine months ended September 30, 2025, as well as an increase in the transaction price from additional funds received under the First Amendment to the Gilead Collaboration Agreement.
Research and Development Expenses
The following table summarizes the period-over-period changes in our research and development expenses (in thousands, except for percentages):
|
Nine Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
External program expenses: |
||||||||||||||||
|
5366 |
$ |
7,158 |
$ |
3,933 |
$ |
3,225 |
82 |
% |
||||||||
|
1179 |
5,047 |
3,531 |
1,516 |
43 |
% |
|||||||||||
|
6250 |
4,617 |
5,361 |
(744 |
) |
(14 |
%) |
||||||||||
|
4334 |
919 |
1,939 |
(1,020 |
) |
(53 |
%) |
||||||||||
|
7272 |
(1) |
2,004 |
- |
2,004 |
100 |
% |
||||||||||
|
Research and discovery |
5,839 |
6,753 |
(914 |
) |
(14 |
%) |
||||||||||
|
VBR |
- |
(67 |
) |
(2) |
67 |
(100 |
%) |
|||||||||
|
Total external program expenses |
25,584 |
21,450 |
4,134 |
19 |
% |
|||||||||||
|
Employee and contractor-related expenses |
19,365 |
18,081 |
1,284 |
7 |
% |
|||||||||||
|
Facility and other expenses |
2,614 |
2,122 |
492 |
23 |
% |
|||||||||||
|
Total research and development expenses |
$ |
47,563 |
$ |
41,653 |
$ |
5,910 |
14 |
% |
||||||||
Research and development expenses were $47.6 million for the nine months ended September 30, 2025 compared to $41.7 million for the same period in 2024. The $5.9 million increase was primarily driven by higher external program expenses as we advance our pipeline. Most notably, our HSV programs incurred additional costs as both the 5366 and 1179 Phase 1a/b studies were underway during the nine months ended September 30, 2025, compared to only the 5366 Phase 1a study in the same period in 2024. Employee and contractor-related expenses also increased, primarily due to the recognition of stock-based compensation expense on previously granted performance-based restricted stock units now deemed probable of vesting.
General and Administrative Expenses
The following table summarizes the period-over-period changes in our general and administrative expenses (in thousands, except for percentages):
|
Nine Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
General and administrative expenses |
$ |
14,188 |
$ |
13,398 |
$ |
790 |
6 |
% |
||||||||
General and administrative expenses were $14.2 million for the nine months ended September 30, 2025 compared to $13.4 million for the same period in 2024. The $0.8 million increase was primarily driven by higher professional fees related to patent filings as well as an increase in stock-based compensation expense from the recognition of expense on previously granted performance-based restricted stock units now deemed probable of vesting.
Interest and Other Income, Net
The following table summarizes the period-over-period changes in our interest and other income, net (in thousands, except for percentages):
|
Nine Months Ended September 30, |
$ Change |
% Change |
||||||||||||||
|
2025 |
2024 |
2025 vs. 2024 |
2025 vs. 2024 |
|||||||||||||
|
Interest and other income, net |
$ |
3,705 |
$ |
4,452 |
$ |
(747 |
) |
-17 |
% |
|||||||
Interest and other income, net was $3.7 million for the nine months ended September 30, 2025, compared to $4.5 million for the same period in 2024. The $0.7 million decrease was primarily due to a smaller portfolio balance for most of the period, as we used maturing investments to fund operations prior to our financing transaction in August 2025. Lower interest rates during the nine months ended September 30, 2025 compared to the same period in 2024 also contributed to the decrease.
Liquidity and Capital Resources
Sources of Liquidity
As a result of our significant research and development expenditures and the lack of any FDA-approved products to generate product sales revenue, we have not been profitable and have generated operating losses since we were incorporated in October 2005. We have funded our operations through September 30, 2025 principally through equity financings, raising an aggregate of $821.8 million in net proceeds, and strategic collaborations, raising an aggregate of $200.9 million.
In August 2025, we sold to various investors an aggregate of 5,591,840 shares of common stock and pre-funded warrants to purchase up to 1,040,820 shares of common stock. The securities were issued together with accompanying Class A and Class B warrants to purchase up to an aggregated total of 6,632,660 shares of common stock. The combined price per share of common stock with the accompanying Class A and Class B warrants was $19.60, while the combined price per pre-funded warrant with accompanying Class A and Class B warrants was $19.599. In addition, the Company entered into the August 2025 Private Placement with Gilead for the issuance and sale, in a private placement, of 2,295,920 shares of common stock and accompanying Class A and Class B warrants to purchase up to an aggregate of 2,295,920 shares of common stock. The securities were sold together with accompanying warrants at a price of $19.60 per unit. Each pre-funded warrant sold has a nominal exercise price of $0.001 per share, while each Class A and Class B warrant has an exercise price of $21.60 per share. We received aggregate net proceeds of approximately $166.4 million from this financing transaction, after deducting underwriting discounts and commissions and offering expenses payable.
Cash Flows for the Nine Months Ended September 30, 2025 and 2024
The following table summarizes our cash flow activities (in thousands):
|
Nine Months Ended September 30, |
||||||||
|
2025 |
2024 |
|||||||
|
Net cash used in operating activities |
$ |
(55,330 |
) |
$ |
(50,710 |
) |
||
|
Net cash (used in) provided by investing activities |
(134,691 |
) |
46,867 |
|||||
|
Net cash provided by financing activities |
174,211 |
12,454 |
||||||
|
Net (decrease) increase in cash and cash equivalents |
$ |
(15,810 |
) |
$ |
8,611 |
|||
Operating Activities
Net cash used in operating activities was $55.3 million for the nine months ended September 30, 2025, compared to $50.7 million for the same period in 2024. The increase in cash used in operations was primarily attributable to increases in operating expenses from the advancement of our clinical pipeline.
Investing Activities
Net cash used in investing activities was $134.7 million for the nine months ended September 30, 2025, compared to net cash provided by investing activities of $46.9 million for the same period in 2024. The change was primarily due to our purchases of marketable securities following our financing in August 2025, as we invested the proceeds from those offerings.
Financing Activities
Net cash provided by financing activities was $174.2 million for the nine months ended September 30, 2025, compared to $12.5 million for the same period in 2024. The increase was due to larger proceeds from our financing in August 2025, and the subsequent exercise of warrants issued in that transaction, compared to our financing in June 2024.
Funding Requirements
We have generated significant losses to date, and we expect to continue to generate losses as we develop our product candidates. As of September 30, 2025, we had an accumulated deficit of $854.1 million. Because we do not generate revenue from any of our product candidates, our losses will continue as we further develop and seek regulatory approval for, and commercialize, our product candidates. We expect our future operating expenses to increase over the coming years as we continue to advance our candidates. As a result, our operating losses are likely to be substantial over the next several years if none of our product candidates are approved or successfully launched. We are unable to predict the extent of any future losses or when we will become profitable, if at all.
As of September 30, 2025, we held cash, cash equivalents and marketable securities of $232.6 million. Based on our current operating plan, we believe we have sufficient funds to meet our operating requirements into late 2027. This cash runway guidance does not include potential future payments to us under our collaboration with Gilead or from potential warrant exercises, which would further extend our cash runway beyond 2028. We have based our estimate on assumptions that may prove to be wrong, and we may utilize our available capital resources sooner than we currently expect.
Our future capital requirements will depend on many factors, including:
Critical Accounting Estimates
Our critical accounting policies and significant estimates are detailed in our 2024 Annual Report. There have been no material changes to our significant estimates from those previously disclosed in our 2024 Annual Report.