MedPath

AFA-281 Advanced Drug Monograph

Published:Jun 4, 2025

Generic Name

AFA-281

AFA-281: A Novel Dual Modulator for Neuropathic Pain and CNS Disorders – Preclinical Profile and Clinical Development Trajectory

1. Executive Summary

AFA-281 is an orally bioavailable, small molecule New Chemical Entity (NCE) developed by AfaSci, Inc., a biotechnology company based in Burlingame, California.[1] The compound's primary pharmacological distinction is its dual mechanism of action, functioning as a modulator of T-type calcium channels (Cav3) and an inhibitor of soluble epoxide hydrolase (sEH).[3] This dual activity is hypothesized to normalize neuronal excitability and enhance endogenous anti-inflammatory processes, respectively.[1]

AFA-281 has successfully completed Phase I clinical trials, and its Investigational New Drug (IND) application (IND 157314) for the treatment of neuropathic pain received acceptance from the U.S. Food and Drug Administration (FDA) in November 2022.[1] Building on this, AfaSci is advancing AFA-281 into Phase II clinical development for painful lumbosacral radiculopathy (under trial NCT06649747) and Alcohol Use Disorder (AUD) (trial NCT06710431). A dedicated Phase I drug-alcohol interaction study (NCT06719908) is also planned to support the AUD program.[2] Preclinical evidence further supports its potential utility in Cocaine Use Disorder (CUD), osteoarthritis, cancer pain, and various forms of inflammatory pain.[3]

The development of AFA-281 as a non-opioid, non-steroidal therapeutic agent is significant, given the substantial unmet medical need for safer and more effective treatments for chronic pain and other central nervous system (CNS) disorders.[1] The compound's dual-target mechanism represents a novel therapeutic strategy, potentially offering broader efficacy and an improved side-effect profile compared to single-mechanism agents.

AfaSci's development strategy for AFA-281 reflects a common trajectory in pharmaceutical R&D, moving from a broad exploration of potential indications—including osteoarthritis, various pain types (cancer, inflammatory, neuropathic), substance use disorders (AUD, CUD), seizures, anxiety, dyssomnias, and depression [1]—towards a more focused clinical execution. The FDA's acceptance of the IND for neuropathic pain [4] served as a key validation point. This regulatory milestone appears to be enabling the company to strategically leverage the initial Phase I safety and pharmacokinetic data to explore related indications like lumbosacral radiculopathy (a specific type of neuropathic pain) and mechanistically plausible CNS disorders such as AUD.[2] This funneling from broad potential to specific lead indications is a logical progression, driven by emerging preclinical and early clinical data, assessment of unmet medical needs, and strategic resource allocation.

The cornerstone of AFA-281's therapeutic hypothesis and competitive positioning is its unique dual-target mechanism. The consistent emphasis across company communications and grant applications on AFA-281 as a "dual modulator" or "dual inhibitor" of Cav3 T-type calcium channels and soluble epoxide hydrolase (sEH) [1] underscores this. The scientific rationale is that by concurrently inhibiting Cav3 channels and sEH, AFA-281 can normalize neuronal excitability and bolster the body's natural anti-inflammatory responses.[1] This approach is particularly relevant for complex conditions like chronic pain and addiction, where both neuronal hyperexcitability and neuroinflammatory processes are often intertwined. Addressing both facets with a single molecule could offer advantages over therapies targeting only one pathway or requiring the combination of multiple drugs, potentially leading to enhanced efficacy and a simplified treatment regimen.[4]

2. Introduction to AFA-281 and AfaSci, Inc.

2.1. AFA-281: Nomenclature and Chemical Nature

AFA-281 is identified as a New Chemical Entity (NCE) and is classified as a small molecule drug.[1] Its common synonyms in development literature include AFA 281 and AFA281. As an NCE, AFA-281 benefits from the potential for robust intellectual property protection. The small molecule nature of AFA-281 typically suggests advantages in terms of manufacturing scalability, chemical stability, and the likelihood of achieving oral bioavailability. Preclinical studies have indeed confirmed that AFA-281 can be administered orally and also intravenously.[3]

2.2. AfaSci, Inc.: Company Overview, Mission, and Focus

AfaSci, Inc. is a biotechnology company located in the San Francisco Bay Area, specifically in Burlingame, California, USA.[1] The company is dedicated to the discovery and development of novel peptide and small molecule therapeutics aimed at addressing significant unmet medical needs, with a primary focus on pain and neurological disorders.[1] AFA-281 is currently their lead program targeting the CNS.[1]

According to information available up to May 2025, AfaSci's pipeline extends beyond AFA-281. Other programs include AFA-280, a preclinical candidate for inflammatory pain; AFA-2000, a preclinical candidate targeting G-protein-coupled inwardly-rectifying potassium (GIRK) channels for pain; and AFA-P30, a discovery-stage program targeting Calcitonin Gene-Related Peptide (CGRP) for pain. A project designated AFA-500 for sleep-wake disorders has been discontinued, indicating active portfolio management.[9]

The company's leadership team includes Xinmin Simon Xie, M.D., Ph.D., who serves as CEO and is a Principal Investigator on National Institutes of Health (NIH) grants supporting AFA-281's development, and Jiang-Hong Ye, Ph.D., also a Principal Investigator on an NIH grant related to the company's research.[12] AfaSci employs a strategy of in-house phenotypic drug discovery and navigates its candidates through early clinical development phases. The company has explicitly stated its intention to seek investment and establish partnering or out-licensing agreements to support later-stage clinical trials (Phase II and beyond) and eventual commercialization of its assets.[1] This positions AfaSci as an early-stage innovator aiming to de-risk therapeutic candidates to key clinical milestones before seeking larger collaborations.

While AFA-281 is a small molecule, AfaSci's broader mission encompasses both "peptide and small molecule therapies".[1] This, combined with a pipeline that includes targets like CGRP (for which peptide therapeutics are a common modality, e.g., AFA-P30), suggests a versatile drug discovery platform within the company. This dual-modality capability could provide flexibility in addressing diverse biological targets and may enhance the company's attractiveness to potential partners with varied interests.

The therapeutic focus for AFA-281 appears to have evolved. Earlier company descriptions mentioned "chronic pain and seizures" as primary targets.[1] However, more recent clinical development activities and grant-funded research heavily emphasize neuropathic pain, other specific pain conditions like lumbosacral radiculopathy, and substance use disorders such as AUD and CUD.[2] While epilepsy might remain an area of underlying interest due to the Cav3 mechanism, the current clinical and financial momentum is clearly directed towards pain and addiction. The broad "Pain" indication listed for AFA-281 at Phase 2 in some databases [9] likely encompasses these more specific pain types.

3. Mechanism of Action: A Novel Dual Modulator

AFA-281's therapeutic potential is rooted in its unique ability to simultaneously modulate two distinct biological targets: T-type calcium channels (Cav3) and soluble epoxide hydrolase (sEH). This dual mechanism is central to its development for a range of CNS disorders.

3.1. Target 1: T-type Calcium Channels (Cav3)

AFA-281 functions as a modulator, specifically an inhibitor, of T-type calcium channels (Cav3).[3] These channels, which include subtypes Cav3.1, Cav3.2, and Cav3.3, are low voltage-activated calcium channels that play crucial roles in regulating neuronal excitability, particularly in burst firing patterns. AfaSci posits that by inhibiting these channels, especially when they are overactivated in pathological states, AFA-281 can normalize neuronal excitability and thereby restore more balanced nervous system function.[1] Preclinical research indicates AFA-281 potently inhibits the Cav3.2 subtype.[4]

The relevance of T-type calcium channels to the targeted indications is well-supported. In pain states, particularly neuropathic pain, T-type channels in sensory neurons contribute to the generation and propagation of aberrant pain signals. In epilepsy, they are involved in the generation of thalamocortical oscillations that can lead to seizures. More recently, their role in substance use disorders has gained attention. For instance, cocaine has been shown to activate T-type channels in the brain, increasing neuronal excitability. The older T-channel inhibitor, mibefradil, demonstrated an ability to block certain cocaine-induced behavioral and neurophysiological effects in mice.[5] Furthermore, genetic studies in Cav3.1 knockout mice have highlighted the importance of T-type channels in maintaining neuronal excitability in the ventral tegmental area (VTA), a key region in the brain's reward circuitry. Cav3.2-deficient mice have also shown reduced sensitivity to psychostimulants, providing biological validation for Cav3 subunits as therapeutic targets in CUD.[13]

3.2. Target 2: Soluble Epoxide Hydrolase (sEH)

In addition to its action on Cav3 channels, AFA-281 is an inhibitor of soluble epoxide hydrolase (sEH).[3] sEH is an enzyme responsible for the degradation of endogenous lipid signaling molecules known as epoxyeicosatrienoic acids (EETs). EETs are derived from arachidonic acid via cytochrome P450 epoxygenase pathways and possess potent anti-inflammatory, analgesic, vasodilatory, and organ-protective properties. By inhibiting sEH, AFA-281 is expected to increase the concentration and prolong the half-life of these beneficial EETs, thereby enhancing the body's natural anti-inflammatory and pro-resolving capabilities.[1] Preclinical evidence supports this, showing that single sEH inhibitors or genetic deletion of sEH can lead to a reduction in neuroinflammation.[13]

3.3. Rationale and Potential Synergies of Dual Inhibition

AFA-281 is designed as a single chemical entity possessing this dual inhibitory activity.[1] The therapeutic rationale is that many complex CNS disorders, particularly chronic pain and addiction, are driven by both direct neuronal dysfunction (such as hyperexcitability) and a significant, often chronic, neuroinflammatory component. By simultaneously targeting Cav3 channels and sEH, AFA-281 aims to address both of these pathological pillars. For example, in the context of CUD, AfaSci hypothesizes that AFA-281's dual action could suppress cocaine-induced neuronal hyperexcitability (via Cav3 inhibition) and mitigate associated neuroinflammation (via sEH inhibition).[13] This integrated approach with a single molecule may offer advantages over administering multiple single-target drugs, potentially reducing polypharmacy, improving patient adherence by simplifying treatment regimens, minimizing the risk of dose-limiting side effects from individual agents, and reducing the likelihood of complex drug-drug interactions.[4]

The specific activity profile of AFA-281 across the three Cav3 subtypes (Cav3.1, Cav3.2, Cav3.3) is an area for further detailed elucidation. While potent inhibition of Cav3.2 is noted [4], the comparative potencies against Cav3.1 and Cav3.3 are not explicitly provided in the available documentation. These subtypes exhibit distinct tissue distribution and physiological roles; for example, Cav3.2 is prominently involved in peripheral nociception, whereas Cav3.1 and Cav3.3 are more associated with thalamocortical rhythms and conditions like absence epilepsy. A comprehensive understanding of AFA-281's subtype selectivity will be crucial for predicting its precise therapeutic spectrum and potential side-effect profile across its diverse target indications.

The dual mechanism of AFA-281 positions it as a modulator of the neuro-immune axis. Cav3 inhibition directly influences neuronal firing and excitability, while sEH inhibition indirectly modulates neuronal function by dampening neuroinflammatory signals and promoting a more favorable microenvironment. This capacity to address both direct neuronal pathology and associated inflammatory processes aligns with contemporary understanding of many CNS disorders, where a bidirectional and often detrimental interplay exists between neuronal hyperexcitability and inflammation. By intervening at both levels, AFA-281 may disrupt these pathological cycles more effectively than agents targeting only one component, potentially leading to more robust and sustained therapeutic outcomes.

4. Pharmacological Profile

4.1. Pharmacodynamics (PD)

Preclinical pharmacodynamic studies have provided evidence for AFA-281's activity across several domains relevant to its target indications:

  • Analgesic Effects: AFA-281 has demonstrated broad analgesic properties in preclinical models of both neuropathic and inflammatory pain.[4] A key study in the spared nerve injury (SNI) rat model, a well-established paradigm for neuropathic pain, showed that AFA-281 administration resulted in significant pain relief. Notably, this analgesic effect was accompanied by a reduction in brain microglia activation, providing in vivo support for the anti-neuroinflammatory component of its dual mechanism, likely mediated by sEH inhibition, in addition to its expected neuronal effects through Cav3 channel modulation.[4]
  • Neuromodulatory Effects in Substance Use Disorder Models:
  • Cocaine Use Disorder (CUD): Preclinical investigations indicate that AFA-281 can block cocaine-enhanced neuronal excitability in ex vivo rat thalamocortical slices. In behavioral models, AFA-281 suppressed cocaine-seeking behavior, as measured by intravenous self-administration paradigms, and reduced cocaine-induced hyperlocomotion in rats.[4] These findings form a strong basis for its development in CUD.
  • Alcohol Use Disorder (AUD): In rodent models of AUD, AFA-281 has been shown to decrease alcohol intake, reduce binge-drinking patterns, diminish preference for alcohol over non-alcoholic alternatives, and suppress alcohol-seeking behaviors.[4] Further mechanistic studies, supported by an NIH grant, are planned to investigate AFA-281's influence on alcohol-induced alterations in neural activity (using c-Fos expression as a marker), brain neuroinflammation, and Cav3 channel activity and cellular excitability, with a specific focus on the lateral habenula, a brain region critically involved in reward processing and aversive signaling.[4]
  • Anti-inflammatory Properties: The inhibition of sEH by AFA-281 is designed to augment the body's endogenous anti-inflammatory mechanisms.[1] This is supported by preclinical observations of reduced brain microglia activation in pain models [4] and is a central hypothesis in its planned investigation for neuroinflammation in AUD models.[4]

Collectively, these pharmacodynamic effects provide a compelling rationale for AFA-281's development across a spectrum of pain and addiction disorders, directly linking its dual mechanism of action to tangible therapeutic outcomes in relevant animal models.

4.2. Pharmacokinetics (PK)

The pharmacokinetic profile of AFA-281 has been characterized in preclinical species, with human PK data from the completed Phase I trial informing its progression, although specific human parameters are not detailed in the provided documents.

  • Absorption and Bioavailability: AFA-281 is suitable for both oral and intravenous administration.[3] Preclinical studies have demonstrated excellent oral bioavailability, reported as 70% in rats and 92% in dogs.[4] This high oral bioavailability is a significant advantage for a drug intended for chronic administration.
  • Distribution: A critical feature for its intended CNS applications, AFA-281 has shown an ability to penetrate the central nervous system in preclinical models.[4] The extent and regional distribution within the CNS will be important factors for its efficacy in different neurological disorders.
  • Metabolism and Excretion: Animal studies indicate an acceptable plasma half-life (t1/2​) ranging from 2.5 to 4.3 hours.[4] This half-life suggests that manageable oral dosing schedules (e.g., once or twice daily) might be achievable in humans. Human pharmacokinetics were assessed during the completed Phase I trial, and further characterization will occur in the upcoming Phase I alcohol interaction study and Phase II trials.[2]

The favorable preclinical PK profile, particularly its high oral bioavailability and CNS penetration, supports the continued development of AFA-281 as an oral therapeutic for chronic, centrally-mediated conditions. However, the successful translation of these animal PK parameters to humans is a critical step. While the completion of Phase I trials implies acceptable initial human safety and some PK understanding [1], detailed human PK data (such as Cmax​, AUC, t1/2​, and inter-subject variability) and clear pharmacodynamic markers of target engagement at various doses are not yet publicly available from the provided information. The planned Phase I alcohol interaction study (NCT06719908) [8] and the Phase II efficacy studies in lumbosacral radiculopathy (NCT06649747) [2] and AUD (NCT06710431) [2] will be instrumental in comprehensively defining the human PK/PD relationships, establishing optimal dosing regimens, and confirming target engagement across diverse patient populations. The drug-alcohol interaction study is particularly vital for de-risking the AUD development program.

Furthermore, while "CNS penetration" is confirmed preclinically [4], a more nuanced understanding of the extent and regional specificity of this penetration, as well as the mechanisms governing its transit across the blood-brain barrier (e.g., passive diffusion, active transport, P-glycoprotein substrate/inhibitor status), would be beneficial. This information is key for optimizing dosing strategies for different CNS disorders and for anticipating potential drug-drug interactions at the level of the BBB. While the AUD mechanistic studies plan to investigate effects in the lateral habenula [4], a broader characterization of CNS distribution and target occupancy at clinically relevant doses would significantly strengthen the overall development program.

5. Therapeutic Indications and Development Pipeline

AFA-281 is being investigated across a range of indications, primarily focusing on pain and substance use disorders, leveraging its dual mechanism of action.

5.1. Lead Indication: Neuropathic Pain (IND 157314)

The most advanced program for AFA-281 is in neuropathic pain. An Investigational New Drug application (IND 157314) was accepted by the FDA on November 11, 2022, allowing clinical trials in this indication to proceed in the United States.[4] Phase I clinical trials for neuropathic pain have been successfully completed.[1] Industry benchmarks from GlobalData suggest that Phase I drugs for neuropathic pain have a 76% phase transition success rate (PTSR) for progressing into Phase II, indicating a generally positive outlook for drugs reaching this stage in this therapeutic area.[3] Neuropathic pain continues to be an area with significant unmet medical need, characterized by often inadequate efficacy and problematic side effects of current treatments.

5.2. Expansion Indications (Clinical - Not Yet Recruiting or Early Phase)

Building on the foundation of the neuropathic pain IND, AfaSci is pursuing several other indications:

  • Lumbosacral Radiculopathy: A Phase II, multi-center, double-blind, placebo-controlled study (NCT06649747) is planned to assess the safety, tolerability, pharmacokinetics, and efficacy of oral AFA-281 in patients suffering from painful lumbosacral radiculopathy.[2] This condition is a specific type of neuropathic pain stemming from nerve root compression or irritation in the lumbar or sacral spine.
  • Alcohol Use Disorder (AUD): A Phase IIa human laboratory study (NCT06710431) is planned to evaluate the safety and exploratory efficacy of oral AFA-281 in patients with AUD.[2] This program is supported by NIH funding through an SBIR Phase I U43 project (PAR-22-102), which focuses on preclinical proof-of-concept studies.[4] Preclinical data in rodent models have shown that AFA-281 can reduce alcohol intake, binge drinking, alcohol preference, and suppress alcohol-seeking behaviors.[4]
  • Other Potential Phase 2 Indications: According to Synapse database information, AFA-281 is also considered for Phase 2 development in broader categories such as Back Pain, general Pain, Anxiety Disorders, Dyssomnias (sleep disorders), and Endogenous Depression.[2] Phase 1 development is listed for Inflammatory pain and Neuralgia (a general term for nerve pain).[2]

5.3. Expansion Indications (Preclinical/Early Development)

  • Cocaine Use Disorder (CUD): AfaSci is conducting preclinical proof-of-concept studies for AFA-281 in CUD, supported by an NIH SBIR Phase I grant (R43DA059524).[4] The therapeutic hypothesis is that the dual inhibition of Cav3 channels and sEH by AFA-281 can effectively suppress both cocaine-induced neuronal hyperexcitability and associated neuroinflammation.[13]
  • Osteoarthritis, Cancer Pain, Inflammatory Pain, Epilepsy: These conditions have been listed as areas of development or potential indications for AFA-281 in various documents, reflecting the broad applicability of its dual mechanism.[1]

5.4. Scientific Rationale for Key Indications

The selection of these indications is underpinned by the known roles of Cav3 channels and sEH in their respective pathophysiologies:

  • Pain (neuropathic, inflammatory, radiculopathy, osteoarthritis, cancer): Cav3 T-type calcium channels are crucial in pain signaling pathways and the generation of neuronal hyperexcitability, a hallmark of neuropathic pain. sEH inhibition, by preserving anti-inflammatory EETs, addresses the inflammatory component often present in chronic pain states.[1]
  • Substance Use Disorders (AUD, CUD): Cav3 channels are implicated in the neuronal adaptations and reward pathway dysregulation associated with addiction. sEH inhibition may counteract the neuroinflammatory processes that are increasingly recognized as contributing to the development and maintenance of substance use disorders.[4]
  • Epilepsy/Seizures: T-type calcium channels are validated targets for anti-epileptic drugs due to their contribution to neuronal burst firing and the generation of seizures, particularly absence seizures.[1]

AfaSci's strategy of leveraging the active IND for neuropathic pain (IND 157314) and the associated safety and PK data from the completed Phase I program to explore AUD and CUD [4] is an efficient approach. It allows the company to build upon existing regulatory groundwork and safety information, potentially accelerating the path to clinical proof-of-concept for these new indications. The NIH grant support for the AUD and CUD preclinical programs provides external validation of the scientific rationale and crucial non-dilutive funding for these exploratory efforts.

While the Synapse database [2] outlines a broad range of potential Phase 1 and 2 indications, a company of AfaSci's size will likely need to maintain a strategic focus on a few lead indications for advancement into more resource-intensive later-stage trials. The current explicit clinical focus on lumbosacral radiculopathy and AUD for Phase II, alongside the Phase I alcohol interaction study, appears to represent a pragmatic initial step in this direction. Showcasing a wider potential pipeline can be attractive for partnerships, which AfaSci is actively seeking [1], but successful execution will depend on careful prioritization and resource allocation.

6. Clinical Development Program

The clinical development of AFA-281 is progressing from initial safety assessments to exploratory efficacy studies in targeted patient populations.

6.1. Summary of AFA-281 Clinical Trials

The following table summarizes the known clinical trials for AFA-281 based on the provided information:

NCT IDPhaseIndicationStatusSponsorKey ObjectivesNo. of Patients (Est.)Key Endpoints (examples)
(Implied)Phase INeuropathic PainCompleted 1AfaSci, Inc.Safety, Tolerability, PharmacokineticsN/AAdverse events, PK parameters
NCT06719908Phase IHealthy Volunteers (Drug-Alcohol Interaction)Not Yet Recruiting/Begins enrollment soon 2AfaSci, Inc.PK/PD effects of AFA-281 & ethanol co-administration, Safety20Blood concentrations of AFA-281 & ethanol, Vital signs, Clinical pathology, Symptom surveys
NCT06649747Phase IIPainful Lumbosacral RadiculopathyNot Yet Recruiting 2AfaSci, Inc.Safety, Tolerability, Pharmacokinetics, EfficacyN/APain scores, Functional improvement, Safety parameters, PK parameters
NCT06710431Phase IIaAlcohol Use DisorderNot Yet Recruiting 2AfaSci, Inc.Safety, Exploratory EfficacyN/AAlcohol cravings, Drinking-related feelings, Pain sensitivity, Anxiety, Depression, Sleep quality, Safety parameters

6.2. Completed Phase I Studies (Neuropathic Pain)

AFA-281 has successfully completed Phase I clinical trials.[1] These studies were foundational for the acceptance of IND 157314 by the FDA for neuropathic pain on November 11, 2022. The IND submission was supported by comprehensive Good Laboratory Practice (GLP) safety pharmacology, toxicology, and genotoxicity studies, which reportedly indicated no cardiac toxicity concerns and a generally good safety profile for AFA-281.[4] An SBIR grant proposal also references the plan to conduct Phase I trials under this active IND.[4] The successful completion of these initial human studies is a critical milestone, providing essential data on safety, tolerability, and pharmacokinetics to guide the design and dosing for subsequent Phase II trials.

6.3. Ongoing and Planned Clinical Trials

  • NCT06719908 (Phase I, Drug-Alcohol Interaction Study): [2]
  • Status: Not yet recruiting, with enrollment anticipated to begin in approximately two months from early 2025.
  • Sponsor: AfaSci, Inc.
  • Purpose: This study aims to evaluate the pharmacokinetic (PK) and pharmacodynamic (PD) effects of co-administering a single oral dose of AFA-281 (40 mg) with ethanol in healthy adult volunteers (aged 21-55 years) who are social or moderate drinkers. A key objective is to assess any potential side effects arising from this combination.
  • Design: The trial is a randomized, double-blind, placebo-controlled, 4-way crossover study. Each of the estimated 20 healthy male and female participants will receive four treatment regimens in a randomized order: AFA-281 plus ethanol; AFA-281 plus ethanol placebo; AFA-281 placebo plus ethanol; and AFA-281 placebo plus ethanol placebo. A washout period of at least two days will separate treatment sessions.
  • Duration: Participants will be inpatients for 9 days (8 nights), with a final follow-up visit 3 to 5 days after discharge from the clinical unit.
  • Outcome Measures: Primary outcomes will include blood concentration levels of AFA-281 and ethanol at pre-dose and multiple timepoints post-treatment. Safety parameters such as vital signs and clinical pathology will be monitored, and participants will complete symptom assessment surveys.
  • NCT06649747 (Phase II, Painful Lumbosacral Radiculopathy): [2]
  • Status: Not yet recruiting.
  • Sponsor: AfaSci, Inc.
  • Purpose: To evaluate the safety, tolerability, pharmacokinetics, and efficacy of orally administered AFA-281 in patients with painful lumbosacral radiculopathy.
  • Design: This will be a double-blind, placebo-controlled, multi-center study.
  • NCT06710431 (Phase IIa, Alcohol Use Disorder): [2]
  • Status: Not yet recruiting.
  • Sponsor: AfaSci, Inc.
  • Purpose: To assess the safety and exploratory efficacy of oral AFA-281 in patients diagnosed with Alcohol Use Disorder.
  • Design: This is planned as a double-blind, placebo-controlled, human laboratory study.
  • Population: The study aims to enroll adults aged 21-65 years who meet DSM-5 diagnostic criteria for moderate or severe AUD and report specific levels of recent alcohol consumption (at least 28 drinks per week for males, 21 for females, in the 28 days prior to consent). Exclusion criteria include current treatment-seeking for alcohol use, dependence on other psychoactive substances (except nicotine), current severe psychiatric disorders (e.g., schizophrenia, bipolar disorder, major depression with suicidal ideation), clinically significant alcohol withdrawal symptoms, pregnancy, and various other medical conditions or laboratory abnormalities that could confound results or pose safety risks.[11]
  • Outcome Measures: The study will evaluate AFA-281's effects on alcohol cravings, subjective feelings related to drinking, pain sensitivity, anxiety levels, depressive symptoms, and sleep quality, alongside standard safety assessments.

AfaSci's clinical strategy for the AUD indication demonstrates a prudent, phased approach. The initial Phase I drug-alcohol interaction study in healthy volunteers (NCT06719908) is designed to establish a safety and PK/PD interaction profile before progressing to the Phase IIa exploratory efficacy study in AUD patients (NCT06710431). This sequence is standard practice for CNS drugs intended for populations where alcohol consumption is prevalent, as it helps to de-risk later-phase studies by understanding potential interactions that could impact safety or efficacy.

The eligibility criteria for the AUD patient trial (NCT06710431), such as the exclusion of individuals currently seeking treatment or those with severe comorbid psychiatric conditions [11], are typical for early-phase exploratory studies in complex psychiatric indications. These criteria aim to enroll a relatively homogenous patient population to obtain a clearer initial signal of the drug's efficacy by minimizing potential confounding variables common in AUD.

7. Preclinical Evidence Supporting AFA-281 Development

AFA-281's progression into clinical trials is supported by a body of preclinical research demonstrating its efficacy in relevant animal models and a favorable pharmacological and safety profile.

7.1. Summary of Key Preclinical Studies and Findings for AFA-281

Animal Model (Species, Strain)Indication ModeledAFA-281 Administration (Dose, Route, Duration)Key Efficacy OutcomesKey Safety/PK OutcomesSource Snippet(s)
Rats (Spared Nerve Injury - SNI)Neuropathic PainN/A (details not in snippets)Significant analgesia, reduction of brain microglia activationN/A4
RatsCocaine Use DisorderN/ASuppressed cocaine-seeking (self-administration), reduced cocaine-elicited hyperlocomotion, blocked cocaine-enhanced neural excitability (thalamocortical slices)N/A4
RodentsAlcohol Use DisorderN/ADecreased alcohol intake, binge drinking, preference; suppressed alcohol-seeking behaviorN/A4
Rats & DogsPharmacokineticsOral, IVN/AExcellent oral bioavailability (70% rats, 92% dogs), acceptable t1/2​ (2.5-4.3h), CNS penetration4
Various (in vitro, ex vitro)Safety/ToxicologyN/AN/ANo cardiac toxicity concerns (human cardiomyocytes ex vitro, CEREP 81 off-target screening), good safety margins, 50-500-fold weaker inhibition against 81 off-targets vs Cav3.2/sEH4

7.2. Efficacy in Animal Models of Pain

Preclinical studies have indicated that AFA-281 possesses broad analgesic effects, showing activity in models of both neuropathic and inflammatory pain.[4] A particularly relevant finding comes from the spared nerve injury (SNI) model in rats, a standard and well-validated paradigm for studying neuropathic pain. In this model, AFA-281 administration led to significant analgesia. Importantly, this pain-relieving effect was associated with a reduction in brain microglia activation.[4] This latter observation is significant as it provides in vivo evidence supporting the anti-neuroinflammatory component of AFA-281's dual mechanism of action, likely mediated through sEH inhibition, complementing its direct neuronal effects via Cav3 channel modulation.

7.3. Efficacy in Animal Models of Substance Use Disorders (AUD, CUD)

AFA-281 has also demonstrated promising activity in preclinical models of addiction:

  • Alcohol Use Disorder (AUD): Studies in rodents have shown that AFA-281 can decrease voluntary alcohol intake, reduce binge-drinking patterns, diminish preference for alcohol over alternative fluids, and suppress alcohol-seeking behaviors.[4] Further research, funded by NIH grants, aims to elucidate the pharmacokinetics/pharmacodynamics of AFA-281 in reducing alcohol consumption and seeking via operant self-administration paradigms. These studies will also investigate its effects on comorbid conditions often seen with AUD, such as hyperalgesia, anxiety, and depression, in rats undergoing alcohol withdrawal. Mechanistic explorations will focus on AFA-281's modulation of alcohol-altered neural activity (using c-Fos as a marker of neuronal activation), brain neuroinflammation, and Cav3 channel activity/cellular excitability, with a specific focus on the lateral habenula, a brain region increasingly implicated in addiction and mood regulation.[4]
  • Cocaine Use Disorder (CUD): In rat models, AFA-281 has been shown to suppress cocaine-seeking behavior (assessed through intravenous self-administration) and reduce cocaine-induced hyperlocomotion.[4] Furthermore, ex vivo studies using rat thalamocortical slices demonstrated that AFA-281 could block cocaine-enhanced neuronal excitability.[5] Ongoing NIH-funded SBIR Phase I research will further investigate the PK/PD of AFA-281 in suppressing cocaine-seeking behavior and hyperlocomotion.[4]

These preclinical findings provide a robust rationale for advancing AFA-281 into clinical trials for AUD and CUD, conditions with high unmet medical needs and limited effective pharmacological treatments. The planned mechanistic studies in AUD models are particularly important for deepening the understanding of how AFA-281 exerts its therapeutic effects within the brain.

7.4. Safety Pharmacology and Toxicology

A comprehensive preclinical safety assessment has been conducted for AFA-281. This includes Good Laboratory Practice (GLP) safety pharmacology, general toxicology, and genotoxicity studies.[4] A key finding from these evaluations is the absence of cardiac safety concerns, as demonstrated in studies using human cardiomyocytes ex vitro and through broad off-target screening (CEREP panel of 81 targets).[4] AFA-281 exhibited 50- to 500-fold weaker inhibition against these 81 off-targets compared to its activity on Cav3.2 and sEH, indicating a favorable selectivity profile.[4] Overall, good safety margins have been reported in preclinical species.[4]

The robust preclinical package, encompassing efficacy in relevant animal models (SNI for pain, self-administration for AUD/CUD), comprehensive PK characterization (including good oral bioavailability and CNS penetration in multiple species), and thorough GLP safety and toxicology assessments (notably, the favorable cardiac safety and off-target profile) [4], formed a strong basis for the FDA's acceptance of the IND for neuropathic pain. This extensive preclinical groundwork significantly de-risks the early stages of clinical development.

The planned mechanistic studies for AUD, with a specific focus on the lateral habenula [4], are particularly noteworthy. The lateral habenula is increasingly recognized for its critical role in processing aversive stimuli, regulating reward pathways, and its involvement in the pathophysiology of addiction and depression. Investigating AFA-281's effects within this specific and highly relevant brain circuit demonstrates a sophisticated neurobiological approach to understanding its central mechanisms of action in AUD, moving beyond generalized CNS effects.

8. Competitive Landscape and Market Potential

8.1. Unmet Medical Needs in Target Indications

AFA-281 is being developed for conditions with substantial unmet medical needs and large patient populations:

  • Chronic pain, including neuropathic pain, affects a significant portion of the population. Estimates suggest that chronic pain and seizures impact approximately 100 million and 4 million people in the US, respectively.[1] Neuropathic and neuroinflammatory pain conditions are prevalent, affecting millions in the United States and an estimated 7-8% of adults globally.[1] Current treatments for these conditions often provide inadequate relief and can be associated with significant side effects or risks, such as those associated with long-term opioid use.
  • Alcohol Use Disorder (AUD) is described as the most prevalent substance use disorder worldwide.[4] Existing pharmacological treatments for AUD have limited efficacy for many individuals and can also have undesirable side effects, highlighting a clear need for novel therapeutic options.
  • Cocaine Use Disorder (CUD) also lacks broadly effective pharmacotherapies, representing another area of high unmet need.

8.2. Differentiation of AFA-281

AFA-281 is positioned to differentiate itself from existing and emerging therapies through several key attributes:

  • Novel Dual Mechanism of Action: As a single small molecule that concurrently modulates Cav3 T-type calcium channels and inhibits soluble epoxide hydrolase (sEH), AFA-281 offers a unique therapeutic approach.[1] This dual action is hypothesized to address both neuronal hyperexcitability and neuroinflammatory components of its target diseases.
  • Non-Opioid, Non-Steroidal Analgesic: For pain indications, AFA-281 is being developed as a non-opioid and non-steroidal analgesic.[1] This is a critical differentiator given the ongoing opioid crisis and the desire for pain relief options that avoid the risks of addiction, tolerance, and other opioid-related adverse events, as well as the side effects associated with long-term steroid use.
  • Potential for Reduced Polypharmacy: By targeting multiple pathological mechanisms with a single molecule, AFA-281 could potentially reduce the need for polypharmacy, thereby simplifying treatment regimens, potentially improving patient adherence, and reducing the risk of drug-drug interactions and cumulative side effects.[4]

8.3. Potential Advantages Over Existing Therapies

Based on its proposed mechanism and preclinical profile, AFA-281 may offer several advantages:

  • Improved Efficacy: The dual mechanism targeting both neuronal excitability and inflammation may lead to superior efficacy in complex conditions where both pathways are implicated, compared to agents targeting only one pathway.
  • Enhanced Safety and Tolerability: As a non-opioid, AFA-281 avoids the significant risks associated with opioid analgesics. Its good off-target selectivity profile observed preclinically [4] also suggests a potential for a favorable side-effect profile.
  • Convenience: Its development as an oral medication offers convenience for patients requiring long-term treatment for chronic conditions.

The "non-opioid" characteristic of AFA-281 for pain management [1] is a particularly strong market differentiator in the current healthcare landscape. The urgent need for effective opioid alternatives is driven by the profound public health impact of opioid addiction and overdose. Should AFA-281 demonstrate robust efficacy and safety in clinical trials for pain, its non-opioid nature alone could drive significant adoption by clinicians and preference from patients and payers.

Furthermore, the potential of AFA-281 to address not only substance use disorders (AUD and CUD) but also commonly co-occurring conditions such as chronic pain, anxiety, and depression is a compelling aspect of its therapeutic profile.[12] Substance use disorders frequently present with these comorbidities, which can exacerbate addiction and complicate treatment. A single therapeutic agent capable of ameliorating both the core addiction-related behaviors and these associated symptoms would represent a significant advance in patient care, potentially leading to more holistic and effective treatment outcomes. The planned investigation of AFA-281's effects on hyperalgesia, anxiety, and depression in alcohol-withdrawn rats [12], alongside its primary development for pain and potential for anxiety/depression [2], underscores this multifaceted therapeutic ambition.

9. Intellectual Property and Regulatory Milestones

9.1. Patented Dual Modulators

AfaSci, Inc. has developed and patented a series of dual modulators of Cav3 T-type calcium channels and soluble epoxide hydrolase (sEH), from which AFA-281 was identified and selected for clinical development.[4] According to Patsnap Synapse, there are "100 Patents (Medical) associated with AFA-281," although detailed access to this patent portfolio requires a login.[2] A strong and broad intellectual property position is crucial for securing investment, forming strategic partnerships, and ensuring future commercial exclusivity.

9.2. FDA Investigational New Drug (IND 157314) Status

A significant regulatory milestone was achieved when the U.S. Food and Drug Administration (FDA) accepted AfaSci's Investigational New Drug application (IND 157314) for AFA-281. This acceptance, dated November 11, 2022, was for a primary indication in neuropathic pain.[4] The IND application was supported by a comprehensive package of preclinical data, including GLP safety pharmacology, toxicology, and genotoxicity studies.[4] Some documentation also refers to AfaSci developing an "FDA-accepted investigational new drug (IND) AFA-281 for alcohol use disorder".[12] This likely indicates the company's intent to leverage the existing IND 157314 framework to expand into the AUD indication, a common practice, or potentially a separate IND filing for AUD, though the former is more typical for early exploratory work in new indications with an already active IND for the same compound.

The acceptance of an IND by the FDA allows a company to initiate clinical trials in human subjects in the United States. It signifies that the agency has reviewed the available preclinical data on the drug's pharmacology, toxicology, and manufacturing and has deemed it sufficiently safe to proceed with human testing for the proposed indication.

AfaSci's strategy of utilizing the active IND for neuropathic pain (IND 157314) as a foundation to explore additional CNS indications such as AUD and CUD [4] is a resource-efficient approach. It allows the company to build upon the established safety database and regulatory interactions associated with the initial IND. This can potentially streamline and accelerate the pathway to clinical proof-of-concept for these new, mechanistically related indications, as the foundational safety data from Phase I studies under the primary IND can often support early-phase exploratory trials in other relevant patient populations. This approach implies a degree of confidence from the FDA in the general safety profile of AFA-281 as established through the initial IND review process.

10. Expert Analysis, Future Outlook, and Recommendations

AFA-281, developed by AfaSci, Inc., stands at a promising juncture in its clinical development. Having successfully completed Phase I trials for neuropathic pain and secured FDA acceptance of IND 157314, the compound is poised for further evaluation in Phase II studies for lumbosacral radiculopathy and Alcohol Use Disorder, alongside a critical Phase I drug-alcohol interaction study. The novel dual mechanism of action, targeting both Cav3 T-type calcium channels and soluble epoxide hydrolase, offers a scientifically compelling rationale for its potential efficacy in complex CNS disorders characterized by neuronal hyperexcitability and neuroinflammation. The preclinical data package appears robust, demonstrating analgesic and anti-addictive properties in relevant animal models, favorable oral pharmacokinetics including CNS penetration, and a good preclinical safety profile, notably lacking cardiac toxicity signals often associated with ion channel modulators.

The strategic decision to pursue indications such as neuropathic pain, AUD, and CUD aligns with significant unmet medical needs. The non-opioid, non-steroidal nature of AFA-281 is a particularly strong differentiator in the pain space, given the ongoing opioid crisis. Furthermore, its potential to address comorbidities often associated with substance use disorders, such as chronic pain and negative affective states, could offer a significant therapeutic advantage if validated in clinical settings.

Key Future Milestones:

The trajectory of AFA-281 will be significantly shaped by several upcoming milestones:

  1. Successful Enrollment and Execution of Planned Clinical Trials: Timely recruitment and completion of the Phase II trial in painful lumbosacral radiculopathy (NCT06649747) and the Phase IIa trial in AUD (NCT06710431) are paramount.
  2. Positive Outcomes from the Drug-Alcohol Interaction Study: The results from NCT06719908 will be critical for the AUD program, informing safety and dosing in patients who may consume alcohol.
  3. Demonstration of Clinical Proof-of-Concept: Clear evidence of efficacy and continued safety in the target patient populations from the Phase II studies will be essential for further development and for attracting partners.
  4. Securing Strategic Partnerships and Funding: As AfaSci has indicated [1], securing partnerships or additional funding will be crucial for financing the larger and more expensive Phase IIb and pivotal Phase III trials required for regulatory approval.

Potential Challenges:

Despite the promising profile, AFA-281 faces several challenges inherent in drug development:

  1. Translation of Preclinical Efficacy: The translation of efficacy observed in animal models, particularly for complex CNS disorders like AUD and CUD, to human clinical success is a significant hurdle for many investigational drugs.
  2. Long-Term Safety and Tolerability: While initial Phase I and preclinical safety data are positive, the long-term safety and tolerability of a dual-target modulator in larger, more diverse patient populations over extended treatment durations need to be established.
  3. Regulatory Pathway Navigation: For novel mechanisms and indications like AUD/CUD, which may have evolving regulatory endpoints and expectations, proactive and clear communication with regulatory agencies (FDA, EMA, etc.) will be essential.
  4. Market Access and Competition: Demonstrating a compelling clinical and pharmacoeconomic value proposition will be necessary to gain market access and compete effectively against existing therapies and other emerging agents.

AfaSci has successfully navigated AFA-281 through early discovery and initial human safety testing. The transition to Phase II, often termed the "valley of death" in biotechnology due to high attrition rates and significant costs, represents the next major challenge. The company's explicit strategy of seeking investment and partnerships for these Phase II studies [1] is a pragmatic recognition of these hurdles. The quality and strength of the data emerging from the planned Phase II trials will be the primary determinant in attracting such collaborations and securing the resources needed for late-stage development.

While the preclinical pharmacokinetic and pharmacodynamic data are encouraging, the establishment of robust human PK/PD relationships and optimal dose ranges across different indications remains a critical ongoing task. The completed Phase I trial would have provided initial human PK data, but the specifics are not available in the reviewed materials. The upcoming Phase I drug-alcohol interaction study and the Phase II trials are therefore vital for confirming the PK profile in relevant patient populations, understanding pharmacodynamic effects (including potential biomarkers of target engagement), and refining dose selection for any future pivotal studies. Establishing a clear therapeutic window that balances efficacy with tolerability will be key to AFA-281's ultimate success.

Strategic Recommendations:

To maximize the potential of AFA-281, the following strategic considerations are recommended:

  1. Rigorous Phase II Trial Design: Ensure that the Phase II studies (NCT06649747, NCT06710431) are robustly designed with clearly defined, clinically meaningful primary and secondary endpoints, appropriate patient selection criteria, and adequate statistical power to provide a clear go/no-go decision for Phase III.
  2. Mechanistic Elucidation in Humans: Where feasible, incorporate biomarker studies into clinical trials to further elucidate AFA-281's dual mechanism of action in human subjects, confirm target engagement, and potentially identify patient subpopulations most likely to respond.
  3. Proactive Regulatory Engagement: Maintain open and proactive dialogue with regulatory authorities (e.g., FDA, EMA) regarding the clinical development plans, particularly for the novel indications like AUD and CUD, to ensure alignment on trial designs and endpoints.
  4. Pharmacoeconomic Value Proposition: Begin developing a comprehensive pharmacoeconomic model to clearly articulate the value proposition of AFA-281 to payers and healthcare systems, especially highlighting its potential to reduce opioid use or manage complex comorbidities.
  5. Aggressively Pursue Strategic Partnerships: Continue to actively seek strategic collaborations or licensing opportunities with larger pharmaceutical companies that possess the resources and expertise for late-stage development, regulatory submission, and global commercialization, aligning with AfaSci's stated business model.[1]

In conclusion, AFA-281 is a promising NCE with a novel dual mechanism of action targeting significant unmet needs in pain and CNS disorders. Its development is supported by a solid preclinical foundation and a clear regulatory path into Phase II studies. The successful execution of these upcoming trials and the ability to secure strategic partnerships will be critical determinants of its future therapeutic and commercial success.

Works cited

  1. About - AfaSci, accessed June 4, 2025, https://www.afasci.com/index.php/about-menu-2
  2. AFA-281 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed June 4, 2025, https://synapse.patsnap.com/drug/dbec121c03dd4b619c6de434cbabfc66
  3. AFA-281 by AfaSci for Neuropathic Pain (Neuralgia): Likelihood of Approval, accessed June 4, 2025, https://www.pharmaceutical-technology.com/data-insights/afa-281-afasci-neuropathic-pain-neuralgia-likelihood-of-approval/
  4. Company - | SBIR, accessed June 4, 2025, https://www.sbir.gov/portfolio/80919
  5. Developing a novel treatment of cocaine use disorder using an IND dual inhibitor of Cav3 channel and soluble epoxide hydrolase - NIH RePORTER, accessed June 4, 2025, https://reporter.nih.gov/project-details/10786151
  6. Clinical Phase I trials on an IND single molecule ... - NIH RePORTER, accessed June 4, 2025, https://reporter.nih.gov/search/Q16CRa99xUK-bwp7-IaBhg/project-details/10923984
  7. NINDS Exploratory Clinical Trials Small Business (R43 R44 Clinical, accessed June 4, 2025, https://www.highergov.com/grant-opportunity/ninds-exploratory-clinical-trials-for-small-business-r43-r44-clinical-trial-req-334776/
  8. A Phase I Study of the Interaction of Alcohol With Oral AFA-281 in Healthy Volunteers, accessed June 4, 2025, https://ctv.veeva.com/study/a-phase-i-study-of-the-interaction-of-alcohol-with-oral-afa-281-in-healthy-volunteers
  9. AfaSci, Inc. - Drug pipelines, Patents, Clinical trials - Patsnap Synapse, accessed June 4, 2025, https://synapse.patsnap.com/organization/4be06ae904e19a1d3cf6c2902947de31
  10. Efficacy Study With AFA-281 for the Treatment of Back Pain, accessed June 4, 2025, https://ctv.veeva.com/study/efficacy-study-with-afa-281-for-the-treatment-of-back-pain
  11. A Study of the Safety and Exploratory Efficacy of Oral AFA-281 in Patients with Alcohol Use Disorder - ClinConnect, accessed June 4, 2025, https://clinconnect.io/trials/NCT06710431
  12. Development of a Novel Medication for Alcohol Use Disorder with an Active IND Dual Inhibitor of T-Type Calcium Channel and Soluble Epoxide Hydrolase - NIH RePORTER, accessed June 4, 2025, https://reporter.nih.gov/search/15E1C00B4E8AC4D47598B8961CAA4A01A2FFCEB861BF/project-details/10933482
  13. Developing a novel treatment of cocaine use disorder using an IND dual inhibitor of Cav3 channel and soluble epoxide hydrolase - HigherGov, accessed June 4, 2025, https://www.highergov.com/grant/R43DA059524/
  14. Xinmin XIE | CEO | M.D., Ph.D. | Therapeutics | Research profile - ResearchGate, accessed June 4, 2025, https://www.researchgate.net/profile/Xinmin-Xie
  15. Development of a Novel Medication for Alcohol Use Disorder with an Active IND Dual Inhibitor of T-Type Calcium Channel and Soluble Epoxide Hydrolase - NIH RePORTER, accessed June 4, 2025, https://reporter.nih.gov/project-details/10815882

Published at: June 4, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.