MedPath

kamuvudine-9 Advanced Drug Monograph

Published:May 28, 2025

Generic Name

kamuvudine-9

Kamuvudine-9: A Comprehensive Report on a Novel Inflammasome Inhibitor

I. Introduction and Overview of Kamuvudine-9

Kamuvudine-9, a novel therapeutic agent under development, represents a focused effort to address a range of debilitating degenerative and inflammatory diseases. This report synthesizes available information on its chemical identity, the rationale behind its development, its proposed mechanism of action, preclinical findings, ongoing clinical investigations, and its intellectual property and regulatory landscape. The primary developer of Kamuvudine-9 is Inflammasome Therapeutics.[1]

A. Nomenclature and Chemical Identity

Kamuvudine-9 is the primary designation for this investigational compound, often referred to by the shorthand K-9 in scientific literature and company communications.[2] An important synonym identified in patent literature is 3Et-3TC [14], which indicates its nature as a tri-ethyl derivative of lamivudine (3TC), a well-known nucleoside reverse transcriptase inhibitor (NRTI).

Chemically, Kamuvudine-9 is classified as an NRTI derivative; specifically, it is an alkylated derivative of lamivudine.[1] The definitive chemical structure and name for 3Et-3TC (Kamuvudine-9) are provided in patent documents, such as WO2016138425A1 and EP4215527NWB1, as 4-(diethylamino)-1-[2-(ethoxymethyl)-1,3-oxathiolan-5-yl]-1,2-dihydropyrimidin-2-one.[14]

The molecular weight (MW) of Kamuvudine-9 (K-9), consistent with its identity as an alkylated lamivudine derivative, is reported as approximately 271.34 g/mol.[6] This is distinct from the parent compound lamivudine, which has a molecular weight of 229.26 g/mol.[18]

A specific CAS (Chemical Abstracts Service) Registry Number for Kamuvudine-9 or its synonym 3Et-3TC has not been explicitly identified in the reviewed research materials.[3] While CAS numbers for related compounds, such as lamivudine itself (134678-17-4 [18]), are available, a unique CAS identifier for Kamuvudine-9 was not found. Such information would typically be detailed in comprehensive synthesis publications or dedicated chemical databases, which were not fully accessible or did not contain this specific information within the scope of the provided snippets.

The unambiguous chemical identification of a novel therapeutic agent like Kamuvudine-9 is of paramount importance in drug development. It ensures precise communication among researchers, regulators, and clinicians, and is essential for patent protection, manufacturing consistency, and accurate scientific reporting. The consolidation of its various names, chemical class, full chemical name, structural information [14], and molecular weight provides a clear chemical fingerprint for the compound.

Table 1: Kamuvudine-9 - Key Identifying Information

Identifier TypeInformationSource Snippet(s)
Primary NameKamuvudine-92
Common AbbreviationK-92
Synonym3Et-3TC14
Chemical ClassNucleoside Reverse Transcriptase Inhibitor (NRTI) Derivative; Alkylated Lamivudine Derivative1
Full Chemical Name (for 3Et-3TC / Kamuvudine-9)4-(diethylamino)-1-[2-(ethoxymethyl)-1,3-oxathiolan-5-yl]-1,2-dihydropyrimidin-2-one14
Molecular Weight~271.34 g/mol6
CAS Registry NumberNot explicitly found in provided snippets for Kamuvudine-9/3Et-3TC14
StructureDescribed and depicted in patent WO2016138425A114

B. Developer

Kamuvudine-9 is being developed by Inflammasome Therapeutics.[1] The company was co-founded by Dr. Jayakrishna Ambati, a prominent researcher in ophthalmology and inflammasome biology, and Dr. Paul Ashton, who serves as President and CEO.[4] This leadership, rooted in scientific discovery, suggests a strong research-driven approach to the company's therapeutic programs. Identifying the developer is essential for understanding the strategic direction, resource allocation, and potential for collaborations or licensing related to Kamuvudine-9's progression through the development pipeline.

C. Rationale for Development

The development of Kamuvudine-9 and the broader class of Kamuvudines is predicated on the hypothesis that chronic, aberrant inflammasome activation is a central pathological driver in a multitude of prevalent and often debilitating degenerative diseases.[4] Inflammasome Therapeutics is targeting a wide array of conditions, including ocular diseases such as age-related macular degeneration (AMD), particularly its advanced form Geographic Atrophy (GA), and Thyroid Eye Disease (TED, also known as Graves' Ophthalmopathy). The pipeline also extends to neurodegenerative disorders like Alzheimer's disease (AD), Parkinson's disease (PD), Multiple Sclerosis (MS), and Amyotrophic Lateral Sclerosis (ALS), as well as autoimmune conditions such as lupus.[1]

This broad therapeutic strategy stems from the understanding that inflammasome activation acts as a "final common pathway" for cellular damage triggered by diverse toxic stimuli in these multifactorial diseases.[4] This approach is distinct from many previous therapeutic attempts that focused on single causative elements (e.g., amyloid-β in AD or specific complement factors in GA), which have often yielded disappointing results in large-scale clinical trials.[4] By targeting the convergent point of inflammasome activation, Kamuvudines aim to offer a more comprehensive and potentially more effective treatment modality.

The genesis of the Kamuvudine program is a noteworthy example of rational drug discovery evolving from observations of existing medicines. Research from Dr. Ambati's laboratory, published in Science in 2014, first identified that certain FDA-approved NRTIs, used for HIV and hepatitis B treatment, possess potent inflammasome-inhibiting properties.[24] This discovery was further supported by epidemiological studies analyzing large health claims databases, which revealed a significantly reduced risk of developing AD, GA, type 2 diabetes, and MS among patients taking NRTIs for their primary viral indications.[24]

However, the clinical utility of parent NRTIs for these chronic degenerative and inflammatory conditions is severely hampered by their known side effects, primarily mitochondrial toxicity.[6] This toxicity is understood to be linked to their intracellular metabolism and off-target inhibition of host cellular polymerases, mechanisms crucial for their antiviral efficacy but detrimental for long-term systemic use in non-viral diseases. Recognizing this limitation, Inflammasome Therapeutics embarked on a medicinal chemistry program to create derivatives of NRTIs – the Kamuvudines. These new chemical entities, including Kamuvudine-9 (an alkylated derivative of lamivudine), were specifically engineered to retain the desirable anti-inflammasome activity of the parent NRTIs while being devoid of the metabolic pathways or structural features that lead to mitochondrial toxicity and reverse transcriptase inhibition.[6] Preclinical studies have reportedly demonstrated that Kamuvudines are over 1,000 times less toxic than their parent NRTI molecules, suggesting a significantly improved therapeutic index and suitability for chronic administration.[24] This strategic chemical modification to uncouple desired therapeutic effects from unwanted toxicities forms the core rationale for the development of Kamuvudine-9.

II. Mechanism of Action

The therapeutic strategy underpinning Kamuvudine-9 centers on its ability to modulate the innate immune response by inhibiting inflammasome activation, a key process in the generation of inflammation.

A. Primary Target: Inflammasome Inhibition

Kamuvudine-9, along with other members of the Kamuvudine class, functions as a potent inhibitor of inflammasome activation.[2] These compounds are specifically described as "dual inflammasome inhibitors" [4], targeting two key inflammasome sensor proteins: NLRC4 (NOD-like receptor family CARD domain containing 4) and NLRP3 (NOD-like receptor family pyrin domain containing 3).[2] This dual inhibitory action is a distinguishing feature of the Kamuvudine class, including K-8 (SOM-401), which also targets both NLRP3 and NLRC4.[26]

Inflammasomes are multi-protein complexes that, upon activation by various pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs), lead to the activation of caspase-1. Activated caspase-1 then cleaves pro-inflammatory cytokines pro-IL-1β and pro-IL-18 into their mature, active forms, which are subsequently secreted and drive inflammatory responses.[6] By inhibiting both NLRP3 and NLRC4, Kamuvudine-9 aims to block this critical cascade at an upstream point, thereby preventing the release of these potent inflammatory mediators. It is important to note that this anti-inflammatory activity is independent of the reverse transcriptase inhibition characteristic of the parent NRTI molecules.[6]

The strategy of dual NLRP3 and NLRC4 inhibition is based on an evolving understanding of inflammasome biology. While NLRP3 has long been recognized as a key player in sterile inflammation and various chronic diseases, research highlighted by Inflammasome Therapeutics (citing publications in Science Immunology) suggests that NLRC4 can cooperate with NLRP3, potentially forming a "dual inflammasome" complex, and that this combined activity may be essential for the pathogenesis of certain inflammasome-mediated diseases.[27] Consequently, targeting only NLRP3, as some other therapeutic approaches do, might not be sufficient for optimal therapeutic effect in diseases where multiple inflammasome pathways are active.[10] Kamuvudines, by inhibiting both sensor proteins, are designed to offer a more comprehensive blockade of this "final common pathway" of inflammation driven by diverse upstream triggers such as complement activation products, retrotransposons, amyloid-β, iron overload, and reactive oxygen species, particularly relevant in multifactorial diseases like GA.[4]

B. Decoupling from Antiviral Activity and Toxicity

A pivotal aspect of Kamuvudine-9's design is the successful separation of its anti-inflammasome activity from the antiviral mechanism and associated toxicities of its parent NRTI compounds. Kamuvudines are chemically modified NRTIs; for instance, K-9 is an alkylated derivative of lamivudine.[6] These modifications were specifically introduced to prevent the intracellular metabolism or interaction with host cellular polymerases that lead to mitochondrial toxicity, a well-documented side effect of long-term NRTI use.[6] The antiviral activity of NRTIs relies on their ability to be phosphorylated and subsequently inhibit viral reverse transcriptase and/or host DNA polymerases (leading to toxicity). Kamuvudines are engineered to largely bypass these steps, thus retaining their ability to inhibit inflammasome activation without exerting significant antiviral effects or causing the related mitochondrial damage.[6]

Preclinical studies have reportedly demonstrated that Kamuvudines are more than 1,000 times less toxic than their parent NRTI molecules.[24] This dramatic improvement in the safety profile is fundamental to their therapeutic rationale, as it potentially allows for chronic administration in non-life-threatening degenerative and inflammatory conditions, where the risk-benefit assessment for unmodified NRTIs would be unfavorable. This successful uncoupling of a desired pharmacological effect from the primary mechanism and toxicity of a parent drug class through rational chemical modification represents a significant advance. It suggests that therapeutically relevant inflammasome inhibition can be achieved with Kamuvudines at exposures far below those that would induce the characteristic toxicities of NRTIs, thereby widening the therapeutic window considerably. This achievement could serve as a model for "detoxifying" or repurposing other drug classes where off-target effects or primary mechanism-related toxicities limit their broader clinical application.

C. Molecular Interactions and Binding Sites

The precise molecular binding sites of Kamuvudine-9 on the NLRP3 and NLRC4 proteins are not explicitly detailed in the reviewed research snippets. The mechanism is primarily described in functional terms: preventing the assembly and activation of these inflammasome complexes.[6]

For the parent NRTIs, some insights into potential interaction sites with NLRP3 have been hypothesized. For example, research on fluoxetine, another compound found to inhibit the NLRP3 inflammasome, suggested that it binds within the NACHT domain of NLRP3, specifically in a nucleotide-binding cavity that can accommodate ATP/ADP.[29] Given that NRTIs are modified nucleoside analogs, it has been speculated that they too might interact with such nucleotide-binding sites on inflammasome proteins.[29] However, this remains a hypothesis for NRTIs in the context of inflammasome inhibition and has not been directly confirmed for Kamuvudines in the provided materials.

A more detailed understanding of the specific molecular interactions and binding pockets of Kamuvudines on NLRP3 and NLRC4 would be valuable for several reasons. It would further solidify the mechanistic understanding, could guide the structure-based design of even more potent or selective next-generation Kamuvudines, and might help in predicting or understanding potential off-target interactions. Currently, the available information focuses on the functional outcome of inflammasome inhibition rather than the atomistic details of the drug-target interaction. Further research, potentially involving co-crystallography, computational docking studies specifically with Kamuvudines, or detailed biochemical binding assays, would be necessary to elucidate these precise molecular interactions.

III. Preclinical Development

The preclinical development of Kamuvudine-9 has provided foundational evidence for its anti-inflammasome activity and therapeutic potential across various disease models.

A. In Vitro Studies

Cell culture studies have been instrumental in demonstrating the direct effects of Kamuvudines on inflammasome activation. Inflammasome Therapeutics reports that their Kamuvudine series successfully inhibits inflammasome activation in such models.[24] More specifically, patent documentation (WO2016138425A1) details experiments with 3Et-3TC, the chemical entity identified as Kamuvudine-9. In these studies, 3Et-3TC was shown to inhibit ATP-induced Caspase-1 activation in mouse J774 iGLuc cells in a dose-dependent manner. Notably, the patent suggests that 3Et-3TC provided a greater reduction in inflammasome activation compared to the parent NRTI, d4T (stavudine), indicating potentially enhanced potency or efficacy of the derivative in directly blocking this key inflammatory pathway.[14] These in vitro findings established the initial proof-of-concept for Kamuvudine-9's mechanism and its superiority over at least one parent NRTI, justifying its progression to more complex in vivo models.

B. In Vivo Animal Models and Efficacy Data

Kamuvudine-9 has been evaluated in several in vivo animal models, yielding promising results in the context of both retinal and neurodegenerative diseases.

1. Retinal Diseases

  • Rhegmatogenous Retinal Detachment (RRD): RRD is a serious condition where physical separation of the neurosensory retina from the RPE can lead to photoreceptor cell death and vision loss. In a mouse model of RRD, systemic (intraperitoneal, IP) administration of Kamuvudine-9 demonstrated significant neuroprotective and anti-inflammatory effects. Treatment with K-9 led to a reduction in retinal inflammasome activation, as evidenced by decreased levels of cleaved caspase-1 and the pro-inflammatory cytokine IL-18. Concurrently, there was a significant reduction in photoreceptor cell death, measured by TUNEL staining. A high-dose regimen of K-9 (K-9H: 60 mg/kg IP, twice daily) was found to provide superior photoreceptor protection compared to an equimolar dose of the parent NRTI, lamivudine (3TC), with protection rates of 56% ± 7% for K-9H versus 38% ± 10% for 3TC (P < 0.05). Additionally, K-9 treatment inhibited the cleavage of caspase-8, another molecule implicated in apoptosis and inflammasome signaling.6 Further studies using a more clinically relevant mouse model of RRD that incorporates spontaneous retinal reattachment (SRR) showed that K-9 (60 mg/kg IP, twice daily for 10 days post-RRD induction) not only preserved retinal electrical function (assessed by electroretinography a- and b-wave amplitudes) during the period of detachment but also facilitated an improvement in function following spontaneous reattachment, outperforming PBS-treated controls.6 These findings suggest that K-9 could serve as an adjunctive therapy to surgical repair in RRD, protecting retinal cells from irreversible damage during the critical period before and after intervention.
  • Age-Related Macular Degeneration (AMD) / Geographic Atrophy (GA): Dry AMD, particularly its advanced stage GA, is a leading cause of irreversible blindness characterized by RPE cell death. Preclinical research indicates that Kamuvudines can inhibit RPE degeneration induced by amyloid-β oligomers (AβOs), a component of drusen implicated in AMD pathogenesis. This protective effect is linked to the inhibition of the P2RX7 receptor, an upstream mediator of NLRP3 inflammasome activation.[6] Another line of research has identified that the reverse transcription of endogenous Alu RNA to cDNA within RPE cells triggers inflammasome activation and cell death in human GA. This pathological process appears localized to the border of the atrophic lesion. Kamuvudines have been shown in preclinical models to halt this Alu RNA-driven inflammasome activation and subsequent disease progression.[25] These studies highlight K-9's potential to interfere with multiple inflammasome-triggering pathways relevant to dry AMD/GA.

2. Neurodegenerative Diseases

  • Alzheimer's Disease (AD): The potential of Kamuvudine-9 in AD has been investigated using the 5xFAD mouse model, which exhibits age-dependent Aβ deposition and cognitive decline. In 24-week-old 5xFAD mice, a 12-week course of K-9 treatment (60 mg/kg IP, twice daily) resulted in a significant reduction in Aβ plaque burden in key brain regions, including the frontal cortex, motor cortex, and hippocampus. Perhaps more remarkably, K-9 treatment not only prevented further cognitive decline but also reversed existing spatial memory and learning deficits. The cognitive performance of K-9-treated 36-week-old 5xFAD mice was superior to that of age-matched PBS-treated controls and, importantly, improved beyond their own pre-treatment baseline at 24 weeks, reaching levels comparable to those of young, healthy wild-type mice.[3] These striking results suggest that K-9 may offer both disease-modifying (Aβ reduction) and symptom-reversing (cognitive improvement) effects in AD by targeting inflammasome-mediated neuroinflammation.

The consistent efficacy demonstrated by Kamuvudine-9 across these diverse preclinical models—spanning different organs (eye, brain) and pathological triggers (physical injury, Aβ accumulation, Alu RNA)—provides robust support for the therapeutic hypothesis that inflammasome inhibition is a viable strategy for a range of complex diseases.

Table 2: Summary of Key Preclinical Efficacy Data for Kamuvudine-9

Disease ModelKamuvudine-9 Dosage & Administration RouteKey Efficacy Endpoints AssessedSummary of Key Results (vs. Control)Source Snippet(s)
Mouse Rhegmatogenous Retinal Detachment (RRD)K-9H: 60 mg/kg IP, twice daily; K-9L: 90 mg/kg IP, once dailyCleaved caspase-1, IL-18 levels, TUNEL+ photoreceptors, Caspase-8 cleavageSignificant reduction in inflammasome activation markers and photoreceptor death. K-9H showed 56% photoreceptor protection vs. 38% for equimolar 3TC.6
Mouse RRD with Spontaneous Reattachment (RRD/SRR)60 mg/kg IP, twice daily (days 0-10)ERG a- and b-wave amplitudesProtected retinal electrical function during detachment and improved function after reattachment compared to PBS.6
Mouse Model of Alzheimer's Disease (5xFAD mice)60 mg/kg IP, twice daily for 12 weeksAβ deposition (frontal cortex, motor cortex, hippocampus); Cognitive performance (Morris water maze - spatial memory and learning)Reduced Aβ deposition. Reversed existing cognitive deficits; performance improved to levels comparable to young wild-type mice and superior to pre-treatment baseline.3
In vitro RPE degeneration model (AβO-induced)Not specified for K-9, but Kamuvudines generallyRPE degeneration, NLRP3 inflammasome activationKamuvudines inhibit AβO-induced RPE degeneration by blocking P2RX7-NLRP3 pathway.6
In vitro RPE degeneration model (Alu RNA-induced)Not specified for K-9, but Kamuvudines generallyInflammasome activation, RPE cell deathKamuvudines halt inflammasome activation and disease progression.25
In vitro Caspase-1 activation (mouse J774 cells)1 nM - 100 µM (for 3Et-3TC/K-9)Caspase-1 activation (Luciferase cleavage)Dose-dependent inhibition of ATP-induced Caspase-1 activation; greater reduction compared to parent NRTI d4T.14

C. Safety and Toxicology (Preclinical)

A cornerstone of the Kamuvudine development program is the significantly enhanced preclinical safety profile compared to the parent NRTI compounds. Kamuvudines, including K-9, were specifically engineered to circumvent the mechanisms responsible for NRTI-associated mitochondrial toxicity, primarily by altering their interaction with host cellular polymerases and their intracellular metabolic fate.[6] This targeted chemical modification has reportedly resulted in Kamuvudines being over 1,000 times less toxic than their parent molecules in a battery of preclinical tests.[24]

This substantial reduction in toxicity is critically important. It suggests that the anti-inflammasome effects of Kamuvudines can be achieved at concentrations far below those that would elicit the characteristic side effects of NRTIs. This improved therapeutic index is what makes Kamuvudines potentially suitable for long-term administration in chronic, non-life-threatening conditions such as GA, AD, or TED, where the chronic use of original NRTIs would be untenable due to their safety liabilities. The successful decoupling of the desired anti-inflammatory activity from the dose-limiting toxicities of the NRTI scaffold represents a significant medicinal chemistry achievement and underpins the rationale for advancing Kamuvudines into clinical development.

IV. Clinical Development

Inflammasome Therapeutics is advancing its Kamuvudine platform into clinical trials, with distinct strategies for different candidates and indications. K-9, designed for oral administration and CNS/retina penetration, is being evaluated for neuroinflammatory and systemic conditions, while K-8, another Kamuvudine, is formulated as an intravitreous implant for localized treatment of retinal diseases like Geographic Atrophy.[4]

B. Focus: Kamuvudine-9 in Graves' Ophthalmopathy (Thyroid Eye Disease - TED) - Clinical Trial NCT06467435

The lead clinical program for Kamuvudine-9 is a Phase I study in patients with Graves' Ophthalmopathy, also known as Thyroid Eye Disease (TED). This trial is registered under the identifier NCT06467435 and is titled "Evaluation of K9 in Subjects with Thyroid Eye Disease (TED)".[1]

The primary objectives of this study are to evaluate the plasma pharmacokinetics (PK) of K-9 in healthy subjects (Cohort 1) and to assess the safety and preliminary treatment efficacy of K-9 in patients with active TED (Cohort 2).[5] It is a non-randomized, open-label study with a planned duration of 6 weeks per participant.[5] Inflammasome Therapeutics announced the completion of enrollment for this Phase I trial in the USA on September 10, 2024 [2], indicating that initial data may become available in late 2024 or early 2025. The estimated total enrollment is 8 participants, aged 18 to 75 years.[5]

The intervention involves Kamuvudine-9 administered as oral tablets. Healthy volunteers in Cohort 1 receive a single oral dose of 96 mg K-9 (potentially weight-based). Patients with active TED in Cohort 2 receive 96 mg K-9 tablets orally twice a day for 4 weeks.[5]

Key inclusion criteria for TED patients (Cohort 2) include a confirmed diagnosis of TED, symptomatic disease diagnosed no more than 9 months prior, and a Clinical Activity Score (CAS) of ≥ 3 (on a 7-point scale) in the more affected eye. Exclusion criteria include body weight under 55 kg, prior treatment with teprotumumab, orbital radiotherapy or surgery, recent systemic corticosteroid or immunosuppressant use, uncontrolled diabetes or hypertension, significant psychiatric disorders, and hepatic or renal impairment.[5]

Primary outcome measures are the frequency of ocular or systemic adverse events (safety) and the plasma concentrations of K-9 (PK in Cohort 1). Secondary outcome measures, primarily for the TED patient cohort, are comprehensive and designed to assess efficacy across multiple domains. These include changes from baseline in: the Standardized Patient Evaluation of Eye Dryness (SPEED) questionnaire score, diplopia (via Bahn-Gorman Scale), CAS, proptosis (via Hertel exophthalmometry), Graves' Ophthalmopathy-specific Quality-of-Life (GO-QOL) scale score, upper and lower eyelid retraction (MRD1 and MRD2), and levels of inflammasome output cytokines IL-1β and IL-18 in plasma (measured by ELISA).[5]

The lead sponsor for NCT06467435 is listed as Peter Timoney [5], though Inflammasome Therapeutics is the developer of K-9 and is intrinsically linked to its clinical evaluation. No results or publications from this specific trial were available in the provided research materials.

The design of this initial Phase I study, incorporating both healthy volunteer PK assessment and early evaluation of safety and efficacy signals in TED patients, represents an efficient approach to early clinical development. This strategy allows for the concurrent gathering of critical human data on drug disposition and therapeutic potential, which can accelerate decision-making for subsequent, larger Phase II trials. The selection of TED as an initial clinical target for an oral, CNS-penetrant inflammasome inhibitor like K-9 appears strategic. TED is an accessible condition with well-defined and measurable inflammatory and structural outcomes (e.g., CAS, proptosis). Positive data in TED could provide strong proof-of-concept for K-9's anti-inflammatory and potential neuroprotective effects, thereby de-risking its development for more complex and challenging CNS indications like Alzheimer's disease or MS, for which K-9 is also envisioned due to its favorable administration route and penetration characteristics.[4]

Table 3: Detailed Summary of Clinical Trial NCT06467435 for Kamuvudine-9 in Thyroid Eye Disease

FeatureDetailsSource Snippet(s)
Trial IdentifierNCT064674355
Official TitleEvaluation of K9 in Subjects with Thyroid Eye Disease (TED)5
PhasePhase I1
Current StatusEnrollment complete (as of Sep 10, 2024, per Inflammasome Therapeutics)2
Study Objective(s)Evaluate plasma PK of K9 in healthy subjects (Cohort 1); Evaluate safety and treatment efficacy of K9 in active TED patients (Cohort 2)5
Study DesignNon-randomized, open-label, two cohorts; Planned duration 6 weeks per participant5
Patient PopulationHealthy Volunteers (Cohort 1); Active TED patients (Cohort 2); Estimated N=8 total; Ages 18-755
Intervention - Cohort 1 (Healthy Volunteers)Drug: Kamuvudine-9 (K9) oral tablets; Dose: Single 96 mg dose (potentially weight-based); Route: Oral; Duration: Single dose5
Intervention - Cohort 2 (TED Patients)Drug: Kamuvudine-9 (K9) oral tablets; Dose: 96 mg; Route: Oral, twice a day; Duration: 4 weeks5
Primary Outcome Measures1. Frequency of ocular or systemic adverse events (Safety). 2. Plasma concentrations of K9 (PK in Cohort 1).32
Key Secondary Outcome Measures (Cohort 2)Change from baseline in: SPEED score, Diplopia (Bahn-Gorman Scale), CAS, Proptosis (Hertel exophthalmometry), GO-QOL score, Upper/Lower eyelid retraction (MRD1/MRD2), Plasma IL-1β and IL-18 levels.5
Key Inclusion Criteria (TED Cohort)Diagnosed TED; Symptomatic TED ≤ 9 months; CAS ≥ 3 (worse eye).5
Key Exclusion Criteria (TED Cohort)Weight < 55 kg; Prior teprotumumab, radiotherapy, orbital surgery; Recent systemic corticosteroids/immunosuppressants; Uncontrolled diabetes/hypertension; Significant psychiatric history; Hepatic/renal impairment.32
Lead Sponsor / Developer InvolvementLead Sponsor: Peter Timoney; Developer of K-9: Inflammasome Therapeutics.2

C. Other Potential Indications for K-9

Beyond TED, the preclinical efficacy of K-9 in models of Alzheimer's disease and retinal degeneration, coupled with its oral bioavailability and CNS/retina penetration, positions it as a candidate for a wide range of neuroinflammatory and neurodegenerative diseases. Inflammasome Therapeutics has indicated that K-9 is specifically designed for such neurological conditions and that clinical trials in diseases like ALS, Parkinson's disease, and MS are anticipated to commence following the initial Phase I study.[4] Preclinical investigations in broader autoimmune and neurodegenerative disorders were reportedly ongoing or completed prior to September 2024, further supporting this expanded development strategy.[2]

D. Pharmacokinetics (Human Data from NCT06467435)

The evaluation of the plasma pharmacokinetics of Kamuvudine-9 in healthy human subjects constitutes one of the primary objectives of the ongoing NCT06467435 trial.[5] This will provide crucial data on how the drug is absorbed, distributed, metabolized, and eliminated (ADME) in humans following oral administration. Such information is vital for determining appropriate dosing regimens for future studies, understanding potential drug-drug interactions, and assessing inter-subject variability. Specific human PK results from this trial are not yet available within the reviewed materials.

E. Safety and Tolerability (Human Data from NCT06467435)

Establishing the safety and tolerability of Kamuvudine-9 in humans is another primary objective of the NCT06467435 trial, encompassing both healthy volunteers and patients with TED.[5] Given the significantly improved preclinical safety profile of Kamuvudines compared to their parent NRTIs (over 1,000 times less toxic [24]), the human safety data will be closely watched. A favorable safety profile in this initial human study is paramount for the continued development of K-9 across its intended chronic indications. As with the PK data, specific safety and tolerability results from this trial are not yet publicly available in the provided snippets.

V. Intellectual Property

The development of Kamuvudine-9 and the broader Kamuvudine platform is supported by a portfolio of intellectual property.

A. Overview of Patent Landscape

Inflammasome Therapeutics has licensed a series of molecules, identified as Kamuvudines, for development.[24] The foundational patents related to these compounds and their therapeutic applications appear to originate from academic research institutions, with the University of Kentucky Research Foundation and the University of Virginia Patent Foundation listed as assignees on key patents. Prominent inventors consistently named on these patents include Dr. Jayakrishna Ambati, Benjamin Fowler, and Kameshwari Ambati.[3]

Several key patent documents have been identified:

  • WO2016138425A1 (filed February 26, 2016) and its U.S. counterpart US11219623B2 (granted January 11, 2022), titled "Compositions and methods for treating retinal degradation." These patents are central as they describe Kamuvudine-9 (specifically named as 3Et-3TC and 4-(diethylamino)-1-[2-(ethoxymethyl)-1,3-oxathiolan-5-yl]-1,2-dihydropyrimidin-2-one), its synthesis, and its utility in inhibiting inflammasome activation for treating retinal damage and degeneration.[14]
  • US10294220B2, titled "Compounds and compositions for inhibiting inflammasome activation".[41]
  • US12097201B2 (granted September 24, 2024), titled "Compositions and methods for treating retinal degradation".[42]
  • US11998547B2 (granted June 4, 2024), titled "Compositions and methods for treating multiple sclerosis".[42]

This patent estate is critical for protecting the novel chemical entities (the Kamuvudines) and their diverse therapeutic applications, forming the commercial foundation for Inflammasome Therapeutics' pipeline. The intellectual property strategy appears to encompass both the composition of matter for these modified NRTIs and their methods of use in a range of inflammasome-driven diseases, including retinal and neurodegenerative conditions. This broad protection is evident from the patent titles and abstracts which consistently refer to compounds, compositions, and methods for treating various pathologies linked to inflammasome activation.

B. Focus of Patents

The patents related to Kamuvudines generally cover:

  • The novel modified nucleoside reverse transcriptase inhibitors themselves (the Kamuvudine class of compounds).
  • Pharmaceutical compositions containing these compounds.
  • Methods of using these compounds and compositions for treating a variety of conditions associated with inflammasome activation. These include, but are not limited to, retinal damage and degeneration (such as dry and wet AMD, and GA), neurodegenerative diseases (like Alzheimer's disease, Parkinson's disease, Multiple Sclerosis), and methods for inhibiting inflammasome activation triggered by various pathological stimuli such as Alu RNA and ATP.[14]

This comprehensive patent coverage aims to secure exclusive rights to the Kamuvudine platform technology across its intended spectrum of therapeutic indications.

VI. Regulatory Status

Information regarding specific regulatory designations for Kamuvudine-9 is limited in the provided materials.

A. Orphan Drug Designation

According to AdisInsight, as of September 28, 2024, Kamuvudine-9 (K-9) does not hold Orphan Drug Designation from either the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA).[2] General FDA Freedom of Information Act (FOIA) logs [47] did not yield specific designation information for K-9.

Orphan Drug Designation is typically granted to drugs intended for rare diseases (e.g., prevalence <5 in 10,000 in the EU [46]) and provides various development and marketing incentives. The absence of this designation for Kamuvudine-9 for its current lead indication, Graves' Ophthalmopathy, could imply that either the company has not sought this status or that the defined patient population for TED does not meet the formal criteria for orphan designation in the targeted regulatory regions. Alternatively, the company's broader strategic focus on more prevalent neurodegenerative diseases might influence its pursuit of orphan designations at this early stage.

B. Other Regulatory Designations (FDA, EMA)

The provided research snippets do not mention any other specific regulatory designations for Kamuvudine-9, such as Fast Track, Breakthrough Therapy, or Priority Review, from the FDA or EMA.[4] While K-9 is described as a "newly developed inflammasome-blocking drug" with researchers calling for clinical trials [12], and K-8 (a related Kamuvudine) is noted in the context of GA where other drugs have received FDA approval [4], no formal expedited pathway designations for K-9 itself are apparent from the data. In the absence of such designations, Kamuvudine-9 would be expected to follow standard regulatory review pathways.

VII. Summary and Future Outlook

Kamuvudine-9 (K-9, 3Et-3TC) emerges as a promising investigational therapeutic with a novel mechanism of action and a potentially broad range of applications.

A. Recap of Kamuvudine-9 Profile

Kamuvudine-9 is an orally bioavailable, brain- and retina-penetrant derivative of the nucleoside reverse transcriptase inhibitor lamivudine. Developed by Inflammasome Therapeutics, it is engineered to function as a dual inhibitor of the NLRP3 and NLRC4 inflammasomes. This dual inhibition is believed to confer a comprehensive anti-inflammatory effect by targeting a common pathological pathway in various degenerative and inflammatory diseases. A key feature of Kamuvudine-9 is its significantly improved preclinical safety profile compared to parent NRTIs, particularly the mitigation of mitochondrial toxicity, which makes it a more viable candidate for chronic administration.

Strong preclinical efficacy has been demonstrated in diverse animal models, including those for rhegmatogenous retinal detachment (showing neuroprotection and functional improvement) and Alzheimer's disease (showing reduction in Aβ pathology and reversal of cognitive deficits). Kamuvudine-9 is currently in Phase I clinical development (NCT06467435) for Graves' Ophthalmopathy (Thyroid Eye Disease), with patient enrollment reported as complete in September 2024.

B. Potential Advantages and Differentiation

Kamuvudine-9 offers several potential advantages that differentiate it from existing therapies and other investigational agents:

  • Dual Inflammasome Inhibition: By targeting both NLRP3 and NLRC4 inflammasomes, K-9 may provide a more robust and comprehensive anti-inflammatory effect in complex, multifactorial diseases compared to inhibitors that target only a single inflammasome pathway.[10] This approach addresses a "final common pathway" implicated in numerous conditions.
  • Improved Safety Profile: The chemical modifications designed to eliminate the mitochondrial toxicity associated with parent NRTIs are a critical advantage. A reported >1000-fold reduction in preclinical toxicity [24] could translate to a favorable safety profile in humans, suitable for long-term treatment of chronic diseases.
  • Oral Administration and CNS/Retina Penetration: The oral tablet formulation of K-9 and its ability to penetrate the blood-brain and blood-retina barriers make it a particularly attractive candidate for a wide range of neuroinflammatory, neurodegenerative, and systemic autoimmune diseases where CNS or ocular involvement is common.[4] This contrasts with many biologic therapies that require parenteral administration.
  • Broad Therapeutic Potential: The targeting of a fundamental pathological mechanism (inflammasome activation) common to many diseases suggests that K-9 could have utility across a wide spectrum of indications with significant unmet medical needs.

C. Challenges and Next Steps

Despite its promise, the development of Kamuvudine-9 faces several challenges and requires clear next steps:

  • Clinical Validation: The immediate and most critical challenge is to demonstrate safety and efficacy in human clinical trials. The ongoing Phase I study in TED (NCT06467435) will provide the first human data on K-9's PK, safety, and preliminary efficacy signals. Positive results from this trial will be crucial for advancing to later-stage development.
  • Translation of Preclinical Efficacy: A common hurdle in drug development is ensuring that the robust efficacy observed in animal models translates to meaningful clinical benefit in human patients. The complexity of human diseases and inter-species differences can impact this translation.
  • Dose Optimization and Patient Selection: As development progresses, refining optimal dosing regimens and identifying specific patient populations or disease subtypes most likely to respond to K-9 will be essential for maximizing therapeutic benefit and designing successful pivotal trials. Biomarker development may play a role here.
  • Competitive Landscape: The field of inflammasome inhibition is an active area of research and development, with multiple companies pursuing various strategies and molecules targeting components of this pathway.[12] Kamuvudine-9 will need to demonstrate clear advantages in terms of efficacy, safety, or patient convenience to establish its place.
  • Long-Term Safety: For chronic conditions, establishing a favorable long-term safety profile with continuous oral administration will be paramount.

The successful clinical development of Kamuvudine-9 holds considerable potential. If its efficacy and safety are confirmed in humans, it could validate dual NLRP3/NLRC4 inflammasome inhibition as a significant therapeutic strategy for a host of currently intractable degenerative and inflammatory diseases. Given its oral bioavailability and CNS-penetrant properties, K-9 could be particularly impactful for neurological conditions like Alzheimer's disease, Parkinson's disease, and MS, where effective, conveniently administered treatments are urgently needed. Success in a challenging indication such as TED would provide strong impetus for its broader clinical exploration.

VIII. Conclusion

Kamuvudine-9 (K-9, 3Et-3TC) is an investigational oral medication being developed by Inflammasome Therapeutics. It is a chemically modified derivative of the nucleoside reverse transcriptase inhibitor lamivudine, engineered to function as a dual inhibitor of the NLRP3 and NLRC4 inflammasomes while significantly mitigating the mitochondrial toxicity associated with its parent NRTI class. This innovative mechanism of action targets a common pathogenic pathway implicated in a wide array of degenerative and inflammatory diseases.

Preclinical studies have demonstrated promising efficacy for Kamuvudine-9 in diverse animal models, including those for rhegmatogenous retinal detachment and Alzheimer's disease, where it has shown neuroprotective effects, reduction in disease-specific pathology (e.g., Aβ deposition), and functional improvements, including reversal of cognitive deficits. This robust preclinical data, combined with a markedly improved safety profile compared to traditional NRTIs, underpins its progression into clinical development.

The lead clinical program for Kamuvudine-9 is a Phase I trial (NCT06467435) evaluating its pharmacokinetics, safety, and preliminary efficacy in healthy volunteers and patients with active Thyroid Eye Disease (Graves' Ophthalmopathy). Enrollment for this study was reported as complete in September 2024. The outcomes of this trial will be crucial in establishing the human safety profile of K-9 and providing initial indications of its therapeutic potential.

With its novel dual inflammasome inhibitory mechanism, favorable preclinical safety and efficacy, and convenient oral administration route with CNS penetration, Kamuvudine-9 holds the potential to address significant unmet medical needs across a spectrum of ophthalmic, neurodegenerative, and autoimmune disorders. Successful clinical validation could position Kamuvudine-9 as a first-in-class therapy and validate a broader therapeutic strategy targeting inflammasome-driven pathology. Future development will depend on the results of ongoing and planned clinical trials to confirm its safety and efficacy in human populations.

Works cited

  1. Kamuvudine-9 by Inflammasome Therapeutics for Graves' Ophthalmopathy: Likelihood of Approval - Pharmaceutical Technology, accessed May 28, 2025, https://www.pharmaceutical-technology.com/data-insights/kamuvudine-9-inflammasome-therapeutics-graves-ophthalmopathy-likelihood-of-approval/
  2. K 9 - AdisInsight, accessed May 28, 2025, https://adisinsight.springer.com/drugs/800079780
  3. Reduction of human Alzheimer's disease risk and reversal of mouse ..., accessed May 28, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10055589/
  4. Inflammasome Therapeutics Announces Topline 3-month Data from ..., accessed May 28, 2025, https://www.biospace.com/press-releases/inflammasome-therapeutics-announces-topline-3-month-data-from-a-clinical-trial-of-its-first-in-class-dual-inflammasome-inhibitor-k8-in-geographic-atrophy-demonstrating-rapid-substantial-and-significant-efficacy-and-safety
  5. Evaluation of K9 in Subjects with Thyroid Eye Disease (TED) - MedPath Clinical Trial Search, accessed May 28, 2025, https://trial.medpath.com/clinical-trial/d78222097b324582/evaluation-k9-thyroid-eye-disease
  6. Kamuvudine-9 Protects Retinal Structure and Function in a Novel ..., accessed May 28, 2025, https://iovs.arvojournals.org/article.aspx?articleid=2785585
  7. Inflammasome Therapeutics Announces Topline 3-month Data from a Clinical Trial of its First-in-Class Dual Inflammasome Inhibitor, K8, in Geographic Atrophy, Demonstrating Rapid, Substantial and Significant Efficacy and Safety - FirstWord Pharma, accessed May 28, 2025, https://firstwordpharma.com/story/5928273
  8. Inflammasome Therapeutics Announces Topline 3-month Data from a Clinical Trial of, accessed May 28, 2025, https://inflam.com/images/pdfs-doc/Inflammasome_announces_top_line_3_month_data.pdf
  9. Inflammasome Therapeutics Completes Enrollment in Study for First Oral Dual, accessed May 28, 2025, https://inflam.com/images/pdfs-doc/First_Patient_Dosed_with_K9_-_September_2024.pdf
  10. Inflammasome Therapeutics completes enrollment of Phase I PK ..., accessed May 28, 2025, https://www.modernretina.com/view/inflammasome-therapeutics-completes-enrollment-of-phase-i-pk-safety-for-k9
  11. Inflammasome Therapeutics Announces Topline 3-month Data from a Clinical Trial of its First-in-Class Dual Inflammasome Inhibitor, K8, in Geographic Atrophy, Demonstrating Rapid, Substantial and Significant Efficacy and Safety - Business Wire, accessed May 28, 2025, https://www.businesswire.com/news/home/20250115779420/en/Inflammasome-Therapeutics-Announces-Topline-3-month-Data-from-a-Clinical-Trial-of-its-First-in-Class-Dual-Inflammasome-Inhibitor-K8-in-Geographic-Atrophy-Demonstrating-Rapid-Substantial-and-Significant-Efficacy-and-Safety
  12. kamuvudine-9 - MedPath Clinical Trial Search, accessed May 28, 2025, https://trial.medpath.com/drug/52bc910c928edf33?tab=news&page=1
  13. Inflammasome Therapeutics - News, accessed May 28, 2025, https://www.inflam.com/news
  14. WO2016138425A1 - Compositions and methods for treating retinal ..., accessed May 28, 2025, https://patents.google.com/patent/WO2016138425A1/en
  15. COMPOSITIONS AND METHODS FOR TREATING RETINAL, accessed May 28, 2025, https://data.epo.org/publication-server/rest/v1.2/patents/EP4215527NWB1/document.pdf
  16. (PDF) Nucleoside reverse transcriptase inhibitors and Kamuvudines inhibit amyloid-β induced retinal pigmented epithelium degeneration - ResearchGate, accessed May 28, 2025, https://www.researchgate.net/publication/350857340_Nucleoside_reverse_transcriptase_inhibitors_and_Kamuvudines_inhibit_amyloid-b_induced_retinal_pigmented_epithelium_degeneration
  17. Comparative Study of NRTI and Novel Inhibitor K-9 - Pubcompare, accessed May 28, 2025, https://www.pubcompare.ai/protocol/AQap1YwB4C3bMWOehEo-/
  18. LAMIVUDINE | New Drug Approvals, accessed May 28, 2025, https://newdrugapprovals.org/2016/03/18/lamivudine/
  19. Diethanolamine - Oakwood Chemical, accessed May 28, 2025, https://www.oakwoodchemical.com/ProductsList.aspx?CategoryID=-2&txtSearch=115995&ExtHyperLink=1
  20. 26661-13-2|N-(2-Oxo-1,2-dihydropyrimidin-4-yl)benzamide - BLDpharm, accessed May 28, 2025, https://www.bldpharm.com/products/26661-13-2.html
  21. PubChem, accessed May 28, 2025, https://pubchem.ncbi.nlm.nih.gov/
  22. Lamivudine (CAS 134678-17-4) - Cayman Chemical, accessed May 28, 2025, https://www.caymanchem.com/product/18514/lamivudine
  23. Inflammasome Therapeutics announces first patient dosed in Phase 1 trial evaluating Kamuvudine-8 treatment in patients with GA, accessed May 28, 2025, https://www.optometrytimes.com/view/inflammasome-therapeutics-announces-first-patient-dosed-in-phase-1-trial-evaluating-kamuvudine-8-treatment-in-patients-with-ga
  24. About Inflammasome Therapeutics, accessed May 28, 2025, https://www.inflam.com/about-inflammasome-therapeutics
  25. Inflammasome Therapeutics' Kamuvudines Continue to Show Promise for Future Treatment – and Potentially a Cure – for Blindness in Seniors - Business Wire, accessed May 28, 2025, https://www.businesswire.com/news/home/20210930005161/en/Inflammasome-Therapeutics-Kamuvudines-Continue-to-Show-Promise-for-Future-Treatment-and-Potentially-a-Cure-for-Blindness-in-Seniors
  26. Inflammasome Therapeutics announces first patient dosed in Phase ..., accessed May 28, 2025, https://www.ophthalmologytimes.com/view/inflammasome-therapeutics-announces-first-patient-dosed-in-phase-1-study-of-inflammasome-inhibitor-drug-in-subjects-with-dme
  27. New Study Shows Multiple Inflammasomes Combine to Cause ..., accessed May 28, 2025, https://www.businesswire.com/news/home/20211206005105/en/New-Study-Shows-Multiple-Inflammasomes-Combine-to-Cause-Lupus-Macular-Degeneration-and-Other-Neuroinflammatory-Diseases-Such-as-Alzheimers-Disease-and-Multiple-Sclerosis
  28. (PDF) Lamivudine Inhibits Alu RNA-induced Retinal Pigment Epithelium Degeneration via Anti-inflammatory and Anti-senescence Activities - ResearchGate, accessed May 28, 2025, https://www.researchgate.net/publication/342618997_Lamivudine_Inhibits_Alu_RNA-induced_Retinal_Pigment_Epithelium_Degeneration_via_Anti-inflammatory_and_Anti-senescence_Activities
  29. Identification of fluoxetine as a direct NLRP3 inhibitor to treat atrophic macular degeneration, accessed May 28, 2025, https://www.researchgate.net/publication/355149126_Identification_of_fluoxetine_as_a_direct_NLRP3_inhibitor_to_treat_atrophic_macular_degeneration
  30. Thyroid Ophthalmopathy Recruiting Phase 1 Trials for Thyroid, porcine (DB09100), accessed May 28, 2025, https://go.drugbank.com/indications/DBCOND0141877/clinical_trials/DB09100?phase=1&status=recruiting
  31. Thyroid, porcine Recruiting Phase 1 Trials for Thyroid Eye Disease Treatment - DrugBank, accessed May 28, 2025, https://go.drugbank.com/drugs/DB09100/clinical_trials?conditions=DBCOND0051704&phase=1&purpose=treatment&status=recruiting
  32. Evaluation of K9 in Subjects with Thyroid Eye Disease (TE..., accessed May 28, 2025, https://clinicaltrial.be/en/details/440238?per_page=20&only_recruiting=0&only_eligible=0&only_active=0
  33. Evaluation of K9 in Subjects with Thyroid Eye Disease (TED) - ngram, accessed May 28, 2025, https://www.ngram.com/trials/NCT06467435
  34. Graves Disease - Drugs, Targets, Patents - Synapse, accessed May 28, 2025, https://synapse-patsnap-com.libproxy1.nus.edu.sg/disease/0e1af5cdca874d34b9b87a793d65d466
  35. International Clinical Trials Registry Platform (ICTRP) - World Health Organization (WHO), accessed May 28, 2025, https://www.who.int/tools/clinical-trials-registry-platform
  36. ICTRP search portal - World Health Organization (WHO), accessed May 28, 2025, https://www.who.int/tools/clinical-trials-registry-platform/the-ictrp-search-portal
  37. Evaluation of K9 in Subjects with Thyroid Eye Disease (TED) - ClinicalTrials.Veeva, accessed May 28, 2025, https://ctv.veeva.com/study/evaluation-of-k9-in-subjects-with-thyroid-eye-disease-ted
  38. accessed January 1, 1970, https://clinicaltrials.gov/study/NCT06467435
  39. International Clinical Trials Registry Platform (ICTRP), accessed May 28, 2025, https://www.who.int/clinical-trials-registry-platform
  40. US11219623B2 - Inflammasome inhibition for the treatment of Parkinson's disease, Alzheimer's disease and multiple sclerosis - Google Patents, accessed May 28, 2025, https://patents.google.com/patent/US11219623B2/en
  41. US10294220B2 - Compositions and methods for treating retinal degradation - Google Patents, accessed May 28, 2025, https://patents.google.com/patent/US10294220B2/en
  42. Jayakrishna Ambati Inventions, Patents and Patent Applications - Justia Patents Search, accessed May 28, 2025, https://patents.justia.com/inventor/jayakrishna-ambati
  43. accessed January 1, 1970, https://patents.google.com/patent/US12097201B2/en
  44. accessed January 1, 1970, https://patents.google.com/patent/US20210228578A1/en
  45. accessed January 1, 1970, https://patents.google.com/patent/US11998547B2/en
  46. Orphan designation: Overview | European Medicines Agency (EMA), accessed May 28, 2025, https://www.ema.europa.eu/en/human-regulatory-overview/orphan-designation-overview
  47. Approval Package for: ANDA 090218 - accessdata.fda.gov, accessed May 28, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/090218Orig1s000.pdf
  48. queryResult - FDA, accessed May 28, 2025, https://www.fda.gov/media/185286/download
  49. Kamuvudine-8 - Drug Targets, Indications, Patents - Patsnap Synapse, accessed May 28, 2025, https://synapse.patsnap.com/drug/9fab86d76d7f4d3ca827e2c5cbe7370b

Published at: May 28, 2025

This report is continuously updated as new research emerges.

© Copyright 2025. All Rights Reserved by MedPath