An extensive review of available data reveals that the designation "K-001" is not a unique identifier for a single therapeutic agent. Instead, it is a polysemous term used for multiple, scientifically distinct entities, creating a significant potential for confusion in research, development, and investment analysis. This report provides a definitive disambiguation and a comprehensive, standalone analysis of the primary investigational drug candidates that have been associated with this designation.
The three principal pharmaceutical assets examined in this dossier are:
This report systematically deconstructs the data associated with each of these assets, providing a detailed examination of their respective compositions, mechanisms of action, clinical development programs, and strategic implications. The objective is to deliver a clear, evidence-based analysis that resolves the nomenclature ambiguity and supports informed strategic decision-making within the life sciences sector.
The use of a simple alphanumeric code such as "K-001" as an identifier for multiple, unrelated assets across different companies, research fields, and commercial contexts presents a significant analytical challenge. The source materials conflate data from disparate fields, including oncology [1], immunology [3], metabolic disease [4], chemical reagent catalogs [5], and industrial materials [7], all under the same "K-001" label. This overlap can lead to the erroneous attribution of preclinical data, clinical trial results, or mechanisms of action from one entity to another, fundamentally compromising any strategic assessment. This report serves to systematically untangle these disparate data threads, providing a clear and accurate profile for each distinct entity.
To establish a clear framework for the detailed analysis that follows, this report will focus on the three primary pharmaceutical candidates that are the subject of the most substantive data. Each will be analyzed in its own dedicated section to ensure clarity and depth. Other entities bearing the "K-001" name will be addressed and formally dismissed in a concluding section to provide a complete and exhaustive review.
The following table provides a high-level summary to immediately disambiguate the entities and serve as a reference guide for the remainder of the report.
Table 1.1: Summary of Entities Designated as "K-001"
Designation | Scientific Name / Composition | Developer / Sponsor | Therapeutic Area | Development Status |
---|---|---|---|---|
K-001 | Peptidoglycan complex of Spirulina | RenJi Hospital | Oncology (Advanced Pancreatic Cancer) | Phase II/III (NCT04183478) |
ESK-001 | N-(4-((2-methoxy-3-(1-(methyl-d3)-1H-1,2,4-triazol-3-yl)phenyl)amino)-5-(propanoyl-3,3,3-d3)pyridin-2-yl)cyclopropanecarboxamide hydrochloride (Allosteric TYK2 Inhibitor) | Alumis Inc. | Immunology (Plaque Psoriasis, SLE) | Phase 3 (ONWARD program) |
K-001 | Fixed-dose combination of K-877-ER (Pemafibrate) and CSG452 (Tofogliflozin) | Kowa Research Institute, Inc. | Metabolic Disorders (e.g., MASH) | Phase 1 (Bioequivalence study NCT05722262), Phase 2 for MASH |
K-001 | Karanjin (C18H12O4) | Indofine Chemical Company, Inc. (Catalog Number) | N/A (Chemical Reagent) | N/A |
This investigational agent is a biological compound being developed for advanced pancreatic cancer, an indication with a profound unmet medical need. Its development program, sponsored by RenJi Hospital, is centered on a novel immunomodulatory approach.
K-001 is defined as an orally bioavailable preparation of a peptidoglycan complex derived from the fermentation product of the marine microorganism Spirulina.[1] The primary active component is peptidoglycan (PGN), a large polymer with a molecular weight reported to be greater than 100,000 daltons.[9] This agent is classified as a first-class new drug in China and holds patent licenses in China, America, and Japan.[8]
The proposed mechanism of action involves immunomodulating and antineoplastic activities.[1] Upon oral administration, the peptidoglycan complex is hypothesized to activate the innate immune system. This activation is thought to subsequently re-engage the body's adaptive antitumor immune response, effectively overcoming the immune evasion mechanisms employed by the tumor.[1] This positions K-001 as a form of oral immunotherapy.
This therapeutic rationale is supported by a broader body of research into the anti-cancer properties of Spirulina and its components. For instance, phycocyanin, a pigment-protein complex purified from Spirulina, has been shown to inhibit the proliferation of pancreatic cancer cell lines both in vitro and in vivo. The mechanisms underlying this effect include the induction of G2/M cell cycle arrest, apoptosis (programmed cell death), and autophagic cell death.[10] These preclinical findings provide a scientific foundation for investigating
Spirulina-derived compounds as potential treatments for pancreatic cancer.[11] Preclinical toxicology studies of K-001 itself indicated that it is a non-toxic or only slightly toxic substance, foreshadowing the favorable safety profile later observed in human trials.[9]
The clinical development of K-001 has focused on patients with advanced pancreatic ductal adenocarcinoma (PDAC), specifically those who have progressed on multiple prior lines of therapy. This is a population with a dismal prognosis and for whom no standard of care is approved for third-line treatment, representing a critical unmet need.[2]
An initial Phase I/IIa open-label, dose-escalation trial was conducted to establish the safety, dose-limiting toxicities (DLTs), and maximum tolerated dose (MTD) of K-001.[9] The study followed a standard 3+3 design and evaluated doses escalating from 1350 mg to 2160 mg administered twice daily (BID).[12] A related Phase I tolerability study, also under the NCT02720666 identifier, explored an even broader dose range, with cohorts receiving up to 4320 mg/day (2160 mg BID).[14]
Safety Profile:
The most striking finding from these early-phase studies was the drug's exceptional safety and tolerability. Across all tested doses, no dose-limiting toxicities were observed.9 In an analysis of twelve patients, a total of 47 adverse events (AEs) were reported. The vast majority of these were low-grade, with 27 events (57.4%) being Grade 1 and 17 events (36.2%) being Grade 2. Crucially, only two AEs—indigestion and gastrointestinal flatulence—were definitively attributed to the study drug, and both were Grade 2.13 This remarkably benign safety profile is a critical attribute for a therapy intended for a heavily pre-treated and often frail patient population that may not be able to tolerate further cytotoxic chemotherapy.9
Efficacy Results:
Efficacy was assessed in twelve evaluable patients. According to the Response Evaluation Criteria in Solid Tumors (RECIST), the Objective Response Rate (ORR), which measures tumor shrinkage (complete or partial response), was 0%.9 While this figure might initially seem disappointing, the Disease Control Rate (DCR) was 83.3%. This means that 10 of the 12 patients achieved stable disease (SD), where their tumors did not progress during the treatment period.9
The clinical significance of this high DCR cannot be overstated in this specific therapeutic context. For patients with third-line or later advanced PDAC, who have exhausted standard cytotoxic options, the primary therapeutic goal often shifts from achieving tumor regression to preventing further rapid progression, managing symptoms, and preserving quality of life. In this setting, achieving durable disease stabilization is a meaningful clinical benefit. The combination of a high DCR with an excellent safety profile led investigators to conclude that K-001 demonstrated "satisfactory safety and tolerability, as well as meaningful antitumor activity," warranting further investigation.[9]
Based on the promising results of the Phase I/IIa study, a pivotal, randomized, double-blind, multi-center Phase II/III study was initiated.[2]
Study Design:
The trial was designed to compare K-001 in combination with Best Supportive Care (BSC) against a control arm of placebo plus BSC. The target population consists of patients with metastatic or locally advanced PDAC who have received at least two prior lines of chemotherapy and whose disease has progressed or who could not tolerate further chemotherapy.2
Primary Endpoint:
The primary outcome measure for the study is overall survival (OS), a gold-standard endpoint that directly measures clinical benefit in oncology.16 This endpoint is particularly appropriate given the drug's observed effect on disease stabilization rather than tumor shrinkage.
Status:
The trial record indicates a last verification date of November 2020, with an estimated primary completion date of March 2021.16 However, no results or updates from this pivotal trial have been made publicly available in the provided documentation. This lack of data represents a critical information gap. The future viability of this drug candidate is entirely dependent on the outcome of this trial. Without these results, its potential remains speculative, based solely on the early-phase data. The absence of published findings could be due to various factors, including negative or inconclusive results, termination for futility, or significant delays in data analysis and publication.
ESK-001 is an investigational small molecule therapeutic being developed by Alumis Inc. It represents a distinct and highly promising asset that, while separate from the other "K-001" entities, is included in this report due to its prominence in the available documentation and the potential for nomenclature-based confusion. ESK-001 is positioned at the forefront of oral therapies for autoimmune and inflammatory diseases.
Composition:
ESK-001 is an orally administered small molecule with a molecular weight of less than 500 g/mol, a characteristic favorable for oral bioavailability.17 Its full chemical name is N-(4-((2-methoxy-3-(1-(methyl-d3)-1H-1,2,4-triazol-3-yl)phenyl)amino)-5-(propanoyl-3,3,3-d3)pyridin-2-yl)cyclopropanecarboxamide hydrochloride.18
Mechanism of Action:
ESK-001 is a highly selective, next-generation allosteric inhibitor of tyrosine kinase 2 (TYK2).3 TYK2 is an intracellular signaling enzyme that plays a crucial role in the pathways of several key pro-inflammatory cytokines, including interleukin-23 (IL-23), IL-12, and type 1 interferons (IFN).3 By inhibiting TYK2, ESK-001 is designed to correct the immune dysregulation that drives a spectrum of autoimmune diseases.
A key differentiating feature of ESK-001 is its allosteric mechanism. It binds to the regulatory pseudokinase (JH2) domain of the TYK2 enzyme, rather than the highly conserved active kinase (JH1) domain.[17] This is the same mechanism employed by the approved TYK2 inhibitor, deucravacitinib. This mode of binding confers high selectivity for TYK2 over other Janus kinase (JAK) family members (JAK1, JAK2, JAK3), which is intended to avoid the off-target effects and safety liabilities associated with broader JAK inhibitors.[17]
Pharmacokinetics:
Phase 1 studies in healthy volunteers have characterized the pharmacokinetic profile of ESK-001. It is rapidly absorbed, with a time to maximum plasma concentration (Tmax) of 1 to 3 hours. It exhibits a mean terminal half-life of 8 to 13 hours and shows dose-proportional increases in systemic exposure.17 The half-life supports twice-daily (BID) dosing, with steady-state concentrations achieved by day 3. The bioavailability of ESK-001 is not significantly affected by food, allowing for administration with or without meals.18
The most advanced clinical program for ESK-001 is in the treatment of moderate-to-severe plaque psoriasis.
The STRIDE trial was a randomized, double-blind, placebo-controlled, dose-ranging study that enrolled over 220 patients.[3] The trial successfully demonstrated that ESK-001 produced statistically significant and clinically meaningful improvements in skin clearance compared to placebo across all primary and secondary endpoints.[3] The 40 mg BID dose was identified as the optimal dose, providing the best balance of efficacy and safety for further development.[21] The drug was well-tolerated, with no treatment-related serious adverse events reported. The most common AEs were mild in severity and included headache, upper respiratory tract infection, and nasopharyngitis.[3]
Patients who completed the 12-week STRIDE trial were eligible to enroll in an open-label extension (OLE) study to assess long-term safety and durability of response.[3] The long-term data from this OLE have been particularly compelling. Results presented after 52 weeks of treatment showed not only sustained but
increasing clinical responses over time. For patients on the 40 mg BID dose, 61.3% achieved a 90% improvement in the Psoriasis Area and Severity Index (PASI 90), and a remarkable 38.8% achieved complete skin clearance (PASI 100).[21] The safety profile remained consistent and favorable at the one-year mark, with no new safety signals emerging.[21]
Based on the strong Phase 2 results, Alumis initiated its pivotal ONWARD Phase 3 program in July 2024.[24] This program consists of two identical, global, multi-center, randomized, double-blind trials (ONWARD1 and ONWARD2), each enrolling approximately 840 patients with moderate-to-severe plaque psoriasis.[24]
Design:
The trials are designed to evaluate the efficacy and safety of ESK-001 (40 mg BID) against both placebo and an active comparator, apremilast (Otezla), a widely used oral psoriasis therapy.21
Endpoints:
The co-primary efficacy endpoints are the proportion of patients achieving PASI 75 and a static Physician's Global Assessment (sPGA) score of 0 (clear) or 1 (almost clear) compared to placebo at Week 16.24
Status:
Patient enrollment in the ONWARD1 and ONWARD2 trials is complete. Alumis has guided that topline data are anticipated in the first quarter of 2026.23
ESK-001 is also being evaluated for the treatment of SLE, a complex autoimmune disease where the type 1 interferon pathway is known to be a key driver of pathogenesis. The drug's ability to potently inhibit this pathway provides a strong rationale for its use in this indication.[3] The LUMUS trial is a global, Phase 2b, placebo-controlled study designed to evaluate multiple doses of ESK-001 in adults with moderately-to-severely active SLE.[3]
ESK-001 is an investigational product and has not yet been approved for marketing by any regulatory agency.[22] Its primary competitor in the TYK2 inhibitor class is deucravacitinib (brand name Sotyktu), which was approved by the FDA in 2022 for moderate-to-severe plaque psoriasis.[26]
The development strategy for ESK-001 appears to be centered on establishing a "best-in-class" profile by pushing the efficacy ceiling for oral therapies. The high rates of skin clearance observed in the OLE study, particularly the PASI 100 rate of 38.8%, approach levels typically seen with highly effective injectable biologics. Alumis's positioning suggests that this superior efficacy is a result of achieving maximal (>90%) and sustained TYK2 inhibition with the 40 mg BID dose, a level of target engagement that may not be achieved with the approved dose of deucravacitinib without incurring additional side effects.[21] If the Phase 3 ONWARD program confirms these high efficacy rates with a continued favorable safety profile, ESK-001 could be positioned not just as an alternative to deucravacitinib, but as a compelling oral option for patients who might otherwise require a biologic.
One potential commercial consideration is the current twice-daily (BID) dosing regimen, which may be perceived as less convenient than the once-daily (QD) regimen of deucravacitinib. Recognizing this, Alumis is actively developing a once-daily modified-release (MR) formulation of ESK-001, which is intended to replace the current immediate-release formulation used in the pivotal trials.[25] The successful development and regulatory approval of this QD formulation will be a key factor in its long-term commercial competitiveness.
Table 3.1: Comparative Profile: ESK-001 vs. Deucravacitinib (Sotyktu)
Metric | ESK-001 | Deucravacitinib |
---|---|---|
Target | Allosteric TYK2 inhibitor 3 | Allosteric TYK2 inhibitor 26 |
Claimed Differentiation | Achieves maximal (>90%) target inhibition at well-tolerated doses, aiming for "biologic-like efficacy" 21 | Approved dose provides partial target coverage; higher doses have been associated with increased adverse events 28 |
Efficacy (Psoriasis) | At 52 weeks (OLE):PASI 90: 61.3%PASI 100: 38.8% 21 | At 3-4 years (LTE, mNRI):PASI 75: ~72-74%PASI 90: ~48-49% 29 |
Safety Profile | Generally well-tolerated. Most common AEs: headache, nasopharyngitis, URI. No new safety signals at 1 year.3 | Well-tolerated. Most common AE: nasopharyngitis. Stable safety profile through 4 years with no clinically meaningful lab changes.27 |
Dosing | 40 mg BID (twice daily) in pivotal trials; once-daily formulation in development.21 | 6 mg QD (once daily) 27 |
Note: Direct cross-trial efficacy comparisons should be interpreted with caution due to differences in study design, patient populations, and statistical analysis methods (e.g., open-label extension vs. long-term extension, use of modified non-responder imputation).
A third distinct entity designated as K-001 is a fixed-dose combination (FDC) tablet under development by Kowa Research Institute, Inc. This product combines two metabolic drugs to target complex, multi-factorial diseases.
The K-001 FDC is composed of two active pharmaceutical ingredients:
K-877-ER is an extended-release formulation of pemafibrate.[4] Pemafibrate is a novel, potent, and highly selective peroxisome proliferator-activated receptor alpha (PPARα) modulator, also known as a SPPARMα.[32] It is approved for the treatment of hyperlipidemia in Japan and several other countries.[31] Its primary mechanism involves the regulation of genes involved in lipid metabolism, leading to a significant reduction in serum triglyceride levels.[31]
CSG452 is the development code for tofogliflozin, a potent and highly specific inhibitor of the sodium-glucose cotransporter 2 (SGLT2).[36] SGLT2 is responsible for the majority of glucose reabsorption in the kidneys. By inhibiting SGLT2, tofogliflozin increases the excretion of glucose in the urine, thereby lowering blood glucose levels. It is approved in Japan for the treatment of type 2 diabetes mellitus.[36]
The development of the K-001 FDC represents a strategic approach to product lifecycle management and a targeted effort to address a significant unmet medical need. By combining two approved drugs with complementary mechanisms, Kowa aims to create a new, value-added therapeutic for complex metabolic diseases.
The primary indication being pursued for this combination is metabolic dysfunction-associated steatohepatitis (MASH), a progressive form of fatty liver disease. MASH is characterized by liver steatosis, inflammation, and fibrosis, and is often driven by underlying metabolic dysfunction, including both dyslipidemia and insulin resistance. Preclinical studies in mouse models of NASH have demonstrated that the combination of pemafibrate and tofogliflozin acts synergistically. The combination treatment significantly improved liver histopathology (reducing hepatocyte ballooning), alleviated hyperglycemia and hypertriglyceridemia, and ultimately improved survival rates and reduced the incidence of liver tumors.[41] This strong preclinical evidence provides a compelling scientific rationale for developing the FDC for this indication.
Before efficacy trials can be conducted with the FDC tablet, regulatory agencies require a demonstration that the combination pill provides the same drug exposure as taking the individual components separately. To this end, Kowa conducted a Phase 1, randomized, open-label, crossover study in healthy volunteers.[4]
The primary objective of this trial was to demonstrate the bioequivalence of a single oral administration of the K-001 FDC relative to the co-administration of separate K-877-ER and CSG452 tablets. A secondary objective was to characterize the effect of food on the pharmacokinetics of the FDC.[4] According to clinical trial registries, this study, which was initiated in early 2023, is now completed.[4]
With the foundational bioequivalence data presumably in hand, Kowa is proceeding with mid-stage clinical development. The combination of pemafibrate and tofogliflozin is listed as being in Phase 2 development for MASH in the United States, Japan, and Spain.[45] A protocol for a Phase 2, multicenter, placebo-controlled, randomized, double-blind, 48-week study has been registered. This trial is designed to evaluate the efficacy and safety of the combination therapy in patients with noncirrhotic NASH with liver fibrosis.[47]
The establishment of a formal "K-001 Expanded Access Policy" by Kowa Research Institute is also a noteworthy aspect of this program.[48] While the policy states that expanded access is generally not provided for drugs in early development like K-001, it also establishes a clear framework for considering requests on a case-by-case basis as the program progresses. This proactive approach suggests that the company anticipates potential demand from patients with serious conditions and is preparing the necessary regulatory and logistical infrastructure, signaling confidence in the drug's potential to address a significant unmet need.
To ensure a fully exhaustive report and prevent future confusion, this section briefly addresses other substances, products, and codes that were identified with the "K-001" designation in the source material but are unrelated to the primary pharmaceutical candidates analyzed above.
Several other pharmaceutical products and clinical trials were identified due to similar naming conventions or trial identifiers. These are distinct from the three primary assets and are listed here for clarification.
The phrase "K-001 development code" appeared in several documents. In all cases, this referred to municipal land development, zoning, and building regulations, such as those for the City of Austin, Texas, and had no connection to pharmaceutical development.[57]
This comprehensive analysis has successfully disambiguated the "K-001" designation, revealing it to be a non-specific identifier for several distinct entities. The investigation focused on the three primary pharmaceutical candidates, each with a unique profile, development trajectory, and strategic context.
In summary, the "K-001" designation encompasses a diverse range of investigational therapies at different stages of development. A clear understanding of these distinct assets, as provided in this report, is essential for accurate scientific assessment and strategic decision-making.
Published at: September 22, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.