An Analytical Report on the Investigational IDO1 Inhibitor Linrodostat (BMS-986205)
Section 1: Executive Summary
Linrodostat, also known by its development code BMS-986205, is an investigational, orally administered small molecule engineered as a potent and selective inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1).[1] The therapeutic rationale for Linrodostat is rooted in the central role of the IDO1 enzyme in orchestrating tumor-mediated immune suppression. By catalyzing the degradation of the essential amino acid tryptophan into immunosuppressive metabolites known as kynurenines, IDO1 fosters a tumor microenvironment that is hostile to anti-cancer immune surveillance.[3] The central hypothesis driving Linrodostat's development was that its inhibition of IDO1 would reverse this immunosuppressive state, thereby restoring the ability of the host immune system to attack malignant cells, particularly in synergy with immune checkpoint inhibitors.
Originally developed by Flexus Biosciences, Linrodostat was acquired by Bristol-Myers Squibb (BMS) in a high-value transaction and was co-developed with Ono Pharmaceutical, reflecting significant initial confidence in its potential.[6] This confidence translated into a broad and ambitious clinical development program, primarily investigating Linrodostat in combination with the anti-PD-1 antibody nivolumab across a wide spectrum of solid and hematologic malignancies.
Key findings from its clinical evaluation revealed a complex picture. Pharmacologically, Linrodostat was a success; it demonstrated robust and dose-dependent pharmacodynamic activity, consistently achieving significant suppression of systemic kynurenine levels in patients, confirming successful engagement of its molecular target.[5] Early-phase clinical trials yielded encouraging, albeit inconsistent, signals of anti-tumor activity, with a particularly noteworthy objective response rate observed in heavily pre-treated patients with advanced bladder cancer.[9] However, the broader late-stage development of Linrodostat was abruptly curtailed. This strategic pivot was not primarily driven by negative data from Linrodostat's own trials but was a direct consequence of the high-profile failure of a competing IDO1 inhibitor, epacadostat, in a pivotal Phase 3 trial. The failure of the epacadostat study triggered a profound, class-wide re-evaluation of the therapeutic hypothesis, leading BMS to halt its registrational trials for Linrodostat in melanoma, non-small cell lung cancer, and head and neck cancer.[12]
Currently, Linrodostat remains an investigational agent and has not received regulatory approval from the U.S. Food and Drug Administration (FDA) or the European Medicines Agency (EMA).[15] The trajectory of Linrodostat serves as a critical case study in immuno-oncology drug development. It exemplifies a stark disconnect between successful target engagement and clinical efficacy, and powerfully illustrates the systemic risks within the pharmaceutical industry, where the failure of a lead asset can precipitate the collapse of an entire therapeutic class, irrespective of the distinct mechanistic attributes of follow-on molecules.
Section 2: Molecular Profile and Physicochemical Characteristics
A comprehensive understanding of Linrodostat's therapeutic potential begins with a detailed characterization of its molecular identity and physical properties, which dictate its formulation, delivery, and interaction with its biological target.
2.1 Identification and Nomenclature
Linrodostat is identified by a variety of names and registry numbers across scientific literature, clinical trial databases, and chemical registries.
- Nonproprietary Names: The official names are Linrodostat, assigned as the United States Adopted Name (USAN) and International Nonproprietary Name (INN).[18]
- Development Codes: Throughout its development, it has been referred to as BMS-986205 (by Bristol-Myers Squibb), ONO-7701 (by Ono Pharmaceutical), and F-001287 (by its original developer, Flexus Biosciences).[1]
- Registry Numbers: Key identifiers for database tracking include:
- CAS Number: 1923833-60-6 [1]
- DrugBank ID: DB14986 [1]
- UNII: 0A7729F42K [1]
2.2 Chemical Structure and Properties
Linrodostat is classified as a synthetic organic small molecule.[21] Its precise chemical architecture, including its stereochemistry, is fundamental to its biological activity.
- IUPAC Name: The systematic name is (2R)-N-(4-Chlorophenyl)-2-propanamide.[1] The specific stereoisomers, noted as
cis-(αR), are critical for potent and selective inhibition of IDO1.[19]
- Molecular Formula: C24H24ClFN2O.[1]
- Molecular Weight: The calculated molar mass is consistently reported as approximately 410.92 g·mol⁻¹.[1]
- Structural Identifiers: For computational and database purposes, its structure is represented by:
- SMILES: FC1=CC=2C(=CC=NC2C=C1)[C@@H]3CC[C@]([C@H](C(NC4=CC=C(Cl)C=C4)=O)C)(CC3)[H].[1]
- InChIKey: KRTIYQIPSAGSBP-ZACQAIPSSA-N.[1]
2.3 Physicochemical and Formulation Data
The physical properties of Linrodostat present both challenges and opportunities for its development as an oral therapeutic.
- Appearance: It is formulated as a crystalline solid.[21]
- Solubility: Linrodostat exhibits poor aqueous solubility, being described as insoluble in water (< 0.1 mg/mL). In contrast, it is highly soluble in organic solvents such as dimethyl sulfoxide (DMSO), with reported solubilities ranging from 50 to 90 mg/mL.[20]
- Lipophilicity: The molecule is highly lipophilic, with a calculated partition coefficient (XLogP) of 6.53. This value exceeds the threshold of 5 outlined in Lipinski's rule-of-five, which can sometimes correlate with challenges in oral absorption and bioavailability.[22]
- Stability and Storage: The compound is stable for at least four years when stored as a solid at -20 °C. It is typically shipped at ambient temperature, indicating good stability for short-term handling.[2]
- Salt Form: For clinical development, Linrodostat is often formulated as a mesylate salt, referred to as Linrodostat Mesylate.[5] The parent molecule, Linrodostat, is the active moiety responsible for the pharmacological effect.[18]
The molecule's physicochemical profile, particularly its high lipophilicity and extremely low water solubility, presents a significant hurdle for oral drug development. Such properties often lead to poor dissolution in the gastrointestinal tract and, consequently, low and variable absorption. However, preclinical studies consistently reported that Linrodostat achieved "high oral bioavailability".[5] This apparent contradiction strongly suggests that overcoming these formulation challenges was a critical and successful aspect of its early development. The use of the mesylate salt form and likely the application of advanced formulation technologies (e.g., amorphous solid dispersions, particle size reduction) were almost certainly required to enable sufficient absorption for a once-daily oral therapy. The existence of a dedicated Phase 1 clinical trial (NCT03312426) designed specifically to evaluate the effect of food on its absorption further underscores the importance and focus placed on optimizing its oral delivery.[28]
Table 1: Summary of Linrodostat's Chemical and Physical Properties
Property | Value | Source(s) |
---|
Common Name | Linrodostat | 18 |
Development Codes | BMS-986205, ONO-7701, F-001287 | 1 |
DrugBank ID | DB14986 | 1 |
CAS Number | 1923833-60-6 | 1 |
IUPAC Name | (2R)-N-(4-Chlorophenyl)-2-propanamide | 1 |
Molecular Formula | C24H24ClFN2O | 1 |
Molar Mass | 410.92 g·mol⁻¹ | 1 |
Appearance | Crystalline solid | 21 |
Solubility (DMSO) | 50 - 90 mg/mL | 20 |
Solubility (Water) | Insoluble (< 0.1 mg/mL) | 20 |
Storage | -20 °C (long-term) | 2 |
Stability | ≥ 4 years at -20 °C | 21 |
XLogP | 6.53 | 22 |
Salt Form | Linrodostat Mesylate (for clinical studies) | 5 |
Section 3: Mechanism of Action and Preclinical Pharmacology
Linrodostat was engineered to counteract a fundamental mechanism of tumor immune evasion by targeting the IDO1 metabolic pathway. Its specific and potent mechanism of action, validated through extensive preclinical research, formed the scientific bedrock for its advancement into human clinical trials.
3.1 The IDO1 Pathway: A Key Immune Checkpoint
Indoleamine 2,3-dioxygenase 1 (IDO1) is a heme-containing intracellular enzyme that serves as a critical regulator of immune tolerance. It catalyzes the initial and rate-limiting step in the catabolism of the essential amino acid L-tryptophan along the kynurenine pathway.[4] In the context of cancer, tumor cells and other cells within the tumor microenvironment (TME) frequently overexpress IDO1. This overexpression is a key strategy for immune escape, exerting its immunosuppressive effects through two primary mechanisms:
- Tryptophan Depletion: The enzymatic activity of IDO1 locally depletes tryptophan, an amino acid vital for the proliferation and function of effector immune cells, particularly T-lymphocytes. This starvation state activates cellular stress response kinases like GCN2 and inhibits the master metabolic regulator mTOR, leading to T-cell cycle arrest (anergy) and programmed cell death (apoptosis).[10]
- Kynurenine Accumulation: The catabolism of tryptophan produces a series of bioactive metabolites, collectively known as kynurenines. Kynurenine acts as an endogenous ligand for the Aryl Hydrocarbon Receptor (AhR), a transcription factor present on various immune cells. Activation of AhR by kynurenine promotes the differentiation of naïve T-cells into immunosuppressive regulatory T-cells (Tregs) and skews dendritic cells (DCs) towards a tolerogenic phenotype, further dampening the anti-tumor immune response.[2]
Consequently, high IDO1 expression in tumors is strongly correlated with poor prognosis and resistance to immunotherapy, establishing it as a high-priority target for therapeutic intervention.[3]
3.2 Linrodostat's Unique Inhibitory Mechanism
Linrodostat is characterized as a highly potent, selective, and irreversible inhibitor of the IDO1 enzyme.[2] Its mechanism of action is distinct from other clinically investigated IDO1 inhibitors, such as epacadostat. While epacadostat is a reversible, competitive inhibitor that vies with tryptophan for the catalytic site of the active, heme-containing enzyme (holo-IDO1), Linrodostat operates through a different and more permanent mechanism.[10]
Linrodostat specifically targets and binds to the heme-free (apo) form of IDO1. It occupies the pocket where the essential heme cofactor would normally bind, thereby preventing the formation of the catalytically active holo-enzyme.[5] This mechanism is effectively irreversible, as the enzyme cannot be reactivated without the synthesis of entirely new protein. This irreversible nature provides a durable pharmacodynamic effect that can persist even as plasma drug concentrations decline, a property that supports a convenient once-daily dosing schedule.[8]
This unique mode of targeting the apo-enzyme was a key point of differentiation for Linrodostat. Emerging research has suggested that in addition to its enzymatic function, apo-IDO1 may possess non-catalytic, pro-tumorigenic signaling capabilities through interactions with other intracellular proteins.[4] Some catalytic inhibitors have been hypothesized to paradoxically stabilize this signaling-competent apo-form, potentially undermining their own therapeutic benefit.[4] By binding to the apo-enzyme's heme pocket, Linrodostat was theoretically positioned to not only prevent catalytic activity but also potentially disrupt these non-enzymatic functions, representing a potential mechanistic advantage that likely contributed to the high level of investment in its development.
3.3 In Vitro Potency and Selectivity
Preclinical in vitro studies consistently demonstrated Linrodostat's exceptional potency and selectivity for its intended target. In a variety of cell-based assays designed to measure the inhibition of kynurenine production, Linrodostat exhibited half-maximal inhibitory concentrations (IC50) in the low nanomolar range:
- IC50 of 1.1 nM in HEK293 cells engineered to overexpress human IDO1.[20]
- IC50 of 1.7 nM in HeLa cervical cancer cells, which endogenously express IDO1 upon stimulation.[21]
This high potency was matched by excellent selectivity. Linrodostat showed no meaningful inhibitory activity against the related tryptophan-catabolizing enzymes Tryptophan 2,3-dioxygenase (TDO) and Indoleamine 2,3-dioxygenase 2 (IDO2).[5] The functional consequence of this potent and selective inhibition was confirmed in co-culture assays, where Linrodostat successfully restored the proliferation of T-cells that had been suppressed by IDO1-expressing dendritic cells.[5]
3.4 In Vivo Preclinical Efficacy
The promising in vitro profile of Linrodostat was successfully translated into in vivo models. In mice bearing human tumor xenografts (e.g., the SKOV3 ovarian cancer cell line), oral administration of Linrodostat led to a significant and dose-dependent reduction of kynurenine levels within the tumors, providing clear evidence of target engagement and pharmacodynamic activity in a living system.[5] In one such model, a 60 mg/kg daily dose of Linrodostat achieved a 61% reduction in tumor kynurenine, a pharmacodynamic effect that was comparable to that achieved with a 100 mg/kg twice-daily dose of epacadostat, underscoring its high
in vivo potency.[29]
Table 2: Summary of Key Preclinical Pharmacology Data
Assay / Model | Target(s) | Key Metric | Result | Source(s) |
---|
Cellular Assay (HEK293) | Human IDO1 | IC50 | 1.1 nM | 20 |
Cellular Assay (HeLa) | Human IDO1 | IC50 | 1.7 nM | 21 |
Cellular Assay (HEK293) | Human TDO | IC50 | > 2000 nM | 25 |
Cellular Assay (HEK293) | Murine IDO2 | - | No activity detected | 5 |
Mixed-Lymphocyte Reaction | IDO1-expressing DCs | T-cell Proliferation | Proliferation restored | 5 |
SKOV3 Xenograft Model | Human IDO1 | Tumor Kynurenine | Significant reduction | 5 |
Section 4: Human Pharmacokinetics and Pharmacodynamics
The transition of Linrodostat from preclinical models to human subjects in Phase 1 trials was crucial for establishing its clinical viability. These studies aimed to define its pharmacokinetic (PK) profile—how the body processes the drug—and its pharmacodynamic (PD) effects—how the drug affects its biological target in patients.
4.1 Pharmacokinetics (PK)
Linrodostat was developed for oral administration, with a once-daily (QD) dosing regimen being the standard in clinical trials.[1]
- Absorption and Bioavailability: While preclinical studies in animals suggested high oral bioavailability, its absorption characteristics in humans were a key area of investigation, as evidenced by a dedicated Phase 1 study (NCT03312426) designed to assess the impact of food on its uptake.[5] Clinical data from the Phase 1/2 study (NCT02658890) demonstrated that therapeutic exposures were achieved and exceeded predicted target concentrations starting at a dose of 50 mg QD.[5]
- Key Pharmacokinetic Parameters: Data from patients receiving a 100 mg single oral dose of Linrodostat while on a stable regimen of nivolumab provide a snapshot of its clinical PK profile [18]:
- Maximum Plasma Concentration (Cmax): Peak concentrations in plasma ranged from approximately 596 ng/mL after a single dose to 781 ng/mL at steady-state.
- Total Drug Exposure (AUC): The area under the concentration-time curve was approximately 3,380 ng·h/mL after a single dose, increasing to around 6,000 ng·h/mL at steady-state.
- Elimination Half-Life (T1/2): The terminal half-life was determined to be approximately 22.9 hours at steady-state. This long half-life provides strong pharmacokinetic justification for the convenience of a once-daily dosing schedule.
4.2 Pharmacodynamics (PD)
The primary pharmacodynamic objective for Linrodostat was to demonstrate successful inhibition of the IDO1 enzyme in patients. This was primarily assessed by measuring the levels of kynurenine, the direct product of IDO1's enzymatic activity.
- Evidence of Target Engagement: Clinical data provided unequivocal evidence of potent and sustained target engagement. Administration of Linrodostat, particularly in combination with nivolumab, resulted in a marked and dose-dependent reduction in kynurenine concentrations in both patient serum and tumor biopsy samples.[5] This confirmed that the drug was reaching its target
in situ and effectively inhibiting its function.
- The PK/PD-Efficacy Disconnect: A critical finding, and one that encapsulates the central challenge for Linrodostat and the entire IDO1 inhibitor class, emerged from these studies. While the drug was pharmacologically successful—it achieved therapeutic concentrations (PK) and robustly inhibited its target (PD)—this did not consistently translate into clinical efficacy (i.e., tumor shrinkage). The pivotal Phase 1/2 study report explicitly noted that the decrease in kynurenine levels occurred regardless of whether a patient's tumor responded to the therapy and that these PD changes did not correlate with response.[5]
This disconnect between successful pharmacology and inconsistent clinical benefit is the core paradox of the Linrodostat story. It strongly suggests that the foundational therapeutic hypothesis—that simply blocking the catalytic activity of IDO1 is a sufficient condition to overcome tumor immune resistance—was either incomplete or fundamentally flawed. This finding forced a re-evaluation of the underlying biology, raising critical questions about the potential roles of redundant metabolic pathways (like TDO), the importance of non-catalytic IDO1 functions, the adequacy of the achieved level of target inhibition, and the critical need for predictive biomarkers to identify the small subset of patients who might actually benefit.
Section 5: Clinical Development Program: A Comprehensive Review
The clinical development of Linrodostat was characterized by its broad scope and ambitious strategy, reflecting the initial high expectations for IDO1 inhibition as a cornerstone of combination cancer immunotherapy. The program spanned multiple phases and a diverse range of malignancies before its late-stage efforts were largely curtailed.
5.1 Overview of Clinical Strategy
The central pillar of Linrodostat's clinical strategy was its evaluation in combination with immune checkpoint inhibitors. The prevailing scientific hypothesis was that by dismantling the IDO1-mediated immunosuppressive shield, Linrodostat would sensitize tumors to the effects of anti-PD-1/PD-L1 and anti-CTLA-4 therapies, thereby increasing response rates and durability.[9] Consequently, the vast majority of trials investigated Linrodostat in combination with nivolumab, with some three-drug regimens also including ipilimumab.[1]
5.2 Foundational Phase 1/2 Study (NCT02658890)
This large, multi-part study served as the foundation for the entire clinical program. It was designed to establish the safety, tolerability, recommended dose, and preliminary efficacy of Linrodostat in combination with nivolumab, with or without ipilimumab, in patients with a wide array of advanced cancers.[5]
- Safety and Dosing: The study successfully established a manageable safety profile for the combination. The maximum tolerated dose (MTD) of Linrodostat was determined to be 200 mg once daily. As anticipated for a combination of immunomodulatory agents, the dose-limiting toxicities were primarily immune-related in nature.[5] Overall rates of Grade 3/4 adverse events were substantial, ranging from 50.1% to 63.4% across the study parts.[5]
- Efficacy Signals: The study demonstrated preliminary signs of anti-tumor activity across several cohorts, with responses being more pronounced in patients who were naïve to prior immunotherapy.[5] A particularly strong and promising signal emerged from the cohort of patients with heavily pre-treated, immunotherapy-naïve advanced bladder cancer. In this group, the combination of Linrodostat and nivolumab achieved an objective response rate (ORR) of 37%.[9] This result was a clear standout within the early-phase program and provided the primary rationale for advancing the combination into a pivotal Phase 3 trial for bladder cancer.[33]
- Biomarker Findings: This trial was instrumental in uncovering the disconnect between pharmacodynamics and efficacy. While confirming robust kynurenine suppression, it revealed that this biomarker did not predict clinical response. Instead, the study identified a potential predictive biomarker signature: an IFN-γ gene expression signature was associated with response, and in patients with non-melanoma tumors, a composite biomarker of low TDO gene expression combined with a high IFN-γ signature showed the strongest association with clinical benefit.[5]
5.3 Late-Stage Programs and Their Discontinuation
Buoyed by early signals, BMS launched an aggressive late-stage development program. However, this program was largely dismantled following a strategic re-evaluation of the entire IDO1 inhibitor class.
- Advanced Melanoma (NCT03329846): This was a large, randomized, double-blind Phase 3 trial designed to definitively test the efficacy of adding Linrodostat to nivolumab versus nivolumab alone in first-line metastatic or unresectable melanoma.[40] The trial was initiated in late 2017 but was halted by BMS in 2018 before its completion. This decision was a direct reaction to the failure of the analogous ECHO-301 trial of epacadostat, which showed no benefit for the addition of an IDO1 inhibitor in the same patient population.[12] As a result, no conclusive efficacy data from this pivotal trial are available.
- NSCLC and Head & Neck Cancer: Registrational Phase 3 trials in non-small cell lung cancer (e.g., NCT03417037) and squamous cell carcinoma of the head and neck (e.g., NCT03386838) were also initiated or planned. These, too, were withdrawn by BMS, with the company citing changes in "business strategy," a decision widely understood to be a direct consequence of the ECHO-301 fallout.[6]
- Muscle-Invasive Bladder Cancer (ENERGIZE, NCT03661320): Based on the strong early signal, a Phase 3 study was launched to evaluate Linrodostat in the neoadjuvant setting for muscle-invasive bladder cancer, in combination with chemotherapy and nivolumab.[33] For a time, this trial represented the last major ongoing Phase 3 effort for an IDO1 inhibitor. However, the overall development program for Linrodostat was eventually discontinued, including the bladder cancer indication.[16]
The timing of these events reveals a critical aspect of the program's trajectory. The ambitious, large-scale Phase 3 trials were designed and launched based on early, promising ORR data from unselected patient populations. The more sophisticated biomarker analysis from the Phase 1/2 study, which suggested that only a specific subset of patients (e.g., those with low TDO and high IFN-γ expression) were likely to respond, emerged after these resource-intensive trials were already underway. The class-wide failure of epacadostat in an unselected melanoma population occurred before this biomarker-driven strategy could be implemented in a pivotal setting for Linrodostat, representing a case of clinical development proceeding faster than the translational science could guide it.
Table 3: Overview of Major Clinical Trials for Linrodostat (BMS-986205)
NCT Identifier | Phase | Indication(s) | Intervention (Combination Agents) | Status | Key Findings / Rationale |
---|
NCT02658890 | 1 / 2 | Advanced Cancers (incl. Melanoma, NSCLC, Bladder) | Nivolumab ± Ipilimumab | Completed | Foundational dose-escalation/expansion. Established MTD of 200 mg. Showed promising ORR (37%) in bladder cancer. Identified potential predictive biomarkers (low TDO, high IFN-γ). |
NCT03329846 | 3 | Advanced Melanoma | Nivolumab | Terminated | Pivotal trial vs. nivolumab monotherapy. Halted by sponsor following failure of competitor's (epacadostat) analogous trial. |
NCT03661320 | 3 | Muscle-Invasive Bladder Cancer (MIBC) | Neoadjuvant Chemotherapy + Nivolumab | Discontinued | "ENERGIZE" study. Based on strong Phase 1/2 signal. Program was ultimately discontinued. |
NCT03417037 | 3 | Non-Small Cell Lung Cancer (NSCLC) | Nivolumab | Withdrawn | Pivotal trial. Withdrawn by sponsor due to "business strategy changes" post-epacadostat failure. |
NCT03386838 | 3 | Head and Neck Cancer | Nivolumab | Withdrawn | Pivotal trial. Withdrawn by sponsor due to "business strategy changes" post-epacadostat failure. |
NCT04047706 | 1 | Glioblastoma | Nivolumab + Radiation | Active, Not Recruiting | Early-phase study in newly diagnosed glioblastoma. |
NCT04106414 | 2 | Endometrial Cancer / Carcinosarcoma | Nivolumab | Active, Not Recruiting | Phase 2 study in recurrent or persistent disease. |
NCT03312426 | 1 | Healthy Volunteers | Monotherapy | Completed | Assessed the effect of food on absorption of Linrodostat. |
Section 6: Strategic Analysis and Future Outlook
The clinical development arc of Linrodostat is a compelling narrative in modern oncology, defined less by its own intrinsic data and more by the seismic impact of a competitor's failure. A thorough analysis reveals crucial lessons about therapeutic hypothesis validation, portfolio risk, and the future of targeting metabolic pathways in immuno-oncology.
6.1 The "Epacadostat Effect": A Class-Wide Catastrophe
The trajectory of Linrodostat cannot be understood in isolation. Its fate was inextricably linked to that of epacadostat, the lead IDO1 inhibitor developed by Incyte. In early 2018, the Phase 3 ECHO-301 trial, which combined epacadostat with pembrolizumab in advanced melanoma, failed dramatically. The combination showed no improvement in progression-free survival or overall survival compared to pembrolizumab alone.[43]
The industry's reaction was immediate and decisive. The failure was interpreted not just as a failure of one drug, but as a potential invalidation of the entire therapeutic hypothesis for IDO1 inhibition. Within weeks, Bristol-Myers Squibb confirmed that it was halting its own pivotal trials of Linrodostat in melanoma, head and neck cancer, and non-small cell lung cancer, explicitly citing the "emerging data on the IDO pathway".[6] This event powerfully demonstrates the concept of class-level risk in drug development. BMS had invested significantly in Linrodostat, including an $800 million upfront payment in its acquisition of Flexus Biosciences, and had constructed a vast and expensive late-stage clinical program.[6] This entire portfolio was effectively dismantled based on the negative results of a competitor's asset, highlighting how shared confidence in a novel biological mechanism can create systemic risk that cascades across companies when the lead program fails.
6.2 Deconstructing the Failure: Why Didn't Target Engagement Translate to Efficacy?
The central question arising from the clinical experience with Linrodostat and other IDO1 inhibitors is why clear, potent target inhibition failed to produce consistent clinical benefit. Several non-mutually exclusive hypotheses have emerged to explain this disconnect:
- Redundant Metabolic Pathways: The immunosuppressive metabolite kynurenine is produced not only by IDO1 but also by TDO. In tumors that co-express both enzymes, selectively inhibiting IDO1 may be insufficient to meaningfully lower total kynurenine levels in the TME to a therapeutically relevant degree.[10] This hypothesis is strongly supported by the biomarker data from the NCT02658890 trial, where clinical response to Linrodostat correlated with
low baseline expression of TDO.[8]
- The Non-Enzymatic Role of IDO1: There is growing evidence that the apo-form of IDO1 (lacking its heme cofactor) can function as a signaling scaffold, promoting a pro-tumorigenic and immunosuppressive phenotype independent of its catalytic activity.[4] It is therefore plausible that simply blocking the enzyme's catalytic function is insufficient. This raises complex questions about Linrodostat's unique mechanism; while it was hoped that targeting the apo-enzyme would be an advantage, the ultimate clinical outcomes suggest this was not enough to overcome the pathway's resistance.
- Inadequate Patient Selection: As previously noted, the large pivotal trials for both epacadostat and Linrodostat were designed for broad, "all-comer" patient populations. They were not designed to enrich for patients with the specific biological profile (e.g., high IFN-γ signature, low TDO expression) that was later identified as potentially predictive of response.[8] The failure in an unselected population may have masked potential benefit in a small, well-defined subset of patients.
6.3 The Future of IDO1 Inhibition and Linrodostat
Following the termination of its major trials, BMS indicated a commitment to continued early-phase research to better understand the biology of the IDO1 pathway and identify relevant biomarkers.[13] However, the subsequent comprehensive discontinuation of nearly all active programs suggests that the asset was ultimately deprioritized within the company's portfolio.[16]
The broader field of IDO1 inhibition has been forced to evolve. The lessons learned from the first generation of inhibitors, including Linrodostat, have shifted focus toward more sophisticated strategies. These include the development of dual IDO1/TDO inhibitors to address pathway redundancy, the targeting of downstream effectors like the Aryl Hydrocarbon Receptor (AhR), and the exploration of novel modalities such as proteolysis-targeting chimeras (PROTACs). PROTACs are designed to induce the complete degradation of the IDO1 protein, a strategy that would eliminate both its catalytic and non-catalytic signaling functions.[11]
In conclusion, Linrodostat stands as a well-characterized, potent, and mechanistically differentiated molecule. It successfully demonstrated target engagement in clinical trials, yet its therapeutic potential was never fully realized. Its story is a cautionary tale of the complexities of cancer immunology and the significant risks of pursuing novel mechanisms of action. The ultimate failure to bring Linrodostat to market was a result of a confluence of factors: a therapeutic hypothesis that proved to be overly simplistic, a clinical strategy that outpaced the supporting translational science, and the profound, cascading impact of a high-profile competitor failure that precipitated a strategic retreat from an entire class of promising, yet challenging, therapeutic agents.
Works cited
- Linrodostat - Wikipedia, accessed September 7, 2025, https://en.wikipedia.org/wiki/Linrodostat
- Linrodostat free base | BMS-986205 | CAS#1923833-60-6 - MedKoo Biosciences, accessed September 7, 2025, https://www.medkoo.com/products/13500
- Definition of linrodostat - NCI Drug Dictionary, accessed September 7, 2025, https://www.cancer.gov/publications/dictionaries/cancer-drug/def/linrodostat
- Epacadostat stabilizes the apo-form of IDO1 and signals a pro-tumorigenic pathway in human ovarian cancer cells - Frontiers, accessed September 7, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2024.1346686/full
- Preclinical Characterization of Linrodostat Mesylate, a Novel, Potent ..., accessed September 7, 2025, https://pubmed.ncbi.nlm.nih.gov/33298590/
- Linrodostat BMS 986205, ONO 7701 - New Drug Approvals, accessed September 7, 2025, https://newdrugapprovals.org/2018/06/25/bms-986205-ono-7701/
- linrodostat | Ligand page | IUPHAR/BPS Guide to MALARIA PHARMACOLOGY, accessed September 7, 2025, https://www.guidetomalariapharmacology.org/GRAC/LigandDisplayForward?ligandId=9707
- Phase 1/2 Study of the Indoleamine 2,3-Dioxygenase 1 Inhibitor Linrodostat Mesylate Combined with Nivolumab or Nivolumab and Ipilimumab in Advanced Solid Tumors or Hematologic Malignancies - PubMed, accessed September 7, 2025, https://pubmed.ncbi.nlm.nih.gov/39670852/
- BMS-986205, an indoleamine 2, 3-dioxygenase 1 inhibitor (IDO1i), in combination with nivolumab (nivo): Updated safety across all tumor cohorts and efficacy in advanced bladder cancer (advBC). - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/publication/331598750_BMS-986205_an_indoleamine_2_3-dioxygenase_1_inhibitor_IDO1i_in_combination_with_nivolumab_nivo_Updated_safety_across_all_tumor_cohorts_and_efficacy_in_advanced_bladder_cancer_advBC
- IDO Inhibitor Development Shows Fresh Signs of Life Across Tumor Types - OncLive, accessed September 7, 2025, https://www.onclive.com/view/ido-inhibitor-development-shows-fresh-signs-of-life-across-tumor-types
- (PDF) Indoleamine 2,3-dioxygenase 1 in cancer immunotherapy: from small-molecule inhibition to PROTAC-mediated degradation - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/publication/394504946_Indoleamine_23-dioxygenase_1_in_cancer_immunotherapy_from_small-molecule_inhibition_to_PROTAC-mediated_degradation
- Bristol-Myers Squibb halts pivotal studies of IDO inhibitor BMS-986205, accessed September 7, 2025, https://firstwordpharma.com/story/4578649
- BMS pulls the plug on Phase 3 trials of IDO1 inhibitor - Pharmafile, accessed September 7, 2025, https://pharmafile.com/news/bms-pulls-plug-phase-3-trials-ido1-inhibitor/
- Bristol-Myers Squibb Abandons 2 Phase III Trials of its IDO1 Inhibitor Program - BioSpace, accessed September 7, 2025, https://www.biospace.com/bristol-myers-squibb-abandons-2-phase-iii-trials-of-its-ido1-inhibitor-program
- Oncology (Cancer)/Hematologic Malignancies Approval Notifications - FDA, accessed September 7, 2025, https://www.fda.gov/drugs/resources-information-approved-drugs/oncology-cancerhematologic-malignancies-approval-notifications
- Linrodostat - Bristol-Myers Squibb - AdisInsight - Springer, accessed September 7, 2025, https://adisinsight.springer.com/drugs/800044556
- European Medicines Agency (EMA), accessed September 7, 2025, https://www.ema.europa.eu/en/medicines
- LINRODOSTAT - Inxight Drugs, accessed September 7, 2025, https://drugs.ncats.io/substance/0A7729F42K
- LINRODOSTAT - gsrs, accessed September 7, 2025, https://gsrs.ncats.nih.gov/ginas/app/ui/substances/68cf50d5-0b4c-456e-8e98-1f890433adb4
- BMS-986205 | CAS NO.:1923833-60-6 - GlpBio, accessed September 7, 2025, https://www.glpbio.com/bms-986205.html
- BMS 986205 (Linrodostat, CAS Number: 1923833-60-6) | Cayman Chemical, accessed September 7, 2025, https://www.caymanchem.com/product/25025/bms-986205
- linrodostat | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 7, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId=9707
- Linrodostat - - Sigma-Aldrich, accessed September 7, 2025, https://www.sigmaaldrich.com/US/en/substance/bbelinrodostat410921923833606
- Linrodostat | 1923833-60-6 | MOLNOVA, accessed September 7, 2025, https://www.molnova.com/en/ProductsThr/Linrodostat.html
- Linrodostat (BMS-986205) | 99.89%(HPLC) | IDO/TDO inhibitor - Selleck Chemicals, accessed September 7, 2025, https://www.selleckchem.com/products/bms-986205.html
- Linrodostat | Indoleamine 2,3-Dioxygenase (IDO) - TargetMol, accessed September 7, 2025, https://www.targetmol.com/compound/linrodostat
- Preclinical Characterization of Linrodostat Mesylate, a Novel, Potent, and Selective Oral Indoleamine 2,3-Dioxygenase 1 Inhibitor - Consensus, accessed September 7, 2025, https://consensus.app/papers/preclinical-characterization-of-linrodostat-mesylate-a-maley-lin/ab2f1332cc3e59cfaca15de5decad8f4/
- Linrodostat Completed Phase 1 Trials for Healthy Volunteers (HV) Basic Science, accessed September 7, 2025, https://go.drugbank.com/drugs/DB14986/clinical_trials?conditions=DBCOND0114743&phase=1&purpose=basic_science&status=completed
- Discovery and Preclinical Evaluation of BMS-986242, a Potent ..., accessed September 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7883469/
- Indoleamine 2,3-dioxygenase 1 in cancer immunotherapy: from small-molecule inhibition to PROTAC-mediated degradation, accessed September 7, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC12394918/
- Discovery of IDO1 Inhibitors: From Bench to Bedside | Cancer ..., accessed September 7, 2025, https://aacrjournals.org/cancerres/article/77/24/6795/624889/Discovery-of-IDO1-Inhibitors-From-Bench-to
- Preclinical Characterization of Linrodostat Mesylate, a Novel, Potent, and Selective Oral Indoleamine 2,3-Dioxygenase 1 Inhibitor - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/publication/347527521_Preclinical_Characterization_of_Linrodostat_Mesylate_a_Novel_Potent_and_Selective_Oral_Indoleamine_23-Dioxygenase_1_Inhibitor
- A phase III randomized study of neoadjuvant chemotherapy (NAC) alone or in combination with nivolumab (NIVO) ± linrodostat mesylate, followed by adjuvant postsurgical NIVO ± linrodostat, in cisplatin-eligible muscle invasive bladder cancer (MIBC). - ASCO Publications, accessed September 7, 2025, https://ascopubs.org/doi/10.1200/JCO.2020.38.15_suppl.TPS5091
- The catalytic inhibitor epacadostat can affect the non-enzymatic function of IDO1 - Frontiers, accessed September 7, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2023.1134551/full
- The catalytic inhibitor epacadostat can affect the non-enzymatic function of IDO1 - PubMed, accessed September 7, 2025, https://pubmed.ncbi.nlm.nih.gov/37122718/
- NSCLC Completed Phase 1 / 2 Trials for Linrodostat (DB14986) | DrugBank Online, accessed September 7, 2025, https://go.drugbank.com/indications/DBCOND0022283/clinical_trials/DB14986?phase=1%2C2&status=completed
- Encouraging Response Observed with ... - Bristol Myers Squibb, accessed September 7, 2025, https://news.bms.com/news/details/2017/Encouraging-Response-Observed-with-Opdivo-nivolumab-Plus-Investigational-IDO1-Inhibitor-BMS-986205-in-Heavily-Pre-Treated-Patients-with-Advanced-Cancers-in-Phase-12a-Study-CA017-003/default.aspx
- ENERGIZE: A Phase III study of neoadjuvant chemotherapy alone or with nivolumab with/without linrodostat mesylate for muscle-invasive bladder cancer | Request PDF - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/publication/337903517_ENERGIZE_A_Phase_III_study_of_neoadjuvant_chemotherapy_alone_or_with_nivolumab_withwithout_linrodostat_mesylate_for_muscle-invasive_bladder_cancer
- Linrodostat mesylate concentration pharmacokinetic profile in patients... - ResearchGate, accessed September 7, 2025, https://www.researchgate.net/figure/Linrodostat-mesylate-concentration-pharmacokinetic-profile-in-patients-treated-with_fig1_387030482
- Linrodostat Completed Phase 3 Trials for Skin Cancers / Melanoma Treatment - DrugBank, accessed September 7, 2025, https://go.drugbank.com/drugs/DB14986/clinical_trials?conditions=DBCOND0020306%2CDBCOND0046599&phase=3&purpose=treatment&status=completed
- linrodostat | Ligand page | IUPHAR/BPS Guide to PHARMACOLOGY, accessed September 7, 2025, https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?tab=clinical&ligandId=9707
- NCT03661320 | A Study to Compare Chemotherapy Alone Versus Chemotherapy Plus Nivolumab or Nivolumab and BMS-986205, Followed by Continued Therapy After Surgery With Nivolumab or Nivolumab and BMS-986205 in Participants With Muscle Invasive Bladder Cancer | ClinicalTrials.gov, accessed September 7, 2025, https://www.clinicaltrials.gov/study/NCT03661320
- Is There a Clinical Future for IDO1 Inhibitors After the Failure of Epacadostat in Melanoma?, accessed September 7, 2025, https://www.annualreviews.org/content/journals/10.1146/annurev-cancerbio-030419-033635
- Epacadostat (E) plus pembrolizumab (P) versus pembrolizumab alone in patients (pts) with unresectable or metastatic melanoma: Results of the phase 3 ECHO-301/KEYNOTE-252 study. - ASCO Publications, accessed September 7, 2025, https://ascopubs.org/doi/10.1200/JCO.2018.36.15_suppl.108
- Life After ECHO-301: Lessons Learned About Modulating IDO/TDO ..., accessed September 7, 2025, https://lifescivc.com/2018/04/life-after-echo-301-lessons-learned-about-modulating-ido-tdo-to-treat-cancer/
- Is There a Clinical Future for IDO1 Inhibitors After the Failure of Epacadostat in Melanoma? - Semantic Scholar, accessed September 7, 2025, https://pdfs.semanticscholar.org/cc1b/addb3aa16de771a2a832a408ebd87f98294a.pdf
- Reimagining IDO Pathway Inhibition in Cancer Immunotherapy via Downstream Focus on the Tryptophan–Kynurenine–Aryl Hydrocarbon Axis - AACR Journals, accessed September 7, 2025, https://aacrjournals.org/clincancerres/article/25/5/1462/82368/Reimagining-IDO-Pathway-Inhibition-in-Cancer