Indoximod (DB12827): A Comprehensive Monograph on a First-in-Class IDO Pathway Modulator
Executive Summary
Indoximod (DrugBank ID: DB12827) is an investigational, orally administered, small-molecule immunometabolic agent designed to counteract tumor-mediated immune suppression. It represents a first-in-class approach to targeting the indoleamine 2,3-dioxygenase (IDO) pathway, a critical mechanism of immune escape in cancer. Unlike the class of direct IDO1 enzymatic inhibitors that failed in late-stage clinical trials, Indoximod possesses a distinct and nuanced mechanism of action. It does not directly inhibit the IDO1 enzyme but instead functions downstream as a tryptophan mimetic. This action creates a "tryptophan sufficiency signal" that reactivates the mTORC1 signaling pathway in T-cells, thereby rescuing them from the anergic state induced by tryptophan depletion in the tumor microenvironment. This mechanistic differentiation theoretically confers resistance to tumor bypass pathways that may have contributed to the failure of direct enzymatic inhibitors, positioning Indoximod as a scientifically unique compound.
Clinically, Indoximod has demonstrated a favorable safety and tolerability profile, with a maximum tolerated dose not being reached in early-phase studies. It has shown compelling signals of efficacy, most notably in a Phase 2 trial in combination with pembrolizumab for advanced melanoma, where it produced a high objective response rate of 51% and a complete response rate of 20%. Despite this promise, its development trajectory has been convoluted. The high-profile failure of a competitor's direct IDO1 inhibitor, Epacadostat, created a negative perception of the entire target class, leading the primary developer, NewLink Genetics, to halt its pivotal melanoma program and restructure. Following a merger with Lumos Pharma, which shifted corporate focus to rare endocrine diseases, the stewardship of Indoximod's development has largely transitioned to academic institutions and the National Cancer Institute.
Currently, the most active area of investigation is in pediatric neuro-oncology, where Indoximod is being evaluated in combination with chemotherapy and radiation for recurrent brain tumors and newly diagnosed diffuse intrinsic pontine glioma (DIPG). Early results from these studies have been encouraging, supporting a continued, albeit more niche, development path. The story of Indoximod serves as a critical case study in pharmaceutical development, illustrating how a scientifically distinct agent with clinical promise can be profoundly impacted by external market forces and the oversimplification of drug classes. Its future now hinges on the success of ongoing, academically-led trials in high-unmet-need populations, where it may yet fulfill its potential as a valuable immunometabolic adjuvant.
Section 1: Drug Identity and Physicochemical Profile
1.1. Nomenclature, Synonyms, and Key Identifiers
Indoximod is a small molecule that has been investigated under various names and identifiers throughout its development history. Establishing a clear nomenclature is essential for cross-referencing scientific literature and regulatory documents.
- Generic Name: Indoximod [1]
- Systematic/IUPAC Name: (2R)-2-amino-3-(1-methyl-1H-indol-3-yl)propanoic acid [1]
- Common Synonyms and Developmental Codes: The compound is widely known as 1-methyl-D-tryptophan, a name that reflects its chemical structure as a methylated D-isomer of the amino acid tryptophan. Other common synonyms include D-(+)-1-methyltryptophan, D-1-methyltryptophan, and the abbreviation D-1MT. During its development, it has also been referred to by the codes NLG-8189 and NSC-721782.[1]
- Registry Identifiers: To ensure unambiguous identification, Indoximod is cataloged in numerous chemical and pharmacological databases under specific identifiers:
- DrugBank Accession Number: DB12827 [1]
- CAS (Chemical Abstracts Service) Number: 110117-83-4 [2]
- PubChem Compound ID (CID): 405012 [2]
- FDA Unique Ingredient Identifier (UNII): TX5CYN1KMZ [2]
- ChEMBL ID: CHEMBL571209 [2]
- KEGG ID: D10640 [2]
1.2. Molecular Structure and Chemical Classification
Indoximod's biological activity is intrinsically linked to its chemical structure as an analog of the essential amino acid L-tryptophan.
- Chemical Formula: The empirical formula for Indoximod is C12H14N2O2.[1]
- Molecular Weight: The average molecular weight is 218.256 g/mol, with a monoisotopic mass of 218.105527699 Da.[1]
- Structural Representations: Standardized chemical notations provide a precise description of its structure:
- SMILES (Simplified Molecular Input Line Entry System): CN1C=C(C2=CC=CC=C21)C[C@H](C(=O)O)N [2]
- InChI (International Chemical Identifier): InChI=1S/C12H14N2O2/c1-14-7-8(6-10(13)12(15)16)9-4-2-3-5-11(9)14/h2-5,7,10H,6,13H2,1H3,(H,15,16)/t10-/m1/s1 [2]
- InChIKey: ZADWXFSZEAPBJS-SNVBAGLBSA-N [2]
- Chemical Classification: Indoximod is classified as an investigational small molecule.[1] More specifically, it belongs to the class of organic compounds known as indolyl carboxylic acids and derivatives. It is a D-alpha-amino acid, characterized by an amino group on the alpha-carbon relative to the carboxyl group, with R-stereochemistry. Its structure features an indole ring system, making it an aromatic and cyclic amino acid derivative, and the methyl group on the indole nitrogen classifies it as an N-alkylindole.[1] This structural similarity to L-tryptophan is the foundation of its mechanism of action.
1.3. Physicochemical Properties and Pharmacopharmaceutical Considerations
The physicochemical properties of a drug candidate are critical determinants of its formulation, oral bioavailability, and overall clinical developability. Indoximod's profile presents a mixture of favorable and challenging characteristics.
- Physical Appearance: It is described as a white to off-white solid powder.[4]
- Solubility: A significant challenge in the development of Indoximod is its poor solubility in common pharmaceutical solvents. Its calculated water solubility is low, at 0.721 mg/mL.[1] Experimental data confirm its insolubility in water, ethanol, and even DMSO under certain conditions, though it shows some solubility in 0.1M NaOH and 5% TFA solutions.[5] This property has direct implications for its formulation and absorption characteristics.
- Lipophilicity: The compound is relatively hydrophilic, as indicated by its negative partition coefficient (logP) values, which range from -0.82 to -0.86.[1]
- Ionization: With a strongly acidic pKa of 2.58 (carboxylic acid) and a strongly basic pKa of 9.39 (primary amine), Indoximod exists predominantly as a zwitterion at physiological pH, with a calculated physiological charge of 0.[1]
- Drug-Likeness Assessment: Indoximod's properties have been evaluated against several empirical rules used to predict oral bioavailability and drug-likeness:
- Lipinski's Rule of Five: It passes this rule, suggesting that its fundamental properties (molecular weight < 500, logP < 5, hydrogen bond donors ≤ 5, hydrogen bond acceptors ≤ 10) are within a range conducive to oral absorption.[1]
- Other Rules: It fails the Ghose Filter, Veber's Rule, and the MDDR-like Rule, which may indicate potential liabilities related to its structural features or flexibility (e.g., rotatable bond count of 3).[1]
The physicochemical profile, particularly the poor aqueous solubility, is a central theme in the story of Indoximod's development. This fundamental property is a likely contributor to the pharmacokinetic observation of saturated absorption at higher clinical doses.[8] When a drug's dissolution rate in the gastrointestinal tract is slower than its absorption rate, solubility becomes the rate-limiting step for bioavailability. As the dose increases, the gastrointestinal fluid becomes saturated, and further increases in dose do not lead to a proportional increase in the amount of drug absorbed into the bloodstream. This phenomenon directly influenced the selection of the recommended Phase 2 dose, which was based on this absorption plateau rather than toxicity. Furthermore, this solubility challenge likely provided the primary motivation for preclinical research into prodrugs, such as NLG802, which were synthesized with the explicit goal of improving oral bioavailability.[9]
Table 1: Key Identifiers and Physicochemical Properties of Indoximod | |
---|
Property | Value / Identifier |
DrugBank ID | DB12827 1 |
CAS Number | 110117-83-4 2 |
IUPAC Name | (2R)-2-amino-3-(1-methyl-1H-indol-3-yl)propanoic acid 1 |
Chemical Formula | C12H14N2O2 1 |
Average Molecular Weight | 218.256 g/mol 1 |
Appearance | White to off-white solid powder 4 |
Water Solubility | 0.721 mg/mL 1 |
logP | -0.82 1 |
pKa (Strongest Acidic) | 2.58 1 |
pKa (Strongest Basic) | 9.39 1 |
Rule of Five Compliance | Yes 1 |
Hydrogen Bond Acceptors | 3 1 |
Hydrogen Bond Donors | 2 1 |
Polar Surface Area | 68.25 A˚2 1 |
Section 2: Nonclinical Pharmacology and Mechanism of Action
2.1. The Indoleamine 2,3-Dioxygenase (IDO) Pathway in Oncogenesis
The rationale for developing Indoximod is rooted in the critical role of the indoleamine 2,3-dioxygenase (IDO) pathway as a mechanism of cancer immune evasion. IDO1 is a cytosolic, heme-containing enzyme that catalyzes the first and rate-limiting step in the catabolism of the essential amino acid L-tryptophan along the kynurenine pathway.[1] While this pathway is involved in normal physiological processes, including maternal-fetal tolerance and prevention of autoimmunity, it is frequently hijacked by tumors to create an immunosuppressive microenvironment.[1]
In the context of cancer, IDO1 is often overexpressed by tumor cells themselves or by antigen-presenting cells (APCs), such as dendritic cells, within the tumor microenvironment (TME) and tumor-draining lymph nodes.[3] This overexpression leads to two primary immunosuppressive consequences:
- Tryptophan Depletion: The enzymatic degradation of tryptophan creates a localized state of amino acid starvation. T-cells are highly sensitive to tryptophan levels, and its depletion causes them to arrest in the G1 phase of the cell cycle, become anergic (unresponsive), and ultimately undergo apoptosis.[1]
- Accumulation of Immunosuppressive Metabolites: The catabolism of tryptophan produces a cascade of metabolites, most notably kynurenine. Kynurenine and its derivatives are not inert byproducts; they actively suppress the function of effector immune cells, such as CD8+ T-cells and Natural Killer (NK) cells, while promoting the induction, activation, and expansion of immunosuppressive cell populations, including regulatory T-cells (Tregs) and myeloid-derived suppressor cells (MDSCs).[10]
Together, these effects establish a powerful "immune checkpoint" that shields the tumor from immune surveillance and attack, thereby facilitating tumor growth, progression, and metastasis.[3] This understanding established the IDO pathway as a high-priority target for cancer immunotherapy.
2.2. Indoximod's Distinct Mechanism: A Downstream Immunometabolic Modulator
The central and most critical aspect of Indoximod's pharmacology is that it is fundamentally different from the first generation of IDO-targeting drugs. While commonly referred to as an "IDO inhibitor," this term is functionally imprecise and masks its unique mechanism.
A crucial distinction must be made: Indoximod, which is the D-isomer of 1-methyltryptophan, does not directly bind to or inhibit the catalytic activity of the purified IDO1 enzyme.[11] This has been confirmed in cell-free enzymatic assays, which show that while the L-isomer of 1-methyltryptophan has weak competitive inhibitory activity (with reported
IC50 and Ki values of 7 µM and 19 µM, respectively), the D-isomer (Indoximod) is ineffective as a direct enzymatic inhibitor.[3] Therefore, its anti-tumor effects are not mediated by blocking the active site of the IDO1 enzyme.
Instead, Indoximod functions as an "IDO pathway inhibitor" or an "immunometabolic adjuvant".[4] Its primary mechanism of action occurs downstream of the IDO1 enzyme, where it reverses the functional consequences of IDO1 activity on immune cells, particularly T-cells.[11] This mechanistic divergence is not merely an academic detail; it is the cornerstone of the scientific rationale for its continued development, especially in light of the clinical failures of direct enzymatic inhibitors.
2.3. The Tryptophan Mimetic Hypothesis: Reactivation of mTORC1 Signaling
The leading hypothesis for Indoximod's mechanism of action centers on its ability to act as a molecular mimic of tryptophan, thereby counteracting the starvation signals induced by IDO1.
Tryptophan depletion in the TME triggers a stress response in T-cells that is sensed by intracellular signaling pathways. One of the master regulators of cellular growth and metabolism is the mammalian target of rapamycin complex 1 (mTORC1), which functions as a critical sensor of amino acid availability.[11] When tryptophan levels are low, mTORC1 activity is suppressed. This downregulation shifts the T-cell's metabolic program from growth and proliferation towards a state of conservation and autophagy, leading to the observed T-cell anergy and arrest.[5]
Indoximod, as a D-tryptophan analog, is interpreted by the mTORC1 signaling machinery as a high-potency L-tryptophan mimetic. It effectively generates a false "tryptophan sufficiency signal" within the T-cell.[5] This signal is sufficient to reactivate mTORC1, even in an environment where actual L-tryptophan concentrations remain low. The reactivation of mTORC1 restores the T-cell's metabolic programming, reverses the autophagic response, and allows the T-cell to overcome the IDO-induced immunosuppressive block, thereby restoring its proliferative and effector functions.[5] This mechanism effectively "rescues" or "re-awakens" T-cells that have been silenced by the tumor's metabolic warfare.
This downstream mechanism has profound strategic implications. The failure of direct, competitive IDO1 inhibitors like Epacadostat in large Phase 3 trials was a major setback for the field. One plausible reason for this failure is the existence of tumor bypass mechanisms. For instance, tumors could upregulate other tryptophan-catabolizing enzymes, such as tryptophan 2,3-dioxygenase (TDO) or IDO2, rendering a highly specific IDO1 inhibitor ineffective.[12] Because Indoximod acts on the convergent downstream effector pathway (mTORC1 suppression) that is common to all sources of tryptophan depletion, its action is theoretically agnostic to which specific enzyme (IDO1, IDO2, or TDO) is active in the TME.[11] This provides a strong scientific rationale for why Indoximod might succeed where direct inhibitors have failed and represents its most significant potential advantage.
However, this scientific nuance was largely lost in the aftermath of the Epacadostat failure. The investment and pharmaceutical communities widely perceived a "class effect" failure, leading to a broad retreat from the entire IDO-targeting space.[17] NewLink Genetics explicitly cited the "failure of a competitor's trial" as the primary reason for halting its own promising Phase 3 melanoma program with Indoximod.[18] In this context, Indoximod became collateral damage; its development in mainstream immuno-oncology was derailed not by its own data, but by the failure of a mechanistically dissimilar competitor targeting the same pathway. The subsequent pivot of Indoximod's development into academically-driven trials in pediatric neuro-oncology can be viewed as a strategic retreat to a less commercially competitive space where its unique science could continue to be explored.
2.4. Additional and Secondary Mechanisms
The biological effects of Indoximod are complex and likely involve multiple complementary mechanisms beyond mTORC1 reactivation.
- Aryl Hydrocarbon Receptor (AhR) Modulation: Kynurenine, the primary product of IDO1 activity, is a natural ligand for the Aryl Hydrocarbon Receptor (AhR), a transcription factor that can promote an immunosuppressive Treg phenotype.[11] Indoximod has been shown to modulate AhR-dependent transcriptional activity and, in some preclinical models, can even lead to the downregulation of IDO1 protein expression in dendritic cells through a mechanism involving AhR signaling.[16] This suggests Indoximod may not only rescue effector T-cells but also reprogram the local immune environment.
- IDO2 Inhibition: There is some preclinical evidence to suggest that Indoximod may also function as an indirect inhibitor of IDO2, another tryptophan-catabolizing enzyme.[11]
- Functional Destabilization of the IDO Pathway: Rather than a simple on/off switch, the overall effect of Indoximod appears to be a broader functional destabilization of the IDO pathway within key APCs. This leads to a shift away from a tolerogenic state and towards an immunogenic presentation of tumor antigens, ultimately enhancing anti-tumor immune responses.[20]
Section 3: Clinical Pharmacokinetics
3.1. Absorption, Distribution, and Elimination in Human Subjects
The clinical pharmacokinetic (PK) profile of Indoximod has been characterized in several early-phase studies, providing a basis for its dosing regimen in later trials.
- Administration and Formulation: Indoximod is an orally available small molecule, administered as capsules or tablets.[8] Efforts to optimize its formulation and bioavailability have been a part of its development, as evidenced by a Phase 1 trial in Acute Myeloid Leukemia (AML) that was designed to compare the serum concentrations of a freebase formulation versus two different hydrochloride (HCl) salt formulations (F1 and F2).[22]
- Absorption: Following oral administration, Indoximod is absorbed, with the time to reach maximum plasma concentration (Tmax) occurring at a median of 2.9 hours post-dose in a Phase 1 study of patients with advanced solid tumors (NCT00567931).[8] A dedicated Phase 0 study (NCT03852446) was also conducted in healthy male volunteers to more formally assess single ascending dose PK, absolute oral bioavailability, and the effect of food on absorption, though specific results from this trial are not detailed in the available materials.[23]
- Dose-Proportionality and Saturation: A key pharmacokinetic feature of Indoximod is its non-linear dose-exposure relationship at higher doses. In the Phase 1 dose-escalation study, the area under the curve (AUC) and maximum concentration (Cmax) were observed to plateau at doses above 1200 mg twice daily (BID). The Cmax achieved at the highest tested dose of 2000 mg BID was approximately 12 µM.[8] This absorption ceiling is a classic indicator of solubility-limited or transporter-mediated absorption, consistent with the compound's poor aqueous solubility. This finding was pivotal, as it informed the selection of the recommended Phase 2 dose (RP2D). Since the maximum tolerated dose (MTD) was never reached from a toxicity perspective, the dose for further development (1200 mg BID) was effectively a "pharmacokinetic MTD," chosen based on the principle that escalating the dose further would not result in a meaningful increase in systemic drug exposure.[8]
- Elimination Half-Life: The elimination half-life of Indoximod was determined to be approximately 10.5 hours.[8] This half-life is sufficiently long to support a convenient twice-daily (BID) oral dosing schedule, which has been used in most subsequent clinical trials.
3.2. Pharmacokinetic-Pharmacodynamic Correlations
Connecting the pharmacokinetic profile of Indoximod to its pharmacodynamic (PD) effects—the biological changes it induces in the body—is crucial for understanding its activity and optimizing its use.
- Target Pathway Modulation: Clinical trials have incorporated PD endpoints to confirm that Indoximod engages its intended biological pathway. A primary method has been the measurement of serum levels of tryptophan and its catabolite, kynurenine.[22]
In vitro studies using both murine and human cells have demonstrated that treatment with 1-MT (Indoximod) leads to an increase in tryptophan concentrations and a corresponding decrease in kynurenine levels. The kynurenine-to-tryptophan ratio, often used as a surrogate marker for IDO enzyme activity, was consistently decreased by 1-MT treatment, confirming its ability to functionally inhibit the consequences of the IDO pathway.[4]
- Systemic Inflammatory Response: An interesting and potentially important PD finding from the Phase 1 solid tumor trial was a dose-dependent increase in C-reactive protein (CRP) levels in treated patients.[8] CRP is a general marker of systemic inflammation. This observation suggests that by reversing IDO-mediated immune suppression, Indoximod may induce a pro-inflammatory state, which could be a desired component of its anti-tumor effect.
- Immune Cell Correlates: More advanced PD analyses have been conducted in recent trials. In the Phase 1 pediatric brain tumor study, blood samples were collected for single-cell RNA sequencing and T-cell receptor sequencing. This analysis confirmed the emergence of new circulating CD8+ T-cell clonotypes with a late effector phenotype in treated patients, providing direct evidence of systemic immune activation and diversification of the anti-tumor T-cell repertoire.[20]
Section 4: Clinical Development and Efficacy Analysis
4.1. Corporate and Development History: A Convoluted Path
The clinical development of Indoximod has been characterized by scientific promise, strategic pivots, and significant influence from external market dynamics.
- Origination and Initial Development: The compound originated from research at the Lankenau Institute for Medical Research.[26] Its primary clinical development was subsequently undertaken by NewLink Genetics Corporation, a biopharmaceutical company focused on immuno-oncology.[17]
- Strategic Turmoil and Restructuring: The trajectory of Indoximod was profoundly altered in April 2018 by the high-profile failure of the Phase 3 ECHO-301 trial, which tested a competitor's direct IDO1 enzymatic inhibitor, Epacadostat, in combination with pembrolizumab for melanoma. The failure of Epacadostat cast a pall over the entire IDO inhibitor class, creating significant headwinds for companies like NewLink Genetics.[17] In response to this changing landscape and financial pressures, NewLink Genetics announced it would not proceed with its own planned pivotal trial in melanoma and initiated a corporate restructuring that included a 30% reduction in its workforce. The company's clinical focus was narrowed to ongoing studies in pediatric brain tumors and acute myeloid leukemia.[17]
- Merger and Shift in Corporate Focus: In September 2019, NewLink Genetics announced a merger with Lumos Pharma.[30] The strategic rationale for the merger was to create a new entity focused on developing therapies for rare diseases. The primary focus of the newly formed Lumos Pharma became its lead candidate, LUM-201, an oral therapy for pediatric growth hormone deficiency.[30]
- Current Stewardship: While Lumos Pharma formally holds the asset and provides the drug for ongoing studies, the active clinical development of Indoximod has effectively transitioned to being primarily driven and sponsored by academic institutions, most notably Augusta University, with significant funding from the National Cancer Institute (NCI).[24] This shift has ensured the continued investigation of Indoximod in areas of high unmet need, particularly pediatric neuro-oncology, but has left its broader commercial future uncertain.
4.2. Efficacy in Advanced Melanoma: A Story of Promise and Pivots
The most compelling efficacy data for Indoximod in a large adult population came from its investigation in advanced melanoma, an indication where immuno-oncology has had a transformative impact.
- The NLG2103/INDIGO Study (Phase 2): This open-label, multi-center Phase 2 study (NCT02073123) was designed to evaluate Indoximod (at a dose of 1200 mg BID) in combination with various approved checkpoint inhibitors, including the anti-PD-1 antibodies pembrolizumab and nivolumab, and the anti-CTLA-4 antibody ipilimumab.[14]
- Impressive Clinical Results: The primary results focused on the cohort of 89 efficacy-evaluable patients with advanced, non-ocular melanoma treated with Indoximod plus pembrolizumab. The combination demonstrated a high level of anti-tumor activity [14]:
- Objective Response Rate (ORR): The investigator-assessed ORR was 51%, a figure that compares favorably to the ~40% ORR typically seen with pembrolizumab monotherapy in this setting.
- Complete Response (CR) Rate: Perhaps most impressively, the combination yielded a confirmed CR rate of 20%, suggesting deep and durable responses in a significant subset of patients.
- Disease Control Rate (DCR): The DCR (CR + Partial Response + Stable Disease) was 70%.
- Progression-Free Survival (PFS): The median PFS was 12.4 months (95% CI 6.4 to 24.9).
- The Strategic Halt: These results, presented in 2017 and 2018, were highly encouraging and positioned Indoximod as a potentially best-in-class combination partner for PD-1 inhibitors in melanoma.[29] A pivotal Phase 3 trial was planned. However, as described previously, NewLink Genetics announced in April 2018 that it would not initiate the randomized portion of the study, directly citing the failure of the competitor Epacadostat trial as the reason for re-evaluating the path forward.[17] This decision effectively halted the most promising commercial development program for Indoximod, despite its own strong data and distinct mechanism of action.
4.3. A Niche in Neuro-Oncology: The Pediatric Brain Tumor Program
Following the pivot away from melanoma, the clinical development of Indoximod has become concentrated in pediatric neuro-oncology, an area with a desperate need for new therapeutic options.
- NLG2105 (Phase 1, NCT02502708): This landmark study was the first clinical trial of any IDO pathway inhibitor in a pediatric population. The dose-escalation trial evaluated the safety and preliminary efficacy of Indoximod in combination with standard therapies, primarily temozolomide and/or radiation, in children with recurrent or progressive brain tumors (including glioblastoma, medulloblastoma, and ependymoma) and in a cohort of newly diagnosed diffuse intrinsic pontine glioma (DIPG) patients.[18]
- Key Findings: The study successfully established a pediatric dose of 19.2 mg/kg/dose BID. The combination was found to be safe and well-tolerated. Encouraging signals of efficacy were observed, with a median overall survival (OS) of 13.3 months for the recurrent disease population and 14.4 months for the DIPG cohort. A critical finding was that the subset of patients who achieved an objective response (even partial or mixed) had a more than 3-fold longer median OS of 25.2 months compared to non-responders.[24]
- GCC1949 (Phase 2, NCT04049669): Based on the positive results of the Phase 1 trial, this NCI-funded, open-label Phase 2 study was initiated. It is designed to further evaluate Indoximod-based combination chemo-radio-immunotherapy for patients aged 3 to 21 with progressive brain cancer or newly diagnosed DIPG. The study is currently listed as active but no longer recruiting participants.[34]
- GCC2020 (Phase 1, NCT05106296): Reflecting a continued effort to find synergistic combinations, this active Phase 1 trial is exploring a novel four-drug regimen. It combines Indoximod with the Bruton's Tyrosine Kinase (BTK) inhibitor ibrutinib, plus metronomic cyclophosphamide and etoposide, for pediatric patients with brain cancer. The rationale is based on preclinical data suggesting a close link between the IDO and BTK pathways in creating an immunosuppressive metabolic checkpoint.[26]
4.4. Exploration in Hematologic and Other Solid Tumors
Indoximod has been investigated across a wide range of other malignancies, with varying degrees of success.
- Acute Myeloid Leukemia (AML): A Phase 1 trial (NCT02835729) evaluated Indoximod in combination with standard "7+3" induction chemotherapy (cytarabine and idarubicin) for newly diagnosed AML. The combination was reported to be well-tolerated and induced a high rate of complete remission, with a high proportion of patients achieving minimal residual disease (MRD)-negativity, a key prognostic marker.[22]
- Other Solid Tumors: Numerous other trials have been conducted. These include studies in breast cancer, non-small cell lung cancer (NSCLC), pancreatic cancer, and adult glioma.[1] The status of these trials is mixed, with many having been completed, terminated, or withdrawn for various reasons, including lack of efficacy or strategic reprioritization.[1] For example, a Phase 1b study in metastatic pancreatic cancer combining Indoximod with gemcitabine/nab-paclitaxel showed an encouraging ORR of 42%.[21] However, other studies, such as one in metastatic breast cancer, yielded negative results.[27]
Table 2: Summary of Major Clinical Trials for Indoximod | | | | | |
---|
NCT Identifier | Phase | Indication(s) | Key Interventions | Status | Summary of Key Findings/Rationale |
NCT02073123 | 2 | Advanced Melanoma | Indoximod + Pembrolizumab/Nivolumab/Ipilimumab | Completed | Showed high efficacy with ORR of 51% and CR of 20% with pembrolizumab. Planned Phase 3 was halted due to competitor failure.14 |
NCT02502708 | 1 | Pediatric Brain Tumors (recurrent), DIPG (newly diagnosed) | Indoximod + Temozolomide and/or Radiation | Completed | First trial of an IDO inhibitor in children. Established pediatric dose, confirmed safety, and showed encouraging efficacy signals, leading to Phase 2 follow-up.24 |
NCT04049669 | 2 | Pediatric Brain Tumors (progressive), DIPG (newly diagnosed) | Indoximod + Chemo-Radio-Immunotherapy | Active, Not Recruiting | NCI-funded follow-on study to NCT02502708 to further evaluate efficacy in a larger pediatric cohort.39 |
NCT05106296 | 1 | Pediatric Brain Tumors (progressive/refractory) | Indoximod + Ibrutinib + Cyclophosphamide + Etoposide | Recruiting | Investigating a novel 4-drug combination based on preclinical synergy between IDO and BTK pathway inhibition.44 |
NCT02835729 | 1 | Acute Myeloid Leukemia (newly diagnosed) | Indoximod + Cytarabine + Idarubicin (7+3) | Completed | Combination was well-tolerated and showed a high rate of complete remission with MRD-negativity.22 |
NCT02052648 | 1b/2 | Glioblastoma (recurrent) | Indoximod + Temozolomide +/- Bevacizumab | Completed | Phase 1b/2 study to evaluate safety and preliminary activity in adult patients with recurrent high-grade gliomas.28 |
NCT01191216 | 1 | Metastatic Solid Tumors | Indoximod + Docetaxel | Completed | Early phase trial exploring combination with taxane chemotherapy in a broad range of solid tumors.47 |
NCT00567931 | 1 | Metastatic/Refractory Solid Tumors | Indoximod Monotherapy | Completed | Foundational dose-escalation study that established the safety profile and PK, determining that MTD was not reached.8 |
Section 5: Safety and Tolerability Profile
5.1. Integrated Analysis of Adverse Events from Clinical Trials
Across a broad range of clinical trials involving monotherapy and various combination regimens, Indoximod has consistently demonstrated a favorable safety and tolerability profile. This is one of the compound's most significant and attractive clinical attributes.
- General Tolerability: In studies combining Indoximod with chemotherapy, radiation, or checkpoint inhibitors, the treatment has been repeatedly described as well-tolerated, with minimal added toxicity attributed to Indoximod itself.[8]
- Monotherapy Adverse Event Profile: The foundational Phase 1 dose-escalation study (NCT00567931) in 48 patients with advanced solid tumors provides the clearest picture of Indoximod's intrinsic toxicity profile. In this study, the most commonly reported adverse events (AEs) of any grade, regardless of attribution, were generally mild to moderate (Grade 1 or 2) and included fatigue (56.3%), anemia (37.5%), anorexia (37.5%), dyspnea (35.4%), cough (33.3%), and nausea (29.2%).[8] There were no treatment discontinuations due to toxicity; all discontinuations were due to disease progression.
- Combination Therapy Adverse Event Profile: When used in combination, the AE profile has generally been dominated by the known toxicities of the partner agent(s).
- With Checkpoint Inhibitors: In the Phase 2 melanoma trial, the safety profile of Indoximod plus pembrolizumab was reported to be similar to what would be expected from pembrolizumab monotherapy, with no unexpected or synergistic toxicities observed.[14]
- With Chemotherapy: In the Phase 1 trial for newly diagnosed AML, where Indoximod was combined with intensive "7+3" induction chemotherapy, the most frequent Grade 3 or higher non-hematologic AEs were febrile neutropenia (60%), hypoxia (16%), atrial fibrillation (12%), and pneumonia (12%). These are all well-recognized complications of induction chemotherapy and the underlying disease, and no regimen-limiting toxicities were attributed to Indoximod.[46]
5.2. Significant Safety Signals and Management
While generally safe, the clinical development of Indoximod has identified a few key safety aspects that warrant specific attention.
- Maximum Tolerated Dose (MTD): A critical finding from the Phase 1 monotherapy trial was that the MTD was not reached, even at the highest tested dose of 2000 mg orally twice per day.[8] The absence of a dose-limiting toxicity up to this very high dose indicates a wide therapeutic window and underscores the compound's favorable safety profile. This contrasts with many other oncology agents where dose escalation is limited by toxicity.
- Hypophysitis: The most notable and mechanistically interesting serious adverse event attributed to Indoximod was hypophysitis, an inflammation of the pituitary gland. This occurred in three patients, all of whom were treated at the lowest dose level (200 mg once daily) in the Phase 1 solid tumor study. Critically, all three of these patients had received prior treatment with immune checkpoint inhibitors (e.g., ipilimumab or nivolumab).[8] After the protocol was amended to exclude patients with prior checkpoint inhibitor therapy, no further cases of hypophysitis were observed. This pattern strongly suggests a synergistic toxicity or a "priming" effect. Immune checkpoint inhibitors are known to cause a spectrum of immune-related adverse events, including hypophysitis. The pro-inflammatory and immune-reprogramming effects of Indoximod may lower the threshold for, or exacerbate, this specific autoimmune toxicity in an immune system that has already been modulated by a checkpoint inhibitor. This finding has important implications for patient selection, monitoring (e.g., endocrine function tests), and management in future trials that combine Indoximod with checkpoint inhibitors.
Section 6: Expert Analysis and Future Outlook
6.1. Critical Assessment: Differentiating Indoximod in the Failed Landscape of IDO Inhibitors
The story of Indoximod is inseparable from the broader narrative of IDO pathway inhibition in oncology. To properly assess its potential, it is essential to re-emphasize the fundamental mechanistic distinction that sets it apart. The failure of direct, competitive enzymatic inhibitors like Epacadostat led to a widespread conclusion that targeting IDO1 was a flawed strategy. However, this conclusion may be an oversimplification that overlooks the nuances of the underlying biology and pharmacology.
Indoximod's unique, non-enzymatic, downstream mechanism of action provides a robust scientific rationale for why it should not be summarily dismissed along with the direct inhibitors.[11] Its function as an immunometabolic adjuvant that rescues T-cells from tryptophan starvation by reactivating the convergent mTORC1 signaling pathway is a conceptually different and potentially more robust approach.[11] This mechanism is theoretically resilient to the very tumor escape pathways—such as the upregulation of alternative tryptophan-catabolizing enzymes like TDO or IDO2—that may have contributed to the failure of highly specific IDO1 enzymatic blockers. The impressive efficacy signals seen in the melanoma trial, which were generated in parallel with the failures of other agents, lend clinical support to this mechanistic distinction. Indoximod's development was arguably a casualty of a market-driven "class effect" perception rather than a failure of its own scientific or clinical merit.
6.2. Current Status and Unanswered Questions
Despite its scientific appeal, the commercial development path for Indoximod has been effectively stalled. Following the NewLink Genetics/Lumos Pharma merger and the latter's strategic pivot to rare diseases, Indoximod has become an asset without a dedicated corporate champion.[30] Its continued exploration is now largely dependent on the efforts of academic investigators and NCI funding, primarily focused on the niche indication of pediatric neuro-oncology.[26] This situation leaves several critical questions unanswered:
- Can the encouraging efficacy signals observed in the Phase 1 and ongoing Phase 2 pediatric brain tumor trials be validated and translated into a viable registrational pathway?
- Is there a potential to resurrect the melanoma program, perhaps in a more targeted setting, such as in patients who have failed prior checkpoint inhibitor therapy, an area of profound unmet medical need?
- Can a predictive biomarker be identified to select patients most likely to benefit from Indoximod's unique mechanism? The complexity of its downstream action suggests that simple IDO1 protein expression may not be a reliable biomarker.
6.3. Future Trajectory: Potential Pathways to Registration and Biomarker Strategies
The future of Indoximod hinges on successfully navigating its current niche and potentially re-engaging with broader oncology applications.
- Most Viable Path to Registration: The most direct and currently active path to a potential regulatory approval lies in pediatric neuro-oncology. Positive, clinically meaningful results from the NCI-funded Phase 2 GCC1949 trial could provide the basis for a pivotal study. Success in this rare disease setting could leverage regulatory incentives such as Orphan Drug Designation, Rare Pediatric Disease Designation, and Priority Review Vouchers, which could, in turn, renew commercial interest in the compound.
- The Need for Biomarker Development: Long-term success, particularly if the drug is to be reconsidered for larger indications like melanoma, will almost certainly require the development of a predictive biomarker. Given its mechanism, an effective biomarker may not be based on IDO1 expression alone. Instead, a more sophisticated approach may be required, potentially involving a gene expression signature related to tryptophan metabolism, amino acid sensing pathways (like GCN2), or mTORC1 signaling status within the tumor microenvironment. The exploratory single-cell sequencing work being conducted in the current pediatric trials is a crucial first step in this direction and may uncover the signatures of response.[20]
In final assessment, Indoximod remains a scientifically compelling immunometabolic agent with a distinct mechanism of action, a favorable safety profile, and demonstrated clinical activity. Its development was unfortunately curtailed by broader market dynamics and strategic corporate shifts that were disconnected from its own data. Its future now rests on the success of academically led trials in high-unmet-need pediatric populations. If successful, these studies could not only provide a new therapeutic option for a vulnerable patient group but also serve as the proof-of-concept needed to vindicate its unique scientific rationale and potentially resurrect its development for broader applications in oncology.
Works cited
- Indoximod: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed September 13, 2025, https://go.drugbank.com/drugs/DB12827
- Indoximod | C12H14N2O2 | CID 405012 - PubChem, accessed September 13, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/1-MT
- 1-methyl-D-Tryptophan (D-1MT, Indoximod, CAS Number - Cayman Chemical, accessed September 13, 2025, https://www.caymanchem.com/product/16456/1-methyl-d-tryptophan
- Indoximod | NLG8189 | CAS#110117-83-4 | IDO inhibitor | MedKoo Biosciences, accessed September 13, 2025, https://www.medkoo.com/products/5539
- Indoximod | 99.83%(HPLC) | In Stock | IDO/TDO inhibitor - Selleck Chemicals, accessed September 13, 2025, https://www.selleckchem.com/products/indoximod-nlg-8189.html
- INDOXIMOD - precisionFDA, accessed September 13, 2025, https://precision.fda.gov/ginas/app/ui/substances/TX5CYN1KMZ
- Indoximod, Indoleamine 2,3-dioxygenase (IDO) inhibitor (CAS 110117-83-4) | Abcam, accessed September 13, 2025, https://www.abcam.com/en-us/products/biochemicals/indoximod-indoleamine-23-dioxygenase-ido-inhibitor-ab231506
- A phase I study of indoximod in patients with advanced ..., accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC5008412/
- Comparison of indoximod pharmacokinetics after single dose of indoximod or NLG802 in mice. - ResearchGate, accessed September 13, 2025, https://www.researchgate.net/figure/Comparison-of-indoximod-pharmacokinetics-after-single-dose-of-indoximod-or-NLG802-in-mice_tbl1_341087694
- Limitations and Off-Target Effects of Tryptophan-Related IDO Inhibitors in Cancer Treatment, accessed September 13, 2025, https://www.frontiersin.org/journals/immunology/articles/10.3389/fimmu.2019.01801/full
- Discovery of IDO1 Inhibitors: From Bench to Bedside | Cancer Research - AACR Journals, accessed September 13, 2025, https://aacrjournals.org/cancerres/article/77/24/6795/624889/Discovery-of-IDO1-Inhibitors-From-Bench-to
- Indoximod: An Immunometabolic Adjuvant That Empowers T Cell ..., accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6141803/
- Indoximod | Indoleamine 2,3-Dioxygenase (IDO) - TargetMol, accessed September 13, 2025, https://www.targetmol.com/compound/indoximod
- Phase II trial of the IDO pathway inhibitor indoximod plus pembrolizumab for the treatment of patients with advanced melanoma | Journal for ImmunoTherapy of Cancer, accessed September 13, 2025, https://jitc.bmj.com/content/9/6/e002057
- Preclinical investigations and a first-in-human phase I trial of M4112, the first dual inhibitor of indoleamine 2,3-dioxygenase 1 and tryptophan 2,3-dioxygenase 2, in patients with advanced solid tumors - The Journal for ImmunoTherapy of Cancer, accessed September 13, 2025, https://jitc.bmj.com/content/8/2/e000870
- Indoximod opposes the immunosuppressive effects mediated by IDO and TDO via modulation of AhR function and activation of mTORC1 - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC7321702/
- NewLink cuts 30% of staff to continue indoximod development ..., accessed September 13, 2025, https://www.biopharmadive.com/news/newlink-cuts-30-of-staff-to-continue-indoximod-development/529131/
- NewLink Genetics Announces Initial Phase 1 Data with, accessed September 13, 2025, https://www.globenewswire.com/news-release/2018/04/15/1471681/0/en/NewLink-Genetics-Announces-Initial-Phase-1-Data-with-Indoximod-Plus-Radiation-and-Chemotherapy-for-Pediatric-Patients-with-Diffuse-Intrinsic-Pontine-Glioma-DIPG-Presented-During-AA.html
- NewLink Genetics Announces Initial Phase 1 Data with Indoximod Plus Radiation and Chemotherapy for Pediatric Patients with Diffuse Intrinsic Pontine Glioma (DIPG) Presented During AACR Plenary, accessed September 13, 2025, http://investors.linkp.com/news-releases/news-release-details/newlink-genetics-announces-initial-phase-1-data-indoximod-plus
- Indoximod-based chemo-immunotherapy for pediatric brain tumors: A first-in-children phase I trial | Neuro-Oncology | Oxford Academic, accessed September 13, 2025, https://academic.oup.com/neuro-oncology/article/26/2/348/7275413
- NewLink Genetics Presents Phase 1b Data of Indoximod in Combination With Gemcitabine/Nab-Paclitaxel for Patients With Metastatic Pancreatic Cancer That Show Encouraging Durable Responses With Delayed Pattern | Lumos Pharma, accessed September 13, 2025, http://investors.linkp.com/news-releases/news-release-details/newlink-genetics-presents-phase-1b-data-indoximod-combination
- A Study of Indoximod in Combination With (7+3) Chemotherapy in Patients With Newly Diagnosed Acute Myeloid Leukemia | ClinicalTrials.gov, accessed September 13, 2025, https://clinicaltrials.gov/study/NCT02835729?term=AREA%5BConditionSearch%5D(%22Leukemia%22)%20AND%20AREA%5BInterventionSearch%5D(%22Tryptophan%22)&rank=1
- Indoximod Completed Phase 0 Trials for Healthy Lifestyle Behaviors Treatment - DrugBank, accessed September 13, 2025, https://go.drugbank.com/drugs/DB12827/clinical_trials?conditions=DBCOND0059969&phase=0&purpose=treatment&status=completed
- Indoximod-based chemo-immunotherapy for pediatric brain tumors: A first-in-children phase I trial - PubMed Central, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC10836763/
- Lumos Pharma, Inc. - Drug pipelines, Patents, Clinical trials - Patsnap Synapse, accessed September 13, 2025, https://synapse.patsnap.com/organization/2163d9465d00117579c58dff902129b8
- Indoximod - Lumos Pharma - AdisInsight - Springer, accessed September 13, 2025, https://adisinsight.springer.com/drugs/800027736
- Indoximod - Drug Targets, Indications, Patents - Synapse, accessed September 13, 2025, https://synapse.patsnap.com/drug/f86b4979b4a54c848bc3afa3289893b0
- NCT02052648 | Study of IDO Inhibitor and Temozolomide for Adult Patients With Primary Malignant Brain Tumors | ClinicalTrials.gov, accessed September 13, 2025, https://www.clinicaltrials.gov/study/NCT02052648?term=AREA%5BBasicSearch%5D(AREA%5BNCTIdSearch%5D(nct02052648))&rank=1
- NewLink surges on fresh indoximod data - BioPharma Dive, accessed September 13, 2025, https://www.biopharmadive.com/news/newlink-indoximod-data-ido-competition/504569/
- NewLink Genetics and Lumos Pharma Enter into Merger Agreement ..., accessed September 13, 2025, https://lumos-pharma.com/newlink-genetics-and-lumos-pharma-enter-into-merger-agreement-to-form-biopharmaceutical-company-focused-on-developing-therapies-to-treat-rare-diseases/
- Our Pipeline | Lumos Pharma, accessed September 13, 2025, https://lumos-pharma.com/our-pipeline/
- Lumos Pharma Homepage, accessed September 13, 2025, https://lumos-pharma.com/
- LUM-201 Program Pipeline Slide – Mar 2023 - Lumos Pharma, accessed September 13, 2025, https://lumos-pharma.com/our-pipeline/lum-201-program-pipeline-slide-mar-2023/
- Phase 2 trial using indoximod-based chemo-immunotherapy for patients with childhood brain cancer: Interim analysis of the GCC1949 - Augusta University, accessed September 13, 2025, https://www.augusta.edu/cancer/clinical-trials/pediatric-immunotherapy/documents/2024johnsonispno.pdf
- A Phase 2/3 Study of Indoximod or Placebo Plus Pembrolizumab or Nivolumab in Subjects With Unresectable or Metastatic Melanoma - Mayo Clinic, accessed September 13, 2025, https://www.mayo.edu/research/clinical-trials/cls-20368635
- Phase II trial of the IDO pathway inhibitor indoximod plus pembrolizumab for the treatment of patients with advanced melanoma - PMC, accessed September 13, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8202104/
- A phase I/II study of the combination of indoximod and temozolomide for adult patients with temozolomide-refractory primary malignant brain tumors. - ASCO Publications, accessed September 13, 2025, https://ascopubs.org/doi/10.1200/jco.2014.32.15_suppl.tps2107
- Clinical Trials Using Indoximod - NCI, accessed September 13, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/indoximod?pn=1
- Pediatric Trial of Indoximod With Chemotherapy and Radiation for Relapsed Brain Tumors or Newly Diagnosed DIPG - National Cancer Institute, accessed September 13, 2025, https://www.cancer.gov/research/participate/clinical-trials-search/v?id=NCI-2019-08285
- Study Details | NCT04049669 | Pediatric Trial of Indoximod With Chemotherapy and Radiation for Relapsed Brain Tumors or Newly Diagnosed DIPG | ClinicalTrials.gov, accessed September 13, 2025, https://clinicaltrials.gov/study/NCT04049669
- Phase 2 trial of indoximod with chemotherapy and radiation for children with progressive brain tumors or newly diagnosed DIPG - Augusta University, accessed September 13, 2025, https://www.augusta.edu/research/studies/trial.php?study=1441827
- GCC1949: Phase 2 trial of indoximod with chemotherapy and radiation for children with progressive brain tumors or newly diagnosed DIPG, accessed September 13, 2025, https://www.mdanderson.org/patients-family/diagnosis-treatment/clinical-trials/clinical-trials-index/clinical-trials-detail.ID2024-0303.html
- Clinical Trials Using IDO1 Inhibitor - NCI, accessed September 13, 2025, https://www.cancer.gov/research/participate/clinical-trials/intervention/ido1-inhibitor?pn=1
- Study Details | NCT05106296 | Chemo-immunotherapy Using Ibrutinib Plus Indoximod for Patients With Pediatric Brain Cancer | ClinicalTrials.gov, accessed September 13, 2025, https://www.clinicaltrials.gov/study/NCT05106296?intr=INDOXIMOD&aggFilters=studyType:int&viewType=Table&rank=10
- Chemo-immunotherapy Using Ibrutinib Plus Indoximod for Patients With Pediatric Brain Cancer, accessed September 13, 2025, https://www.georgiacancerinfo.org/trialDetailPrint.aspx?trialID=5766
- Indoximod Combined with Standard Induction Chemotherapy Is Well Tolerated and Induces a High Rate of Complete Remission with MRD-Negativity in Patients with Newly Diagnosed AML: Results from a Phase 1 Trial | Blood - ASH Publications, accessed September 13, 2025, https://ashpublications.org/blood/article/132/Supplement%201/332/264878/Indoximod-Combined-with-Standard-Induction
- Indoximod Completed Phase 1 Trials for Unspecified Adult Solid Tumor, Protocol Specific Treatment - DrugBank, accessed September 13, 2025, https://go.drugbank.com/drugs/DB12827/clinical_trials?conditions=DBCOND0028453&phase=1&purpose=treatment&status=completed
- Indoximod Withdrawn Phase 1 Trials for Advanced Solid Tumors Treatment - DrugBank, accessed September 13, 2025, https://go.drugbank.com/drugs/DB12827/clinical_trials?conditions=DBCOND0030110&phase=1&purpose=treatment&status=withdrawn