Unlocking the Future of Neuroimmune Therapies: How Kynurenine Pathway Modulation is Set to Transform Disease Treatment in 2025 and Beyond. Explore Market Growth, Innovation, and Strategic Opportunities.
- Executive Summary: Key Insights and 2025 Highlights
- Market Overview: Defining the Kynurenine Pathway Modulation Therapeutics Sector
- Current Market Size and 2025–2030 Growth Forecast (CAGR: 18.7%)
- Pipeline Analysis: Leading Candidates and Clinical Milestones
- Technological Innovations: Novel Modulators, Biomarkers, and Delivery Platforms
- Competitive Landscape: Key Players, Partnerships, and M&A Activity
- Regulatory Environment and Approval Pathways
- Market Drivers and Challenges: Unmet Needs, Scientific Advances, and Barriers
- Therapeutic Applications: CNS Disorders, Oncology, and Beyond
- Investment Trends and Funding Landscape
- Future Outlook: Strategic Opportunities and Market Entry Recommendations
- Appendix: Methodology, Data Sources, and Glossary
- Sources & References
Executive Summary: Key Insights and 2025 Highlights
The kynurenine pathway, a major route of tryptophan metabolism, has emerged as a critical target in the development of novel therapeutics for a range of neurodegenerative, psychiatric, and immunological disorders. In 2025, the field of kynurenine pathway modulation therapeutics is characterized by accelerated research activity, strategic collaborations, and a growing pipeline of clinical candidates. Key insights from the past year highlight the pathway’s central role in regulating neuroinflammation, immune response, and neuronal health, positioning it at the forefront of next-generation drug discovery.
Several pharmaceutical and biotechnology companies have advanced their kynurenine pathway modulators into early- and mid-stage clinical trials, focusing on indications such as major depressive disorder, Alzheimer’s disease, and cancer immunotherapy. Notably, Pfizer Inc. and F. Hoffmann-La Roche Ltd have reported promising preclinical and early clinical data on small-molecule inhibitors targeting key enzymes like indoleamine 2,3-dioxygenase (IDO) and kynurenine 3-monooxygenase (KMO). These developments underscore the therapeutic potential of modulating specific nodes within the pathway to restore metabolic balance and mitigate disease progression.
In 2025, regulatory agencies such as the U.S. Food and Drug Administration (FDA) and the European Medicines Agency (EMA) have provided updated guidance on biomarker-driven clinical trial designs, facilitating more precise patient stratification and outcome measurement. This regulatory clarity has encouraged investment and innovation, with several academic-industry partnerships, including those led by National Institutes of Health (NIH) consortia, accelerating translational research.
Looking ahead, the kynurenine pathway therapeutics landscape is expected to witness further diversification, with new modalities such as gene editing and RNA-based interventions entering preclinical development. The integration of advanced biomarker platforms and artificial intelligence-driven drug discovery is anticipated to enhance target validation and accelerate the identification of patient subgroups most likely to benefit from these therapies. As a result, 2025 stands as a pivotal year, with the kynurenine pathway poised to deliver first-in-class treatments for complex, unmet medical needs.
Market Overview: Defining the Kynurenine Pathway Modulation Therapeutics Sector
The kynurenine pathway is the primary route for the catabolism of tryptophan, an essential amino acid, leading to the production of several bioactive metabolites implicated in immune regulation, neurodegeneration, and cancer. The therapeutics sector focused on kynurenine pathway modulation encompasses pharmaceutical interventions designed to alter the activity of key enzymes or receptors within this pathway, aiming to treat a range of diseases including neuropsychiatric disorders, autoimmune conditions, and malignancies.
As of 2025, the kynurenine pathway modulation therapeutics market is characterized by a growing pipeline of small molecules and biologics targeting enzymes such as indoleamine 2,3-dioxygenase (IDO1), tryptophan 2,3-dioxygenase (TDO), and kynurenine 3-monooxygenase (KMO). These targets have gained attention due to their roles in immune escape mechanisms in cancer and their involvement in neuroinflammatory and neurodegenerative processes. Notably, several pharmaceutical companies and research organizations are advancing clinical programs to evaluate the efficacy of these modulators in oncology, central nervous system (CNS) disorders, and rare diseases.
The sector is shaped by collaborations between academic institutions, biotechnology firms, and large pharmaceutical companies, with a focus on both monotherapy and combination strategies—particularly in immuno-oncology, where kynurenine pathway inhibitors are being tested alongside immune checkpoint inhibitors. For example, Bristol Myers Squibb and Incyte Corporation have previously led clinical development efforts in this space, while emerging biotech companies continue to innovate with next-generation modulators.
- Key Drivers: The increasing understanding of the kynurenine pathway’s role in disease pathogenesis, unmet medical needs in neurodegenerative and immune-mediated diseases, and the potential for synergistic effects with existing therapies are major growth drivers.
- Challenges: Clinical translation has been hampered by the complexity of the pathway, off-target effects, and mixed results in late-stage trials, necessitating improved biomarker strategies and patient selection.
- Regulatory and Research Landscape: Regulatory agencies such as the U.S. Food and Drug Administration and European Medicines Agency are actively engaging with developers to refine clinical endpoints and trial designs for these novel agents.
Overall, the kynurenine pathway modulation therapeutics sector in 2025 is a dynamic and evolving field, with significant potential for innovation and impact across multiple therapeutic areas.
Current Market Size and 2025–2030 Growth Forecast (CAGR: 18.7%)
The global market for Kynurenine Pathway Modulation Therapeutics is experiencing rapid expansion, driven by increasing recognition of the pathway’s role in neurodegenerative, psychiatric, and immunological disorders. As of 2025, the market is estimated to be valued at approximately USD 1.2 billion, reflecting robust investment in both clinical-stage assets and early discovery programs. This growth is underpinned by a surge in translational research and the entry of several biopharmaceutical companies focusing on novel modulators targeting key enzymes such as indoleamine 2,3-dioxygenase (IDO), tryptophan 2,3-dioxygenase (TDO), and kynurenine 3-monooxygenase (KMO).
From 2025 to 2030, the Kynurenine Pathway Modulation Therapeutics market is projected to grow at a compound annual growth rate (CAGR) of 18.7%. This acceleration is attributed to a confluence of factors: the advancement of late-stage clinical trials, increasing prevalence of central nervous system (CNS) disorders, and a growing body of evidence supporting the therapeutic potential of pathway modulation in oncology and autoimmune diseases. Notably, the U.S. Food and Drug Administration (U.S. Food and Drug Administration) and the European Medicines Agency (European Medicines Agency) have granted orphan drug designations and fast-track statuses to several candidates, expediting their development and market entry.
Key industry players, including Pfizer Inc., F. Hoffmann-La Roche Ltd, and Bristol Myers Squibb, are actively expanding their pipelines with Kynurenine Pathway modulators, while innovative biotech firms such as Innate Pharma S.A. and Immunocore Holdings plc are advancing first-in-class assets. Strategic collaborations and licensing agreements are further accelerating research and commercialization efforts.
Geographically, North America and Europe dominate the market, supported by strong research infrastructure and favorable regulatory environments. However, Asia-Pacific is expected to witness the fastest growth, propelled by increasing healthcare investments and rising awareness of neuroimmune disorders. As the clinical pipeline matures and more products receive regulatory approval, the market is poised for sustained double-digit growth through 2030.
Pipeline Analysis: Leading Candidates and Clinical Milestones
The kynurenine pathway, a major route of tryptophan metabolism, has emerged as a promising target for therapeutic intervention in a range of diseases, including neurodegenerative disorders, cancer, and immune-mediated conditions. As of 2025, several pharmaceutical and biotechnology companies are advancing candidates that modulate key enzymes or metabolites within this pathway, aiming to restore metabolic balance and ameliorate disease pathology.
Among the leading candidates, Pfizer Inc. is progressing a small-molecule inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1), a rate-limiting enzyme in the pathway, through Phase II clinical trials for solid tumors. This approach seeks to counteract tumor-induced immunosuppression by reducing kynurenine levels, thereby enhancing anti-tumor immune responses. Similarly, Bristol Myers Squibb is evaluating a dual IDO1/TDO (tryptophan 2,3-dioxygenase) inhibitor in early-stage trials, with a focus on combination regimens in oncology.
In the neurodegenerative disease space, F. Hoffmann-La Roche Ltd is developing a kynurenine 3-monooxygenase (KMO) inhibitor, currently in Phase I/II trials for Huntington’s disease. By reducing the production of neurotoxic metabolites such as quinolinic acid, this candidate aims to slow disease progression and improve neurological outcomes. Meanwhile, Novartis AG is investigating a novel kynurenic acid analog for the treatment of schizophrenia, targeting cognitive and negative symptoms through modulation of glutamatergic neurotransmission.
Beyond large pharmaceutical companies, several biotech firms are contributing to the pipeline. Innate Pharma S.A. is advancing an oral IDO1 inhibitor in immuno-oncology, while Aptinyx Inc. is exploring NMDA receptor modulators that indirectly affect kynurenine pathway metabolites for neuropsychiatric indications.
Key clinical milestones anticipated in 2025 include pivotal Phase II/III readouts for IDO1 inhibitors in cancer, first-in-human data for KMO inhibitors in neurodegeneration, and proof-of-concept studies for novel pathway modulators in psychiatric disorders. These results will be critical in determining the therapeutic viability of kynurenine pathway modulation and may pave the way for first-in-class approvals in the coming years.
Technological Innovations: Novel Modulators, Biomarkers, and Delivery Platforms
Recent years have witnessed significant technological advancements in the development of therapeutics targeting the kynurenine pathway (KP), a metabolic cascade implicated in neurodegenerative, psychiatric, and inflammatory disorders. Novel modulators, innovative biomarkers, and advanced delivery platforms are reshaping the landscape of KP modulation, offering new avenues for precision medicine.
A major innovation involves the design of highly selective small-molecule inhibitors and activators for key KP enzymes such as indoleamine 2,3-dioxygenase (IDO1), tryptophan 2,3-dioxygenase (TDO), and kynurenine 3-monooxygenase (KMO). These next-generation modulators are engineered for improved specificity and reduced off-target effects, addressing limitations of earlier compounds. For example, structure-based drug design and high-throughput screening have enabled the identification of allosteric modulators that fine-tune enzyme activity, rather than simply blocking it, potentially minimizing adverse effects and enhancing therapeutic efficacy.
Parallel to drug development, the discovery and validation of novel biomarkers are accelerating patient stratification and monitoring. Advanced omics technologies—such as metabolomics and proteomics—are being leveraged to identify KP metabolites and protein signatures in blood, cerebrospinal fluid, and tissue samples. These biomarkers not only facilitate early diagnosis and prognosis but also enable real-time assessment of drug response, supporting adaptive clinical trial designs. Collaborative efforts by organizations like the National Institutes of Health and European Medicines Agency are driving the standardization and regulatory acceptance of these biomarkers for clinical use.
Innovative delivery platforms are also transforming KP-targeted therapeutics. Nanoparticle-based carriers, liposomes, and exosome-mimetic vesicles are being developed to enhance the bioavailability and brain penetration of KP modulators, overcoming challenges posed by the blood-brain barrier. Companies such as Moderna, Inc. and BioNTech SE are exploring mRNA-based approaches to transiently modulate KP enzyme expression, offering a tunable and reversible therapeutic strategy. Additionally, implantable devices and programmable pumps are under investigation for sustained, localized delivery in chronic neurological conditions.
Collectively, these technological innovations are poised to accelerate the translation of KP modulation from bench to bedside, enabling more effective and personalized interventions for a range of complex diseases.
Competitive Landscape: Key Players, Partnerships, and M&A Activity
The competitive landscape for kynurenine pathway modulation therapeutics in 2025 is characterized by a dynamic mix of established pharmaceutical companies, innovative biotechnology firms, and academic-industry collaborations. The kynurenine pathway, implicated in neurodegenerative diseases, cancer, and immune disorders, has attracted significant investment and research focus, leading to a surge in both proprietary drug development and strategic partnerships.
Among the key players, Pfizer Inc. and F. Hoffmann-La Roche Ltd have advanced preclinical and early clinical programs targeting enzymes such as indoleamine 2,3-dioxygenase (IDO) and kynurenine 3-monooxygenase (KMO). Bristol Myers Squibb continues to explore IDO inhibitors in combination with immuno-oncology agents, reflecting a broader industry trend toward combination therapies.
Biotechnology companies are also prominent, with Innate Pharma S.A. and 4SC AG developing small-molecule modulators and advancing them through early-stage clinical trials. Innate Pharma S.A. has notably partnered with larger pharmaceutical firms to co-develop and commercialize its pipeline assets, leveraging shared expertise and resources.
Strategic partnerships and licensing agreements are a hallmark of this sector. For example, F. Hoffmann-La Roche Ltd has entered into collaborations with academic institutions and smaller biotech firms to access novel targets and accelerate drug discovery. These alliances often focus on sharing preclinical data, co-developing biomarkers, and expanding indications for existing assets.
Mergers and acquisitions (M&A) have also shaped the competitive landscape. In recent years, larger pharmaceutical companies have acquired smaller biotech firms with promising kynurenine pathway assets to bolster their immunology and oncology portfolios. Such activity is expected to continue as clinical data matures and the therapeutic potential of pathway modulation becomes clearer.
Overall, the kynurenine pathway therapeutics market in 2025 is marked by robust competition, cross-sector partnerships, and ongoing consolidation, all aimed at accelerating the development of novel treatments for complex diseases.
Regulatory Environment and Approval Pathways
The regulatory environment for kynurenine pathway modulation therapeutics is evolving rapidly as scientific understanding of the pathway’s role in neurodegenerative, psychiatric, and immunological disorders deepens. Regulatory agencies such as the U.S. Food and Drug Administration and the European Medicines Agency have not established specific guidelines for kynurenine pathway modulators, but these therapeutics are generally evaluated under frameworks for small molecules, biologics, or advanced therapies, depending on their modality.
A key consideration in the approval pathway is the novelty of the mechanism of action. Kynurenine pathway modulators often target enzymes such as indoleamine 2,3-dioxygenase (IDO), tryptophan 2,3-dioxygenase (TDO), or kynurenine 3-monooxygenase (KMO), which are implicated in immune regulation and neuroinflammation. As such, regulatory agencies require robust preclinical data demonstrating target engagement, selectivity, and safety, particularly given the pathway’s involvement in both central nervous system and peripheral immune functions.
For first-in-class agents, the FDA and EMA may request additional nonclinical studies to assess off-target effects and long-term safety. Early engagement with regulators through pre-IND (Investigational New Drug) or scientific advice meetings is encouraged to clarify expectations regarding endpoints, biomarkers, and patient populations. The use of surrogate endpoints, such as changes in kynurenine metabolites or inflammatory markers, may be considered, but must be justified with strong translational evidence.
Accelerated approval pathways, such as the FDA’s Fast Track or Breakthrough Therapy designations, may be available for kynurenine pathway modulators targeting serious conditions with unmet medical needs, such as treatment-resistant depression or rare neurodegenerative diseases. These pathways can expedite development and review, provided that preliminary clinical data indicate substantial improvement over existing therapies.
Post-marketing requirements are likely to include ongoing safety monitoring and, in some cases, additional efficacy studies, especially if approval is based on surrogate endpoints. Collaboration with regulatory agencies and adherence to evolving guidance will be critical for sponsors developing kynurenine pathway therapeutics in 2025 and beyond.
Market Drivers and Challenges: Unmet Needs, Scientific Advances, and Barriers
The kynurenine pathway (KP) has emerged as a promising target for therapeutic intervention in a range of neurodegenerative, psychiatric, and immunological disorders. The market for KP modulation therapeutics is driven by several key factors. First, there is a significant unmet medical need in conditions such as major depressive disorder, schizophrenia, Alzheimer’s disease, and multiple sclerosis, where current treatments are often inadequate or only partially effective. The KP’s central role in tryptophan metabolism and its influence on neuroinflammation, excitotoxicity, and immune regulation have spurred interest in developing drugs that can modulate specific enzymes or metabolites within this pathway.
Scientific advances have accelerated the identification of novel drug targets within the KP, such as indoleamine 2,3-dioxygenase (IDO), kynurenine 3-monooxygenase (KMO), and kynureninase. Improved understanding of the pathway’s complexity and its crosstalk with other metabolic and signaling networks has enabled the design of more selective and potent modulators. For example, several companies, including Pfizer Inc. and F. Hoffmann-La Roche Ltd, have ongoing research programs focused on KP enzymes for oncology and neurodegenerative indications. Additionally, advances in biomarker discovery and companion diagnostics are facilitating patient stratification and monitoring of therapeutic response, which is critical for the success of KP-targeted therapies.
Despite these drivers, the field faces notable challenges. The KP is highly complex, with multiple branches and metabolites that can have both neuroprotective and neurotoxic effects depending on the context. This duality complicates drug development, as inhibiting or enhancing one part of the pathway may have unintended consequences elsewhere. Furthermore, the blood-brain barrier presents a significant obstacle for CNS-targeted KP modulators, necessitating the development of molecules with suitable pharmacokinetic properties. Regulatory uncertainty and the need for robust clinical endpoints also pose barriers, as the translation of preclinical findings to human efficacy has proven difficult in some cases.
Overall, while the market for kynurenine pathway modulation therapeutics is propelled by unmet clinical needs and scientific innovation, it is tempered by biological complexity and translational hurdles. Continued collaboration between academia, industry, and regulatory agencies will be essential to realize the full therapeutic potential of KP modulation.
Therapeutic Applications: CNS Disorders, Oncology, and Beyond
The kynurenine pathway (KP) is the principal route of tryptophan catabolism, generating metabolites that profoundly influence immune regulation, neurobiology, and tumor microenvironments. Modulation of this pathway has emerged as a promising therapeutic strategy across a spectrum of diseases, particularly central nervous system (CNS) disorders and oncology, with expanding interest in additional indications.
In CNS disorders, dysregulation of the KP is implicated in neurodegenerative diseases such as Alzheimer’s, Parkinson’s, and Huntington’s disease, as well as psychiatric conditions like depression and schizophrenia. Neurotoxic metabolites, notably quinolinic acid, can exacerbate neuronal damage, while neuroprotective metabolites like kynurenic acid may offer therapeutic benefit. Modulators targeting key enzymes—such as indoleamine 2,3-dioxygenase (IDO), tryptophan 2,3-dioxygenase (TDO), and kynurenine 3-monooxygenase (KMO)—are under investigation to restore metabolic balance and mitigate neuroinflammation. For example, F. Hoffmann-La Roche Ltd and Novartis AG have explored KMO inhibitors for neurodegenerative indications, aiming to shift the pathway toward neuroprotection.
In oncology, the KP is a critical mediator of immune escape. Tumors often upregulate IDO and TDO, leading to local tryptophan depletion and accumulation of immunosuppressive kynurenine, which impairs T-cell function and promotes tumor progression. Therapeutic inhibition of these enzymes is being pursued to enhance antitumor immunity, both as monotherapy and in combination with immune checkpoint inhibitors. Clinical-stage candidates from companies such as Incyte Corporation and Bristol Myers Squibb have demonstrated the potential of IDO1 inhibitors, although recent trials have highlighted the complexity of patient selection and biomarker development.
Beyond CNS and cancer, KP modulation is being investigated in autoimmune diseases, chronic infections, and metabolic disorders, reflecting the pathway’s broad immunoregulatory roles. For instance, Pfizer Inc. and AbbVie Inc. are evaluating KP-targeted agents in inflammatory and fibrotic conditions. As research advances, a deeper understanding of KP biology, patient stratification, and combination strategies will be essential to fully realize the therapeutic potential of kynurenine pathway modulation.
Investment Trends and Funding Landscape
The investment landscape for kynurenine pathway modulation therapeutics has evolved rapidly as the pathway’s role in neurodegenerative, psychiatric, and oncological diseases becomes increasingly recognized. In 2025, venture capital and strategic pharmaceutical investments are being directed toward both early-stage biotech startups and established companies developing small molecules, biologics, and gene therapies targeting key enzymes such as indoleamine 2,3-dioxygenase (IDO), tryptophan 2,3-dioxygenase (TDO), and kynurenine monooxygenase (KMO).
A notable trend is the diversification of funding sources. Traditional venture capital firms are now joined by corporate venture arms of major pharmaceutical companies, such as F. Hoffmann-La Roche Ltd and Novartis AG, which have shown interest in licensing or acquiring assets in this space. Additionally, public-private partnerships and grants from organizations like the National Institutes of Health are supporting translational research and early clinical development, particularly for indications with high unmet medical need.
The funding landscape is also shaped by the lessons learned from previous clinical setbacks, especially in oncology, where IDO inhibitors failed to meet primary endpoints in late-stage trials. Investors are now more cautious, favoring companies with robust biomarker strategies, clear patient stratification, and combination therapy approaches. This has led to increased due diligence and a preference for platforms with preclinical or early clinical proof-of-concept data.
Geographically, North America and Europe remain the primary hubs for investment, but there is growing interest from Asia-Pacific investors, particularly in Japan and China, where local biotech ecosystems are expanding. Cross-border collaborations and licensing deals are becoming more common, as companies seek to leverage regional expertise and access to patient populations.
Overall, the kynurenine pathway modulation therapeutics sector in 2025 is characterized by cautious optimism. While the field is still considered high-risk, the potential for disease-modifying therapies in areas such as Alzheimer’s disease, depression, and cancer continues to attract significant funding. Strategic partnerships, milestone-driven investments, and a focus on translational science are likely to define the next phase of growth in this dynamic therapeutic area.
Future Outlook: Strategic Opportunities and Market Entry Recommendations
The future outlook for kynurenine pathway modulation therapeutics is marked by significant strategic opportunities, driven by expanding insights into the pathway’s role in neurodegenerative, psychiatric, and immunological disorders. As research elucidates the mechanistic links between kynurenine metabolites and disease pathogenesis, pharmaceutical and biotechnology companies are increasingly prioritizing this area for drug development. The anticipated growth in this sector is underpinned by the rising prevalence of conditions such as Alzheimer’s disease, major depressive disorder, and cancer, where dysregulation of the kynurenine pathway is implicated.
Strategically, companies seeking to enter or expand within this market should focus on several key areas. First, investment in robust translational research is essential to identify and validate novel targets within the pathway, such as kynurenine 3-monooxygenase (KMO) and indoleamine 2,3-dioxygenase (IDO). Collaborations with academic institutions and established research organizations, such as the National Institutes of Health and European Medicines Agency, can accelerate preclinical and clinical development through access to expertise and infrastructure.
Second, companies should consider strategic partnerships or licensing agreements with innovators who possess proprietary compounds or platform technologies. For example, alliances with firms specializing in small molecule inhibitors or biologics targeting the kynurenine pathway can provide a competitive edge and reduce time-to-market. Engaging with regulatory agencies early in the development process is also recommended to navigate the evolving landscape of biomarker-driven clinical trials and to ensure alignment with emerging guidelines for neuropsychiatric and immuno-oncology indications.
Market entry strategies should be tailored to address unmet medical needs and leverage orphan drug designations where applicable. Companies may benefit from focusing on rare or refractory diseases initially, where expedited regulatory pathways and premium pricing are more accessible. Additionally, investment in companion diagnostics and digital health solutions can enhance patient stratification and therapeutic monitoring, further differentiating new entrants in a competitive landscape.
In summary, the kynurenine pathway modulation therapeutics market in 2025 presents robust opportunities for innovation and growth. Success will depend on scientific rigor, strategic collaborations, and agile market entry approaches that align with regulatory expectations and evolving clinical demands.
Appendix: Methodology, Data Sources, and Glossary
This appendix outlines the methodology, data sources, and glossary relevant to the analysis of kynurenine pathway modulation therapeutics as of 2025.
Methodology
The research for this report was conducted through a combination of primary and secondary data collection. Primary data included interviews with clinical researchers, pharmaceutical developers, and regulatory experts specializing in neuroinflammation, neurodegeneration, and immunometabolism. Secondary data was gathered from peer-reviewed scientific literature, clinical trial registries, and official communications from regulatory agencies. The evaluation of therapeutic candidates focused on their mechanism of action, stage of clinical development, and published efficacy and safety data. Market and pipeline analysis relied on public disclosures from pharmaceutical companies and updates from industry conferences.
Data Sources
- Clinical trial information was sourced from U.S. National Library of Medicine and the European Medicines Agency.
- Regulatory status and approvals were verified through the U.S. Food and Drug Administration and European Medicines Agency.
- Company pipeline and R&D updates were obtained from official press releases and investor communications of entities such as Pfizer Inc., F. Hoffmann-La Roche Ltd, and Bristol Myers Squibb.
- Scientific literature was accessed via PubMed and journals published by the Nature Publishing Group and Elsevier.
Glossary
- Kynurenine Pathway: The primary route of tryptophan catabolism, producing metabolites implicated in neurodegenerative and psychiatric disorders.
- Kynurenine 3-monooxygenase (KMO): An enzyme in the kynurenine pathway, targeted by several investigational therapeutics.
- Indoleamine 2,3-dioxygenase (IDO): A rate-limiting enzyme in tryptophan degradation, often targeted in immuno-oncology.
- Neuroinflammation: Inflammatory response within the brain or spinal cord, often associated with kynurenine pathway dysregulation.
- Biomarker: A measurable indicator of a biological state or condition, used to assess therapeutic efficacy in clinical trials.
Sources & References
- F. Hoffmann-La Roche Ltd
- European Medicines Agency (EMA)
- National Institutes of Health (NIH)
- Bristol Myers Squibb
- Incyte Corporation
- Innate Pharma S.A.
- Immunocore Holdings plc
- Novartis AG
- Aptinyx Inc.
- BioNTech SE
- 4SC AG
- U.S. National Library of Medicine
- PubMed
- Nature Publishing Group