Open Access

Tryptophan metabolism: From physiological functions to key roles and therapeutic targets in cancer (Review)

  • Authors:
    • Jiawei Zhao
    • Xiaohui Bai
    • Jingjing Du
    • Yujing Chen
    • Xiaotong Guo
    • Juzheng Zhang
    • Jinfeng Gan
    • Peitao Wu
    • Siqi Chen
    • Xinwen Zhang
    • Jinfeng Yang
    • Jiamin Jin
    • Li Gao
  • View Affiliations

  • Published online on: May 28, 2025     https://doi.org/10.3892/or.2025.8919
  • Article Number: 86
  • Copyright: © Zhao et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Tryptophan (Trp) metabolism is a complex and important biochemical process in humans. It is vital in protein synthesis and is a precursor of various bioactive molecules. Trp is metabolized through the kynurenine, serotonin and indole pathways, mediating diverse physiological functions, including neurotransmitter synthesis, immune regulation, antioxidant effects, and biosynthesis of niacin and melatonin. These metabolic pathways maintain essential functions under normal physiological conditions. However, they are significantly affected by various types of cancers. Trp metabolites regulate tumor angiogenesis, affect the self‑renewal of cancer stem cells, and participate in immune evasion and cell death through complex mechanisms. As the mechanisms underlying Trp metabolism in diseases are increasingly being elucidated, targeting Trp metabolic pathways has emerged as a promising therapeutic strategy. Further investigation of the molecular mechanisms underlying Trp metabolism and its role in diseases may provide new perspectives and approaches for diagnosing and treating diseases.

Introduction

Tryptophan (Trp) is an essential aromatic amino acid in the human body that performs various key physiological functions (13). Trp metabolism primarily involves three pathways: the kynurenine (Kyn) pathway, catalyzed by indoleamine 2,3-dioxygenase (IDO) or Trp 2,3-dioxygenase (TDO); the serotonin [or 5-hydroxytryptamine (5-HT)] pathway, mediated by the gut microbiota; and the indole pathway, which is largely dependent on the gut microbiota. A total of ~95% of Trp is metabolized through the Kyn pathway, catalyzed by TDO in the liver and IDO in immune cells such as macrophages and dendritic cells (DCs). The products of this pathway include Kyn, nicotinic acid, and nicotinamide, which ultimately enter the nicotinamide adenine dinucleotide (NAD+) synthesis pathway to produce essential redox-regulating molecules for cells (4). The 5-HT pathway consumes ~1% Trp to produce 5-HT, which is an important neurotransmitter and vasoactive substance. The gut microbiota metabolizes Trp to indole derivatives via the indole pathway. These metabolites activate the host aryl hydrocarbon receptor (AhR), thereby regulating gut barrier function and immune responses (5). Dysregulated Trp metabolism is associated with cancer stem cell (CSC) self-renewal, tumor angiogenesis, and immune evasion. For example, tumors can use the Trp-Kyn pathway to suppress local immune responses and promote the formation of an immunosuppressive tumor microenvironment (TME) that facilitates cancer progression (6). IDO is overexpressed in various cancers and associated with poor prognosis (7). Therefore, targeting the Trp metabolism has emerged as a promising therapeutic strategy in oncology. Previous studies have focused on the therapeutic potential of inhibiting key enzymes in the Trp metabolic pathway, such as IDO and TDO, to enhance antitumor immune responses and improve the efficacy of cancer immunotherapy (8). New compounds targeting Trp metabolism are being investigated to assess their ability to modulate immune responses and inhibit tumor growth, highlighting significant opportunities and challenges (9). Given the central role of Trp metabolism in cancer biology, recent advances have highlighted the potential of multi-targeted approaches, such as combining Trp pathway modulators with immune checkpoint inhibitors (ICIs), to overcome drug resistance and enhance antitumor efficacy. Additionally, emerging insights into the crosstalk between Trp metabolism and the gut microbiome have opened new avenues for drug discovery with the development of novel compounds aimed at modulating these interactions. The aim of the present review was to provide a comprehensive overview of Trp metabolism and its dysregulation in cancer, focusing on its therapeutic implications. Herein, the key metabolic pathways of Trp, their roles in cancer development and progression, and the latest advances in targeting these pathways for cancer therapy are discussed. By understanding the complex mechanisms underlying Trp metabolism, effective therapeutic strategies can be developed to treat cancer and improve patient outcomes.

Trp metabolism pathways and their clinical biomarker implications

Trp metabolism involves three key amino acid pathways (Fig. 1). These metabolites reflect the metabolic status of the body and can serve as potential disease biomarkers with significant clinical importance.

Kynurenine pathway

Trp is an essential substrate for protein synthesis and is involved in various biological processes. In the human body, the Kyn pathway is the predominant metabolic route accounting for the majority of Trp metabolism, in which Trp is oxidized to N-formyl-L-Kyn by TDO or IDO. Subsequently, N-formyl-L-kyn is converted to L-kyn using formylase. L-Kyn is further metabolized to 3-hydroxy-L-Kyn in a reaction catalyzed by Kyn 3-monooxygenase (KMO). 3-Hydroxy-L-Kyn is then transformed into 3-hydroxyanthranilic (3-HAA) acid via oxidation by 3-HAA oxidase, which generates anthranilic acid. Kyn can be converted into kynurenic acid (KYNA) by Kyn aminotransferases. KYNA exerts neuroprotective effects by inhibiting glutamate receptors and reducing neuroexcitatory toxicity (1012). The Kyn pathway is the predominant pathway for Trp metabolism, accounting for over 95% of Trp catabolism. Various metabolites generated in this metabolic pathway are closely associated with immune regulation, inflammation and neurodegenerative diseases. For instance, an elevated Kyn/Trp ratio often correlates with disease progression and poor prognosis in patients with cancer (such as lung cancer) (1315). Studies have shown that Trp metabolism is altered in the early adenoma stage and persists throughout colorectal cancer (CRC) progression in patients with CRC. Moreover, compared with the control group, the activities of IDO1 and Trp hydroxylase (TPH) enzymes were significantly increased in patients with CRC, suggesting that the Kyn and serotonin pathways may play important roles in immune regulation. Further analysis revealed that patients with colon cancer are more prone to Trp catabolism than patients with rectal cancer. These findings indicated that early abnormalities in Trp metabolism may help colon cancer evade immune surveillance and resist immunosuppression (16). Abnormalities in Trp metabolism are significant in non-cancer-related diseases. For example, elevated Kyn levels are considered to be closely related to neuroinflammation and neurotoxicity in patients with depression (17), and increased quinolinic acid (QA) levels in the cerebrospinal fluid are significantly associated with cognitive decline in patients with neurodegenerative diseases (18). These studies reveal the potential pathological mechanisms of Trp metabolic abnormalities in various diseases and their importance as biomarkers.

5-HT pathway

The 5-HT pathway is another important pathway involved in Trp metabolism. This pathway converts Trp to the neurotransmitter 5-HT, also known as serotonin, where Trp is hydroxylated to form 5-hydroxyTrp (5-HTP) by TPH (19). The conversion of Trp to 5-HTP by TPH is followed by decarboxylation of 5-HTP to 5-HT by aromatic L-amino acid decarboxylase (AADC). TPH, the rate-limiting enzyme in this pathway, has two isoforms: TPH1, which is predominantly expressed in peripheral tissues, and TPH2, which is primarily expressed in the central nervous system (CNS) (20). The decarboxylation of 5-HTP to 5-HT by AADC occurs in the neuronal cytoplasm, and 5-HT is subsequently stored in synaptic vesicles and released into the synaptic cleft to exert its neurotransmitter effects (21). 5-HT binds various receptors in the synaptic cleft to mediate its physiological effects. After the action of 5-HT is terminated, primarily through reuptake by the 5-HT transporter (SERT), it is restored, metabolized, or degraded (22). Within neurons, it is mainly metabolized by monoamine oxidase to form 5-hydroxyindoleacetaldehyde, which is then converted to 5-hydroxyindoleacetic acid (5-HIAA) by aldehyde dehydrogenase and ultimately excreted as a metabolic product (23). 5-HT is acetylated by N-acetyltransferase to form N-acetyl-5-HT (NAc-5-HT), which is then converted into melatonin via O-methylation by hydroxy-indole-O-methyltransferase (24). The 5-HT pathway of Trp metabolism and its metabolites have garnered widespread attention in clinical studies as potential cancer biomarkers. Studies have demonstrated that 5-HT plays a crucial role in the susceptibility to esophageal cancer (EC) and may serve as a potential EC biomarker (25). Moreover, 5-HT metabolism is implicated in the development and progression of various tumors, and relevant research has revealed the therapeutic potential of targeting key enzymes, metabolites and receptors within the 5-HT metabolic pathway (26). Specifically, Trp and its metabolites interact with 5-HT receptors (5-HTR1A, 1B, 2A and 2B) to regulate cancer cell proliferation and metastasis (27). Under normal physiological conditions, Trp metabolism occurs through both 5-HT and indole pathways, with these enzymes and their metabolites widely distributed in various cells and tissues (28). In addition, the metabolic pathway of the end product of Trp metabolism, 5-HIAA, in malignant melanoma is an important direction for future studies (29).

Indole pathway

The indole-synthesis pathway is an essential metabolic process. This pathway occurs primarily in the gut microbiota, where Trp is converted into various bioactive molecules via enzymatic reactions. These molecules play crucial roles in host physiological and pathological processes (30). In the indole pathway, Trp is catalytically degraded by tryptophanase (Trpase) secreted by gut bacteria to produce tryptamine. Subsequently, in the presence of gut bacteria, tryptamine undergoes a series of complex enzymatic reactions to generate indoles. Tryptamine transaminase plays a key role in this process by efficiently converting tryptamine into indole (31,32). Indoles can be further oxidized to indoxyl sulfate, an important intermediate in the indole pathway. Additionally, indole can be metabolized into other derivatives, such as indole-3-acetaldehyde and indole-3-pyruvic acid, via alternative pathways (33,34). Indole-3-acetic acid (IAA) is an auxin essential for plant growth regulation (35). Indole and its derivatives have various biological functions in the cells that produce them, primarily in gut health and immune regulation. Indole compounds (for example, indole-3-propionic acid) activate AhR, modulate gut barrier function, reduce the production of inflammatory cytokines, and enhance the gut immune balance (30). Regarding the gut microbiota, indole metabolites protect gut health by influencing gut barrier function and inflammatory responses (31). Trp metabolism plays a significant role in the interaction between the host and pathogens and involves multiple metabolic pathways. Indole and its derivatives are unique metabolites produced by human gut microbiota. Trp metabolism undergoes significant changes in CRC, and studies have shown that this is related to alterations in the populations of indole-producing bacteria. Indole exhibits anti-inflammatory effects and demonstrates potential therapeutic value through specific anticancer mechanisms and may become part of future anticancer adjuvant strategies (36). Inflammation can induce changes in both the host and microbial Trp metabolism in colon cancer. For example, the overexpression of IDO1 (indoleamine-2,3-dioxygenase 1) shifts Trp metabolism towards the Kyn pathway, thereby promoting tumor immune evasion. By contrast, Trp metabolites, such as indole, can inhibit tumor occurrence and development. However, changes in the gut microbiota often lead to decreased indole levels, disrupting the symbiotic relationship between the host and microbes, consequently enhancing inflammation and exacerbating the vicious cycle (37). Therefore, abnormal levels of indole metabolites are associated with diseases, such as inflammatory bowel disease, metabolic syndrome and gut microbiota dysbiosis, and thus have potential value as biomarkers. In summary, Trp metabolism via the indole pathway is vital for physiological and pathological processes in cells. Elucidating the specific mechanisms of action of indole and its derivatives on cell health is essential for exploring their potential applications in disease prevention and treatment.

Disruption of Trp metabolism in cancer

Disruption of Trp metabolism can induce alterations in various hallmark features of cancer across multiple systems, including the digestive, nervous, respiratory and hematological systems. Its role and underlying mechanisms in cancer are gradually elucidated (Fig. 2). The effects of Trp metabolism in cancer are summarized in Table I.

Table I.

Role and mechanism of tryptophan in tumors.

Table I.

Role and mechanism of tryptophan in tumors.

First author, yearTumor typeTryptophan metabolismFunctionsRelative molecules(Refs.)
Chen et al, 2015;CRCKyn, AhRSelf-renewal of colon CSCsPI3K/Akt,(43,44,48)
Vermeulen et al, 2010; β-catenin
Pham et al, 2018
Zhu et al, 2022CRC5-HTSelf-renewal of colon CSCs and promotion of tumor angiogenesisMMP-12(45)
Shi et al, 2022CRCIDO1Cause of T-cell dysfunction-(52)
Zheng et al, 2017;GliomaMelatoninInhibition of the proliferation andEZH2, Notch(46,47)
Gürsel et al, 2012 tumorigenicity of glioma stem cells
Guastella et al, 2018GliomaKynurenic acid, AhRPromotion of immunosuppression-(66)
Panitz et al, 2021GliomaTDO2, Kyn, AhRPromotion of the motility of tumor cells and inhibit immune cellsAkt(67)
Sahm et al, 2013Glioma NAD+Enhancement of resistance to oxidative stress-(71)
Sadik et al, 2020GliomaI3P, Kyn, AhRPromotion of the motility and malignant phenotype of cancer cellsIL4I1(74)
Bosnyák et al, 2015GlioblastomaKyn, AhRPromotion of the motility of tumor cells and inhibit immune cellsTrp catabolic enzymes(77)
Panitz et al, 2021GlioblastomaTDO2, Kyn, 3-HAAFacilitation of the preservation of tryptophan in tumor cellsHIF-1α(67)
Zhang et al, 2022;EsophagealTDO2Participation in the formation ofOct4, EGFR(39,48)
Pham et al, 2018cancer tumor spheres of esophageal CSCs
Zhao et al, 2012Liver cancerIDOFacilitation of the immune escape of tumorsIFN-γ(55)
Tummala et al, 2014Liver cancer NAD+Prevention of DNA damage and dysregulation of cell proliferationURI(56)
Yu et al, 2021Liver cancerTDO2Inhibition of the proliferation of HCC cellsp21, p27, CDK2, CDK4(57)
Wu et al, 2021Liver cancerTDO2, Kyn, AhRPromotion of the progression of HCC cellsIL-6, STAT3, NF-kB(58)
Li et al, 2021Liver cancerTDO2Promotion of EMT of cancer cells-(59)
Jin et al, 2015Liver cancerKMOPromotion of the progression of HCC cells-(60)
Shi et al, 2021Liver cancer3-HAAInduction of apoptosis of HCC cellsYY1(61)
Zhang et al, 2017;PancreaticIDOReflection of the malignancy of-(63,65)
Koblish et al, 2010cancer pancreatic adenocarcinoma
Hezaveh et al, 2022Pancreatic cancerIndole, AhRPromotion of tumor growth and metastasis-(64)
Talari et al, 2016;MeningiomaIDO2, TDO2,Promotion of tumor growth and-(76,77)
Bosnyák et al, 2015 Kynmetastasis
Ino et al, 2008;EndometrialIDOPromotion of the motility of tumor-(7880)
Yoshida et al, 2008;cancer cells and inhibit immune cells
Ino et al, 2006
Inaba et al, 2009;OvarianIDO, NAD+Promotion of the motility of tumor-(8183)
Odunsi et al, 2022;cancer cells and inhibit immune cells
Gostner et al, 2018
Liu et al, 2014Non-Hodgkin's lymphomaIDO1Facilitation of the immune escape of tumorsLactate dehydrogenase(87)
El Kholy et al, 2011;AcuteIDO,Promotion of the motility of tumor-(88,89)
Curti et al, 2007myeloid leukemiakynurenic acidcells and inhibit immune cells
Zhang et al, 2019Pulmonary adenocarcinomaIDO1Promotion of the motility of tumor cellsEGFR(90)
Tang et al, 2017; Hsu et al, 2016Lung cancerTDO2, Kyn, AhRPromotion of the proliferation of CAFs and EMT in lung cancer cellsAkt/CREB, Akt/WNK1(91,92)
Feng et al, 2022Lung cancerTDO, Kyn, AhREnhancement of the proliferation ability of NSCLC cells and their resistance to EGFR tyrosine kinase inhibitorsAkt, ERK(93)
Tang et al, 2017Lung cancerIDO1Promotion of the motility of tumor cellsP53(91)
Karayama et al, 2021NSCLC3-HAAInhibition of the therapeutic efficacy of immune checkpoint inhibitors-(94)
Levina et al, 2012;Breast cancerIDO1, TDO2,Promotion of tumor growth and-(95,96)
D'Amato et al, 2015 AhRmetastasis
Li et al, 2021Prostate cancerAhRIncrease of the apoptosis rate of prostate cancer cells treated with Abi or Docc-Myc(102)
Zhang et al, 2019Bladder cancerIDO1Promotion of tumor angiogenesismicroRNA-153(111)
Cecchi et al, 2024MelanomaTDOPromotion of tumor angiogenesis-(112)

[i] Kyn, kynurenine; AhR, aryl hydrocarbon receptor; 5-HT, 5-hydroxytryptamine; IDO1, indoleamine 2,3-dioxygenase 1; IDO, indoleamine 2,3-dioxygenase; TDO2, tryptophan 2,3-dioxygenase; NAD+, nicotinamide adenine dinucleotide; 3-HAA, 3-hydroxyanthranilic acid; KMO, kynurenine 3-monooxygenase; I3P, indole-3-pyruvate; EZH2, enhancer of Zeste Homolog 2; IL4I1, interleukin 4-induced 1; HIF-1α, hypoxia-inducible factor 1-alpha; Oct4, octamer-binding transcription factor 4; EGFR, epidermal growth factor receptor; IFN-γ, interferon-gamma; URI, unconventional prefoldin RPB5 interactor; IL-6, interleukin-6; STAT3, signal transducer and activator of transcription 3; NF-Kb, nuclear factor kappa-light-chain-enhancer of activated B cells; Akt, protein kinase B; CREB, cAMP response element-binding protein; ERK, extracellular signal-regulated kinase; P53, tumor protein P53; c-Myc, Myc proto-oncogene protein; Abi, abiraterone; Doc, docetaxel; HCC, hepatocellular carcinoma; EMT, epithelial-mesenchymal transition; CSCs, cancer stem cells; CRC, colorectal cancer; NSCLC, non-small cell lung cancer.

Correlation between Trp metabolism and CSC self-renewal

Human pluripotent stem cells (hPSCs), which can differentiate into various cell types, hold great promise as a source of cells for regenerative therapies and drug discovery. Trp metabolism is critical for promoting hPSC proliferation without compromising their pluripotency (38). Several studies focused on the relationship between Trp metabolism and CSCs. Modulation of the Trp metabolic pathway can influence the self-renewal ability of CSCs. For example, the Trp metabolite Kyn can activate AhR to regulate self-renewal and differentiation of CSCs (39). In some diseases, colorectal CSCs represent a small subpopulation of cells with self-renewal and differentiation capabilities within CRC (40). These pathways are beneficial for the self-renewal of CSCs. Abnormal hyperactivation of specific signaling pathways in CSCs, such as the phosphoinositide 3-kinase/protein kinase B (PI3K/Akt) and β-catenin pathways, may induce uncontrolled cell proliferation and abnormal differentiation, leading to tissue-specific tumorigenesis (4144). The neurotransmitter 5-HT promotes self-renewal and tumorigenesis of colorectal CSCs (45). The Trp-derivative melatonin may inhibit the proliferation and tumorigenicity of glioma stem-like cells by suppressing the EZH2 and Notch pathways, which are crucial for the survival of glioma stem cells (46,47). Overexpression of TDO2 is associated with tumor staging and recurrence status, as well as the CD44 CSC marker in esophageal squamous cell carcinoma (ESCC) and is involved in the formation of tumor spheres in esophageal CSCs. TDO2 may promote the generation of esophageal CSCs by inducing the expression of Oct4 and CD44, activating the epidermal growth factor receptor (EGFR) pathway, stimulating epithelial-mesenchymal transition (EMT) and invasion of esophageal CSCs, which are associated with poor prognosis in patients with ESCC (39,48). Additionally, indole-3-pyruvate, a Trp metabolite, reduces the expression of Oct4 in CSCs by activating the AhR transcriptional pathway, inducing CSC differentiation, and decreasing CSC tumorigenicity (39). Moreover, activating the Trp metabolic pathway helps CSCs evade attacks from the host immune system through immune modulation and metabolic reprogramming, thereby maintaining survival. For example, Trp depletion and hypoxia preserve CSC phenotype by enhancing OCT4 transcription (49). Kyn inhibits the activity of effector T-cells via the AhR pathway, enhances Treg function, and suppresses antitumor immune responses. This immunosuppressive effect provides a favorable environment for CSC survival (7,39). In addition, Trp metabolites regulate the metabolic state of CSCs to adapt to hypoxia and nutrient deficiency in the TME (50). In summary, Trp metabolism and its key enzymes are vital for the maintenance and function of CSCs, promoting tumor progression and drug resistance by directly affecting CSC behavior and indirectly regulating immune responses in the TME.

Cancer cell proliferation, invasion and immune evasion
Digestive system diseases CRC

Kyn, a bioactive metabolite of the Trp metabolism, acts as an ‘oncometabolite’ in CRC cells. It maintains the proliferation of CRC cells by activating the transcription factor AhR and regulating genes that promote cell proliferation (51). Additionally, Kyn accumulation affects the TME, facilitating the proliferation and spread of CRC cells. USP14 promotes Trp metabolism in CRC by stabilizing the expression of IDO1, leading to T-cell dysfunction. This metabolic alteration enables tumor cells to evade immune surveillance, enhancing their proliferative capacity and metastatic potential (52). High expression of IDO is associated with the local depletion of Trp, which inhibits T-cell proliferation and activity, allowing tumor cells to escape immune attack and promote tumor proliferation and metastasis (53). The activity of IDO1 promotes immune tolerance in the TME and directly enhances the proliferation of CRC cells by activating the β-catenin signaling pathway (42). These studies indicate that modulation of Trp metabolism influences the development and progression of CRC. Therefore, in-depth studies on the relationship between Trp metabolism and CRC may facilitate the development of novel therapeutic strategies.

Hepatocellular carcinoma (HCC)

In HCC, IDO inhibits the proliferation of cytotoxic T-cells by degrading Trp and promoting tumor immune evasion. High IDO expression is significantly associated with the metastasis rate and poor prognosis in patients with HCC, suggesting that IDO may play a crucial role in tumor cell proliferation, invasion and metastasis (54). CD69+ T-cells in the TME can induce high expression of IDO in tumor-associated macrophages (TAMs) through the secretion of interferon-γ (IFN-γ), which, in turn, inhibits T-cell proliferation and function, promoting tumor immune evasion (55). The role of Trp metabolism in HCC is reflected by its effect on NAD+ synthesis and DNA damage mechanisms. Overexpression of URI (an unconventional prefoldin RPB5-interacting factor) inhibits the Trp metabolism pathway, leading to decreased NAD+ levels, DNA damage and dysregulated cell proliferation, promoting the malignant transformation of hepatocytes and tumorigenesis (56). Overexpression of TDO2 inhibits HCC cell proliferation and induces cell cycle arrest by upregulating p21 and p27 and downregulating CDK2 and CDK4. The absence of TDO2 may promote tumor proliferation and metastasis, indicating that TDO2 is a potential biomarker and therapeutic target for HCC (57). High TDO2 expression is associated with poor prognosis in patients with HCC. TDO2 promotes the conversion of Trp to Kyn, activates AhR, upregulates the secretion of interleukin-6 (IL-6), activating signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa B (NF-Κb) signaling pathways, and promotes HCC cell proliferation and metastasis (58). TDO2 promotes EMT in cancer cells by activating the Kyn-AhR pathway, thereby enhancing HCC cell proliferation, invasion and metastasis (59). High KMO expression in HCC tissues is associated with poor prognosis. In vitro experiments have shown that KMO overexpression promotes HCC cell proliferation, migration and invasion, whereas KMO knockdown inhibits these processes (60). 3-HAA, a derivative of Trp, is present at low concentrations in tumor cells. Exogenous 3-HAA induces apoptosis in HCC cells by binding to the transcription factor YY1. The mechanism involves protein kinase C Zeta (PKCζ)-mediated phosphorylation of YY1, enhancing its binding to target genes, and regulating HCC cell proliferation, invasion and metastasis through modulation of 3-HAA levels (61). 5-HT1D is significantly upregulated in HCC tissues and cell lines, with its expression associated with poor clinical pathological features. 5-HT1D stabilizes PIK3R1 to activate the PI3K/Akt signaling pathway, enhances FoxO6 expression, and promotes cancer cell proliferation, EMT and metastasis. Additionally, 5-HT1D inhibits the expression of TPH1 via the PI3K/Akt/CUX1 axis, thereby affecting 5-HT synthesis (62). In-depth research into the relationship between Trp metabolism and HCC may facilitate the development of novel therapeutic strategies against HCC.

Pancreatic cancer

High IDO expression is closely related to the malignancy of pancreatic adenocarcinoma, particularly in patients with poorly differentiated tumors, lymph node metastasis, and advanced TNM staging, and is associated with a poor prognosis. This suggests that IDO is involved in pancreatic cancer progression, making it a potential therapeutic target (63). The gut microbiota metabolizes Trp to produce indole compounds that activate the AhR, inhibit antitumor immunity, and promote tumor growth and metastasis. Macrophages lacking AhR function exhibit a stronger inflammatory phenotype, increased CD8+ T-cell infiltration and tumor growth inhibition (64). IDO catalyzes the conversion of Trp to Kyn, depleting Trp levels, causing immune suppression, and facilitating tumor cell evasion during immune attacks. Inhibition of IDO activity can reduce Kyn levels in the TME, enhance the function of tumor-infiltrating lymphocytes (TILs), and inhibit tumor growth and metastasis (65). These results indicate that modulation of Trp metabolism may be an effective strategy for cancer therapy.

CNS diseases
Glioma

Dysregulation of the Kyn pathway in primary brain tumors, such as gliomas, leads to local Trp depletion within cancer cells and promotes an immunosuppressive TME, affecting tumor cell proliferation and invasion capabilities. This pathway may also affect tumor metastasis by modulating the AhR signaling pathway (66). The upregulation of Trp catabolic enzymes in glioblastoma (GBM) promotes Trp degradation to produce metabolites, such as Kyn, which activate AhR and subsequently enhance tumor cell motility while suppressing immune cell function (67). TDO2 facilitates the generation of Kyn from Trp metabolism, which activates the AhR and Akt signaling pathways, enhancing tumor cell proliferation and invasion. Moreover, Kyn inhibits the proliferation of functional T-cells, leading to immunosuppression and the promotion of glioma cell proliferation and metastasis (68). Under hypoxic conditions, hypoxia-inducible factor-1α in GBM downregulates the expression of TDO2, reducing Trp degradation and consequently decreasing the production of Kyn and 3-HAA. This mechanism may help tumor cells conserve Trp in the hypoxic microenvironment and maintain their proliferative and survival properties (69). IDO expression in the brain increases with age. This enzyme generates immunosuppressive metabolites from Trp metabolism, weakening the efficacy of immunotherapy and resulting in poor treatment outcomes for GBM in elderly patients (70). Glioma cells generate QA via microglia and utilize it to synthesize NAD+, enhancing resistance to oxidative stress. Activation of this metabolic pathway is closely related to the malignant phenotype of tumors, and the accumulation of Quin endows glioma cells with a greater survival capacity when subjected to radiotherapy and chemotherapy (for example, temozolomide), promoting tumor invasion and metastasis (71). Positron emission tomography (PET) using α-11C-methyl-L-Trp (AMT) can be used to assess 5-HT synthesis in the brain and track the upregulation of the Kyn pathway in tumor tissues in patients with malignant gliomas. Increased AMT uptake in the tumor tissues of patients with high-grade gliomas (grades III–IV) suggests that alterations in Trp metabolism may be closely related to the biological behavior of tumors such as proliferation and metastasis (72). Increased Trp metabolism, mainly through the Kyn pathway, observed using PET, positively correlates with tumor proliferative activity (assessed using the Ki-67 labeling index) (73). Finally, IL4I1 catalyzes Trp metabolism to generate indole-3-propionic acid (I3P) and its derivatives (for example, Quin and KynA), which activate AhR and promote cancer cell motility and a malignant phenotype, thereby correlating with reduced survival rates in patients with glioma (74). In low-grade brain tumors, such as dysplastic neuroepithelial tumors, widespread expression of the Trp-catabolic enzyme IDO may lead to local Trp depletion and inhibition of cell proliferation, as evidenced by a lower proliferative index (Ki-67). By contrast, IDO expression is less frequent in high-grade tumors such as GBM. It is mainly confined to endothelial cells and is possibly associated with tumor invasiveness and metastatic capability (75). Trp metabolism is important for immune evasion in gliomas and provides a survival advantage to glioma cells; thus, it is emerging as a potential therapeutic target.

Meningioma

Meningioma cells evade immune responses via the Trp metabolic pathway, particularly the Kyn pathway, resulting in elevated levels of Kyn and lower levels of Trp and its metabolites, thereby promoting tumor progression. This process is accompanied by the upregulation of enzymes such as IDO2 and INOS, highlighting the significant role of Trp metabolism in cancer cell proliferation and invasion (76). The grading of meningiomas is positively correlated with PET imaging parameters of Trp metabolism (for example, the k3′ ratio). These parameters can be used to effectively distinguish meningiomas of different grades. The key enzyme in Trp metabolism, TDO2, exhibits significant immunostaining in meningiomas, suggesting its potential role in tumor immune tolerance and proliferation (77). These findings indicate that Trp metabolism is associated with the biological characteristics of tumors and may provide new strategies for improving meningioma treatment outcomes.

Reproductive system diseases
Endometrial cancer

IDO is highly expressed in endometrial cancer and is significantly associated with tumor aggressiveness, lymph node metastasis and involvement of vascular lymphatic spaces. IDO exerts its effects by depleting Trp and generating toxic metabolites that inhibit the function of TILs and natural killer (NK) cells and promote tumor immune evasion (78). However, in mouse xenograft models, tumors overexpressing IDO exhibited faster growth rates, which correlated with a decrease in the number and function of NK cells, suggesting that IDO inhibits NK cell activity through Trp metabolism, thereby facilitating tumor progression (79). High IDO expression in endometrial cancer cells positively correlated with the clinicopathological features of the tumor. Elevated IDO expression is significantly associated with reduced overall and progression-free survival (PFS) (80). IDO influences endometrial cancer cell proliferation, invasion and metastasis by regulating Trp metabolism, making it a potential therapeutic target.

Ovarian cancer

High IDO expression is associated with an immunosuppressive state in tumor cells, reducing the number of TILs and affecting patient survival. Although in vitro experiments have shown no significant differences in the proliferation, migration and invasion capabilities of IDO-overexpressing ovarian cancer cells, mouse xenograft models have demonstrated a substantial increase in peritoneal metastasis in IDO-overexpressing cells (81). IDO1 inhibition alters Trp metabolism and suppresses immune evasion in tumor cells. It also induces metabolic adaptation by increasing NAD+ synthesis and inhibiting T-cell proliferation and function. This metabolic adaptation may limit the antitumor immune response and promote the proliferation, invasion and metastasis of ovarian cancer cells (82). An elevated Kyn/Trp ratio is associated with disease progression in patients with ovarian cancer. The presence of circulating tumor cells (CTCs) correlates with alterations in Trp metabolism and elevated levels of immune activation markers (such as neopterin), suggesting that CTCs continuously stimulate the immune system by releasing tumor antigens or cytokines, facilitating tumor immune evasion and metastasis (83).

Hematological system diseases
Non-Hodgkin's lymphoma (NHL)

NHL is a hematological malignancy originating from lymph nodes and other extranodal lymphoid tissues (8486). IDO1 expression is significantly upregulated in NHL tissues and is associated with the clinical stage of the tumor, tumor size and serum lactate dehydrogenase levels, indicating a poor prognosis. IDO1 promotes local immune tolerance by depleting Trp and its metabolites (for example L-kyn) and inhibiting the proliferation and activation of antigen-specific T-cells. Upregulation of IDO1 enhances the generation and infiltration of Tregs, possibly representing a mechanism by which NHL evades immune control (87), making it a potential target for treatment.

Acute myeloid leukemia (AML)

IDO is expressed in patients with AML. It suppresses T-cell proliferation by reducing local Trp concentrations and generating immunosuppressive metabolites (for example, Quin), thereby promoting tumor immune tolerance. This mechanism may enhance the proliferation and survival of tumor cells, facilitating their invasion and metastasis (88). IDO expression is associated with increased circulating CD4+CD25+FOXP3+ regulatory T cells. AML cells directly convert CD4+CD25 T cells into CD4+CD25+ Treg cells via Trp metabolism, inhibiting T-cell proliferation and activity and promoting tumor immune evasion. This process affects the TME and may contribute to cancer cell proliferation, invasion and metastasis (89). The inhibition of IDO may represent a novel therapeutic strategy against AML.

Other disease systems

The expression of IDO1 is significantly associated with tumor aggressiveness, smoking history, and the abundance of tumor-infiltrating CD8+ and T-bet+ cells; in some cases, it is related to EGFR mutations. Although IDO1 expression is associated with tumor growth and metastasis, its independent expression does not significantly affect patient survival, suggesting that its immunosuppressive role in lung adenocarcinoma may involve other mechanisms (90). High expression of IDO1 in lung cancer tissues correlates with clinical stage and lymph node metastasis. It enhances cancer cell migratory and invasive capacities, which are partially attenuated by p53 through suppression of the IDO signaling pathway (91). Lung cancer cells activate cancer-associated fibroblasts (CAFs) to promote Trp metabolism and generate Kyn via TDO2. Kyn inhibits DC function, promotes proliferation and EMT in lung cancer cells, and activates Akt/CREB and Akt/WNK1 signaling pathways to enhance cancer cell proliferation and migration (92). Kyn production is closely related to the activation in CAFs. It promotes activation of the AhR signaling pathway, activates Akt and ERK signaling, and enhances the proliferative capacity and resistance to EGFR tyrosine kinase inhibitors in non-small cell lung cancer (NSCLC) cells (93). Changes in Trp metabolite 3-HAA in the plasma of patients with NSCLC are associated with the efficacy of ICIs. Lower levels of 3-HAA correlate with improved treatment responses and longer PFS, indicating that 3-HAA may influence tumor growth and metastasis by modulating immune responses (94). IDO1 plays a significant role in Trp metabolism in breast cancer (BC), promoting tumor cell proliferation and metastasis through both immune and non-immune pathways (95). Upregulation of TDO2 and AhR renders triple-negative BC (TNBC) cells more resistant, allowing them to survive in the absence of matrix attachment, which is crucial for BC metastasis (96).

The complex relationship between cancer cells and cell death: Evasion and manipulation

Some metabolites generated during Trp metabolism can influence the immune system by regulating immune cell apoptosis, potentially promoting tumor progression (97).

Apoptosis

Kyn, the first metabolite of Trp degradation via IDO, also functions as an immunosuppressive molecule owing to the generation of its derivatives 3-HAA and Quin, which induce selective apoptosis in murine Th1 cells by activating caspase-8 in vitro (9). T-cell apoptosis occurs at relatively low Kyn concentrations, independent of Fas/Fas ligand interactions, and is associated with caspase-8 activation and mitochondrial release of cytochromes (97,98). IDO and TDO deplete local Trp and accumulate Kyn in tumors and antigen-presenting cells, leading to T-cell anergy and apoptosis via the GCN2 pathway (99). Trp metabolites further enhance their immunosuppressive effects by inhibiting T-cell proliferation and inducing DC-mediated T-cell apoptosis (97). In addition to T-cell apoptosis, Trp metabolism also affects cancer cell apoptosis to some extent. Trp deprivation significantly increases the expression of ERRFI1 in sensitive HCC cells, activating the apoptotic pathway and inducing cell death (100). Enhanced Kyn pathway activation leads to a substantial accumulation of Quin in the CNS in several inflammatory neurological diseases. By contrast, 3OH-Kyn and 3OH-anthranilic acids may induce neuronal apoptosis or necrosis. Kyn hydroxylase inhibitors reduce neuronal death in both in vitro and in vivo models of cerebral ischemia and excitotoxicity (101). AhR can cooperate with NF-κB to promote c-Myc activation in prostate cancer cells, and overexpression of c-Myc upregulates the expression of Trp transporters and ABC transporters, further increasing the apoptosis rate of prostate cancer cells treated with Abi or Doc (102).

Autophagy

In addition to apoptosis, Trp metabolism, which involves various metabolic pathways involving Trp and its metabolites, is associated with autophagy. Trp metabolites modulate the autophagy pathway by activating AMPK and SIRT1, thereby affecting intestinal inflammation (103). For instance, Trp metabolites, such as IAA, regulate autophagy and ameliorate pulmonary fibrosis by inhibiting the PI3K/Akt signaling pathway (104). Trp metabolites also modulate autophagy via the mechanistic target of rapamycin (mTOR) signaling pathway. L-Trp enhances susceptibility to nonalcoholic fatty liver disease via the mTOR pathway (105). Trp metabolism is essential to the immune system, particularly in T-cells, where Trp metabolites modulate immune responses by affecting the autophagy pathway (106). However, Trp metabolism plays different roles in cancer cell autophagy. For example, extracellular vesicles derived from IDO1-high ovarian cancer cells upregulate SIRT3 expression in endothelial cells by increasing its acetylation, which is essential for promoting endothelial mitochondrial autophagy associated with tumor angiogenesis (107).

Necrosis

Trp metabolism generates Kyn and its metabolites (3-hydroxykynurenine and Quin), which may induce necrosis in tumor cells through multiple mechanisms. Metabolites in the Kyn metabolic pathway may induce oxidative stress, damaging the DNA and organelles of tumor cells and ultimately leading to cell necrosis (108). Although IDO/TDO primarily promote tumor growth through immune suppression, certain Kyn metabolites may activate immune cells under specific conditions, exerting cytotoxic effects on tumor cells, which may lead to alterations in the immune microenvironment. For example, Quin activates specific immune cell subsets and promotes tumor necrosis (109).

Ferroptosis

Ferroptosis is a newly discovered type of regulated cell death that is distinct from apoptosis and autophagy. The Trp metabolites, 5-HT and 3-hydroxykynurenine, are potent ferroptosis inhibitors capable of suppressing ferroptosis in tumor cells and promoting their proliferation (110). These effects jointly influence cancer cell proliferation, survival and death and have significant implications for cancer progression and treatment.

Induction of cancer angiogenesis by Trp metabolites

Some metabolites formed during Trp metabolism promote tumor angiogenesis, which is crucial for tumor growth and metastasis. MicroRNA (miR)-153 inhibits tumor angiogenesis by suppressing the expression of IDO1. Downregulation of miR-153 leads to increased IDO1 expression in bladder cancer cells, promoting tumor angiogenesis (111). Angiogenesis is essential for the progression and metastasis of melanoma and is based on the production and release of pro-angiogenic molecules in the TME. The interaction between melanoma cells and endothelial cells affects the molecular signaling pathways involved in tumor growth and progression. TDO affects different melanoma cell lines, and its expression positively correlates with endothelial cell infiltration (112). The Trp metabolite 5-HT can modulate the function of tumor-infiltrating macrophages by regulating the expression of matrix metalloproteinase 12, which influences tumor angiogenesis. This mechanism has been validated in colorectal and lung cancers (113). Indole compounds related to Trp metabolism, such as melatonin, IAA, 5-hydroxytryptophan and 5-HT, significantly inhibit vascular endothelial growth factor (VEGF)-induced VEGF receptor-2 activation and subsequent angiogenesis in human umbilical vein endothelial cells, thereby suppressing tumor angiogenesis. These compounds provide potential molecular targets for developing anti-VEGF signaling pathway drugs that may help control tumor growth and progression (114,115). In summary, Trp metabolites induce tumor angiogenesis through significant tumor growth and metastasis.

Discoveries in targeting Trp metabolism in cancer

Recently, targeting Trp metabolism has led to significant progress in cancer therapy. The Trp metabolic pathway plays a crucial role in the TME, immune evasion and cancer progression, providing a wealth of targets for developing novel therapeutic strategies. It also offers theoretical support and practical evidence for applying multi-target strategies, combination therapies with ICIs, interactions with the microbiome, and the use of AhR antagonists.

Progress in multi-target strategies targeting Trp metabolism in cancer therapy

In recent years, therapeutic strategies targeting Trp metabolism have garnered widespread attention, with the potential for multi-target combination therapies gradually emerging as a research hotspot. The small-molecule IDO1 inhibitor navoximod (synonyms: GDC-0919, NLG-919) has demonstrated significant activity in combination therapy in various tumor models. It has been revealed that this inhibitor is well-tolerated and capable of reducing plasma Kyn levels, which is consistent with its pharmacokinetic (PK) half-life. In addition, a stable disease response has been observed in some patients (116). Further research has revealed that navoximod in combination with cisplatin can effectively reverse immune resistance mediated by the Kyn-AhR-IL6 axis induced by IDO1-positive CAFs and chemoresistance triggered by tumor lymphoid structures by targeting IDO1. This mechanism has exhibited great potential in evaluating chemoresistance and biosafety in oral squamous cell carcinoma (117). Moreover, when navoximod is used in combination with another IDO1 inhibitor, indoximod, it exhibits synergistic effects, prolonging patient survival and enhancing therapeutic efficacy when combined with chemotherapy. Currently, research is underway to optimize prodrug formulations to improve their therapeutic effects (118). 1-Methyl-L-Trp (1-MT-L-Trp) is a non-specific competitive IDO1 inhibitor that is widely used in basic research (119). Notably, even in the absence of IDO expression, 1-MT can induce a strong inflammatory molecular genetic response by activating AhR. These data provide important insights into the potential clinical indications of 1-MT as a cancer immunotherapy and suggest that 1-MT may exert therapeutic effects through AhR-related mechanisms, even in IDO-deficient tumors (120). This finding also suggests that 1-MT may have a multi-target mechanism of action, thereby providing a new research direction for further exploration of its potential value in cancer therapy. Compared with 1-MT-L-Trp, 1-methyl-D-Trp (1-D-MT) is currently undergoing clinical trials for patients with recurrent or refractory solid tumors, with the aim of inhibiting tumor immune evasion mediated by IDO. However, studies have reported that 1-D-MT promotes immune evasion by upregulating the expression of IDO1 in cancer cells. This off-target effect has raised concerns regarding its safety and efficacy in clinical trials (121). Future research should combine high-throughput screening technologies to develop more selective IDO1 inhibitors and employ drug delivery systems (such as nanoparticles or prodrugs) to reduce non-specific effects on normal cells, which may help mitigate such off-target effects. TDO has long been considered constitutively expressed only in the liver. It has been identified that TDO is significantly expressed in various cancers, including BC, ovarian cancer and gliomas. TDO is involved in tumor progression and immune suppression through the TDO-L-Kyn-AhR pathway, and its inhibition is considered to help reverse immune evasion (29,36). Although TDO inhibitors remain in the preclinical stage, research is moving towards the development of dual-target inhibitors (IDO1 and TDO) that may enhance cancer immunotherapy efficacy by blocking L-kyn synthesis. However, the clinical feasibility of multi-target strategies faces challenges such as adverse reactions caused by drug interactions and heterogeneous responses of tumors. In the future, precise diagnostic tools need to be developed to identify tumor metabolic phenotypes and optimize multi-target therapeutic combination strategies.

Synergistic effects of Trp metabolism and ICIs

Recently, the synergistic effects of Trp metabolism and ICIs have become a popular research topic in the field of cancer immunotherapy. The Trp metabolic pathway, particularly the metabolic processes mediated by IDO and TDO, plays a crucial role in tumor immune evasion (99). It has been found that, in primary pulmonary squamous cell carcinoma, the co-expression of IDO1 and programmed death-ligand 1 (PD-L1) has significant prognostic implications and is closely related to TILs. Co-expression of IDO1 and PD-L1 may be an important target for immunotherapy in pulmonary squamous cell carcinoma (122). The IDO1 enzyme inhibitor, BGB-5777, in combination with anti-PD-1 monoclonal antibodies and radiotherapy, demonstrated a robust immune-enhancing effect in GBM model studies. This mechanism may be related to an increase in tumor-infiltrating T-cells, reduction in immunosuppressive IDO1 levels, restoration of immune cell function following PD-1 blockade, and the pro-inflammatory effects of radiation therapy (123). Additionally, the novel IDO1 inhibitor PF-06840003 reversed the T-cell non-responsive state, reduced intra-tumoral Kyn levels, and inhibited tumor growth. It has been shown that the antitumor effect of this drug in combination with anti-PD-L1 antibody therapy is more pronounced, and preclinical data have supported Phase I clinical trials (27). Another IDO1 inhibitor, LY3381916, demonstrated high selectivity and efficacy. LY3381916, either as monotherapy or in combination with PD-L1 inhibitors, significantly inhibited IDO1 activity in tumors and increased CD8+ T-cell infiltration in studies on treating advanced solid tumors. However, the clinical activity of combination therapy is limited, and significant hepatotoxicity has been observed in patients with TNBC, highlighting the importance of dose adjustment and optimization (124). NLG-919, in combination with indoximod and ICIs (such as inhibitors of the PD-1/PD-L1/PD-L2 pathway), has shown synergistic antitumor effects in multiple models. For example, in the B16F10 melanoma model, NLG-919, in combination with indoximod, ICIs, chemotherapy and hgp100 peptide vaccine therapy, significantly inhibited tumor growth, further clarifying the mechanistic basis of such combination strategies. Future studies of the synergistic effects of Trp metabolism and ICIs should focus on several aspects. It has been previously revealed that tumors induce an immunosuppressive microenvironment by upregulating IDO1 expression. Specifically, IDO1 promotes immune tolerance under inflammatory stimulation by depleting Trp and generating metabolites, leading to the loss of effector T-cell function and enhanced activity of Tregs, making it an important target for immunotherapy (28). For example, epacadostat, a novel IDO1 inhibitor, can significantly inhibit Trp catabolism and has multifaceted effects on the maturation of DCs, activation of tumor antigen-specific cytotoxic T lymphocytes, regulation of Tregs and function of peripheral blood mononuclear cells, thereby enhancing the efficacy of immunotherapy (37). Additionally, linifanib mesylate, which occupies the heme cofactor-binding site, prevents the activation of IDO1. In vitro studies have shown that it can inhibit Kyn production and restore T-cell proliferation, while significantly reducing Kyn levels in tumor xenograft models in vivo, with favorable PK and pharmacodynamic properties, providing a basis for clinical development (125). The synergistic effects of Trp metabolism and ICIs offer new insights into cancer immunotherapy. However, their efficacy is influenced by the differential expression of IDO1 and PD-L1. Future studies should focus on patient stratification by using these biomarkers. This combination therapy has great potential to overcome the limitations of monotherapy and provide more efficient antitumor strategies. Further research is expected to drive the development of novel combination therapies.

Interactions between Trp metabolism and the microbiome

Tight interactions exist between Trp metabolism and the gut microbiome, which has garnered widespread attention recently because of its implications in disease pathogenesis and treatment. The gut microbiota metabolizes Trp through various pathways into indole and its derivatives, which exhibit significant biological functions in immune regulation, metabolic homeostasis and suppression of inflammation. For instance, it has been identified that moderate dietary supplementation with Trp can generate indole compounds via microbial metabolism, modulating immune responses and effectively treating various immune-related diseases (126). Additionally, indole and its derivatives have shown considerable potential in treating metabolic and hepato-intestinal diseases, with mechanisms possibly involving multi-target regulation (127,128). Microbial indole metabolism is considered a potential therapeutic strategy for CRC (129). Recently, the discovery of novel microbial strains associated with Trp metabolism has increased the depth of research in this area. For example, certain strains isolated from the gut generate bioactive metabolites by modulating the Trp metabolic pathway, thereby influencing disease onset and progression. A representative example is the gut bacterium Clostridium sporogenes, which captures energy via the Stickland reaction. Its metabolic product, indolepropionic acid, which is known for its antioxidant and anti-inflammatory properties, is considered a probiotic with therapeutic potential for the prevention and treatment of inflammatory bowel disease (130,131). Similarly, Lactobacillus plantarum DPUL-S164 (S164) and its Trp metabolite, indole-lactic acid, demonstrated significant repair effects in mouse models of intestinal barrier damage induced by antibiotic mixtures and dextran sulfate sodium (132,133). The development of next-generation probiotics offers new insights into Trp-metabolism-targeted therapies. For example, by comparing the microbiota composition of healthy and diseased individuals and designing recombinant microbes that overexpress target genes, it is possible to achieve the local delivery of metabolites and regulation of signaling pathways. However, this strategy faces challenges such as regulatory requirements, manufacturing difficulties, complexity of disease models, and diversity of individual gut microbiota, which can lead to variable therapeutic outcomes. Current research has primarily focused on short-term efficacy, whereas long-term safety and effectiveness require further validation. The interactions between Trp metabolism and the gut microbiome provide new directions for disease treatment. An in-depth exploration of their metabolic pathways and microbial regulatory mechanisms can promote the development of novel compounds and optimize therapeutic strategies. However, future studies should address the challenges posed by complex disease models and the need for personalized treatments to achieve broader clinical applications and enhanced therapeutic outcomes.

AhR antagonists targeting Trp metabolism

Trp metabolites are closely related to AhR, and the potential role of AhR antagonists in targeting Trp metabolism in cancer therapy warrants further investigation. The AhR is a ligand-activated transcription factor that plays a key role in maintaining important physiological functions in the body. It exhibits dual pro- and antitumorigenic activities during tumorigenesis, with its expression and activity varying depending on the tumor type and individual differences among patients (134136). It has been revealed that Trp metabolites such as Kyn and indole, generated by endogenous enzymes or microbial metabolism, can bind to and activate AhR, forming the Trp-AhR pathway, which is closely related to cancer progression (137). For example, in melanoma and glioma, the continuous transcriptional activation of AhR is driven by ligands produced by the TME and the tumor itself, promoting tumor growth and suppressing immune defense functions. Moreover, IL4I1, a metabolic pathway parallel to IDO1 and TDO, can generate AhR ligands, further demonstrating the difficulty of completely inhibiting AhR ligand production (138). Based on these mechanisms, the potential applications of AhR antagonists in cancer therapy have attracted widespread attention. Various AhR antagonists have shown significant antitumor potential. For example, CH223191 is a selective competitive AhR antagonist that lacks antagonistic activity against non-aromatic hydrocarbon ligands. Although related research is still mainly focused on the basic scientific field, preliminary therapeutic effects have been demonstrated in pancreatic cancer models (139,140). Additionally, StemRegenin-1, an exogenously applied AhR antagonist, reversed the drug resistance of MCF-7/ADR cells through the AhR/ABC transport and AhR/UGT pathways (141). Furthermore, the antagonistic properties of BAY 2416964 were confirmed using transactivation assays. It effectively and selectively inhibits AhR activation in human and mouse head, head and neck squamous cell carcinoma, NSCLC and CRC cells in vitro, while restoring immune cell function and enhancing antitumor responses. BAY 2416964 exhibited favorable drug tolerance and significant antitumor efficacy in vivo by inducing a pro-inflammatory TME (142). CB7993113 inhibited the invasive capacity of human BC cells under three-dimensional culture conditions and blocked tumor cell migration in two-dimensional culture without affecting cell viability or proliferation. Moreover, this compound effectively inhibits bone marrow ablation induced by 7,12-dimethylbenz[a]anthracene in vivo, further proving that its absorption and distribution can produce pharmacological effects (143). Current research has clarified the dual role of AhR in tumorigenesis; however, the development of AhR antagonists remains in its early stages, with numerous obstacles facing clinical translation, such as balancing the pro-inflammatory and anti-inflammatory effects of AhR and avoiding off-target effects. Research targeting Trp metabolism is at a critical stage of transitioning from basic science to clinical applications, and there is still a need to address core issues such as optimizing drug targeting and safety, dealing with tumor heterogeneity, and integrating multi-target combination therapy strategies.

Concluding remarks

Trp metabolites are potential biomarkers for clinical features, such as inflammation, mental state and cognitive function, which can guide clinical decision-making. These metabolites may facilitate the development of novel therapeutic strategies for treating various diseases. Rate-limiting enzymes, including IDO, TDO, KMO and TPH, are critical for Trp metabolism, and research on their inhibitors has progressed. Although IDO/TDO inhibitors have shown limited efficacy as monotherapies for cancer treatment, they can significantly enhance the efficacy of conventional therapeutic agents when used in combination with traditional drugs. Moreover, KMO inhibitors can reduce the neurological damage caused by acute pancreatitis, and the combination of telotristat ethyl with somatostatin analogs has been approved by the FDA for the treatment of carcinoid-associated diarrhea. Thus, specific inhibitors targeting TPH1 or TPH2 may be safer and more effective. Direct supplementation with indoles and their derivatives is a promising therapeutic strategy. Dysregulation of Trp metabolism in cancer is closely associated with clinical features such as tumor stage, size and lymph node metastasis, and the levels of Trp metabolites can thus serve as predictive biomarkers. Therefore, targeting Trp metabolism has emerged as a promising therapeutic approach for cancer treatment. However, despite preclinical evidence demonstrating the anticancer effects of Trp metabolism inhibition, its translation into clinical success remains challenging, as exemplified by the failure of IDO1 inhibitors in clinical trials. Similarly, although niacin can significantly increase HDL-C levels and has various lipid-modulating effects, it has not shown the expected clinical benefits of reducing the risk of cardiovascular events (144). Future research should delve into the resistance mechanisms, utilize multi-omic approaches to identify new biomarkers and therapeutic targets, and design combination therapies to enhance therapeutic efficacy and minimize resistance. Continued research on Trp metabolism combined with advanced technologies and innovative strategies holds promise for advancing cancer treatments and patient outcomes.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Natural Science Foundation of Guangxi (grant nos. 2023GXNSFAA026061, 2024GXNSFAA010335, 2023GXNSFBA026313 and 2025GXNSFAA069989), the National Natural Science Foundation of China (grant nos. 32360170, 82160590, 81802884, 82460677, 82260602, 82460677 and 82204208) and the Independent project of Guangxi Key Laboratory of Tumor Immunity and Microenvironment Regulation (grant no. 203030302415).

Availability of data and materials

Not applicable.

Authors' contributions

JiZ, XB and JD contributed to conception and manuscript writing. YC and XG contributed to the proofreading and bioanalysis. JuZ and JG contributed to acquisition of data. PW, SC and XZ contributed to table and figure production. JY, JJ and LG contributed to manuscript editing. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Barik S: The uniqueness of tryptophan in biology: Properties, metabolism, interactions and localization in proteins. Int J Mol Sci. 21:87762020. View Article : Google Scholar : PubMed/NCBI

2 

Palego L, Betti L, Rossi A and Giannaccini G: Tryptophan biochemistry: Structural, nutritional, metabolic, and medical aspects in humans. J Amino Acids. 2016:89525202016. View Article : Google Scholar : PubMed/NCBI

3 

Kałużna-Czaplińska J, Gątarek P, Chirumbolo S, Chartrand MS and Bjørklund G: How important is tryptophan in human health? Crit Rev Food Sci Nutr. 59:72–88. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Schwarcz R, Bruno JP, Muchowski PJ and Wu HQ: Kynurenines in the mammalian brain: When physiology meets pathology. Nat Rev Neurosci. 13:465–477. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Sharma L and Riva A: Intestinal barrier function in health and disease - Any role of SARS-CoV-2? Microorganisms. 8:17442020.https://doi.org/10.3390/microorganisms8111744 View Article : Google Scholar : PubMed/NCBI

6 

Liu XH and Zhai XY: Role of tryptophan metabolism in cancers and therapeutic implications. Biochimie. 182:131–139. 2021. View Article : Google Scholar : PubMed/NCBI

7 

Yan J, Chen D, Ye Z, Zhu X, Li X, Jiao H, Duan M, Zhang C, Cheng J, Xu L, et al: Molecular mechanisms and therapeutic significance of Tryptophan Metabolism and signaling in cancer. Mol Cancer. 23:2412024. View Article : Google Scholar : PubMed/NCBI

8 

Perez-Castro L, Garcia R, Venkateswaran N, Barnes S and Conacci-Sorrell M: Tryptophan and its metabolites in normal physiology and cancer etiology. FEBS J. 290:7–27. 2023. View Article : Google Scholar : PubMed/NCBI

9 

Zhang HL, Zhang AH, Miao JH, Sun H, Yan GL, Wu FF and Wang XJ: Targeting regulation of tryptophan metabolism for colorectal cancer therapy: A systematic review. RSC Adv. 9:3072–3080. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Badawy AA: Kynurenine pathway of tryptophan metabolism: Regulatory and functional aspects. Int J Tryptophan Res. 10:11786469176919382017. View Article : Google Scholar : PubMed/NCBI

11 

Stone TW and Darlington LG: Endogenous kynurenines as targets for drug discovery and development. Nat Rev Drug Discov. 1:609–620. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Lugo-Huitrón R, Ugalde Muñiz P, Pineda B, Pedraza-Chaverrí J, Ríos C and Pérez-de la Cruz V: Quinolinic acid: An endogenous neurotoxin with multiple targets. Oxid Med Cell Longev. 2013:1040242013. View Article : Google Scholar : PubMed/NCBI

13 

Mandarano M, Orecchini E, Bellezza G, Vannucci J, Ludovini V, Baglivo S, Tofanetti FR, Chiari R, Loreti E, Puma F, et al: Kynurenine/tryptophan ratio as a potential blood-based biomarker in non-small cell lung cancer. Int J Mol Sci. 22:44032021. View Article : Google Scholar : PubMed/NCBI

14 

Kwiatkowska I, Hermanowicz JM, Przybyszewska-Podstawka A and Pawlak D: Not only immune escape-the confusing role of the TRP metabolic pathway in carcinogenesis. Cancers (Basel). 13:26672021. View Article : Google Scholar : PubMed/NCBI

15 

Suzuki Y, Suda T, Furuhashi K, Suzuki M, Fujie M, Hahimoto D, Nakamura Y, Inui N, Nakamura H and Chida K: Increased serum kynurenine/tryptophan ratio correlates with disease progression in lung cancer. Lung Cancer. 67:361–365. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Crotti S, D'Angelo E, Bedin C, Fassan M, Pucciarelli S, Nitti D, Bertazzo A and Agostini M: Tryptophan metabolism along the kynurenine and serotonin pathways reveals substantial differences in colon and rectal cancer. Metabolomics. 13:1482017. View Article : Google Scholar

17 

Ogyu K, Kubo K, Noda Y, Iwata Y, Tsugawa S, Omura Y, Wada M, Tarumi R, Plitman E, Moriguchi S, et al: Kynurenine pathway in depression: A systematic review and meta-analysis. Neurosci Biobehav Rev. 90:16–25. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Maddison DC and Giorgini F: The kynurenine pathway and neurodegenerative disease. Semin Cell Dev Biol. 40:134–141. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Schwarcz R and Pellicciari R: Manipulation of brain kynurenines: Glial targets, neuronal effects, and clinical opportunities. J Pharmacol Exp Ther. 303:1–10. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Walther DJ and Bader M: A unique central tryptophan hydroxylase isoform. Biochem Pharmacol. 66:1673–1680. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Patel PD, Pontrello C and Burke S: Robust and tissue-specific expression of TPH2 versus TPH1 in rat raphe and pineal gland. Biol Psychiatry. 55:428–433. 2004. View Article : Google Scholar : PubMed/NCBI

22 

Kalinichenko LS, Kornhuber J, Sinning S, Haase J and Müller CP: Serotonin signaling through lipid membranes. ACS Chem Neurosci. 15:1298–1320. 2024. View Article : Google Scholar : PubMed/NCBI

23 

Murphy DL and Lesch KP: Targeting the murine serotonin transporter: Insights into human neurobiology. Nat Rev Neurosci. 9:85–96. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Shi X, Zhao G, Li H, Zhao Z, Li W, Wu M and Du YL: Hydroxytryptophan biosynthesis by a family of heme-dependent enzymes in bacteria. Nat Chem Biol. 19:1415–1422. 2023. View Article : Google Scholar : PubMed/NCBI

25 

Jung KH, LoRusso P, Burris H, Gordon M, Bang YJ, Hellmann MD, Cervantes A, Ochoa de Olza M, Marabelle A, Hodi FS, et al: Phase I study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) administered with PD-L1 inhibitor (Atezolizumab) in advanced solid tumors. Clin Cancer Res. 25:3220–3228. 2019. View Article : Google Scholar : PubMed/NCBI

26 

Ebata T, Shimizu T, Fujiwara Y, Tamura K, Kondo S, Iwasa S, Yonemori K, Shimomura A, Kitano S, Koyama T, et al: Phase I study of the indoleamine 2,3-dioxygenase 1 inhibitor navoximod (GDC-0919) as monotherapy and in combination with the PD-L1 inhibitor atezolizumab in Japanese patients with advanced solid tumours. Invest New Drugs. 38:468–477. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Gomes B, Driessens G, Bartlett D, Cai D, Cauwenberghs S, Crosignani S, Dalvie D, Denies S, Dillon CP, Fantin VR, et al: Characterization of the selective indoleamine 2,3-dioxygenase-1 (IDO1) catalytic inhibitor EOS200271/PF-06840003 Supports IDO1 as a critical resistance mechanism to PD-(L)1 blockade therapy. Mol Cancer Ther. 17:2530–2542. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Crosignani S, Bingham P, Bottemanne P, Cannelle H, Cauwenberghs S, Cordonnier M, Dalvie D, Deroose F, Feng JL, Gomes B, et al: Discovery of a novel and selective indoleamine 2,3-dioxygenase (IDO-1) inhibitor 3-(5-Fluoro-1H-indol-3-yl)pyrrolidine-2,5-dione (EOS200271/PF-06840003) and Its characterization as a potential clinical candidate. J Med Chem. 60:9617–9629. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Kim M and Tomek P: Tryptophan: A rheostat of cancer immune escape mediated by immunosuppressive enzymes IDO1 and TDO. Front Immunol. 12:6360812021. View Article : Google Scholar : PubMed/NCBI

30 

Roager HM and Licht TR: Microbial tryptophan catabolites in health and disease. Nat Commun. 9:32942018. View Article : Google Scholar : PubMed/NCBI

31 

Madella AM, Van Bergenhenegouwen J, Garssen J, Masereeuw R and Overbeek SA: Microbial-derived tryptophan catabolites, kidney disease and gut inflammation. Toxins (Basel). 14:6452022. View Article : Google Scholar : PubMed/NCBI

32 

Medvedev A and Buneeva O: Tryptophan metabolites as mediators of microbiota-gut-brain communication: Focus on isatin. Front Behav Neurosci. 16:9222742022. View Article : Google Scholar : PubMed/NCBI

33 

Ghiboub M, Boneh RS, Sovran B, Wine E, Lefèvre A, Emond P, Verburgt CM, Benninga MA, de Jonge WJ and Van Limbergen JE: Sustained diet-induced remission in pediatric Crohn's disease is associated with kynurenine and serotonin pathways. Inflamm Bowel Dis. 29:684–694. 2023. View Article : Google Scholar : PubMed/NCBI

34 

Vanholder R, Nigam SK, Burtey S and Glorieux G: What if not all metabolites from the uremic toxin generating pathways are toxic? A hypothesis. Toxins (Basel). 14:2212022. View Article : Google Scholar : PubMed/NCBI

35 

Spaepen S and Vanderleyden J: Auxin and plant-microbe interactions. Cold Spring Harb Perspect Biol. 3:a0014382011. View Article : Google Scholar : PubMed/NCBI

36 

Ye Z, Yue L, Shi J, Shao M and Wu T: Role of IDO and TDO in cancers and related diseases and the therapeutic implications. J Cancer. 10:2771–2782. 2019. View Article : Google Scholar : PubMed/NCBI

37 

Jochems C, Fantini M, Fernando RI, Kwilas AR, Donahue RN, Lepone LM, Grenga I, Kim YS, Brechbiel MW, Gulley JL, et al: The IDO1 selective inhibitor epacadostat enhances dendritic cell immunogenicity and lytic ability of tumor antigen-specific T cells. Oncotarget. 7:37762–37772. 2016. View Article : Google Scholar : PubMed/NCBI

38 

Someya S, Tohyama S, Kameda K, Tanosaki S, Morita Y, Sasaki K, Kang MI, Kishino Y, Okada M, Tani H, et al: Tryptophan metabolism regulates proliferative capacity of human pluripotent stem cells. iScience. 24:1020902021. View Article : Google Scholar : PubMed/NCBI

39 

Zhang Q and Li W: Correlation between amino acid metabolism and self-renewal of cancer stem cells: Perspectives in cancer therapy. World J Stem Cells. 14:267–286. 2022. View Article : Google Scholar : PubMed/NCBI

40 

Munro MJ, Wickremesekera SK, Peng L, Tan ST and Itinteang T: Cancer stem cells in colorectal cancer: A review. J Clin Pathol. 71:110–116. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Bishnupuri KS, Alvarado DM, Khouri AN, Shabsovich M, Chen B, Dieckgraefe BK and Ciorba MA: IDO1 and kynurenine pathway metabolites activate PI3K-Akt signaling in the neoplastic colon epithelium to promote cancer cell proliferation and inhibit apoptosis. Cancer Res. 79:1138–1150. 2019. View Article : Google Scholar : PubMed/NCBI

42 

Thaker AI, Rao MS, Bishnupuri KS, Kerr TA, Foster L, Marinshaw JM, Newberry RD, Stenson WF and Ciorba MA: IDO1 metabolites activate β-catenin signaling to promote cancer cell proliferation and colon tumorigenesis in mice. Gastroenterology. 145:416–425.e1-e4. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Chen J, Shao R, Li F, Monteiro M, Liu JP, Xu ZP and Gu W: PI 3K/Akt/mTOR pathway dual inhibitor BEZ 235 suppresses the stemness of colon cancer stem cells. Clin Exp Pharmacol Physiol. 42:1317–1326. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Vermeulen L, De Sousa E, Melo F, Van Der Heijden M, Cameron K, De Jong JH, Borovski T, Tuynman JB, Todaro M, Merz C, et al: Wnt activity defines colon cancer stem cells and is regulated by the microenvironment. Nat Cell Biol. 12:468–476. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Zhu P, Lu T, Chen Z, Liu B, Fan D, Li C, Wu J, He L, Zhu X, Du Y, et al: 5-hydroxytryptamine produced by enteric serotonergic neurons initiates colorectal cancer stem cell self-renewal and tumorigenesis. Neuron. 110:2268–2282. e42022. View Article : Google Scholar : PubMed/NCBI

46 

Zheng X, Pang B, Gu G, Gao T, Zhang R, Pang Q and Liu Q: Melatonin inhibits glioblastoma stem-like cells through suppression of EZH2-NOTCH1 signaling axis. Int J Biol Sci. 13:245–253. 2017. View Article : Google Scholar : PubMed/NCBI

47 

Gürsel DB, Berry N and Boockvar JA: The contribution of Notch signaling to glioblastoma via activation of cancer stem cell self-renewal: the role of the endothelial network. Neurosurgery. 70:N19–N21. 2012. View Article : Google Scholar : PubMed/NCBI

48 

Pham QT, Oue N, Sekino Y, Yamamoto Y, Shigematsu Y, Sakamoto N, Sentani K, Uraoka N and Yasui W: TDO2 overexpression is associated with cancer stem cells and poor prognosis in esophageal squamous cell carcinoma. Oncology. 95:297–308. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Liu J, Qin X, Pan D, Zhang B and Jin F: Amino acid-mediated metabolism: A new power to influence properties of stem cells. Stem Cells Int. 2019:69194632019. View Article : Google Scholar : PubMed/NCBI

50 

Zhang R, Hu P, Zang Q, Yue X, Zhou Z, Xu X, Xu J, Li S, Chen Y, Qiang B, et al: LC-MS-based metabolomics reveals metabolic signatures related to glioma stem-like cell self-renewal and differentiation. RSC Adv. 7:24221–24232. 2017. View Article : Google Scholar

51 

Venkateswaran N and Conacci-Sorrell M: Kynurenine: An oncometabolite in colon cancer. Cell Stress. 4:24–26. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Shi D, Wu X, Jian Y, Wang J, Huang C, Mo S, Li Y, Li F, Zhang C, Zhang D, et al: USP14 promotes tryptophan metabolism and immune suppression by stabilizing IDO1 in colorectal cancer. Nat Commun. 13:56442022. View Article : Google Scholar : PubMed/NCBI

53 

Brandacher G, Perathoner A, Ladurner R, Schneeberger S, Obrist P, Winkler C, Werner ER, Werner-Felmayer G, Weiss HG, Göbel G, et al: Prognostic value of indoleamine 2, 3-dioxygenase expression in colorectal cancer: Effect on tumor-infiltrating T cells. Clin Cancer Res. 12:1144–1151. 2006. View Article : Google Scholar : PubMed/NCBI

54 

Pan K, Wang H, Chen MS, Zhang HK, Weng DS, Zhou J, Huang W, Li JJ, Song HF and Xia JC: Expression and prognosis role of indoleamine 2, 3-dioxygenase in hepatocellular carcinoma. J Cancer Res Clin Oncol. 134:1247–1253. 2008. View Article : Google Scholar : PubMed/NCBI

55 

Zhao Q, Kuang DM, Wu Y, Xiao X, Li XF, Li TJ and Zheng L: Activated CD69+ T cells foster immune privilege by regulating IDO expression in tumor-associated macrophages. J Immunol. 188:1117–1124. 2012. View Article : Google Scholar : PubMed/NCBI

56 

Tummala KS, Gomes AL, Yilmaz M, Graña O, Bakiri L, Ruppen I, Ximénez-Embún P, Sheshappanavar V, Rodriguez-Justo M, Pisano DG, et al: Inhibition of de novo NAD+ synthesis by oncogenic URI causes liver tumorigenesis through DNA damage. Cancer Cell. 26:826–839. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Yu C, Rao D, Zhu H, Liu Q, Huang W, Zhang L, Liang H, Song J and Ding Z: TDO2 was downregulated in hepatocellular carcinoma and inhibited cell proliferation by upregulating the expression of p21 and p27. Biomed Res Int. 2021:47084392021. View Article : Google Scholar : PubMed/NCBI

58 

Wu Z, Yan L, Lin J, Ke K and Yang W: Constitutive TDO2 expression promotes liver cancer progression by an autocrine IL-6 signaling pathway. 2 Cancer Cell Int. 21:5382021. View Article : Google Scholar : PubMed/NCBI

59 

Li L, Wang T, Li S, Chen Z, Wu J, Cao W, Wo Q, Qin X and Xu J: TDO2 promotes the EMT of hepatocellular carcinoma through Kyn-AhR pathway. Front Oncol. 10:5628232021. View Article : Google Scholar : PubMed/NCBI

60 

Jin H, Zhang Y, You H, Tao X, Wang C, Jin G, Wang N, Ruan H, Gu D, Huo X, et al: Prognostic significance of kynurenine 3-monooxygenase and effects on proliferation, migration and invasion of human hepatocellular carcinoma. Sci Rep. 5:104662015. View Article : Google Scholar : PubMed/NCBI

61 

Shi Z, Gan G, Xu X, Zhang J, Yuan Y, Bi B, Gao X, Xu P, Zeng W, Li J, et al: Kynurenine derivative 3-HAA is an agonist ligand for transcription factor YY1. J Hematol Oncol. 14:1532021. View Article : Google Scholar : PubMed/NCBI

62 

Zuo X, Chen Z, Cai J, Gao W, Zhang Y, Han G, Pu L, Wu Z, You W, Qin J, et al: 5-Hydroxytryptamine receptor 1D aggravates hepatocellular carcinoma progression through FoxO6 in AKT-dependent and independent manners. Hepatology. 69:2031–2047. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Zhang T, Tan XL, Xu Y, Wang ZZ, Xiao CH and Liu R: Expression and prognostic value of indoleamine 2, 3-dioxygenase in pancreatic cancer. Chin Med J (Engl). 130:710–716. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Hezaveh K, Shinde RS, Klötgen A, Halaby MJ, Lamorte S, Ciudad MT, Quevedo R, Neufeld L, Liu ZQ, Jin R, et al: Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity. 55:324–40. e82022. View Article : Google Scholar : PubMed/NCBI

65 

Koblish HK, Hansbury MJ, Bowman KJ, Yang G, Neilan CL, Haley PJ, Burn TC, Waeltz P, Sparks RB, Yue EW, et al: Hydroxyamidine inhibitors of indoleamine-2, 3-dioxygenase potently suppress systemic tryptophan catabolism and the growth of IDO-expressing tumors. Mol Cancer Ther. 9:489–498. 2010. View Article : Google Scholar : PubMed/NCBI

66 

Guastella AR, Michelhaugh SK, Klinger NV, Fadel HA, Kiousis S, Ali-Fehmi R, Kupsky WJ, Juhász C and Mittal S: Investigation of the aryl hydrocarbon receptor and the intrinsic tumoral component of the kynurenine pathway of tryptophan metabolism in primary brain tumors. J Neurooncol. 139:239–249. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Panitz V, Končarević S, Sadik A, Friedel D, Bausbacher T, Trump S, Farztdinov V, Schulz S, Sievers P, Schmidt S, et al: Tryptophan metabolism is inversely regulated in the tumor and blood of patients with glioblastoma. Theranostics. 11:9217–9233. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Zhong C, Peng L, Tao B, Yin S, Lyu L, Ding H, Yang X, Peng T, He H and Zhou P: TDO2 and tryptophan metabolites promote kynurenine/AhR signals to facilitate glioma progression and immunosuppression. Am J Cancer Res. 12:2558–2575. 2022.PubMed/NCBI

69 

Mohapatra SR, Sadik A, Tykocinski LO, Dietze J, Poschet G, Heiland I and Opitz CA: Hypoxia inducible factor 1α inhibits the expression of immunosuppressive tryptophan-2, 3-dioxygenase in glioblastoma. Front Immunol. 10:27622019. View Article : Google Scholar : PubMed/NCBI

70 

Ladomersky E, Zhai L, Lauing KL, Bell A, Xu J, Kocherginsky M, Zhang B, Wu JD, Podojil JR, Platanias LC, et al: Advanced age increases immunosuppression in the brain and decreases immunotherapeutic efficacy in subjects with glioblastoma. Clin Cancer Res. 26:5232–5245. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Sahm F, Oezen I, Opitz CA, Radlwimmer B, Von Deimling A, Ahrendt T, Adams S, Bode HB, Guillemin GJ, Wick W and Platten M: The endogenous tryptophan metabolite and NAD+ precursor quinolinic acid confers resistance of gliomas to oxidative stress. Cancer Res. 73:3225–3234. 2013. View Article : Google Scholar : PubMed/NCBI

72 

Kamson DO, Lee TJ, Varadarajan K, Robinette NL, Muzik O, Chakraborty PK, Snyder M, Barger GR, Mittal S and Juhász C: Clinical significance of tryptophan metabolism in the nontumoral hemisphere in patients with malignant glioma. J Nucl Med. 55:1605–1610. 2014. View Article : Google Scholar : PubMed/NCBI

73 

Juhász C, Chugani DC, Barger GR, Kupsky WJ, Chakraborty PK, Muzik O and Mittal S: Quantitative PET imaging of tryptophan accumulation in gliomas and remote cortex: Correlation with tumor proliferative activity. Clin Nucl Med. 37:838–842. 2012. View Article : Google Scholar : PubMed/NCBI

74 

Sadik A, Somarribas Patterson LF, Öztürk S, Mohapatra SR, Panitz V, Secker PF, Pfänder P, Loth S, Salem H, Prentzell MT, et al: IL4I1 is a metabolic immune checkpoint that activates the AHR and promotes tumor progression. Cell. 182:1252–70. e342020. View Article : Google Scholar : PubMed/NCBI

75 

Batista CE, Juhász C, Muzik O, Kupsky WJ, Barger G, Chugani HT, Mittal S, Sood S, Chakraborty PK and Chugani DC: Imaging correlates of differential expression of indoleamine 2, 3-dioxygenase in human brain tumors. Mol Imaging Biol. 11:460–466. 2009. View Article : Google Scholar : PubMed/NCBI

76 

Talari NK, Panigrahi M, Madigubba S, Challa S and Phanithi PB: Altered tryptophan metabolism in human meningioma. J Neurooncol. 130:69–77. 2016. View Article : Google Scholar : PubMed/NCBI

77 

Bosnyák E, Kamson DO, Guastella AR, Varadarajan K, Robinette NL, Kupsky WJ, Muzik O, Michelhaugh SK, Mittal S and Juhász C: Molecular imaging correlates of tryptophan metabolism via the kynurenine pathway in human meningiomas. Neuro Oncol. 17:1284–1292. 2015.PubMed/NCBI

78 

Ino K, Yamamoto E, Shibata K, Kajiyama H, Yoshida N, Terauchi M, Nawa A, Nagasaka T, Takikawa O and Kikkawa F: Inverse correlation between tumoral indoleamine 2, 3-dioxygenase expression and tumor-infiltrating lymphocytes in endometrial cancer: Its association with disease progression and survival. Clin Cancer Res. 14:2310–2317. 2008. View Article : Google Scholar : PubMed/NCBI

79 

Yoshida N, Ino K, Ishida Y, Kajiyama H, Yamamoto E, Shibata K, Terauchi M, Nawa A, Akimoto H, Takikawa O, et al: Overexpression of indoleamine 2, 3-dioxygenase in human endometrial carcinoma cells induces rapid tumor growth in a mouse xenograft model. Clin Cancer Res. 14:7251–7259. 2008. View Article : Google Scholar : PubMed/NCBI

80 

Ino K, Yoshida N, Kajiyama H, Shibata K, Yamamoto E, Kidokoro K, Takahashi N, Terauchi M, Nawa A, Nomura S, et al: Indoleamine 2, 3-dioxygenase is a novel prognostic indicator for endometrial cancer. Br J Cancer. 95:1555–1561. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Inaba T, Ino K, Kajiyama H, Yamamoto E, Shibata K, Nawa A, Nagasaka T, Akimoto H, Takikawa O and Kikkawa F: Role of the immunosuppressive enzyme indoleamine 2, 3-dioxygenase in the progression of ovarian carcinoma. Gynecol Oncol. 115:185–192. 2009. View Article : Google Scholar : PubMed/NCBI

82 

Odunsi K, Qian F, Lugade AA, Yu H, Geller MA, Fling SP, Kaiser JC, Lacroix AM, D'Amico L, Ramchurren N, et al: Metabolic adaptation of ovarian tumors in patients treated with an IDO1 inhibitor constrains antitumor immune responses. Sci Transl Med. 14:eabg84022022. View Article : Google Scholar : PubMed/NCBI

83 

Gostner JM, Obermayr E, Braicu IE, Concin N, Mahner S, Vanderstichele A, Sehouli J, Vergote I, Fuchs D and Zeillinger R: Immunobiochemical pathways of neopterin formation and tryptophan breakdown via indoleamine 2, 3-dioxygenase correlate with circulating tumor cells in ovarian cancer patients-A study of the OVCAD consortium. Gynecol Oncol. 149:371–380. 2018. View Article : Google Scholar : PubMed/NCBI

84 

Singh R, Shaik S, Negi BS, Rajguru JP, Patil PB, Parihar AS and Sharma U: Non-Hodgkin's lymphoma: A review. J Family Med Prim Care. 9:1834–1840. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Shankland KR, Armitage JO and Hancock BW: Non-hodgkin lymphoma. Lancet. 380:848–857. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Feller AC and Diebold J: Histopathology of Nodal and Extranodal Non-Hodgkin's Lymphomas. Springer Science & Business Media; Heidelberg: 2003

87 

Liu XQ, Lu K, Feng LL, Ding M, Gao JM, Ge XL and Wang X: Up-regulated expression of indoleamine 2, 3-dioxygenase 1 in non-Hodgkin lymphoma correlates with increased regulatory T-cell infiltration. Leuk Lymphoma. 55:405–414. 2014. View Article : Google Scholar : PubMed/NCBI

88 

El Kholy NM, Sallam MM, Ahmed MB, Sallam RM, Asfour IA, Hammouda JA, Habib HZ and Abu-Zahra F: Expression of indoleamine 2, 3-dioxygenase in acute myeloid leukemia and the effect of its inhibition on cultured leukemia blast cells. Med Oncol. 28:270–278. 2011. View Article : Google Scholar : PubMed/NCBI

89 

Curti A, Pandolfi S, Valzasina B, Aluigi M, Isidori A, Ferri E, Salvestrini V, Bonanno G, Rutella S, Durelli I, et al: Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25− into CD25+ T regulatory cells. Blood. 109:2871–2877. 2007. View Article : Google Scholar : PubMed/NCBI

90 

Zhang ML, Kem M, Mooradian MJ, Eliane JP, Huynh TG, Iafrate AJ, Gainor JF and Mino-Kenudson M: Differential expression of PD-L1 and IDO1 in association with the immune microenvironment in resected lung adenocarcinomas. Mod Pathol. 32:511–523. 2019. View Article : Google Scholar : PubMed/NCBI

91 

Tang D, Yue L, Yao R, Zhou L, Yang Y, Lu L and Gao W: P53 prevent tumor invasion and metastasis by down-regulating IDO in lung cancer. Oncotarget. 8:545482017. View Article : Google Scholar : PubMed/NCBI

92 

Hsu YL, Hung JY, Chiang SY, Jian SF, Wu CY, Lin YS, Tsai YM, Chou SH, Tsai MJ and Kuo PL: Lung cancer-derived galectin-1 contributes to cancer associated fibroblast-mediated cancer progression and immune suppression through TDO2/kynurenine axis. Oncotarget. 7:27584–27598. 2016. View Article : Google Scholar : PubMed/NCBI

93 

Feng H, Cao B, Peng X and Wei Q: Cancer-associated fibroblasts strengthen cell proliferation and EGFR TKIs resistance through aryl hydrocarbon receptor dependent signals in non-small cell lung cancer. BMC Cancer. 22:7642022. View Article : Google Scholar : PubMed/NCBI

94 

Karayama M, Masuda J, Mori K, Yasui H, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, Enomoto N, Nakamura Y, et al: Comprehensive assessment of multiple tryptophan metabolites as potential biomarkers for immune checkpoint inhibitors in patients with non-small cell lung cancer. Clin Transl Oncol. 23:418–423. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Levina V, Su Y and Gorelik E: Immunological and nonimmunological effects of indoleamine 2, 3-dioxygenase on breast tumor growth and spontaneous metastasis formation. Clin Dev Immunol. 2012:1730292012. View Article : Google Scholar : PubMed/NCBI

96 

D'Amato NC, Rogers TJ, Gordon MA, Greene LI, Cochrane DR, Spoelstra NS, Nemkov TG, D'Alessandro A, Hansen KC and Richer JK: A TDO2-AhR signaling axis facilitates anoikis resistance and metastasis in triple-negative breast cancer. Cancer Res. 75:4651–4664. 2015. View Article : Google Scholar : PubMed/NCBI

97 

Fallarino F, Grohmann U, Vacca C, Bianchi R, Orabona C, Spreca A, Fioretti MC and Puccetti P: T cell apoptosis by tryptophan catabolism. Cell Death Differ. 9:1069–1077. 2002. View Article : Google Scholar : PubMed/NCBI

98 

Fallarino F, Grohmann U, Vacca C, Orabona C, Spreca A, Fioretti MC and Puccetti P: T cell apoptosis by kynurenines. Adv Exp Med Biol. 527:183–190. 2003. View Article : Google Scholar : PubMed/NCBI

99 

Platten M, Wick W and Van den Eynde BJ: Tryptophan catabolism in cancer: Beyond IDO and tryptophan depletion. Cancer Res. 72:5435–5440. 2012. View Article : Google Scholar : PubMed/NCBI

100 

Cui M, Liu D, Xiong W, Wang Y and Mi J: ERRFI1 induces apoptosis of hepatocellular carcinoma cells in response to tryptophan deficiency. Cell Death Discov. 7:2742021. View Article : Google Scholar : PubMed/NCBI

101 

Moroni F: Tryptophan metabolism and brain function: focus on kynurenine and other indole metabolites. Eur J Pharmacol. 375:87–100. 1999. View Article : Google Scholar : PubMed/NCBI

102 

Li F, Zhao Z, Zhang Z, Zhang Y and Guan W: Tryptophan metabolism induced by TDO2 promotes prostatic cancer chemotherapy resistance in a AhR/c-Myc dependent manner. BMC Cancer. 21:11122021. View Article : Google Scholar : PubMed/NCBI

103 

Gao N, Yang Y, Liu S, Fang C, Dou X, Zhang L and Shan A: Gut-derived metabolites from dietary tryptophan supplementation quench intestinal inflammation through the AMPK-SIRT1-autophagy pathway. J Agric Food Chem. 70:16080–16095. 2022. View Article : Google Scholar : PubMed/NCBI

104 

Zhuo J, Chu L, Liu D, Cai S and Dong H: Tryptophan metabolite indole-3-acetic acid ameliorates pulmonary fibrosis by regulating lung microbiota and inducing autophagy via inhibition of the PI3K/AKT/MTOR pathway. Am J Respir Crit Care Med. A24592024.

105 

Osawa Y, Kanamori H, Seki E, Hoshi M, Ohtaki H, Yasuda Y, Ito H, Suetsugu A, Nagaki M, Moriwaki H, et al: L-tryptophan-mediated enhancement of susceptibility to nonalcoholic fatty liver disease is dependent on the mammalian target of rapamycin. J Biol Chem. 286:34800–34808. 2011. View Article : Google Scholar : PubMed/NCBI

106 

Rathmell JC: Metabolism and autophagy in the immune system: Immunometabolism comes of age. Immunol Rev. 249:52012. View Article : Google Scholar : PubMed/NCBI

107 

Liu X, Zhou W, Zhang X, Ding Y, Du Q and Hu R: 1-L-MT, an IDO inhibitor, prevented colitis-associated cancer by inducing CDC20 inhibition-mediated mitotic death of colon cancer cells. Int J Cancer. 143:1516–1529. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Prendergast GC, Malachowski WJ, Mondal A, Scherle P and Muller AJ: Indoleamine 2, 3-dioxygenase and its therapeutic inhibition in cancer. Int Rev Cell Mol Biol. 336:175–203. 2018. View Article : Google Scholar : PubMed/NCBI

109 

Opitz CA, Litzenburger UM, Sahm F, Ott M, Tritschler I, Trump S, Schumacher T, Jestaedt L, Schrenk D, Weller M, et al: An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 478:197–203. 2011. View Article : Google Scholar : PubMed/NCBI

110 

Liu D, Liang CH, Huang B, Zhuang X, Cui W, Yang L, Yang Y, Zhang Y, Fu X, Zhang X, et al: Tryptophan metabolism acts as a new anti-ferroptotic pathway to mediate tumor growth. Adv Sci (Weinh). 10:22040062023. View Article : Google Scholar : PubMed/NCBI

111 

Zhang W, Mao S, Shi D, Zhang J, Zhang Z, Guo Y, Wu Y, Wang R, Wang L, Huang Y and Yao X: MicroRNA-153 decreases tryptophan catabolism and inhibits angiogenesis in bladder cancer by targeting indoleamine 2, 3-dioxygenase 1. Front Oncol. 9:6192019. View Article : Google Scholar : PubMed/NCBI

112 

Cecchi M, Anceschi C, Silvano A, Coniglio ML, Chinnici A, Magnelli L, Lapucci A, Laurenzana A and Parenti A: Unveiling the role of tryptophan 2, 3-dioxygenase in the angiogenic process. Pharmaceuticals (Basel). 17:5582024. View Article : Google Scholar : PubMed/NCBI

113 

Nocito A, Dahm F, Jochum W, Jang JH, Georgiev P, Bader M, Graf R and Clavien PA: Serotonin regulates macrophage-mediated angiogenesis in a mouse model of colon cancer allografts. Cancer Res. 68:5152–5158. 2008. View Article : Google Scholar : PubMed/NCBI

114 

Cerezo AB, Hornedo-Ortega R, Álvarez-Fernández MA, Troncoso AM and García-Parrilla MC: Inhibition of VEGF-induced VEGFR-2 activation and HUVEC migration by melatonin and other bioactive indolic compounds. Nutrients. 9:2492017. View Article : Google Scholar : PubMed/NCBI

115 

Peters MA, Walenkamp AM, Kema IP, Meijer C, de Vries EG and Oosting SF: Dopamine and serotonin regulate tumor behavior by affecting angiogenesis. Drug Resist Updat. 17:96–104. 2014. View Article : Google Scholar : PubMed/NCBI

116 

Nayak-Kapoor A, Hao Z, Sadek R, Dobbins R, Marshall L, Vahanian NN, Jay Ramsey W, Kennedy E, Mautino MR, Link CJ, et al: Phase Ia study of the indoleamine 2,3-dioxygenase 1 (IDO1) inhibitor navoximod (GDC-0919) in patients with recurrent advanced solid tumors. J Immunother Cancer. 6:612018. View Article : Google Scholar : PubMed/NCBI

117 

Chen F, Zhao D, Huang Y, Wen X and Feng S: Synergetic impact of combined navoximod with cisplatin mitigates chemo-immune resistance via blockading IDO1(+) CAFs-secreted Kyn/AhR/IL-6 and pol ζ-prevented CIN in human oral squamous cell carcinoma. Life Sci. 335:1222392023. View Article : Google Scholar : PubMed/NCBI

118 

Wang XX, Sun SY, Dong QQ, Wu XX, Tang W and Xing YQ: Recent advances in the discovery of indoleamine 2,3-dioxygenase 1 (IDO1) inhibitors. Medchemcomm. 10:1740–1754. 2019. View Article : Google Scholar : PubMed/NCBI

119 

Oh JE, Shim KY, Lee JI, Choi SI, Baik SK and Eom YW: 1-Methyl-L-tryptophan promotes the apoptosis of hepatic stellate cells arrested by interferon-γ by increasing the expression of IFN-γRβ, IRF-1 and FAS. Int J Mol Med. 40:576–582. 2017. View Article : Google Scholar : PubMed/NCBI

120 

Lewis HC, Chinnadurai R, Bosinger SE and Galipeau J: The IDO inhibitor 1-methyl tryptophan activates the aryl hydrocarbon receptor response in mesenchymal stromal cells. Oncotarget. 8:91914–91927. 2017. View Article : Google Scholar : PubMed/NCBI

121 

Opitz CA, Litzenburger UM, Opitz U, Sahm F, Ochs K, Lutz C and Wick Wand Platten M: The indoleamine-2,3-dioxygenase (IDO) inhibitor 1-methyl-D-tryptophan upregulates IDO1 in human cancer cells. PLoS One. 6:e198232011. View Article : Google Scholar : PubMed/NCBI

122 

Takada K, Kohashi K, Shimokawa M, Haro A, Osoegawa A, Tagawa T, Seto T, Oda Y and Maehara Y: Co-expression of IDO1 and PD-L1 in lung squamous cell carcinoma: Potential targets of novel combination therapy. Lung Cancer. 128:26–32. 2019. View Article : Google Scholar : PubMed/NCBI

123 

Ladomersky E, Zhai L, Lenzen A, Lauing KL, Qian J, Scholtens DM, Gritsina G, Sun X, Liu Y, Yu F, et al: IDO1 inhibition synergizes with radiation and PD-1 blockade to durably increase survival against advanced glioblastoma. Clin Cancer Res. 24:2559–2573. 2018. View Article : Google Scholar : PubMed/NCBI

124 

Kotecki N, Vuagnat P, O'Neil BH, Jalal S, Rottey S, Prenen H, Benhadji KA, Xia M, Szpurka AM, Saha A, et al: A Phase I study of an IDO-1 inhibitor (LY3381916) as monotherapy and in combination with an Anti-PD-L1 Antibody (LY3300054) in patients with advanced cancer. J Immunother. 44:264–275. 2021. View Article : Google Scholar : PubMed/NCBI

125 

Balog A, Lin TA, Maley D, Gullo-Brown J, Kandoussi EH, Zeng J and Hunt JT: Preclinical characterization of linrodostat mesylate, a novel, potent, and selective oral indoleamine 2,3-dioxygenase 1 inhibitor. Mol Cancer Ther. 20:467–476. 2021. View Article : Google Scholar : PubMed/NCBI

126 

Lin J, Sun-Waterhouse D and Cui C: The therapeutic potential of diet on immune-related diseases: Based on the regulation on tryptophan metabolism. Crit Rev Food Sci Nutr. 62:8793–8811. 2022. View Article : Google Scholar : PubMed/NCBI

127 

Hu W, Yan G, Ding Q, Cai J, Zhang Z, Zhao Z, Lei H and Zhu YZ: Update of indoles: Promising molecules for ameliorating metabolic diseases. Biomed Pharmacother. 150:1129572022. View Article : Google Scholar : PubMed/NCBI

128 

Li X, Zhang B, Hu Y and Zhao Y: New insights into gut-bacteria-derived indole and its derivatives in intestinal and liver diseases. Front Pharmacol. 12:7695012021. View Article : Google Scholar : PubMed/NCBI

129 

Liu Y, Pei Z, Pan T, Wang H, Chen W and Lu W: Indole metabolites and colorectal cancer: Gut microbial tryptophan metabolism, host gut microbiome biomarkers, and potential intervention mechanisms. Microbiol Res. 272:1273922023. View Article : Google Scholar : PubMed/NCBI

130 

Krause FF, Mangold KI, Ruppert AL, Leister H, Hellhund-Zingel A, Lopez Krol A, Pesek J, Watzer B, Winterberg S, Raifer H, et al: Clostridium sporogenes-derived metabolites protect mice against colonic inflammation. Gut Microbes. 16:24126692024. View Article : Google Scholar : PubMed/NCBI

131 

Liu Y, Chen H, Van Treuren W, Hou BH, Higginbottom SK and Dodd D: Clostridium sporogenes uses reductive Stickland metabolism in the gut to generate ATP and produce circulating metabolites. Nat Microbiol. 7:695–706. 2022. View Article : Google Scholar : PubMed/NCBI

132 

Wang A, Guan C, Wang T, Mu G and Tuo Y: Lactiplantibacillus plantarum-derived indole-3-lactic acid ameliorates intestinal barrier integrity through the AhR/Nrf2/NF-κB Axis. J Agric Food Chem. April 10–2024.(Epub ahead of print).

133 

Wang A, Guan C, Wang T, Mu G and Tuo Y: Indole-3-lactic acid, a tryptophan metabolite of lactiplantibacillus plantarum DPUL-S164, improved intestinal barrier damage by activating AhR and Nrf2 signaling pathways. J Agric Food Chem. 71:18792–18801. 2023. View Article : Google Scholar : PubMed/NCBI

134 

Leclerc D, Staats Pires AC, Guillemin GJ and Gilot D: Detrimental activation of AhR pathway in cancer: An overview of therapeutic strategies. Curr Opin Immunol. 70:15–26. 2021. View Article : Google Scholar : PubMed/NCBI

135 

Paris A, Tardif N, Galibert MD and Corre S: AhR and cancer: From gene profiling to targeted therapy. Int J Mol Sci. 22:7522022. View Article : Google Scholar

136 

Wang Z, Monti S and Sherr DH: The diverse and important contributions of the AHR to cancer and cancer immunity. Current Opin Toxicol. 2:93–102. 2017. View Article : Google Scholar

137 

Sun M, Ma N, He T, Johnston LJ and Ma X: Tryptophan (Trp) modulates gut homeostasis via aryl hydrocarbon receptor (AhR). Crit Rev Food Sci Nutr. 60:1760–1768. 2020. View Article : Google Scholar : PubMed/NCBI

138 

Griffith BD and Frankel TL: The aryl hydrocarbon receptor: impact on the tumor immune microenvironment and modulation as a potential therapy. Cancers (Basel). 16:4722024. View Article : Google Scholar : PubMed/NCBI

139 

Leja-Szpak A, Góralska M, Link-Lenczowski P, Czech U, Nawrot-Porąbka K, Bonior J and Jaworek J: The opposite effect of L-kynurenine and Ahr inhibitor Ch223191 on apoptotic protein expression in pancreatic carcinoma cells (Panc-1). Anticancer Agents Med Chem. 19:2079–2090. 2019. View Article : Google Scholar : PubMed/NCBI

140 

Choi EY, Lee H, Dingle RW, Kim KB and Swanson HI: Development of novel CH223191-based antagonists of the aryl hydrocarbon receptor. Mol Pharmacol. 81:3–11. 2012. View Article : Google Scholar : PubMed/NCBI

141 

Zhang Y, Ma YC, Song J, Jin Y and Bao YN: StemRegenin-1 reverses drug resistance of MCF-7/ADR Cells via AhR/ABC transports and AhR/UGTs pathways. Current Proteomics. 21:113–128. 2024. View Article : Google Scholar

142 

Kober C, Roewe J, Schmees N, Roese L, Roehn U, Bader B, Stoeckigt D, Prinz F, Gorjánácz M, Roider HG, et al: Targeting the aryl hydrocarbon receptor (AhR) with BAY 2416964: A selective small molecule inhibitor for cancer immunotherapy. J Immunother Cancer. 11:e0074952023. View Article : Google Scholar : PubMed/NCBI

143 

Parks AJ, Pollastri MP, Hahn ME, Stanford EA, Novikov O, Franks DG, Haigh SE, Narasimhan S, Ashton TD, Hopper TG, et al: In silico identification of an aryl hydrocarbon receptor antagonist with biological activity in vitro and in vivo. Mol Pharmacol. 86:593–608. 2014. View Article : Google Scholar : PubMed/NCBI

144 

Garg A, Sharma A, Krishnamoorthy P, Garg J, Virmani D, Sharma T, Stefanini G, Kostis JB, Mukherjee D and Sikorskaya E: Role of niacin in current clinical practice: A systematic review. Am J Med. 130:173–187. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2025
Volume 54 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhao J, Bai X, Du J, Chen Y, Guo X, Zhang J, Gan J, Wu P, Chen S, Zhang X, Zhang X, et al: Tryptophan metabolism: From physiological functions to key roles and therapeutic targets in cancer (Review). Oncol Rep 54: 86, 2025.
APA
Zhao, J., Bai, X., Du, J., Chen, Y., Guo, X., Zhang, J. ... Gao, L. (2025). Tryptophan metabolism: From physiological functions to key roles and therapeutic targets in cancer (Review). Oncology Reports, 54, 86. https://doi.org/10.3892/or.2025.8919
MLA
Zhao, J., Bai, X., Du, J., Chen, Y., Guo, X., Zhang, J., Gan, J., Wu, P., Chen, S., Zhang, X., Yang, J., Jin, J., Gao, L."Tryptophan metabolism: From physiological functions to key roles and therapeutic targets in cancer (Review)". Oncology Reports 54.1 (2025): 86.
Chicago
Zhao, J., Bai, X., Du, J., Chen, Y., Guo, X., Zhang, J., Gan, J., Wu, P., Chen, S., Zhang, X., Yang, J., Jin, J., Gao, L."Tryptophan metabolism: From physiological functions to key roles and therapeutic targets in cancer (Review)". Oncology Reports 54, no. 1 (2025): 86. https://doi.org/10.3892/or.2025.8919