1
|
Krupa MM, Pienkowski T, Tankiewicz-Kwedlo A, Lyson T. Targeting the kynurenine pathway in gliomas: Insights into pathogenesis, therapeutic targets, and clinical advances. Biochim Biophys Acta Rev Cancer 2025; 1880:189343. [PMID: 40345262 DOI: 10.1016/j.bbcan.2025.189343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Gliomas, the most prevalent primary brain tumors, continue to present significant challenges in oncology due to poor patient prognosis despite advances in treatment such as immunotherapy and cancer vaccines. Recent research highlights the potential of targeting tryptophan metabolism, particularly the kynurenine pathway (KP) and combinatorial approaches with immunotherapies, as a promising strategy in cancer research. The key enzymes of the kynurenine pathway, such as IDO1, IDO2, and TDO, and metabolites like kynurenine, kynurenic acid, and quinolinic acid, are implicated in fostering an immunosuppressive tumor microenvironment and promoting glioma cell survival. In glioblastoma, a highly aggressive glioma subtype, elevated IDO and TDO expression correlates with reduced survival rates. KP metabolites, such as kynurenine (KYN), 3-hydroxykynurenine (3-HK), kynurenic acid (KYNA), and quinolinic acid (QUIN), are involved in modulating immune responses, oxidative stress, neuroprotection, and neurotoxicity. This review synthesizes recent findings on the kynurenine pathway involvement in glioma pathogenesis, examining potential therapeutic targets within this pathway and discussing ongoing clinical trials that draw attention to treatments based on this pathway. Furthermore, it highlights novel findings on the post-translational modifications of kynurenine pathway enzymes and their regulatory roles, presenting their potential as therapeutic targets in gliomas.
Collapse
Affiliation(s)
- Mikolaj Marek Krupa
- Department of Neurosurgery, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Tomasz Pienkowski
- Laboratory of Metabolomics and Proteomics, Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland.
| | - Anna Tankiewicz-Kwedlo
- Department of Pharmacodynamics, Medical University of Bialystok, 15-222 Bialystok, Poland
| | - Tomasz Lyson
- Department of Neurosurgery, Medical University of Bialystok, 15-276 Bialystok, Poland; Department of Interventional Neurology, Medical University of Bialystok, 15-276 Bialystok, Poland
| |
Collapse
|
2
|
Liu ZQ, Ciudad MT, McGaha TL. New insights into tryptophan metabolism in cancer. Trends Cancer 2025:S2405-8033(25)00076-7. [PMID: 40274457 DOI: 10.1016/j.trecan.2025.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025]
Abstract
Tryptophan (Trp) is an essential amino acid and key intermediate in a range of biological processes. Early studies identified altered Trp utilization in cancer cells favoring cancer survival and growth. Seminal findings linking Trp metabolism and suppression of immunity led to an explosion of interest ultimately culminating in clinical trials targeting these pathways in melanoma. The failure of these trials led to a clinical retreat in this approach; however, recent insights into the complex interplay of the various Trp circuits and between tumor cells, immune cells, and the microbiota have shown that reconsideration of Trp metabolism is needed. Here, we discuss recent developments in our understanding of Trp metabolism and apparent contradictions in the field. We also discuss adaptations that occur when Trp pathways are manipulated, which may impact therapy responses.
Collapse
Affiliation(s)
- Zhe Qi Liu
- Tumor Immunotherapy Program Princess Margaret Cancer Centre, Toronto, ON, M5G 2M9, Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - M Teresa Ciudad
- Tumor Immunotherapy Program Princess Margaret Cancer Centre, Toronto, ON, M5G 2M9, Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Tracy L McGaha
- Tumor Immunotherapy Program Princess Margaret Cancer Centre, Toronto, ON, M5G 2M9, Canada; Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
3
|
Stockwell CA, Thang M, Kram DE, Satterlee AB, Hingtgen S. Therapeutic approaches for targeting the pediatric brain tumor microenvironment. Drug Deliv Transl Res 2025:10.1007/s13346-025-01839-3. [PMID: 40257744 DOI: 10.1007/s13346-025-01839-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2025] [Indexed: 04/22/2025]
Abstract
Central nervous system (CNS) tumors are the most frequent solid malignant tumors in pediatric patients and are the leading cause of tumor-related death in children. Treatment for this heterogeneous group of tumors consists of various combinations of safe maximal surgical resection, chemotherapy, and radiation therapy which offer a cure for some children but often cause debilitating adverse late effects in others. While therapies targeting the tumor microenvironment (TME) like immune checkpoint inhibition (ICI) have been successful in treating some cancers, these therapies failed to exhibit treatment efficacy in the majority of pediatric brain tumors in the clinic. Importantly, the pediatric TME is unique and distinct from adult brain tumors and designing therapies to effectively target these tumors requires understanding the unique biology of pediatric brain tumors and the use of translational models that recapitulate the TME. Here we describe the TME of medulloblastoma (MB) and diffuse midline glioma (DMG), specifically diffuse intrinsic pontine glioma (DIPG), and further present the current drug delivery approaches and clinical administration routes targeting the TME in these tumors, including preclinical and clinical studies.
Collapse
Affiliation(s)
- Caroline A Stockwell
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Morrent Thang
- Neuroscience Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David E Kram
- Division of Pediatric Hematology-Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrew B Satterlee
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Eshelman Innovation, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Shawn Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Liu C, Qiao H, Li H, Hu X, Yan M, Fu Z, Zhang H, Wang Y, Du N. Exploring the role of LOX family in glioma progression and immune modulation. Front Immunol 2025; 16:1512186. [PMID: 40270974 PMCID: PMC12014642 DOI: 10.3389/fimmu.2025.1512186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/11/2025] [Indexed: 04/25/2025] Open
Abstract
Background Glioma is a major cause of mortality among central nervous system tumors, with a generally poor prognosis. The lysyl oxidase (LOX) family, a group of copper-dependent amine oxidases, has been implicated in the progression of various cancers, but its specific role in glioma and its relationship with immune infiltration remains insufficiently explored. This study aims to investigate the LOX family's expression, prognostic significance, and immune infiltration dynamics in glioma to identify potential therapeutic targets. Methods A comprehensive analysis was conducted using public databases to assess gene expression, mutation frequency, and immune infiltration patterns related to the LOX family in glioma. The results were validated through survival analysis and immunohistochemistry. Functional assays, including EdU, Transwell, and flow cytometry, were used to evaluate glioma cell proliferation, migration, invasion, and apoptosis. Co-culture experiments with immune cells, ELISA, and a glioma transplantation model were employed to study the immune-modulatory effects of the LOX family. Gene and protein expression levels were further analyzed using qRT-PCR and Western blotting. Results The LOX family was significantly upregulated in low-grade gliomas and strongly associated with poor clinical outcomes. Although mutation frequencies were low, the LOX family contributed to glioma progression through pathways involving metastasis, hypoxia response, angiogenesis, and immune cell infiltration. LOX expression correlated with increased infiltration of macrophages and eosinophils and decreased presence of Treg and CD8+ T cells. Knockdown of LOX genes impaired glioma cell functions, induced apoptosis, and altered immune cell behavior by reducing M2 macrophage polarization and enhancing CD8+ T cell activity. Conclusions The LOX family is overexpressed in glioma and is associated with poor prognosis and altered immune infiltration patterns. These findings highlight the LOX family as a promising prognostic marker and therapeutic target, particularly for enhancing the effectiveness of immunotherapy in glioma treatment.
Collapse
Affiliation(s)
- Chen Liu
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Oncology, the Fifth Medical Center, Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Huilian Qiao
- Department of Pathology, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Hongqi Li
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Xiaolong Hu
- Department of Radiation Oncology, Beijing Geriatric Hospital, Beijing, China
| | - Maohui Yan
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Zhiguang Fu
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Hengheng Zhang
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Yingjie Wang
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| | - Nan Du
- Medical School of Chinese People’s Liberation Army (PLA), Beijing, China
- Department of Radiotherapy, Air Force Medical Center, The Fourth Military Medical University, People’s Liberation Army (PLA), Beijing, China
| |
Collapse
|
5
|
Gao Y, Zhang M, Wang G, Lai W, Liao S, Chen Y, Ning Q, Tang S. Metabolic cross-talk between glioblastoma and glioblastoma-associated microglia/macrophages: From basic insights to therapeutic strategies. Crit Rev Oncol Hematol 2025; 208:104649. [PMID: 39922398 DOI: 10.1016/j.critrevonc.2025.104649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025] Open
Abstract
Glioblastoma (GBM), a highly malignant "cold" tumor of the central nervous system, is characterized by its ability to remodel the GBM immune microenvironment (GME), leading to significant resistance to immunotherapy. GBM-associated microglia/macrophages (GAMs) are essential components of the GME. Targeting GAMs has emerged as a promising strategy against GBM. However, their highly immunosuppressive nature contributes to GBM progression and drug resistance, significantly impeding anti-GBM immunotherapy. Accumulating evidence suggests that metabolic reprogramming accompanies GBM progression and GAM polarization, which are in turn driven by specific metabolic abnormalities and altered cellular signaling pathways. Importantly, metabolic crosstalk between GBM and GAMs further promotes tumor progression. Clarifying and disrupting this metabolic crosstalk is expected to enhance the antitumor phenotype of GAMs and inhibit GBM malignant progression. This review explores metabolism-based interregulation between GBM and GAMs and summarizes recent therapeutic strategies targeting this crosstalk, offering new insights into GBM immunotherapy.
Collapse
Affiliation(s)
- Yuan Gao
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Mengxia Zhang
- Department of Histology and Embryology, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Guihua Wang
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Weiwei Lai
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Shuxian Liao
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Yao Chen
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China
| | - Qian Ning
- Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| | - Shengsong Tang
- Department of Pharmacology, Ningxia Medical University, Yinchuan 750004, China; Hunan Province Key Laboratory for Antibody-Based Drug and Intelligent Delivery System, School of Pharmaceutical Sciences, Hunan University of Medicine, Huaihua 418000, China; College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
6
|
Shen ZP, Zhang ZY, Li N, Xu L, Chen Y. Targeted therapy for pediatric glioma: RAF(t)ing in the molecular era. World J Pediatr 2025; 21:338-351. [PMID: 40227462 DOI: 10.1007/s12519-025-00889-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Pediatric gliomas are the most frequently occurring central nervous system tumors in children. While targeted therapies have been widely applied in the treatment of many adult cancers, their use in pediatric gliomas has lagged behind. However, recent advances in multiomics profiling of pediatric gliomas, coupled with the approval of inhibitors against Raf serine/threonine kinase (RAF), isocitrate dehydrogenase 1/2 (IDH1/2) and neurotrophic receptor tyrosine kinase (NTRK), have spurred significant progress in this field. In light of these developments, this review aims to provide a comprehensive overview of current advancements and the evolving landscape of targeted therapeutic strategies and approaches for pediatric gliomas. DATA SOURCES Data analyzed in this study were obtained from the literature from PubMed, as well as other online databases and websites, including ClinicalTrials.gov and the Pediatric Neuro-Oncology Consortium. RESULTS Based on findings from multiomics profiling, significant insights have been gained into the genetic and molecular landscape of pediatric gliomas, enabling the identification of key mutations and potentially targetable lesions. These advancements provide rationales for the development of more precise treatment strategies and targeted therapies. Recent approvals of targeted therapies and ongoing clinical trials in pediatric gliomas are converging on the targeting of key signaling molecules and metabolic pathways. CONCLUSIONS In the molecular era, targeted therapies offer new hope for more effective and personalized treatment options for pediatric glioma patients. By developing and tailoring treatments to target specific molecular and metabolic vulnerabilities, targeted therapies have the potential to improve the clinical management of pediatric gliomas, ultimately enhancing both the treatment experience and overall prognosis of these patients.
Collapse
Affiliation(s)
- Zhi-Peng Shen
- Department of Neurosurgery, Children's Hospital Zhejiang University School of Medicine, Hangzhou, 310052, China
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Zhong-Yuan Zhang
- Department of Neurosurgery, Children's Hospital Zhejiang University School of Medicine, Hangzhou, 310052, China
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Nan Li
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Liang Xu
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Ye Chen
- Department of Neurosurgery, Children's Hospital Zhejiang University School of Medicine, Hangzhou, 310052, China.
- Pediatric Cancer Research Center, National Clinical Research Center for Child Health, Hangzhou, 310052, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
7
|
Srivastava S, Anbiaee R, Houshyari M, Laxmi, Sridhar SB, Ashique S, Hussain S, Kumar S, Taj T, Akbarnejad Z, Taghizadeh-Hesary F. Amino acid metabolism in glioblastoma pathogenesis, immune evasion, and treatment resistance. Cancer Cell Int 2025; 25:89. [PMID: 40082966 PMCID: PMC11908050 DOI: 10.1186/s12935-025-03721-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/01/2025] [Indexed: 03/16/2025] Open
Abstract
Glioblastoma (GBM) ranks among the most lethal primary tumors of the central nervous system. This is partly due to its complex intracellular metabolism and interactions with the surrounding tumor microenvironment (TME). Compelling evidence represents that altered amino acids (AAs) metabolism plays a crucial role in both areas. The role of AAs and their metabolites in glioma biology is an emerging topic. Therefore, this review was conducted to summarize the current knowledge about the molecular mechanisms by which AAs participate in the GBM pathogenesis. AAs can directly influence tumor progression by affecting tumor cell metabolism or indirectly by releasing bioactive agents through particular metabolic pathways. This review begins by examining the metabolic pathways of essential AAs, such as tryptophan, tyrosine, and phenylalanine, which contribute to synthesizing critical neurotransmitters and shape tumor metabolism signatures. We explore how these pathways impact tumor growth and immune modulation, focusing on how AAs and their metabolites can promote malignant properties in GBM cells. AAs also play a pivotal role in reprogramming the TME, contributing to immune evasion and resistance to therapy. The review further discusses how tumor metabolism signatures, influenced by AA metabolism, can enhance the immunosuppressive microenvironment, providing new avenues for targeted immunotherapies. Finally, we outline potential therapeutic strategies to modulate AA metabolism and emphasize critical opportunities for future research to improve GBM management.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, 203201, India
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi, 110017, India
| | - Robab Anbiaee
- Radio Oncology Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Houshyari
- Radio Oncology Department, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Laxmi
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, 203201, India
| | | | - Sumel Ashique
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, 711316, West Bengal, India
| | - Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, Uttarakhand, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), Sector 3 Pushp Vihar, New Delhi, 110017, India
| | - Tahreen Taj
- Department of Pharmacology, Yenepoya Pharmacy college and research centre, Yenepoya (Deemed to be) university, Mangalore, 575018, India
| | - Zeinab Akbarnejad
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Clinical Oncology Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Tsuboi N, Rivera-Caraballo KA, Sahu U, Pacholczyk R, Douglass E, Johnson TS, Wang Q, Kolhe R, Hedrick CC, Munn DH, Hong B. Blocking Feedback Immunosuppression of Antigen Presentation in Brain Tumor During Oncolytic Virotherapy with oHSV-mshPKR. Mol Cancer Ther 2025; 24:444-452. [PMID: 39711419 PMCID: PMC11879753 DOI: 10.1158/1535-7163.mct-24-0629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
Glioblastoma (GBM) is the most frequent malignant brain tumor. We recently discovered that oncolytic herpes simplex virus engineered to disable tumor-intrinsic protein kinase R (PKR) signaling (oHSV-shPKR) could increase oHSV oncolysis and antitumor immune response. However, in this study, we show that disabling tumor-intrinsic PKR signaling can also induce the activation of the indoleamine 2,3-dioxygenase (IDO) signaling pathway. Both GBM tumor progression and oHSV intratumoral therapy increased infiltration of IDO+CD11c+ dendritic cells (DC) into the tumor. The coculture of oHSV-infected human GBM neurospheres with monocyte-derived DCs (MoDC) dramatically increased IDO signaling activation in MoDCs through type-I IFN signaling. Addition of IDO inhibitor (indoximod) in the coculture significantly increased MoDC activation and reduced the consumption of tryptophan. Combining indoximod and oHSV significantly inhibited tumor growth and induced antigen-specific CD8+ T-cell activation. These results suggest that inhibition of the IDO pathway could significantly block feedback immunosuppression during oncolytic virotherapy of GBM.
Collapse
Affiliation(s)
- Nobushige Tsuboi
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | | | - Upasana Sahu
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Rafal Pacholczyk
- Department of Biochemistry and Molecular Biology, Georgia Cancer Center at Augusta University, 1410 Laney Walker Blvd, Augusta, GA, USA
| | - Eugene Douglass
- Department of Pharmaceutical & Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Theodore S. Johnson
- Department of Pediatrics, Pediatric Immunotherapy Program, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Qin Wang
- Department of Neuroscience & Regenerative Medicine, Augusta University, Augusta, GA, USA
| | - Ravindra Kolhe
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Catherine C. Hedrick
- Department of Medicine, Immunology Center of Georgia, Augusta University, Augusta, GA, USA
| | - David H. Munn
- Department of Pediatrics, Pediatric Immunotherapy Program, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| | - Bangxing Hong
- Department of Pathology, Georgia Cancer Center at Augusta University, Augusta, GA, USA
| |
Collapse
|
9
|
Kumar S, Koseki Y, Tanita K, Shibata A, Mizutani A, Kasai H. SN-38-indoximod conjugate: carrier free nano-prodrug for cancer therapy. Ther Deliv 2025; 16:217-226. [PMID: 39887189 PMCID: PMC11875466 DOI: 10.1080/20415990.2025.2458449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 01/22/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND The integration of immunotherapy alongside chemotherapy represents a crucial approach in the treatment of cancer. Herein we report the SN-38-indoximod conjugate nano-prodrug to address the difficulties encountered by individuals. In this prodrug, SN-38 is connected to indoximod through a specific disulfide linker, which enables the release of the components in response to the tumor microenvironment characterized by elevated levels of glutathione, thereby facilitating programmed chemoimmunotherapy. RESULTS SN-38-indoximod conjugate was synthesized and fabricated to nano-prodrug by reprecipitation method. It showed comparable anti-cancer activity against A549 cells than SN-38 (IC50 = 0.24 ± 0.01 µM) with IC50 value 0.32 ± 0.04 µM. It inhibited 90% A549 cell at very lower concentration (IC90 = 6.07 ± 0.41 µM) as compared with SN-38 (IC90 = 24.60 ± 1.24 µM) and mixture of SN-38: indoximod (1:1, IC90 >30 µM). The nano-prodrug showed better size distribution profile and dispersion stability contains nanoparticles in effective size range (80-160 nm) required for the EPR effect. CONCLUSION This research offers valuable insights into the advancement of conjugate nano-prodrugs exhibiting synergistic pharmacological effects, while also presenting novel opportunities for the design of prodrug molecules capable of releasing drugs in response to diverse triggers.
Collapse
Affiliation(s)
- Sanjay Kumar
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Yoshitaka Koseki
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Keita Tanita
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Aki Shibata
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Asuka Mizutani
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| | - Hitoshi Kasai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Sendai, Japan
| |
Collapse
|
10
|
Kiełbowski K, Bakinowska E, Becht R, Pawlik A. Metabolism of Tryptophan, Glutamine, and Asparagine in Cancer Immunotherapy-Synergism or Mechanism of Resistance? Metabolites 2025; 15:144. [PMID: 40137109 PMCID: PMC11944271 DOI: 10.3390/metabo15030144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025] Open
Abstract
Amino acids are crucial components of proteins, key molecules in cellular physiology and homeostasis. However, they are also involved in a variety of other mechanisms, such as energy homeostasis, nitrogen exchange, further synthesis of bioactive compounds, production of nucleotides, or activation of signaling pathways. Moreover, amino acids and their metabolites have immunoregulatory properties, significantly affecting the behavior of immune cells. Immunotherapy is one of the oncological treatment methods that improves cytotoxic properties of one's own immune system. Thus, enzymes catalyzing amino acid metabolism, together with metabolites themselves, can affect immune antitumor properties and responses to immunotherapy. In this review, we will discuss the involvement of tryptophan, glutamine, and asparagine metabolism in the behavior of immune cells targeted by immunotherapy and summarize results of the most recent investigations on the impact of amino acid metabolites on immunotherapy.
Collapse
Affiliation(s)
- Kajetan Kiełbowski
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Estera Bakinowska
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Rafał Becht
- Department of Clinical Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University, 71-252 Szczecin, Poland;
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| |
Collapse
|
11
|
Wang F, Du R, Shang Y. Biological function of d-tryptophan: a bibliometric analysis and review. Front Microbiol 2025; 15:1455540. [PMID: 39872820 PMCID: PMC11770058 DOI: 10.3389/fmicb.2024.1455540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 12/30/2024] [Indexed: 01/30/2025] Open
Abstract
Background d-Tryptophan is recognised for its unique physiological properties. In this study, we aimed to explore the dynamic trends and emerging topics in d-tryptophan research to offer fresh perspectives for future studies. Methods Employing bibliometric analysis, we examined the literature on d-tryptophan indexed in the Web of Science Core Collection from January 1987 to December 2023. The "Bibliometrix" R package and CiteSpace were utilised for data processing. Results Analyses of 865 publications revealed 2209 keywords, 4068 authors, 2094 institutions, and contributors from 302 regions. The USA was at the forefront of publications concerning d-tryptophan, but the European Journal of Pharmacology, Journal of Biological Chemistry, and Journal of Medicinal Chemistry were notable for their contributions, co-citations, and impact, respectively. This literature review reveals that since 1987, studies have developed from a focus on d-tryptophan metabolism to the exploration of its functions in organic and medicinal chemistry and food science. Recent findings highlight the potential of d-tryptophan as a non-nutritional sweetener and food preservative as well as its role in inhibiting the growth of bacterial biofilms. Additionally, its immunomodulatory properties are being investigated in relation to allergic diseases. Furthermore, d-tryptophan plays a role in the therapy of atherosclerosis, osteoporosis, tuberculosis, and cancer. Conclusion The results of bibliometric analysis highlight that future research should focus on the biological functions of d-tryptophan as a food preservative and its use in immunomodulation and drug development, providing strong guidance for future research.
Collapse
Affiliation(s)
- Fei Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Runyu Du
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Alessi I, Banton KL, J S, C ZM, CH P, RJ R, D BO. Exploring novel non-opioid pathways and therapeutics for pain modulation. Mol Pain 2025; 21:17448069251327840. [PMID: 40070108 PMCID: PMC11938896 DOI: 10.1177/17448069251327840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/26/2025] Open
Abstract
The opioid crisis has highlighted the urgent need for alternative pain management strategies. This review explores novel non-opioid targets and pathways involved in pain modulation, highlighting advancements in understanding and therapeutic potential. Pain, a multifaceted phenomenon with nociceptive, neuropathic, and inflammatory components, involves intricate molecular signaling cascades. Key pathways reviewed include voltage-gated sodium channels (Nav1.7, Nav1.8, Nav1.9), inflammasome complexes (NLRP3), the kynurenine pathway, prostaglandins, and bradykinin-mediated signaling. Emerging therapeutics such as selective Nav channel blockers, NLRP3 inhibitors, kynurenine pathway modulators, EP receptor antagonists, and bradykinin receptor antagonists offer promising alternatives to opioids. Despite challenges in clinical translation, these developments signal a paradigm shift in pain management, with precision-focused therapies poised to address unmet needs. This review emphasizes the importance of integrating molecular insights into the development of safer, more effective analgesics, setting the stage for transformative advancements in non-opioid pain relief.
Collapse
Affiliation(s)
- Isabella Alessi
- College of Osteopathic Medicine, Rocky Vista University, Centennial, CO, USA
| | - Kaysie L Banton
- Trauma Research, Swedish Medical Center, Englewood, CO, USA
- Trauma Services, Swedish Medical Center, Englewood, CO, USA
| | - Sliter J
- Trauma Services, Wesley Medical Center, Wichita, KS, USA
| | - Zaw-mon C
- Trauma Services, Lutheran Medical Center, Wheat Ridge, CO, USA
- Trauma Research, South Texas Health System, McAllen, TX, USA
| | - Palacio CH
- Trauma Research, South Texas Health System, McAllen, TX, USA
- Trauma Services, South Texas Health System McAllen, McAllen, TX, USA
| | - Ryznar RJ
- Department of Molecular Biology, Rocky Vista University, Parker, CO, USA
| | - Bar-Or D
- Trauma Research, Swedish Medical Center, Englewood, CO, USA
- Trauma Research, South Texas Health System, McAllen, TX, USA
- Department of Molecular Biology, Rocky Vista University, Parker, CO, USA
- Trauma Research, Wesley Medical Center, Wichita, KS, USA
| |
Collapse
|
13
|
Li H, Liu J, Wang J, Li Z, Yu J, Huang X, Wan B, Meng X, Zhang X. Improving the Anti-Tumor Effect of Indoleamine 2,3-Dioxygenase Inhibitor CY1-4 by CY1-4 Nano-Skeleton Drug Delivery System. J Funct Biomater 2024; 15:372. [PMID: 39728172 DOI: 10.3390/jfb15120372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
Background: CY1-4, 9-nitropyridine [2',3':4,5] pyrimido [1,2-α] indole -5,11- dione, is an indoleamine 2,3-dioxygenase (IDO) inhibitor and a poorly water-soluble substance. It is very important to increase the solubility of CY1-4 to improve its bioavailability and therapeutic effect. In this study, the mesoporous silica nano-skeleton carrier material Sylysia was selected as the carrier to load CY1-4, and then the CY1-4 nano-skeleton drug delivery system (MSNM@CY1-4) was prepared by coating the hydrophilic polymer material Hydroxypropyl methylcellulose (HPMC) and the lipid material Distearoylphosphatidyl-ethanolamine-poly(ethylene glycol)2000 (DSPE-PEG2000) to improve the anti-tumor effect of CY1-4. Methods: The solubility and dissolution of MSNM@CY1-4 were investigated, and its bioavailability, anti-tumor efficacy, IDO inhibitory ability and immune mechanism were evaluated in vivo. Results: CY1-4 was loaded in MSNM@CY1-4 in an amorphous form, and MSNM@CY1-4 could significantly improve the solubility (up to about 200 times) and dissolution rate of CY1-4. In vivo studies showed that the oral bioavailability of CY1-4 in 20 mg/kg MSNM@CY1-4 was about 23.9-fold more than that in 50 mg/kg CY1-4 suspension. In B16F10 tumor-bearing mice, MSNM@CY1-4 significantly inhibited tumor growth, prolonged survival time, significantly inhibited IDO activity in blood and tumor tissues, and reduced Tregs in tumor tissues and tumor-draining lymph nodes to improve anti-tumor efficacy. Conclusions: The nano-skeleton drug delivery system (MSNM@CY1-4) constructed in this study is a potential drug delivery platform for improving the anti-tumor effect of oral poorly water-soluble CY1-4.
Collapse
Affiliation(s)
- Hui Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Junwei Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingru Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhuoyue Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jianming Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xu Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Bingchuan Wan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiangbao Meng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xuan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
- Ningbo Institute of Marine Medicine, Peking University, Ningbo 315832, China
| |
Collapse
|
14
|
Hu Y, Liu W, Fang W, Dong Y, Zhang H, Luo Q. Tumor energy metabolism: implications for therapeutic targets. MOLECULAR BIOMEDICINE 2024; 5:63. [PMID: 39609317 PMCID: PMC11604893 DOI: 10.1186/s43556-024-00229-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/30/2024] Open
Abstract
Tumor energy metabolism plays a crucial role in the occurrence, progression, and drug resistance of tumors. The study of tumor energy metabolism has gradually become an emerging field of tumor treatment. Recent studies have shown that epigenetic regulation is closely linked to tumor energy metabolism, influencing the metabolic remodeling and biological traits of tumor cells. This review focuses on the primary pathways of tumor energy metabolism and explores therapeutic strategies to target these pathways. It covers key areas such as glycolysis, the Warburg effect, mitochondrial function, oxidative phosphorylation, and the metabolic adaptability of tumors. Additionally, this article examines the role of the epigenetic regulator SWI/SNF complex in tumor metabolism, specifically its interactions with glucose, lipids, and amino acids. Summarizing therapeutic strategies aimed at these metabolic pathways, including inhibitors of glycolysis, mitochondrial-targeted drugs, exploitation of metabolic vulnerabilities, and recent developments related to SWI/SNF complexes as potential targets. The clinical significance, challenges, and future directions of tumor metabolism research are discussed, including strategies to overcome drug resistance, the potential of combination therapy, and the application of new technologies.
Collapse
Affiliation(s)
- Youwu Hu
- The Public Experimental Center of Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou, 563003, China
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wanqing Liu
- The Public Experimental Center of Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou, 563003, China
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - WanDi Fang
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yudi Dong
- The Public Experimental Center of Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou, 563003, China
| | - Hong Zhang
- Department of Pathology, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qing Luo
- The Public Experimental Center of Medicine, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, Guizhou, 563003, China.
- Guizhou Provincial Key Laboratory of Cell Engineering, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
15
|
Meléndez-Vázquez NM, Gomez-Manzano C, Godoy-Vitorino F. Oncolytic Virotherapies and Adjuvant Gut Microbiome Therapeutics to Enhance Efficacy Against Malignant Gliomas. Viruses 2024; 16:1775. [PMID: 39599889 PMCID: PMC11599061 DOI: 10.3390/v16111775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Glioblastoma (GBM) is the most prevalent malignant brain tumor. Current standard-of-care treatments offer limited benefits for patient survival. Virotherapy is emerging as a novel strategy to use oncolytic viruses (OVs) for the treatment of GBM. These engineered and non-engineered viruses infect and lyse cancer cells, causing tumor destruction without harming healthy cells. Recent advances in genetic modifications to OVs have helped improve their targeting capabilities and introduce therapeutic genes, broadening the therapeutic window and minimizing potential side effects. The efficacy of oncolytic virotherapy can be enhanced by combining it with other treatments such as immunotherapy, chemotherapy, or radiation. Recent studies suggest that manipulating the gut microbiome to enhance immune responses helps improve the therapeutic efficacy of the OVs. This narrative review intends to explore OVs and their role against solid tumors, especially GBM while emphasizing the latest technologies used to enhance and improve its therapeutic and clinical responses.
Collapse
Affiliation(s)
- Natalie M. Meléndez-Vázquez
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00918, USA;
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, University of Puerto Rico-Medical Sciences Campus, San Juan, PR 00918, USA;
| |
Collapse
|
16
|
Zhang J, Chen M, Yang Y, Liu Z, Guo W, Xiang P, Zeng Z, Wang D, Xiong W. Amino acid metabolic reprogramming in the tumor microenvironment and its implication for cancer therapy. J Cell Physiol 2024; 239:e31349. [PMID: 38946173 DOI: 10.1002/jcp.31349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/08/2024] [Accepted: 06/14/2024] [Indexed: 07/02/2024]
Abstract
Amino acids are essential building blocks for proteins, crucial energy sources for cell survival, and key signaling molecules supporting the resistant growth of tumor cells. In tumor cells, amino acid metabolic reprogramming is characterized by the enhanced uptake of amino acids as well as their aberrant synthesis, breakdown, and transport, leading to immune evasion and malignant progression of tumor cells. This article reviews the altered amino acid metabolism in tumor cells and its impact on tumor microenvironment, and also provides an overview of the current clinical applications of amino acid metabolism. Innovative drugs targeting amino acid metabolism hold great promise for precision and personalized cancer therapy.
Collapse
Affiliation(s)
- Jiarong Zhang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Mingjian Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Yuxin Yang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Ziqi Liu
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wanni Guo
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Pingjuan Xiang
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Zhaoyang Zeng
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Dan Wang
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| | - Wei Xiong
- NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, China
| |
Collapse
|
17
|
Yan J, Chen D, Ye Z, Zhu X, Li X, Jiao H, Duan M, Zhang C, Cheng J, Xu L, Li H, Yan D. Molecular mechanisms and therapeutic significance of Tryptophan Metabolism and signaling in cancer. Mol Cancer 2024; 23:241. [PMID: 39472902 PMCID: PMC11523861 DOI: 10.1186/s12943-024-02164-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/24/2024] [Indexed: 11/02/2024] Open
Abstract
Tryptophan (Trp) metabolism involves three primary pathways: the kynurenine (Kyn) pathway (KP), the 5-hydroxytryptamine (serotonin, 5-HT) pathway, and the indole pathway. Under normal physiological conditions, Trp metabolism plays crucial roles in regulating inflammation, immunity, and neuronal function. Key rate-limiting enzymes such as indoleamine-2,3-dioxygenase (IDO), Trp-2,3-dioxygenase (TDO), and kynurenine monooxygenase (KMO) drive these metabolic processes. Imbalances in Trp metabolism are linked to various cancers and often correlate with poor prognosis and adverse clinical characteristics. Dysregulated Trp metabolism fosters tumor growth and immune evasion primarily by creating an immunosuppressive tumor microenvironment (TME). Activation of the KP results in the production of immunosuppressive metabolites like Kyn, which modulate immune responses and promote oncogenesis mainly through interaction with the aryl hydrocarbon receptor (AHR). Targeting Trp metabolism therapeutically has shown significant potential, especially with the development of small-molecule inhibitors for IDO1, TDO, and other key enzymes. These inhibitors disrupt the immunosuppressive signals within the TME, potentially restoring effective anti-tumor immune responses. Recently, IDO1 inhibitors have been tested in clinical trials, showing the potential to enhance the effects of existing cancer therapies. However, mixed results in later-stage trials underscore the need for a deeper understanding of Trp metabolism and its complex role in cancer. Recent advancements have also explored combining Trp metabolism inhibitors with other treatments, such as immune checkpoint inhibitors, chemotherapy, and radiotherapy, to enhance therapeutic efficacy and overcome resistance mechanisms. This review summarizes the current understanding of Trp metabolism and signaling in cancer, detailing the oncogenic mechanisms and clinical significance of dysregulated Trp metabolism. Additionally, it provides insights into the challenges in developing Trp-targeted therapies and future research directions aimed at optimizing these therapeutic strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Zi Ye
- Department of Scientific Research, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xuqiang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Xueyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Henan Jiao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengjiao Duan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Chaoli Zhang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan, Zhengzhou, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Hongjiang Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
18
|
Li L, Zhang T, Xiao M, Lu Y, Gao L. Brain macrophage senescence in glioma. Semin Cancer Biol 2024; 104-105:46-60. [PMID: 39098625 DOI: 10.1016/j.semcancer.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/20/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Gliomas are a diverse group of primary central nervous system neoplasms with no curative therapies available. Brain macrophages comprise microglia in the brain parenchyma, border-associated macrophages in the meningeal-choroid plexus-perivascular space and monocyte-derived macrophages infiltrating the brain. With the great improvement of our recognition of brain macrophages, diverse macrophage populations have been found in the context of glioma, which exhibit functional and phenotypic heterogeneity. We have long thought that brain macrophage senescence is detrimental, manifested by specialized forms of persistent cell cycle arrest and chronic low-grade inflammation. Persistent senescence of macrophages may result in immune dysfunction, potentially contributing to glioma initiation and development. Given the crucial roles played by brain macrophages in glioma, we unravel how brain macrophages undergo reprogramming and their contribution to glioma. We outline general molecular alterations and specific biomarkers in senescent brain macrophages, as well as functional changes (such as metabolism, autophagy, phagocytosis, antigen presentation, and infiltration and recruitment). In addition, recent advances in genetic regulation and mechanisms linked to senescent brain macrophages are discussed. In particular, this review emphasizes the contribution of senescent brain macrophages to glioma, which may drive translational efforts to utilize brain macrophages as a prognostic marker or/and treatment target in glioma. An in-depth comprehending of how brain macrophage senescence functionally influences the tumor microenvironment will be key to our development of innovative therapeutics for glioma.
Collapse
Affiliation(s)
- Lu Li
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Tianhe Zhang
- Department of Neurosurgery, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, China
| | - Meiling Xiao
- Department of Rehabilitation, The Central Hospital of Shenyang Medical College, Shenyang, Liaoning 110024, China
| | - Yu Lu
- Rehabilitation Medicine Department, The People's Hospital of China Medical University, The People's Hospital of Liaoning Province, Shenyang, Liaoning 110016, China.
| | - Lin Gao
- Department of Rehabilitation, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
19
|
Rogovskii V. Tumor-produced immune regulatory factors as a therapeutic target in cancer treatment. Front Immunol 2024; 15:1416458. [PMID: 39206193 PMCID: PMC11349530 DOI: 10.3389/fimmu.2024.1416458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Vladimir Rogovskii
- Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
20
|
Bonada M, Pittarello M, De Fazio E, Gans A, Alimonti P, Slika H, Legnani F, Di Meco F, Tyler B. Pediatric Hemispheric High-Grade Gliomas and H3.3-G34 Mutation: A Review of the Literature on Biological Features and New Therapeutic Strategies. Genes (Basel) 2024; 15:1038. [PMID: 39202398 PMCID: PMC11353413 DOI: 10.3390/genes15081038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Pediatric high-grade glioma (pHGG) encompasses a wide range of gliomas with different genomic, epigenomic, and transcriptomic features. Almost 50% of pHGGs present a mutation in genes coding for histone 3, including the subtype harboring the H3.3-G34 mutation. In this context, histone mutations are frequently associated with mutations in TP53 and ATRX, along with PDGFRA and NOTCH2NL amplifications. Moreover, the H3.3-G34 histone mutation induces epigenetic changes in immune-related genes and exerts modulatory functions on the microenvironment. Also, the functionality of the blood-brain barrier (BBB) has an impact on treatment response. The prognosis remains poor with conventional treatments, thus eliciting the investigation of additional and alternative therapies. Promising molecular targets include PDGFRA amplification, BRAF mutation, EGFR amplification, NF1 loss, and IDH mutation. Considering that pHGGs harboring the H3.3-G34R mutation appear to be more susceptible to immunotherapies (ITs), different options have been recently explored, including immune checkpoint inhibitors, antibody mediated IT, and Car-T cells. This review aims to summarize the knowledge concerning cancer biology and cancer-immune cell interaction in this set of pediatric gliomas, with a focus on possible therapeutic options.
Collapse
Affiliation(s)
- Marta Bonada
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
| | - Matilde Pittarello
- Department of Biomedical Sciences, Humanitas University, 20072 Milan, Italy;
| | - Emerson De Fazio
- Department of Medicine, Vita-Salute San Raffaele University School of Medicine, 20132 Milan, Italy;
| | - Alessandro Gans
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
- ASST Ovest Milanese, Neurology and Stroke Unit, Neuroscience Department, 20025 Legnano, Italy
| | - Paolo Alimonti
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02120, USA;
| | - Hasan Slika
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Federico Legnani
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
| | - Francesco Di Meco
- Department of Neurosurgery, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133 Milan, Italy; (M.B.); (F.L.); (F.D.M.)
- Department of Oncology and Hemato-Oncology, University of Milan School of Medicine, Via Rudini 8, 20122 Milan, Italy;
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA;
| |
Collapse
|
21
|
Lin C, Smith C, Rutka J. Current immunotherapeutic approaches to diffuse intrinsic pontine glioma. Front Genet 2024; 15:1349612. [PMID: 38774284 PMCID: PMC11106442 DOI: 10.3389/fgene.2024.1349612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/22/2024] [Indexed: 05/24/2024] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is an aggressive brain tumour that occurs in the pons of the brainstem and accounts for over 80% of all brainstem gliomas. The median age at diagnosis is 6-7 years old, with less than 10% overall survival 2 years after diagnosis and less than 1% after 5 years. DIPGs are surgically inaccessible, and radiation therapy provides only transient benefit, with death ensuing from relentless local tumour infiltration. DIPGs are now the leading cause of brain tumour deaths in children, with a societal cancer burden in years of life lost (YLL) of more than 67 per individual, versus approximately 14 and 16 YLL for lung and breast cancer respectively. More than 95 clinical drug trials have been conducted on children with DIPGs, and all have failed to improve survival. No single or combination chemotherapeutic strategy has been successful to date because of our inability to identify targeted drugs for this disease and to deliver these drugs across an intact blood-brain barrier (BBB). Accordingly, there has been an increased focus on immunotherapy research in DIPG, with explorations into treatments such as chimeric antigen receptor T (CAR-T) cells, immune checkpoint blockades, cancer vaccines, and autologous cell transfer therapy. Here, we review the most recent advances in identifying genetic factors influencing the development of immunotherapy for DIPG. Additionally, we explore emerging technologies such as Magnetic Resonance-guided Focused Ultrasound (MRgFUS) in potential combinatorial approaches to treat DIPG.
Collapse
Affiliation(s)
- Catherine Lin
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Christian Smith
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
| | - James Rutka
- Cell Biology Research Program, The Hospital for Sick Children, Toronto, ON, Canada
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, The Hospital for Sick Children, Toronto, ON, Canada
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
22
|
Holmberg KO, Borgenvik A, Zhao M, Giraud G, Swartling FJ. Drivers Underlying Metastasis and Relapse in Medulloblastoma and Targeting Strategies. Cancers (Basel) 2024; 16:1752. [PMID: 38730706 PMCID: PMC11083189 DOI: 10.3390/cancers16091752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/12/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Medulloblastomas comprise a molecularly diverse set of malignant pediatric brain tumors in which patients are stratified according to different prognostic risk groups that span from very good to very poor. Metastasis at diagnosis is most often a marker of poor prognosis and the relapse incidence is higher in these children. Medulloblastoma relapse is almost always fatal and recurring cells have, apart from resistance to standard of care, acquired genetic and epigenetic changes that correlate with an increased dormancy state, cell state reprogramming and immune escape. Here, we review means to carefully study metastasis and relapse in preclinical models, in light of recently described molecular subgroups. We will exemplify how therapy resistance develops at the cellular level, in a specific niche or from therapy-induced secondary mutations. We further describe underlying molecular mechanisms on how tumors acquire the ability to promote leptomeningeal dissemination and discuss how they can establish therapy-resistant cell clones. Finally, we describe some of the ongoing clinical trials of high-risk medulloblastoma and suggest or discuss more individualized treatments that could be of benefit to specific subgroups.
Collapse
Affiliation(s)
- Karl O. Holmberg
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (K.O.H.); (M.Z.); (G.G.)
| | - Anna Borgenvik
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Miao Zhao
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (K.O.H.); (M.Z.); (G.G.)
| | - Géraldine Giraud
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (K.O.H.); (M.Z.); (G.G.)
- Department of Women and Child Health, Uppsala University, 75124 Uppsala, Sweden
- Department of Pediatric Hematology and Oncology, Uppsala University Children’s Hospital, 75185 Uppsala, Sweden
| | - Fredrik J. Swartling
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (K.O.H.); (M.Z.); (G.G.)
| |
Collapse
|
23
|
Lasser SA, Ozbay Kurt FG, Arkhypov I, Utikal J, Umansky V. Myeloid-derived suppressor cells in cancer and cancer therapy. Nat Rev Clin Oncol 2024; 21:147-164. [PMID: 38191922 DOI: 10.1038/s41571-023-00846-y] [Citation(s) in RCA: 126] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2023] [Indexed: 01/10/2024]
Abstract
Anticancer agents continue to dominate the list of newly approved drugs, approximately half of which are immunotherapies. This trend illustrates the considerable promise of cancer treatments that modulate the immune system. However, the immune system is complex and dynamic, and can have both tumour-suppressive and tumour-promoting effects. Understanding the full range of immune modulation in cancer is crucial to identifying more effective treatment strategies. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of myeloid cells that develop in association with chronic inflammation, which is a hallmark of cancer. Indeed, MDSCs accumulate in the tumour microenvironment, where they strongly inhibit anticancer functions of T cells and natural killer cells and exert a variety of other tumour-promoting effects. Emerging evidence indicates that MDSCs also contribute to resistance to cancer treatments, particularly immunotherapies. Conversely, treatment approaches designed to eliminate cancer cells can have important additional effects on MDSC function, which can be either positive or negative. In this Review, we discuss the interplay between MDSCs and various other cell types found in tumours as well as the mechanisms by which MDSCs promote tumour progression. We also discuss the relevance and implications of MDSCs for cancer therapy.
Collapse
Affiliation(s)
- Samantha A Lasser
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Feyza G Ozbay Kurt
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Ihor Arkhypov
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Jochen Utikal
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Viktor Umansky
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany.
- Skin Cancer Unit, German Cancer Research Center (Deutsches Krebsforschungszentrum (DKFZ)), Heidelberg, Germany.
- DKFZ-Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany.
| |
Collapse
|
24
|
Frumento D, Grossi G, Falesiedi M, Musumeci F, Carbone A, Schenone S. Small Molecule Tyrosine Kinase Inhibitors (TKIs) for Glioblastoma Treatment. Int J Mol Sci 2024; 25:1398. [PMID: 38338677 PMCID: PMC10855061 DOI: 10.3390/ijms25031398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
In the last decade, many small molecules, usually characterized by heterocyclic scaffolds, have been designed and synthesized as tyrosine kinase inhibitors (TKIs). Among them, several compounds have been tested at preclinical and clinical levels to treat glioblastoma multiforme (GBM). GBM is the most common and aggressive type of cancer originating in the brain and has an unfavorable prognosis, with a median survival of 15-16 months and a 5-year survival rate of 5%. Despite recent advances in treating GBM, it represents an incurable disease associated with treatment resistance and high recurrence rates. For these reasons, there is an urgent need for the development of new pharmacological agents to fight this malignancy. In this review, we reported the compounds published in the last five years, which showed promising activity in GBM preclinical models acting as TKIs. We grouped the compounds based on the targeted kinase: first, we reported receptor TKIs and then, cytoplasmic and peculiar kinase inhibitors. For each small molecule, we included the chemical structure, and we schematized the interaction with the target for some representative compounds with the aim of elucidating the mechanism of action. Finally, we cited the most relevant clinical trials.
Collapse
Affiliation(s)
| | | | | | - Francesca Musumeci
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (D.F.); (G.G.); (M.F.); (S.S.)
| | - Anna Carbone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (D.F.); (G.G.); (M.F.); (S.S.)
| | | |
Collapse
|