1
|
Shende S, Rathored J, Budhbaware T. Role of metabolic transformation in cancer immunotherapy resistance: molecular mechanisms and therapeutic implications. Discov Oncol 2025; 16:453. [PMID: 40175681 PMCID: PMC11965038 DOI: 10.1007/s12672-025-02238-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Immunotherapy in the treatment of cancer, with immune inhibitors helps in many cancer types. Many patients still encounter resistance to these treatments, though. This resistance is mediated by metabolic changes in the tumour microenvironment and cancer cells. The development of novel treatments to overcome resistance and boost immunotherapy's effectiveness depends on these metabolic changes. OBJECTIVE This review concentrates on the molecular mechanisms through which metabolic transformation contributes to cancer immunotherapy resistance. Additionally, research therapeutic approaches that target metabolic pathways to enhance immunotherapy for resistance. METHODS We used databases available on PubMed, Scopus, and Web of Science to perform a thorough review of peer-reviewed literature. focusing on the tumor microenvironment, immunotherapy resistance mechanisms, and cancer metabolism. The study of metabolic pathways covers oxidative phosphorylation, glycolysis, lipid metabolism, and amino acid metabolism. RESULTS An immunosuppressive tumour microenvironment is produced by metabolic changes in cancer cells, such as dysregulated lipid metabolism, enhanced glutaminolysis, and increased glycolysis (Warburg effect). Myeloid-derived suppressor cells and regulatory T cells are promoted, immune responses are suppressed, and T cell activity is impaired when lactate and other metabolites build up. changes in the metabolism of amino acids in the pathways for arginine and tryptophan, which are nutrients crucial for immune function. By enhancing their function in the tumour microenvironment, these metabolic alterations aid in resistance to immune checkpoint inhibitors. CONCLUSION Metabolic change plays a key role in cancer immunotherapy resistance. Gaining knowledge of metabolic processes can help develop efficient treatments that improve immunotherapy's effectiveness. In order to determine the best targets for therapeutic intervention, future studies should concentrate on patient-specific metabolic profiling.
Collapse
Affiliation(s)
- Sandesh Shende
- Central Research Laboratory and Molecular Diagnostics, School of Allied Health Sciences, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, 442001, Maharashtra, India
| | - Jaishriram Rathored
- Central Research Laboratory and Molecular Diagnostics, School of Allied Health Sciences, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, 442001, Maharashtra, India.
| | - Tanushree Budhbaware
- Central Research Laboratory and Molecular Diagnostics, School of Allied Health Sciences, Datta Meghe Institute of Higher Education and Research, Sawangi (Meghe), Wardha, 442001, Maharashtra, India
| |
Collapse
|
2
|
Chen W, Wang X, Li X, Wang W, Huang Y, Qin Y, Liu P, Wu K, Li B, He Y, Zeng S, Yi L, Wang L, Zhao M, Ding H, Fan S, Li Z, Chen J. Foot-and-mouth disease virus activates glycolysis and hijacks HK2 to inhibit innate immunity and promote viral replication. Vet Res 2025; 56:71. [PMID: 40170113 PMCID: PMC11963632 DOI: 10.1186/s13567-025-01497-w] [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: 10/22/2024] [Accepted: 01/23/2025] [Indexed: 04/03/2025] Open
Abstract
Foot-and-mouth disease (FMD) severely restricts the healthy development of global animal husbandry, and the unclear pathogenic mechanism of FMD virus (FMDV) leads to difficulty in preventing and purifying FMD. Glycolytic remodelling is considered one of the hallmarks of viral infection, providing energy and precursors for viral assembly and replication. In this work, the interaction and mechanism between FMDV and glycolysis were explored from the perspective of immune metabolism. We found that FMDV infection increased the extracellular acidification rate, lactic acid accumulation, and HK2 level. In addition, during FMDV infection, HK2 enhances glycolytic activity and mediates autophagic degradation of IRF3/7 to antagonize the innate immune response, thereby promoting viral replication. Our findings provide evidence that FMDV is closely correlated with host metabolism, increasing the understanding that glycolysis and HK2 facilitate virus infection, and provide new ideas for further elucidating the pathogenic mechanism of FMDV.
Collapse
Affiliation(s)
- Wenxian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xinyan Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaowen Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Weijun Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yaoyao Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuwei Qin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Pengfei Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Keke Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Bingke Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yintao He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Sen Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Lin Yi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | | | - Mingqiu Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Hongxing Ding
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Shuangqi Fan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhaoyao Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
- Wen's Foodstuffs Group Co., Ltd., Yunfu, Xinxing, China.
| | - Jinding Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
3
|
Xiao J, Liu T, Zeng F, Zhang J. New insights into T cell metabolism in liver cancer: from mechanism to therapy. Cell Death Discov 2025; 11:118. [PMID: 40122853 PMCID: PMC11930970 DOI: 10.1038/s41420-025-02397-w] [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: 07/08/2024] [Revised: 02/19/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025] Open
Abstract
Liver cancer is the sixth most common cancer worldwide and the third most common cause of cancer mortality. The development and progression of liver cancer and metastases is a multifaceted process involving numerous metabolic pathways. T cells have a protective role in the defense against cancer, and manipulating metabolic pathways in T cells can alter their antitumor activity. Furthermore, Liver cancer and T cell nutrition competition lead to T cell dysfunction through various molecular mechanisms. Some nanomaterials and drugs can improve T cell metabolism and promote the anti-liver cancer function of T cells. This review discusses the current literature regarding metabolic changes in liver cancer, the role of T cells in liver cancer, T cell metabolism in liver cancer, and targeted T cell metabolism therapy for liver cancer. The promise and challenges of studying target T cell metabolism for treating liver cancer are also addressed. Targeting T cell metabolism is a promising approach for treating liver cancer.
Collapse
Affiliation(s)
- Jie Xiao
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China
| | - Ting Liu
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
- School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Fanxin Zeng
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichua, China.
| | - Jinhua Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, China.
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China.
| |
Collapse
|
4
|
Qiu L, Qiu X, Yang X. PLOD3 as a novel oncogene in prognostic and immune infiltration risk model based on multi-machine learning in cervical cancer. Discov Oncol 2025; 16:294. [PMID: 40067513 PMCID: PMC11896957 DOI: 10.1007/s12672-025-02031-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
Cervical carcinoma (CC) remains a significant global health issue despite advancements in screening and treatment. To improve prognostic accuracy and therapeutic strategies, we developed a multi-machine learning prognostic model based on metabolic-associated genes. This study integrated genomic, transcriptomic, and spatial data from multiple databases to identify key metabolic genes with a causal relationship to CC. We identified 112 key metabolic genes, which were used to construct and validate a prognostic model through various machine learning algorithms. GO and KEGG enrichment analysis revealed the MAPK cascade plays a crucial role in metabolic processes. To pinpoint key metabolic genes, we constructed WGCNA and extracted 337 key genes. Supervised principal component analysis and random survival forests were incorporated into the final model, which showed strong predictive ability in classifying patients. Furthermore, the model demonstrated notable variations in immune cell infiltration among risk categories, which shown regulatory T cells may be involved in immune suppression, and natural killer cells might have a limited effect in tumor clearance. Spatial transcriptomics and single-cell analyses further validated the model, uncovering tumor heterogeneity and distinct intercellular communication patterns associated with different risk levels. The functional experiment results indicated that down expression of PLOD3 could suppress the proliferation of CC cell. In this study, offer a precision medicine methods for predicting patient outcomes as well as fresh insights into the metabolic foundations, which may contribute to the prognosis and immunotherapy of CC. Additionally, we discovered PLOD3 to be a novel oncogene in CC. These findings imply that this model may be applied to assess prognostic risk and identify potential therapeutic targets for CC patients.
Collapse
Affiliation(s)
- Lingling Qiu
- Department of Reproductive Medicine, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| | - Xiuchai Qiu
- Department of Public Health, Jinjiang City Hospital Jinnan Branch, Fujian Province, China
| | - Xiaoyi Yang
- Department of Oncology, Shanghai General Hospital, , Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| |
Collapse
|
5
|
Suri C, Pande B, Suhasini Sahithi L, Swarnkar S, Khelkar T, Verma HK. Metabolic crossroads: unravelling immune cell dynamics in gastrointestinal cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:7. [PMID: 40051496 PMCID: PMC11883236 DOI: 10.20517/cdr.2024.164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/09/2025]
Abstract
Metabolic reprogramming within the tumor microenvironment (TME) plays a critical role in driving drug resistance in gastrointestinal cancers (GI), particularly through the pathways of fatty acid oxidation and glycolysis. Cancer cells often rewire their metabolism to sustain growth and reshape the TME, creating conditions such as nutrient depletion, hypoxia, and acidity that impair antitumor immune responses. Immune cells within the TME also undergo metabolic alterations, frequently adopting immunosuppressive phenotypes that promote tumor progression and reduce the efficacy of therapies. The competition for essential nutrients, particularly glucose, between cancer and immune cells compromises the antitumor functions of effector immune cells, such as T cells. Additionally, metabolic by-products like lactate and kynurenine further suppress immune activity and promote immunosuppressive populations, including regulatory T cells and M2 macrophages. Targeting metabolic pathways such as fatty acid oxidation and glycolysis presents new opportunities to overcome drug resistance and improve therapeutic outcomes in GI cancers. Modulating these key pathways has the potential to reinvigorate exhausted immune cells, shift immunosuppressive cells toward antitumor phenotypes, and enhance the effectiveness of immunotherapies and other treatments. Future strategies will require continued research into TME metabolism, the development of novel metabolic inhibitors, and clinical trials evaluating combination therapies. Identifying and validating metabolic biomarkers will also be crucial for patient stratification and treatment monitoring. Insights into metabolic reprogramming in GI cancers may have broader implications across multiple cancer types, offering new avenues for improving cancer treatment.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton AB T6G 1Z2, Canada
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India
| | | | | | - Tuneer Khelkar
- Department of Botany and Biotechnology, Govt. Kaktiya P G College, Jagdalpur 494001, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lung Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Munich 85764, Germany
| |
Collapse
|
6
|
Hindes MT, McElligott AM, Best OG, Ward MP, Selemidis S, Miles MA, Nturubika BD, Gregory PA, Anderson PH, Logan JM, Butler LM, Waugh DJ, O'Leary JJ, Hickey SM, Thurgood LA, Brooks DA. Metabolic reprogramming, malignant transformation and metastasis: Lessons from chronic lymphocytic leukaemia and prostate cancer. Cancer Lett 2025; 611:217441. [PMID: 39755364 DOI: 10.1016/j.canlet.2025.217441] [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: 10/17/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
Metabolic reprogramming is a hallmark of cancer, crucial for malignant transformation and metastasis. Chronic lymphocytic leukaemia (CLL) and prostate cancer exhibit similar metabolic adaptations, particularly in glucose and lipid metabolism. Understanding this metabolic plasticity is crucial for identifying mechanisms contributing to metastasis. This review considers glucose and lipid metabolism in CLL and prostate cancer, exploring their roles in healthy and malignant states and during disease progression. In CLL, lipid metabolism supports cell survival and migration, with aggressive disease characterised by increased fatty acid oxidation and altered sphingolipids. Richter's transformation and aggressive lymphoma, however, exhibit a metabolic shift towards increased glycolysis. Similarly, prostate cell metabolism is unique, relying on citrate production in the healthy state and undergoing metabolic reprogramming during malignant transformation. Early-stage prostate cancer cells increase lipid synthesis and uptake, and decrease glycolysis, whereas metastatic cells re-adopt glucose metabolism, likely driven by interactions with the tumour microenvironment. Genetic drivers including TP53 and ATM mutations connect metabolic alterations to disease severity in these two malignancies. The bone microenvironment supports the metabolic demands of these malignancies, serving as an initiation niche for CLL and a homing site for prostate cancer metastases. By comparing these malignancies, this review underscores the importance of metabolic plasticity in cancer progression and highlights how CLL and prostate cancer may be models of circulating and solid tumours more broadly. The metabolic phenotypes throughout cancer cell transformation and metastasis, and the microenvironment in which these processes occur, present opportunities for interventions that could disrupt metastatic processes and improve patient outcomes.
Collapse
Affiliation(s)
- Madison T Hindes
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia.
| | - Anthony M McElligott
- Discipline of Haematology, School of Medicine, Trinity Translational Medicine Institute, St. James's Hospital and Trinity College, Dublin, Ireland
| | - Oliver G Best
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Mark P Ward
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Stavros Selemidis
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Mark A Miles
- Centre for Respiratory Science and Health, School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University, Bundoora, Victoria, Australia
| | - Bukuru D Nturubika
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - Paul H Anderson
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lisa M Butler
- South Australian ImmunoGENomics Cancer Institute and Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, Australia; Solid Tumour Program, Precision Cancer Medicine theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - David J Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia
| | - Lauren A Thurgood
- Molecular Medicine and Genetics, College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, Australia
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, Adelaide, Australia; Department of Histopathology, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
7
|
Hanahan D, Michielin O, Pittet MJ. Convergent inducers and effectors of T cell paralysis in the tumour microenvironment. Nat Rev Cancer 2025; 25:41-58. [PMID: 39448877 DOI: 10.1038/s41568-024-00761-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
Tumorigenesis embodies the formation of a heterotypic tumour microenvironment (TME) that, among its many functions, enables the evasion of T cell-mediated immune responses. Remarkably, most TME cell types, including cancer cells, fibroblasts, myeloid cells, vascular endothelial cells and pericytes, can be stimulated to deploy immunoregulatory programmes. These programmes involve regulatory inducers (signals-in) and functional effectors (signals-out) that impair CD8+ and CD4+ T cell activity through cytokines, growth factors, immune checkpoints and metabolites. Some signals target specific cell types, whereas others, such as transforming growth factor-β (TGFβ) and prostaglandin E2 (PGE2), exert broad, pleiotropic effects; as signals-in, they trigger immunosuppressive programmes in most TME cell types, and as signals-out, they directly inhibit T cells and also modulate other cells to reinforce immunosuppression. This functional diversity and redundancy pose a challenge for therapeutic targeting of the immune-evasive TME. Fundamentally, the commonality of regulatory programmes aimed at abrogating T cell activity, along with paracrine signalling between cells of the TME, suggests that many normal cell types are hard-wired with latent functions that can be triggered to prevent inappropriate immune attack. This intrinsic capability is evidently co-opted throughout the TME, enabling tumours to evade immune destruction.
Collapse
Affiliation(s)
- Douglas Hanahan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), Lausanne, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.
| | - Olivier Michielin
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Medicine, University of Geneva (UNIGE), Geneva, Switzerland
| | - Mikael J Pittet
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Department of Pathology and Immunology, University of Geneva (UNIGE), Geneva, Switzerland
| |
Collapse
|
8
|
Aden D, Sureka N, Zaheer S, Chaurasia JK, Zaheer S. Metabolic Reprogramming in Cancer: Implications for Immunosuppressive Microenvironment. Immunology 2025; 174:30-72. [PMID: 39462179 DOI: 10.1111/imm.13871] [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: 05/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
Cancer is a complex and heterogeneous disease characterised by uncontrolled cell growth and proliferation. One hallmark of cancer cells is their ability to undergo metabolic reprogramming, which allows them to sustain their rapid growth and survival. This metabolic reprogramming creates an immunosuppressive microenvironment that facilitates tumour progression and evasion of the immune system. In this article, we review the mechanisms underlying metabolic reprogramming in cancer cells and discuss how these metabolic alterations contribute to the establishment of an immunosuppressive microenvironment. We also explore potential therapeutic strategies targeting metabolic vulnerabilities in cancer cells to enhance immune-mediated anti-tumour responses. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT02044861, NCT03163667, NCT04265534, NCT02071927, NCT02903914, NCT03314935, NCT03361228, NCT03048500, NCT03311308, NCT03800602, NCT04414540, NCT02771626, NCT03994744, NCT03229278, NCT04899921.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
9
|
Singer M, Hamdy R, Ghonaim JH, Husseiny MI. Metabolic Imbalance in Immune Cells in Relation to Metabolic Disorders, Cancer, and Infections. METABOLIC DYNAMICS IN HOST-MICROBE INTERACTION 2025:187-218. [DOI: 10.1007/978-981-96-1305-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
|
10
|
Pavelescu LA, Enache RM, Roşu OA, Profir M, Creţoiu SM, Gaspar BS. Predictive Biomarkers and Resistance Mechanisms of Checkpoint Inhibitors in Malignant Solid Tumors. Int J Mol Sci 2024; 25:9659. [PMID: 39273605 PMCID: PMC11395316 DOI: 10.3390/ijms25179659] [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: 08/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), defective deoxyribonucleic acid (DNA) mismatch repair (dMMR), microsatellite instability (MSI), and the tumor microenvironment (TME). Over the past decade, several types of ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, anti-programmed cell death 1 (PD-1) antibodies, anti-programmed cell death ligand 1 (PD-L1) antibodies, and anti-lymphocyte activation gene-3 (LAG-3) antibodies have been studied and approved by the Food and Drug Administration (FDA), with ongoing research on others. Recent studies highlight the critical role of the gut microbiome in influencing a positive therapeutic response to ICIs, emphasizing the importance of modeling factors that can maintain a healthy microbiome. However, resistance mechanisms can emerge, such as increased expression of alternative immune checkpoints, T-cell immunoglobulin (Ig), mucin domain-containing protein 3 (TIM-3), LAG-3, impaired antigen presentation, and alterations in the TME. This review aims to synthesize the data regarding the interactions between microbiota and immunotherapy (IT). Understanding these mechanisms is essential for optimizing ICI therapy and developing effective combination strategies.
Collapse
Affiliation(s)
- Luciana Alexandra Pavelescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Robert Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
11
|
Xi Y, Liu R, Zhang X, Guo Q, Zhang X, Yang Z, Zheng H, Song Q, Hua B. A Bibliometric Analysis of Metabolic Reprogramming in the Tumor Microenvironment From 2003 to 2022. Cancer Rep (Hoboken) 2024; 7:e2146. [PMID: 39158178 PMCID: PMC11331499 DOI: 10.1002/cnr2.2146] [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: 11/01/2023] [Revised: 06/23/2024] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Despite considerable progress in cancer immunotherapy, it is not available for many patients. Resistance to immune checkpoint blockers arises from the intricate interactions between cancer and its microenvironment. Metabolic reprogramming in tumor and immune cells in the tumor microenvironment (TME) influences anti-tumor immune responses by remodeling the immune microenvironment. Metabolic reprogramming has emerged as an important hallmark of tumorigenesis. However, few studies have focused on the TME and metabolic reprogramming. Therefore, we aimed to explore the current research status and popular topics in TME-related metabolic reprogramming over a 20 years using a bibliometric approach. METHODS Studies focusing on metabolic reprogramming and TME were searched using the Web of Science Core Collection database. Bibliometric and visual analyses of the articles and reviews were performed using Bibliometrix, VOSviewer, and CiteSpace. RESULTS In total, 4726 articles published between 2003 and 2022 were selected. The number of publications and citations has increased annually. Cooperation network analysis indicated that the United States holds the foremost position in metabolic reprogramming and TME research with the highest volume of publications and citations, thus exerting the greatest influence. Among these institutions, Fudan University displayed the highest level of productivity. Frontiers in Immunology showed the highest degree of productivity in this field. Ho Ping-Chih made the most article contributions, and Pearce Edward J. was the most co-cited author. Four clusters were obtained after a cluster analysis of the authors' keywords: TME, metabolic reprogramming, immunometabolism, and immunity. Immunometabolism, glycolysis, immune cells, and tumor-associated macrophages are relatively recent keywords that have attracted increasing attention. CONCLUSIONS A comprehensive landscape of advancements in metabolic reprogramming and the TME was evaluated, which provided crucial information for scholars to further advance this promising field. Further research should explore new topics related to immunometabolism in the TME using a transdisciplinary approach.
Collapse
Affiliation(s)
- Yupeng Xi
- Department of General Internal Medicine, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Rui Liu
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Xing Zhang
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Qiujun Guo
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Xiwen Zhang
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Zizhen Yang
- Department of General Internal MedicineXi'an Fifth HospitalXi'anShanxiChina
| | - Honggang Zheng
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| | - Baojin Hua
- Department of Oncology, Guang'anmen HospitalChina Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
12
|
Chen L, Xing X, Zhu Y, Chen Y, Pei H, Song Q, Li J, Zhang P. Palmitoylation alters LDHA activity and pancreatic cancer response to chemotherapy. Cancer Lett 2024; 587:216696. [PMID: 38331089 DOI: 10.1016/j.canlet.2024.216696] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Lactate dehydrogenase A (LDHA) serves as a key regulator of the Warburg Effect by catalyzing the conversion of pyruvate to lactate in the final step of glycolysis. Both the expression level and enzyme activity of LDHA are upregulated in cancers, however, the underlying mechanism remains incompletely understood. Here, we show that LDHA is post-translationally palmitoylated by ZDHHC9 at cysteine 163, which promotes its enzyme activity, lactate production, and reduces reactive oxygen species (ROS) generation. Replacement of endogenous LDHA with a palmitoylation-deficient mutant leads to reduced pancreatic cancer cell proliferation, increased T-cell infiltration, and limited tumor growth; it also affects pancreatic cancer cell response to chemotherapy. Moreover, LDHA palmitoylation is upregulated in gemcitabine resistant pancreatic cancer cells. Clinically, ZDHHC9 is upregulated in pancreatic cancer and correlated with poor prognoses for patients. Overall, our findings identify ZDHHC9-mediated palmitoylation as a positive regulator of LDHA, with potentially significant implications for cancer etiology and targeted therapy for pancreatic cancer.
Collapse
Affiliation(s)
- Luojun Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430062, Hubei, China
| | - Xiaoke Xing
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430062, Hubei, China
| | - Yue Zhu
- Department of Radiotherapy, The First Affiliated Hospital, Medical School of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yali Chen
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, 100850, China
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, 20057, DC, USA.
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430062, Hubei, China.
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, 430062, Hubei, China.
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, 430062, Hubei, China.
| |
Collapse
|