1
|
Wu B, Zhang B, Li B, Wu H, Jiang M. Cold and hot tumors: from molecular mechanisms to targeted therapy. Signal Transduct Target Ther 2024; 9:274. [PMID: 39420203 PMCID: PMC11491057 DOI: 10.1038/s41392-024-01979-x] [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: 04/17/2024] [Revised: 08/20/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment, particularly through immune checkpoint blockade (ICB), which has shown notable clinical benefits across various tumor types. Despite the transformative impact of ICB treatment in cancer therapy, only a minority of patients exhibit a positive response to it. In patients with solid tumors, those who respond well to ICB treatment typically demonstrate an active immune profile referred to as the "hot" (immune-inflamed) phenotype. On the other hand, non-responsive patients may exhibit a distinct "cold" (immune-desert) phenotype, differing from the features of "hot" tumors. Additionally, there is a more nuanced "excluded" immune phenotype, positioned between the "cold" and "hot" categories, known as the immune "excluded" type. Effective differentiation between "cold" and "hot" tumors, and understanding tumor intrinsic factors, immune characteristics, TME, and external factors are critical for predicting tumor response and treatment results. It is widely accepted that ICB therapy exerts a more profound effect on "hot" tumors, with limited efficacy against "cold" or "altered" tumors, necessitating combinations with other therapeutic modalities to enhance immune cell infiltration into tumor tissue and convert "cold" or "altered" tumors into "hot" ones. Therefore, aligning with the traits of "cold" and "hot" tumors, this review systematically delineates the respective immune characteristics, influencing factors, and extensively discusses varied treatment approaches and drug targets based on "cold" and "hot" tumors to assess clinical efficacy.
Collapse
Affiliation(s)
- Bo Wu
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bo Zhang
- Department of Youth League Committee, The Fourth Affiliated Hospital, China Medical University, Shenyang, China
| | - Bowen Li
- Department of Pancreatic and Gastrointestinal Surgery, Ningbo No. 2 Hospital, Ningbo, China
| | - Haoqi Wu
- Department of Gynaecology and Obstetrics, The Second Hospital of Dalian Medical University, Dalian, China
| | - Meixi Jiang
- Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Knudsen-Clark AM, Mwangi D, Cazarin J, Morris K, Baker C, Hablitz LM, McCall MN, Kim M, Altman BJ. Circadian rhythms of macrophages are altered by the acidic tumor microenvironment. EMBO Rep 2024:10.1038/s44319-024-00288-2. [PMID: 39415049 DOI: 10.1038/s44319-024-00288-2] [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: 08/01/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/18/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are prime therapeutic targets due to their pro-tumorigenic functions, but varying efficacy of macrophage-targeting therapies highlights our incomplete understanding of how macrophages are regulated within the tumor microenvironment (TME). The circadian clock is a key regulator of macrophage function, but how circadian rhythms of macrophages are influenced by the TME remains unknown. Here, we show that conditions associated with the TME such as polarizing stimuli, acidic pH, and lactate can alter circadian rhythms in macrophages. While cyclic AMP (cAMP) has been reported to play a role in macrophage response to acidic pH, our results indicate pH-driven changes in circadian rhythms are not mediated solely by cAMP signaling. Remarkably, circadian disorder of TAMs was revealed by clock correlation distance analysis. Our data suggest that heterogeneity in circadian rhythms within the TAM population level may underlie this circadian disorder. Finally, we report that circadian regulation of macrophages suppresses tumor growth in a murine model of pancreatic cancer. Our work demonstrates a novel mechanism by which the TME influences macrophage biology through modulation of circadian rhythms.
Collapse
Affiliation(s)
- Amelia M Knudsen-Clark
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Daniel Mwangi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Kristina Morris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
| | - Cameron Baker
- Genomics Research Center, University of Rochester Medical Center, Rochester, NY, USA
| | - Lauren M Hablitz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew N McCall
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA
| | - Brian J Altman
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY, USA.
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
3
|
Lim J, La J, Kim HC, Kang I, Kang BH, Ku KB, Kim Y, Kwon MS, Lee HK. Type I interferon signaling regulates myeloid and T cell crosstalk in the glioblastoma tumor microenvironment. iScience 2024; 27:110810. [PMID: 39286510 PMCID: PMC11404196 DOI: 10.1016/j.isci.2024.110810] [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: 04/08/2024] [Revised: 08/02/2024] [Accepted: 08/21/2024] [Indexed: 09/19/2024] Open
Abstract
Downstream interferon signaling through the type I interferon (IFN) receptor, IFNAR, is crucial for the proper production of type I IFNs in mounting anti-tumor immune responses. Our study investigates the role of type I IFN signaling in the glioblastoma (GBM) tumor microenvironment by leveraging single-cell RNA sequencing to analyze tumor-infiltrating lymphocytes. We investigate how type I IFN signaling within the myeloid compartment contributes to the crosstalk with T cells in the tumor microenvironment. Through the use of the Gl261 murine GBM model, we find that the lack of proper type I IFN response results in enhanced PD-L1 interactions among myeloid cells, thereby affecting T cell functionality. Additionally, we also characterize how anti-PD1 treatment induces transcriptional changes in tumor-associated monocytes and macrophages by analyzing intercellular communication networks and propose how immune checkpoint blockade therapy could possibly relieve some of the immunosuppression derived from the lack of proper type I IFN production.
Collapse
Affiliation(s)
- Juhee Lim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Jeongwoo La
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Hyeon Cheol Kim
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
- Life Science Institute, KAIST, Daejeon 34141, Republic of Korea
| | - In Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Byeong Hoon Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Keun Bon Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
- Center for Infectious Disease Vaccine and Diagnosis Innovation, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Yumin Kim
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Myoung Seung Kwon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
| | - Heung Kyu Lee
- Laboratory of Host Defenses, Department of Biological Sciences, KAIST, Daejeon 34141, Republic of Korea
- KAIST Institute of Health Science and Technology, KAIST, Daejeon 34141, Republic of Korea
| |
Collapse
|
4
|
Aktar T, Modak S, Majumder D, Maiti D. A detailed insight into macrophages' role in shaping lung carcinogenesis. Life Sci 2024; 352:122896. [PMID: 38972632 DOI: 10.1016/j.lfs.2024.122896] [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: 03/05/2024] [Revised: 06/24/2024] [Accepted: 07/03/2024] [Indexed: 07/09/2024]
Abstract
Despite significant advancements in cancer treatment in recent decades, the high mortality rate associated with lung cancer remains a significant concern. The development and proper execution of new targeted therapies needs more deep knowledge regarding the lung cancer associated tumour microenvironment. One of the key component of that tumour microenvironment is the lung resident macrophages. Although in normal physiological condition the lung resident macrophages are believed to maintain lung homeostasis, but they may also initiate a vicious inflammatory response in abnormal conditions which is linked to lung cancer development. Depending on the activation pathway, the lung resident macrophages are either of M1 or M2 sub-type. The M1 and M2 sub-types differ significantly in various prospectuses, from phenotypic markers to metabolic pathways. In addition to this generalized classification, the recent advancement of the multiomics technology is able to identify some other sub-types of lung resident macrophages. Researchers have also observed that these different sub-types can manipulate the pathogenesis of lung carcinogenesis in a context dependent manner and can either promote or inhibit the development of lung carcinogenesis upon receiving proper activation. As proper knowledge about the role played by the lung resident macrophages' in shaping the lung carcinogenesis is limited, so the main purpose of this review is to bring all the available information under the same roof. We also elaborated the different mechanisms involved in maintenance of the plasticity of M1/M2 sub-type, as this plasticity can be a good target for lung cancer treatment.
Collapse
Affiliation(s)
- Tamanna Aktar
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Snehashish Modak
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India
| | - Debabrata Majumder
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India; Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Debasish Maiti
- Immunology Microbiology Lab, Department of Human Physiology, Tripura University, Suryamaninagar, Tripura 799022, India.
| |
Collapse
|
5
|
Song X, Hao C, Li Y, Li Y, Dong H, Wei Q, Wei M, Li H, Zhao L. Chiral inorganic nanomaterials in the tumor microenvironment: A new chapter in cancer therapy. Pharmacol Res 2024; 208:107386. [PMID: 39216840 DOI: 10.1016/j.phrs.2024.107386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 08/20/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Chirality plays a crucial function in the regulation of normal physiological processes and is widespread in organisms. Chirality can be imparted to nanomaterials, whether they are natural or manmade, through the process of asymmetric assembly and/or grafting of molecular chiral groups or linkers. Chiral inorganic nanomaterials possess unique physical and chemical features that set them apart from regular nanomaterials. They also have the ability to interact with cells and tissues in a specific manner, making them useful in various biomedical applications, particularly in the treatment of tumors. Despite the growing amount of research on chiral inorganic nanomaterials in the tumor microenvironment (TME) and their promising potential applications, there is a lack of literature that comprehensively summarizes the intricate interactions between chiral inorganic nanomaterials and TME. In this review, we introduce the fundamental concept, classification, synthesis methods, and physicochemical features of chiral inorganic nanomaterials. Next, we briefly outline the components of TME, such as T cells, macrophages, dendritic cells, and weak acids, and then discuss the anti-tumor effects of several chiral inorganic nanoparticles targeting these components and their potential for possible application during cancer therapy. Finally, the present challenges faced by chiral inorganic nanomaterials in cancer treatment and their future areas of investigation are disclosed.
Collapse
Affiliation(s)
- Xueyi Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Chenjing Hao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Yao Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Yunong Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Hongzhi Dong
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Qian Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| | - Heran Li
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China.
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang 110122, PR China; Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation, China Medical University, Shenyang 110122, PR China.
| |
Collapse
|
6
|
Praharaj M, Shen F, Lee AJ, Zhao L, Nirschl TR, Theodros D, Singh AK, Wang X, Adusei KM, Lombardo KA, Williams RA, Sena LA, Thompson EA, Tam A, Yegnasubramanian S, Pearce EJ, Leone RD, Alt J, Rais R, Slusher BS, Pardoll DM, Powell JD, Zarif JC. Metabolic Reprogramming of Tumor-Associated Macrophages Using Glutamine Antagonist JHU083 Drives Tumor Immunity in Myeloid-Rich Prostate and Bladder Cancers. Cancer Immunol Res 2024; 12:854-875. [PMID: 38701369 PMCID: PMC11217738 DOI: 10.1158/2326-6066.cir-23-1105] [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: 12/27/2023] [Revised: 04/10/2024] [Accepted: 04/30/2024] [Indexed: 05/05/2024]
Abstract
Glutamine metabolism in tumor microenvironments critically regulates antitumor immunity. Using the glutamine-antagonist prodrug JHU083, we report potent tumor growth inhibition in urologic tumors by JHU083-reprogrammed tumor-associated macrophages (TAMs) and tumor-infiltrating monocytes. We show JHU083-mediated glutamine antagonism in tumor microenvironments induced by TNF, proinflammatory, and mTORC1 signaling in intratumoral TAM clusters. JHU083-reprogrammed TAMs also exhibited increased tumor cell phagocytosis and diminished proangiogenic capacities. In vivo inhibition of TAM glutamine consumption resulted in increased glycolysis, a broken tricarboxylic acid (TCA) cycle, and purine metabolism disruption. Although the antitumor effect of glutamine antagonism on tumor-infiltrating T cells was moderate, JHU083 promoted a stem cell-like phenotype in CD8+ T cells and decreased the abundance of regulatory T cells. Finally, JHU083 caused a global shutdown in glutamine-utilizing metabolic pathways in tumor cells, leading to reduced HIF-1α, c-MYC phosphorylation, and induction of tumor cell apoptosis, all key antitumor features. Altogether, our findings demonstrate that targeting glutamine with JHU083 led to suppressed tumor growth as well as reprogramming of immunosuppressive TAMs within prostate and bladder tumors that promoted antitumor immune responses. JHU083 can offer an effective therapeutic benefit for tumor types that are enriched in immunosuppressive TAMs.
Collapse
Affiliation(s)
- Monali Praharaj
- Pathobiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Fan Shen
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Alex J. Lee
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Liang Zhao
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Thomas R. Nirschl
- Pathobiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Debebe Theodros
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Alok K. Singh
- Department of Medicine, Center for Tuberculosis Research, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| | - Xiaoxu Wang
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Kenneth M. Adusei
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Graduate Program in Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Kara A. Lombardo
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Raekwon A. Williams
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Laura A. Sena
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Elizabeth A. Thompson
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Ada Tam
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Srinivasan Yegnasubramanian
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Edward J. Pearce
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Robert D. Leone
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Jesse Alt
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland.
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Rana Rais
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland.
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, Maryland.
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland.
| | - Drew M. Pardoll
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Jonathan D. Powell
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| | - Jelani C. Zarif
- Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
7
|
Krieg S, Fernandes SI, Kolliopoulos C, Liu M, Fendt SM. Metabolic Signaling in Cancer Metastasis. Cancer Discov 2024; 14:934-952. [PMID: 38592405 PMCID: PMC7616057 DOI: 10.1158/2159-8290.cd-24-0174] [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: 02/07/2024] [Revised: 03/05/2024] [Accepted: 03/12/2024] [Indexed: 04/10/2024]
Abstract
Metastases, which are the leading cause of death in patients with cancer, have metabolic vulnerabilities. Alterations in metabolism fuel the energy and biosynthetic needs of metastases but are also needed to activate cell state switches in cells leading to invasion, migration, colonization, and outgrowth in distant organs. Specifically, metabolites can activate protein kinases as well as receptors and they are crucial substrates for posttranslational modifications on histone and nonhistone proteins. Moreover, metabolic enzymes can have moonlighting functions by acting catalytically, mainly as protein kinases, or noncatalytically through protein-protein interactions. Here, we summarize the current knowledge on metabolic signaling in cancer metastasis. SIGNIFICANCE Effective drugs for the prevention and treatment of metastases will have an immediate impact on patient survival. To overcome the current lack of such drugs, a better understanding of the molecular processes that are an Achilles heel in metastasizing cancer cells is needed. One emerging opportunity is the metabolic changes cancer cells need to undergo to successfully metastasize and grow in distant organs. Mechanistically, these metabolic changes not only fulfill energy and biomass demands, which are often in common between cancer and normal but fast proliferating cells, but also metabolic signaling which enables the cell state changes that are particularly important for the metastasizing cancer cells.
Collapse
Affiliation(s)
- Sarah Krieg
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Sara Isabel Fernandes
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Constantinos Kolliopoulos
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Ming Liu
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Herestraat 49, 3000 Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
8
|
Chapman NM, Chi H. Metabolic rewiring and communication in cancer immunity. Cell Chem Biol 2024; 31:862-883. [PMID: 38428418 PMCID: PMC11177544 DOI: 10.1016/j.chembiol.2024.02.001] [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: 11/09/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 03/03/2024]
Abstract
The immune system shapes tumor development and progression. Although immunotherapy has transformed cancer treatment, its overall efficacy remains limited, underscoring the need to uncover mechanisms to improve therapeutic effects. Metabolism-associated processes, including intracellular metabolic reprogramming and intercellular metabolic crosstalk, are emerging as instructive signals for anti-tumor immunity. Here, we first summarize the roles of intracellular metabolic pathways in controlling immune cell function in the tumor microenvironment. How intercellular metabolic communication regulates anti-tumor immunity, and the impact of metabolites or nutrients on signaling events, are also discussed. We then describe how targeting metabolic pathways in tumor cells or intratumoral immune cells or via nutrient-based interventions may boost cancer immunotherapies. Finally, we conclude with discussions on profiling and functional perturbation methods of metabolic activity in intratumoral immune cells, and perspectives on future directions. Uncovering the mechanisms for metabolic rewiring and communication in the tumor microenvironment may enable development of novel cancer immunotherapies.
Collapse
Affiliation(s)
- Nicole M Chapman
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
9
|
Chen S, Zeng J, Li R, Zhang Y, Tao Y, Hou Y, Yang L, Zhang Y, Wu J, Meng X. Traditional Chinese medicine in regulating macrophage polarization in immune response of inflammatory diseases. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117838. [PMID: 38310986 DOI: 10.1016/j.jep.2024.117838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Numerous studies have demonstrated that various traditional Chinese medicines (TCMs) exhibit potent anti-inflammatory effects against inflammatory diseases mediated through macrophage polarization and metabolic reprogramming. AIM OF THE STUDY The objective of this review was to assess and consolidate the current understanding regarding the pathogenic mechanisms governing macrophage polarization in the context of regulating inflammatory diseases. We also summarize the mechanism action of various TCMs on the regulation of macrophage polarization, which may contribute to facilitate the development of natural anti-inflammatory drugs based on reshaping macrophage polarization. MATERIALS AND METHODS We conducted a comprehensive review of recently published articles, utilizing keywords such as "macrophage polarization" and "traditional Chinese medicines" in combination with "inflammation," as well as "macrophage polarization" and "inflammation" in conjunction with "natural products," and similar combinations, to search within PubMed and Google Scholar databases. RESULTS A total of 113 kinds of TCMs (including 62 components of TCMs, 27 TCMs as well as various types of extracts of TCMs and 24 Chinese prescriptions) was reported to exert anti-inflammatory effects through the regulation of key pathways of macrophage polarization and metabolic reprogramming. CONCLUSIONS In this review, we have analyzed studies concerning the involvement of macrophage polarization and metabolic reprogramming in inflammation therapy. TCMs has great advantages in regulating macrophage polarization in treating inflammatory diseases due to its multi-pathway and multi-target pharmacological action. This review may contribute to facilitate the development of natural anti-inflammatory drugs based on reshaping macrophage polarization.
Collapse
Affiliation(s)
- Shiyu Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Rui Li
- The Affiliated Meishan Hospital of Chengdu University of Traditional Chinese Medicine, Meishan, 620010, PR China
| | - Yingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yiwen Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Ya Hou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Lu Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Yating Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jiasi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| |
Collapse
|
10
|
Sheng Y, Hu W, Chen S, Zhu X. Efferocytosis by macrophages in physiological and pathological conditions: regulatory pathways and molecular mechanisms. Front Immunol 2024; 15:1275203. [PMID: 38779685 PMCID: PMC11109379 DOI: 10.3389/fimmu.2024.1275203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 04/17/2024] [Indexed: 05/25/2024] Open
Abstract
Efferocytosis is defined as the highly effective phagocytic removal of apoptotic cells (ACs) by professional or non-professional phagocytes. Tissue-resident professional phagocytes ("efferocytes"), such as macrophages, have high phagocytic capacity and are crucial to resolve inflammation and aid in homeostasis. Recently, numerous exciting discoveries have revealed divergent (and even diametrically opposite) findings regarding metabolic immune reprogramming associated with efferocytosis by macrophages. In this review, we highlight the key metabolites involved in the three phases of efferocytosis and immune reprogramming of macrophages under physiological and pathological conditions. The next decade is expected to yield further breakthroughs in the regulatory pathways and molecular mechanisms connecting immunological outcomes to metabolic cues as well as avenues for "personalized" therapeutic intervention.
Collapse
Affiliation(s)
- Yan−Ran Sheng
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Wen−Ting Hu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Siman Chen
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiao−Yong Zhu
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- Key Laboratory of Reproduction Regulation of NPFPC, SIPPR, IRD, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Wang Z, Wang Q, Cao H, Wang Z, Wang D, Liu J, Gao T, Ren C, Liu J. Mitochondrial Localized In Situ Self-Assembly Reprogramming Tumor Immune and Metabolic Microenvironment for Enhanced Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311043. [PMID: 38190762 DOI: 10.1002/adma.202311043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/19/2023] [Indexed: 01/10/2024]
Abstract
The inherent immune and metabolic tumor microenvironment (TME) of most solid tumors adversely affect the antitumor efficacy of various treatments, which is an urgent issue to be solved in clinical cancer therapy. In this study, a mitochondrial localized in situ self-assembly system is constructed to remodel the TME by improving immunogenicity and disrupting the metabolic plasticity of cancer cells. The peptide-based drug delivery system can be pre-assembled into nanomicelles in vitro and form functional nanofibers on mitochondria through a cascade-responsive process involving reductive release, targeted enrichment, and in situ self-assembly. The organelle-specific in situ self-assemblyeffectively switches the role of mitophagy from pro-survival to pro-death, which finally induces intense endoplasmic reticulum stress and atypical type II immunogenic cell death. Disintegration of the mitochondrial ultrastructure also impedes the metabolic plasticity of tumor cells, which greatly promotes the immunosuppresive TME remodeling into an immunostimulatory TME. Ultimately, the mitochondrial localized in situ self-assembly system effectively suppresses tumor metastases, and converts cold tumors into hot tumors with enhanced sensitivity to radiotherapy and immune checkpoint blockade therapy. This study offers a universal strategy for spatiotemporally controlling supramolecular self-assembly on sub-organelles to determine cancer cell fate and enhance cancer therapy.
Collapse
Affiliation(s)
- Zhilong Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Qian Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Hongmei Cao
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Zhongyan Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Dianyu Wang
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jinjian Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Tongxin Gao
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Chunhua Ren
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| | - Jianfeng Liu
- Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Chinese Academy of Medical Sciences, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, P. R. China
| |
Collapse
|
12
|
Saha P, Ettel P, Weichhart T. Leveraging macrophage metabolism for anticancer therapy: opportunities and pitfalls. Trends Pharmacol Sci 2024; 45:335-349. [PMID: 38494408 DOI: 10.1016/j.tips.2024.02.005] [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: 01/26/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/19/2024]
Abstract
Tumor-associated macrophages (TAMs) constitute an important part of the tumor microenvironment (TME) that regulates tumor progression. Tumor-derived signals, hypoxia, and competition for nutrients influence TAMs to reprogram their cellular metabolism. This altered metabolic profile creates a symbiotic communication between tumor and other immune cells to support tumor growth. In addition, the metabolic profile of TAMs regulates the expression of immune checkpoint molecules. The dynamic plasticity also allows TAMs to reshape their metabolism in response to modern therapeutic strategies. Therefore, over the years, a significant number of approaches have been implicated to reprogram cancer-promoting metabolism in TAMs. In this review, we discuss the current strategies and pitfalls, along with upcoming promising opportunities in leveraging TAM metabolism for developing better therapeutic approaches against cancer.
Collapse
Affiliation(s)
- Piyal Saha
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria
| | - Paul Ettel
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria
| | - Thomas Weichhart
- Institute for Medical Genetics, Center for Pathobiochemistry and Genetics, Medical University of Vienna, Währinger Straße 10, 1090 Vienna, Austria.
| |
Collapse
|
13
|
Soldani C, De Simone G, Polidoro MA, Morabito A, Franceschini B, Colombo FS, Anselmo A, Milana F, Lleo A, Torzilli G, Pastorelli R, Donadon M, Brunelli L. Riboflavin-LSD1 axis participates in the in vivo tumor-associated macrophage morphology in human colorectal liver metastases. Cancer Immunol Immunother 2024; 73:63. [PMID: 38430255 PMCID: PMC10908638 DOI: 10.1007/s00262-024-03645-1] [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: 10/21/2023] [Accepted: 01/29/2024] [Indexed: 03/03/2024]
Abstract
Tumor-associated macrophages (TAMs) are key components of the tumor microenvironment (TME). In colorectal liver metastasis (CLM), TAM morphology correlates with prognosis, with smaller TAMs (S-TAMs) conferring a more favorable prognosis than larger TAMs (L-TAMs). However, the metabolic profile of in vivo human TAM populations remains unknown. Multiparametric flow cytometry was used to freshly isolate S- and L-TAMs from surgically resected CLM patients (n = 14S-, 14L-TAMs). Mass spectrometry-based metabolomics analyses were implemented for the metabolic characterization of TAM populations. Gene expression analysis and protein activity were used to support the biochemical effects of the enzyme-substrate link between riboflavin and (lysine-specific demethylase 1A, LSD1) with TAM morphologies. L-TAMs were characterized by a positive correlation and a strong association between riboflavin and TAM morphologies. Riboflavin in both L-TAMs and in-vitro M2 polarized macrophages modulates LSD1 protein expression and activity. The inflammatory stimuli promoted by TNFα induced the increased expression of riboflavin transporter SLC52A3 and LSD1 in M2 macrophages. The modulation of the riboflavin-LSD1 axis represents a potential target for reprogramming TAM subtypes, paving the way for promising anti-tumor therapeutic strategies.
Collapse
Affiliation(s)
- Cristiana Soldani
- Laboratory of Hepatobiliary Immunopathology, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Giulia De Simone
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
- Department of Biotechnologies and Biosciences, Università degli Studi Milano Bicocca, Milan, Italy
| | - Michela Anna Polidoro
- Laboratory of Hepatobiliary Immunopathology, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Aurelia Morabito
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Barbara Franceschini
- Laboratory of Hepatobiliary Immunopathology, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Federico Simone Colombo
- Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Struttura Complessa di Patologia Clinica, Laboratorio di Citometria, Milano, Italy
| | - Achille Anselmo
- Flow Cytometry Resource, Advanced Cytometry Technical Application Laboratory, IRCCS Ospedale San Raffaele, Milano, Italy
| | - Flavio Milana
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele - Milan, Italy
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Ana Lleo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele - Milan, Italy
- Division of Internal Medicine and Hepatology, Department of Gastroenterology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Guido Torzilli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele - Milan, Italy
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy
| | - Roberta Pastorelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Matteo Donadon
- Laboratory of Hepatobiliary Immunopathology, IRCCS Humanitas Research Hospital, Via Alessandro Manzoni 56, 20089, Rozzano, Milan, Italy.
- Division of Hepatobiliary and General Surgery, Department of Surgery, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy.
- Department of General Surgery, University Maggiore Hospital, Novara, Italy.
| | - Laura Brunelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy.
| |
Collapse
|
14
|
Zhang D, Huang H, Gao X, Yu G, Zhang X, Jin H, Xu R, Wang Z, Zhang G. High expression of B7-H3 on monocyte/macrophages in tumor microenvironment promotes lung cancer progression by inhibiting apoptosis. Transl Oncol 2024; 41:101874. [PMID: 38262113 PMCID: PMC10832491 DOI: 10.1016/j.tranon.2023.101874] [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: 10/16/2023] [Revised: 12/03/2023] [Accepted: 12/22/2023] [Indexed: 01/25/2024] Open
Abstract
Monocyte/macrophages constitute a significant population of tumor-infiltrating immune cells and play a crucial role in tumor growth, invasion, and metastasis. B7-H3, has immune regulatory functions, however, it is unclear whether B7-H3 expressed on monocyte/macrophages plays a significance role in tumor progression. We found B7-H3 was high-expressed on monocyte/macrophages in tumor microenvironment compared with adjacent tissues in lung cancer, and its expression level was positively correlated with the number of monocyte/macrophages. Furthermore, the expression of B7-H3 was related to clinical stage and lymph node metastasis. Moreover, miR-29a-3p negatively regulated B7-H3, and the expression of B7-H3 on THP-1-derived macrophages was regulated by secreting exosomes containing miR-29a-3p. In addition, knockdown of B7-H3 promoted macrophage apoptosis under hypoxia. Mechanistically, B7-H3 enhanced the antiapoptotic ability of macrophage by up-regulating HIF-1ɑ via activating NF-κB. Taken together, these results imply that B7-H3 as a therapeutic target could hold promise for enhancing anti-tumor immune responses in individuals diagnosed with lung cancer.
Collapse
Affiliation(s)
- Dongze Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Haitao Huang
- Department of Thoracic surgery, The First Affiliated Hospital of Soochow University, China
| | - Xin Gao
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, China
| | - Gehua Yu
- Department of Immunology, School of Biology and Basic Medical Sciences, Soochow University, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Haiyan Jin
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Ruyan Xu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China
| | - Zhenxin Wang
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, China.
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, China.
| |
Collapse
|
15
|
Li XM, Yang Y, Jiang FQ, Hu G, Wan S, Yan WY, He XS, Xiao F, Yang XM, Guo X, Lu JH, Yang XQ, Chen JJ, Ye WL, Liu Y, He K, Duan HX, Zhou YJ, Gan WJ, Liu F, Wu H. Histone lactylation inhibits RARγ expression in macrophages to promote colorectal tumorigenesis through activation of TRAF6-IL-6-STAT3 signaling. Cell Rep 2024; 43:113688. [PMID: 38245869 DOI: 10.1016/j.celrep.2024.113688] [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: 07/31/2023] [Revised: 12/06/2023] [Accepted: 01/03/2024] [Indexed: 01/23/2024] Open
Abstract
Macrophages are phenotypically and functionally diverse in the tumor microenvironment (TME). However, how to remodel macrophages with a protumor phenotype and how to manipulate them for therapeutic purposes remain to be explored. Here, we show that in the TME, RARγ is downregulated in macrophages, and its expression correlates with poor prognosis in patients with colorectal cancer (CRC). In macrophages, RARγ interacts with tumor necrosis factor receptor-associated factor 6 (TRAF6), which prevents TRAF6 oligomerization and autoubiquitination, leading to inhibition of nuclear factor κB signaling. However, tumor-derived lactate fuels H3K18 lactylation to prohibit RARγ gene transcription in macrophages, consequently enhancing interleukin-6 (IL-6) levels in the TME and endowing macrophages with tumor-promoting functions via activation of signal transducer and activator of transcription 3 (STAT3) signaling in CRC cells. We identified that nordihydroguaiaretic acid (NDGA) exerts effective antitumor action by directly binding to RARγ to inhibit TRAF6-IL-6-STAT3 signaling. This study unravels lactate-driven macrophage function remodeling by inhibition of RARγ expression and highlights NDGA as a candidate compound for treating CRC.
Collapse
Affiliation(s)
- Xiu-Ming Li
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Yun Yang
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Fu-Quan Jiang
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Guang Hu
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Shan Wan
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Ying Yan
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Xiao-Shun He
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Fei Xiao
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Xue-Mei Yang
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Xin Guo
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Jun-Hou Lu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Jun-Jie Chen
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Wen-Long Ye
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Yue Liu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Kuang He
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Han-Xiao Duan
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Yu-Jia Zhou
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Juan Gan
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China.
| | - Feng Liu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China.
| | - Hua Wu
- Department of Pathology, Medical Center of Soochow University and Suzhou Medical College of Soochow University and YongDing Clinical Institute of Soochow University, Soochow University, Suzhou 215123, China.
| |
Collapse
|
16
|
Jiang G, Hong J, Sun L, Wei H, Gong W, Wang S, Zhu J. Glycolysis regulation in tumor-associated macrophages: Its role in tumor development and cancer treatment. Int J Cancer 2024; 154:412-424. [PMID: 37688376 DOI: 10.1002/ijc.34711] [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: 04/18/2023] [Revised: 07/27/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023]
Abstract
Tumor-associated macrophages constitute the main cell population in the tumor microenvironment and play a crucial role in regulating the microenvironment composition. Emerging evidence has revealed that the metabolic profile determines the tumor-associated macrophage phenotype. Tumor-associated macrophage function is highly dependent on glucose metabolism, with glycolysis being the major metabolic pathway. Recent reports have demonstrated diversity in glucose flux of tumor-associated macrophages and complex substance communication with cancer cells. However, how the glucose flux in tumor-associated macrophages connects with glycolysis to influence tumor progression and the tumor microenvironment is still obscure. Moreover, while the development of single-cell sequencing technology allows a clearer and more accurate classification of tumor-associated macrophages, the metabolic profiles of tumor-associated macrophages from the perspective of single-cell omics has not been well summarized. Here, we review the current state of knowledge on glucose metabolism in tumor-associated macrophages and summarize the metabolic profiles of different tumor-associated macrophage subtypes from the perspective of single-cell omics. Additionally, we describe the current strategies targeting glycolysis in tumor-associated macrophages for cancer therapy.
Collapse
Affiliation(s)
- Guangyi Jiang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Junjie Hong
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Lu Sun
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Haibin Wei
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Wangang Gong
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| | - Shu Wang
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Jianqing Zhu
- Department of Gynecological Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Zhejiang, Hangzhou, China
| |
Collapse
|
17
|
Cao L, Meng X, Zhang Z, Liu Z, He Y. Macrophage heterogeneity and its interactions with stromal cells in tumour microenvironment. Cell Biosci 2024; 14:16. [PMID: 38303024 PMCID: PMC10832170 DOI: 10.1186/s13578-024-01201-z] [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: 10/11/2023] [Accepted: 01/22/2024] [Indexed: 02/03/2024] Open
Abstract
Macrophages and tumour stroma cells account for the main cellular components in the tumour microenvironment (TME). Current advancements in single-cell analysis have revolutionized our understanding of macrophage diversity and macrophage-stroma interactions. Accordingly, this review describes new insight into tumour-associated macrophage (TAM) heterogeneity in terms of tumour type, phenotype, metabolism, and spatial distribution and presents the association between these factors and TAM functional states. Meanwhile, we focus on the immunomodulatory feature of TAMs and highlight the tumour-promoting effect of macrophage-tumour stroma interactions in the immunosuppressive TME. Finally, we summarize recent studies investigating macrophage-targeted therapy and discuss their therapeutic potential in improving immunotherapy by alleviating immunosuppression.
Collapse
Affiliation(s)
- Liren Cao
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Xiaoyan Meng
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Zhonglong Liu
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yue He
- Department of Oral Maxillofacial & Head and Neck Oncology, National Clinical Research Center for Oral Disease, National Center of Stomatology, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
18
|
Chuang YM, Tzeng SF, Ho PC, Tsai CH. Immunosurveillance encounters cancer metabolism. EMBO Rep 2024; 25:471-488. [PMID: 38216787 PMCID: PMC10897436 DOI: 10.1038/s44319-023-00038-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: 02/10/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 01/14/2024] Open
Abstract
Tumor cells reprogram nutrient acquisition and metabolic pathways to meet their energetic, biosynthetic, and redox demands. Similarly, metabolic processes in immune cells support host immunity against cancer and determine differentiation and fate of leukocytes. Thus, metabolic deregulation and imbalance in immune cells within the tumor microenvironment have been reported to drive immune evasion and to compromise therapeutic outcomes. Interestingly, emerging evidence indicates that anti-tumor immunity could modulate tumor heterogeneity, aggressiveness, and metabolic reprogramming, suggesting that immunosurveillance can instruct cancer progression in multiple dimensions. This review summarizes our current understanding of how metabolic crosstalk within tumors affects immunogenicity of tumor cells and promotes cancer progression. Furthermore, we explain how defects in the metabolic cascade can contribute to developing dysfunctional immune responses against cancers and discuss the contribution of immunosurveillance to these defects as a feedback mechanism. Finally, we highlight ongoing clinical trials and new therapeutic strategies targeting cellular metabolism in cancer.
Collapse
Affiliation(s)
- Yu-Ming Chuang
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Sheue-Fen Tzeng
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland.
| | - Chin-Hsien Tsai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
- Department and Graduate Institute of Biochemistry, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
19
|
Yin T, Li X, Li Y, Zang X, Liu L, Du M. Macrophage plasticity and function in cancer and pregnancy. Front Immunol 2024; 14:1333549. [PMID: 38274812 PMCID: PMC10808357 DOI: 10.3389/fimmu.2023.1333549] [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: 11/05/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
As the soil of life, the composition and shaping process of the immune microenvironment of the uterus is worth exploring. Macrophages, indispensable constituents of the innate immune system, are essential mediators of inflammation and tissue remodeling as well. Recent insights into the heterogeneity of macrophage subpopulations have renewed interest in their functional diversity in both physiological and pathological settings. Macrophages display remarkable plasticity and switch from one phenotype to another. Intrinsic plasticity enables tissue macrophages to perform a variety of functions in response to changing tissue contexts, such as cancer and pregnancy. The remarkable diversity and plasticity make macrophages particularly intriguing cells given their dichotomous role in either attacking or protecting tumors and semi-allogeneic fetuses, which of both are characterized functionally by immunomodulation and neovascularization. Here, we reviewed and compared novel perspectives on macrophage biology of these two settings, including origin, phenotype, differentiation, and essential roles in corresponding microenvironments, as informed by recent studies on the heterogeneity of macrophage identity and function, as well as their mechanisms that might offer opportunities for new therapeutic strategies on malignancy and pregnancy complications.
Collapse
Affiliation(s)
- Tingxuan Yin
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xinyi Li
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yanhong Li
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Lu Liu
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | - Meirong Du
- Lab of Reproduction Immunology, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
20
|
Qian Y, Yin Y, Zheng X, Liu Z, Wang X. Metabolic regulation of tumor-associated macrophage heterogeneity: insights into the tumor microenvironment and immunotherapeutic opportunities. Biomark Res 2024; 12:1. [PMID: 38185636 PMCID: PMC10773124 DOI: 10.1186/s40364-023-00549-7] [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: 10/01/2023] [Accepted: 12/12/2023] [Indexed: 01/09/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are a heterogeneous population that play diverse functions in tumors. Their identity is determined not only by intrinsic factors, such as origins and transcription factors, but also by external signals from the tumor microenvironment (TME), such as inflammatory signals and metabolic reprogramming. Metabolic reprogramming has rendered TAM to exhibit a spectrum of activities ranging from pro-tumorigenic to anti-tumorigenic, closely associated with tumor progression and clinical prognosis. This review implicates the diversity of TAM phenotypes and functions, how this heterogeneity has been re-evaluated with the advent of single-cell technologies, and the impact of TME metabolic reprogramming on TAMs. We also review current therapies targeting TAM metabolism and offer new insights for TAM-dependent anti-tumor immunotherapy by focusing on the critical role of different metabolic programs in TAMs.
Collapse
Affiliation(s)
- Yujing Qian
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yujia Yin
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaocui Zheng
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Zhaoyuan Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xipeng Wang
- Department of Obstetrics and Gynecology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
21
|
Demicco M, Liu XZ, Leithner K, Fendt SM. Metabolic heterogeneity in cancer. Nat Metab 2024; 6:18-38. [PMID: 38267631 DOI: 10.1038/s42255-023-00963-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] [Received: 07/12/2022] [Accepted: 12/06/2023] [Indexed: 01/26/2024]
Abstract
Cancer cells rewire their metabolism to survive during cancer progression. In this context, tumour metabolic heterogeneity arises and develops in response to diverse environmental factors. This metabolic heterogeneity contributes to cancer aggressiveness and impacts therapeutic opportunities. In recent years, technical advances allowed direct characterisation of metabolic heterogeneity in tumours. In addition to the metabolic heterogeneity observed in primary tumours, metabolic heterogeneity temporally evolves along with tumour progression. In this Review, we summarize the mechanisms of environment-induced metabolic heterogeneity. In addition, we discuss how cancer metabolism and the key metabolites and enzymes temporally and functionally evolve during the metastatic cascade and treatment.
Collapse
Affiliation(s)
- Margherita Demicco
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Xiao-Zheng Liu
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Katharina Leithner
- Division of Pulmonology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.
- Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
22
|
Saito Y, Iida-Norita R, Afroj T, Refaat A, Hazama D, Komori S, Ohata S, Takai T, Oduori OS, Kotani T, Funakoshi Y, Koma YI, Murata Y, Yakushijin K, Matsuoka H, Minami H, Yokozaki H, Manz MG, Matozaki T. Preclinical evaluation of the efficacy of an antibody to human SIRPα for cancer immunotherapy in humanized mouse models. Front Immunol 2023; 14:1294814. [PMID: 38162643 PMCID: PMC10757636 DOI: 10.3389/fimmu.2023.1294814] [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: 09/15/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are abundant in the tumor microenvironment and are considered potential targets for cancer immunotherapy. To examine the antitumor effects of agents targeting human TAMs in vivo, we here established preclinical tumor xenograft models based on immunodeficient mice that express multiple human cytokines and have been reconstituted with a human immune system by transplantation of human CD34+ hematopoietic stem and progenitor cells (HIS-MITRG mice). HIS-MITRG mice supported the growth of both human cell line (Raji)- and patient-derived B cell lymphoma as well as the infiltration of human macrophages into their tumors. We examined the potential antitumor action of an antibody to human SIRPα (SE12C3) that inhibits the interaction of CD47 on tumor cells with SIRPα on human macrophages and thereby promotes Fcγ receptor-mediated phagocytosis of the former cells by the latter. Treatment with the combination of rituximab (antibody to human CD20) and SE12C3 inhibited Raji tumor growth in HIS-MITRG mice to a markedly greater extent than did rituximab monotherapy. This enhanced antitumor effect was dependent on human macrophages and attributable to enhanced rituximab-dependent phagocytosis of lymphoma cells by human macrophages. Treatment with rituximab and SE12C3 also induced reprogramming of human TAMs toward a proinflammatory phenotype. Furthermore, the combination treatment essentially prevented the growth of patient-derived diffuse large B cell lymphoma in HIS-MITRG mice. Our findings thus support the study of HIS-MITRG mice as a model for the preclinical evaluation in vivo of potential therapeutics, such as antibodies to human SIRPα, that target human TAMs.
Collapse
Affiliation(s)
- Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Rie Iida-Norita
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tania Afroj
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Biosignal Regulation, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Alaa Refaat
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Daisuke Hazama
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Satomi Komori
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Biosignal Regulation, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Ohata
- Division of Medical Oncology and Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoko Takai
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Biosignal Regulation, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Okechi S. Oduori
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Biosignal Regulation, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takenori Kotani
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yohei Funakoshi
- Division of Medical Oncology and Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu-Ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoji Murata
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kimikazu Yakushijin
- Division of Medical Oncology and Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Matsuoka
- Division of Medical Oncology and Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Bioresource Research and Development, Department of Social/Community Medicine and Health Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hironobu Minami
- Division of Medical Oncology and Hematology, Department of Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
- Comprehensive Cancer Center Zurich at the University of Zurich, Zurich, Switzerland
| | - Takashi Matozaki
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Biosignal Regulation, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
23
|
Chaudagar K, Hieromnimon HM, Kelley A, Labadie B, Shafran J, Rameshbabu S, Drovetsky C, Bynoe K, Solanki A, Markiewicz E, Fan X, Loda M, Patnaik A. Suppression of Tumor Cell Lactate-generating Signaling Pathways Eradicates Murine PTEN/p53-deficient Aggressive-variant Prostate Cancer via Macrophage Phagocytosis. Clin Cancer Res 2023; 29:4930-4940. [PMID: 37721526 PMCID: PMC10841690 DOI: 10.1158/1078-0432.ccr-23-1441] [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: 05/23/2023] [Revised: 08/04/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
PURPOSE Phosphatase and tensin homolog (PTEN) loss-of-function/PI3K pathway hyperactivation is associated with poor therapeutic outcomes and immune checkpoint inhibitor resistance across multiple malignancies. Our prior studies in Pb-Cre;PTENfl/flTrp53fl/fl genetically engineered mice (GEM) with aggressive-variant prostate cancer (AVPC) demonstrated tumor growth control in 60% mice following androgen deprivation therapy/PI3K inhibitor (PI3Ki)/programmed cell death protein 1 (PD-1) antibody combination, via abrogating lactate cross-talk between cancer cells and tumor-associated macrophages (TAM), and suppression of histone lactylation (H3K18lac)/phagocytic activation within TAM. Here, we targeted immunometabolic mechanism(s) of PI3Ki resistance, with the goal of durable tumor control in AVPC. EXPERIMENTAL DESIGN Pb-Cre;PTENfl/flTrp53fl/fl GEM were treated with PI3Ki (copanlisib), MEK inhibitor (trametinib) or Porcupine inhibitor (LGK'974) singly or their combinations. MRI was used to monitor tumor kinetics and immune/proteomic profiling/ex vivo coculture mechanistic studies were performed on GEM tumors or corresponding tumor-derived cell lines. RESULTS Given our proteomic profiling showing persistent MEK signaling within tumors of PI3Ki-resistant GEM, we tested whether addition of trametinib to copanlisib enhances tumor control in GEM, and we observed 80% overall response rate via additive suppression of lactate within TME and H3K18lac within TAM, relative to copanlisib (37.5%) monotherapy. The 20% resistant mice demonstrated feedback Wnt/β-catenin activation, resulting in restoration of lactate secretion by tumor cells and H3K18lac within TAM. Cotargeting Wnt/β-catenin signaling with LGK'974 in combination with PI3Ki/MEKi, demonstrated durable tumor control in 100% mice via H3K18lac suppression and complete TAM activation. CONCLUSIONS Abrogation of lactate-mediated cross-talk between cancer cells and TAM results in durable ADT-independent tumor control in PTEN/p53-deficient AVPC, and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Kiranj Chaudagar
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Hanna M. Hieromnimon
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Anne Kelley
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Brian Labadie
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jordan Shafran
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Srikrishnan Rameshbabu
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Catherine Drovetsky
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Kaela Bynoe
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL, USA
| | | | - Xiaobing Fan
- Department of Radiology, University of Chicago, Chicago IL, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Akash Patnaik
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
24
|
Oliveres H, Cascante M, Maurel J. Metabolic interventions to enhance immunotherapy and targeted therapy efficacy in advanced colorectal cancer. Curr Opin Chem Biol 2023; 77:102401. [PMID: 37806262 DOI: 10.1016/j.cbpa.2023.102401] [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: 06/17/2022] [Revised: 07/25/2023] [Accepted: 09/07/2023] [Indexed: 10/10/2023]
Abstract
Current standard-of-care for metastatic colorectal cancer patients includes chemotherapy and anti-angiogenic or anti-epidermal growth factor receptor for microsatellite stable tumors and pembrolizumab for microsatellite instable tumors. However, despite the available therapies, the prognosis remains poor. In recent years, new drugs combined with immune checkpoint inhibitors have been tested in microsatellite stable metastatic colorectal cancer patients, but the benefit was modest. Here, we review the metabolic interactions between the immune microenvironment and cancer cells. More specifically, we highlight potential correlatives of tumor immune and metabolic features with transcriptomic classifications such as the Consensus Molecular Subtype. Finally, we discuss the unmet need of immune-metabolic signatures and the value of a new signature (IMMETCOLS) for guiding new strategies in metastatic colorectal cancer. We conclude that the field is ready to propose customized strategies for modifying metabolism and improving immunotherapy and targeted therapy efficacy.
Collapse
Affiliation(s)
- Helena Oliveres
- Medical Oncology Department, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, Barcelona, Spain
| | - Marta Cascante
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Biochemistry and Molecular Biomedicine and Institute of Biomedicine (IBUB), Universitat de Barcelona, Barcelona, Spain.
| | - Joan Maurel
- Medical Oncology Department, Hospital Clinic of Barcelona, Translational Genomics and Targeted Therapeutics in Solid Tumors Group, IDIBAPS, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
25
|
Liu J, Cao X. Glucose metabolism of TAMs in tumor chemoresistance and metastasis. Trends Cell Biol 2023; 33:967-978. [PMID: 37080816 DOI: 10.1016/j.tcb.2023.03.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 04/22/2023]
Abstract
Tumor-associated macrophages (TAMs) are critical in promoting tumor progression and therapeutic resistance. In adapting to metabolic changes in the tumor microenvironment (TME), TAMs reprogram their metabolisms and acquire immunosuppressive and pro-tumor properties. Increased glucose metabolism in TAMs leads to the accumulation of a variety of oncometabolites that exhibit potent tumor-promoting capacity via regulating gene expression and signaling transduction. Glucose uptake also fuels O-GlcNAcylation and other post-translational modifications to promote pro-tumor polarization and function of TAMs. Glucose metabolism coordinates interactions between TAMs and various types of cells in the TME, creating a complex network that facilitates tumor progression. Targeting glucose metabolism represents a promising strategy to switch TAMs from pro-tumor toward anti-tumor function for cancer therapy.
Collapse
Affiliation(s)
- Juan Liu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai 200433, China.
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai 200433, China; Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China; Institute of Immunology, College of Life Sciences, Nankai University, Tianjin 300071, China.
| |
Collapse
|
26
|
Jin J, Zhao Q, Wei Z, Chen K, Su Y, Hu X, Peng X. Glycolysis-cholesterol metabolic axis in immuno-oncology microenvironment: emerging role in immune cells and immunosuppressive signaling. Cell Biosci 2023; 13:189. [PMID: 37828561 PMCID: PMC10571292 DOI: 10.1186/s13578-023-01138-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023] Open
Abstract
Cell proliferation and function require nutrients, energy, and biosynthesis activity to duplicate repertoires for each daughter. It is therefore not surprising that tumor microenvironment (TME) metabolic reprogramming primarily orchestrates the interaction between tumor and immune cells. Tumor metabolic reprogramming affords bioenergetic, signaling intermediates, and biosynthesis requirements for both malignant and immune cells. Different immune cell subsets are recruited into the TME, and these manifestations have distinct effects on tumor progression and therapeutic outcomes, especially the mutual contribution of glycolysis and cholesterol metabolism. In particularly, glycolysis-cholesterol metabolic axis interconnection plays a critical role in the TME modulation, and their changes in tumor metabolism appear to be a double-edged sword in regulating various immune cell responses and immunotherapy efficacy. Hence, we discussed the signature manifestation of the glycolysis-cholesterol metabolic axis and its pivotal role in tumor immune regulation. We also highlight how hypothetical combinations of immunotherapy and glycolysis/cholesterol-related metabolic interventions unleash the potential of anti-tumor immunotherapies, as well as developing more effective personalized treatment strategies.
Collapse
Affiliation(s)
- Jing Jin
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Qijie Zhao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhigong Wei
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Keliang Chen
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Yonglin Su
- Department of Rehabilitation, Cancer Center, West China Hospital, Sichuan University, Sichuan, People's Republic of China.
| | - Xiaolin Hu
- Department of Nursing, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| |
Collapse
|
27
|
Bennett CL, Perona-Wright G. Metabolic adaption of mucosal macrophages: Is metabolism a driver of persistence across tissues? Mucosal Immunol 2023; 16:753-763. [PMID: 37385586 PMCID: PMC10564628 DOI: 10.1016/j.mucimm.2023.06.006] [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: 02/21/2023] [Revised: 05/27/2023] [Accepted: 06/17/2023] [Indexed: 07/01/2023]
Abstract
Macrophages play essential roles in tissue homeostasis, defense, and repair. Their functions are highly tissue-specific, and when damage and inflammation stimulate repopulation by circulating monocytes, the incoming monocytes rapidly acquire the same, tissue-specific functions as the previous, resident macrophages. Several environmental factors are thought to guide the functional differentiation of recruited monocytes, including metabolic pressures imposed by the fuel sources available in each tissue. Here we discuss whether such a model of metabolic determinism can be applied to macrophage differentiation across barrier sites, from the lung to the skin. We suggest an alternative model, in which metabolic phenotype is a consequence of macrophage longevity rather than an early driver of tissue-specific adaption.
Collapse
Affiliation(s)
- Clare L Bennett
- Department of Haematology, UCL Cancer Institute, University College London, London, UK.
| | | |
Collapse
|
28
|
Zeng W, Li F, Jin S, Ho PC, Liu PS, Xie X. Functional polarization of tumor-associated macrophages dictated by metabolic reprogramming. J Exp Clin Cancer Res 2023; 42:245. [PMID: 37740232 PMCID: PMC10517486 DOI: 10.1186/s13046-023-02832-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
Macrophages are highly plastic in different tissues and can differentiate into functional subpopulations under different stimuli. Tumor-associated macrophages (TAMs) are one of the most important innate immune cells implicated in the establishment of an immunosuppressive tumor microenvironment (TME). Recent evidence pinpoints the critical role of metabolic reprogramming in dictating pro-tumorigenic functions of TAMs. Both tumor cells and macrophages undergo metabolic reprogramming to meet energy demands in the TME. Understanding the metabolic rewiring in TAMs can shed light on immune escape mechanisms and provide insights into repolarizing TAMs towards anti-tumorigenic function. Here, we discuss how metabolism impinges on the functional divergence of macrophages and its relevance to macrophage polarization in the TME.
Collapse
Affiliation(s)
- Wentao Zeng
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Fei Li
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Shikai Jin
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Ping-Chih Ho
- Department of Fundamental Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Ludwig Lausanne Branch, Lausanne, Switzerland
| | - Pu-Ste Liu
- Institute of Cellular and System Medicine, National Health Research Institute, Miaoli, Taiwan, ROC
| | - Xin Xie
- School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
29
|
Heieis GA, Patente TA, Almeida L, Vrieling F, Tak T, Perona-Wright G, Maizels RM, Stienstra R, Everts B. Metabolic heterogeneity of tissue-resident macrophages in homeostasis and during helminth infection. Nat Commun 2023; 14:5627. [PMID: 37699869 PMCID: PMC10497597 DOI: 10.1038/s41467-023-41353-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/01/2023] [Indexed: 09/14/2023] Open
Abstract
Tissue-resident macrophage populations constitute a mosaic of phenotypes, yet how their metabolic states link to the range of phenotypes and functions in vivo is still poorly defined. Here, using high-dimensional spectral flow cytometry, we observe distinct metabolic profiles between different organs and functionally link acetyl CoA carboxylase activity to efferocytotic capacity. Additionally, differences in metabolism are evident within populations from a specific site, corresponding to relative stages of macrophage maturity. Immune perturbation with intestinal helminth infection increases alternative activation and metabolic rewiring of monocyte-derived macrophage populations, while resident TIM4+ intestinal macrophages remain immunologically and metabolically hyporesponsive. Similar metabolic signatures in alternatively-activated macrophages are seen from different tissues using additional helminth models, but to different magnitudes, indicating further tissue-specific contributions to metabolic states. Thus, our high-dimensional, flow-based metabolic analyses indicates complex metabolic heterogeneity and dynamics of tissue-resident macrophage populations at homeostasis and during helminth infection.
Collapse
Affiliation(s)
- Graham A Heieis
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| | - Thiago A Patente
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Luís Almeida
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Frank Vrieling
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708WE, Wageningen, The Netherlands
| | - Tamar Tak
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Georgia Perona-Wright
- School of Infection and Immunity, University of Glasgow, 120 University Place, G12 8TA, Glasgow, UK
| | - Rick M Maizels
- School of Infection and Immunity, University of Glasgow, 120 University Place, G12 8TA, Glasgow, UK
| | - Rinke Stienstra
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, 6708WE, Wageningen, The Netherlands
| | - Bart Everts
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
30
|
Zhang J, Koolmeister C, Han J, Filograna R, Hanke L, Àdori M, Sheward DJ, Teifel S, Gopalakrishna S, Shao Q, Liu Y, Zhu K, Harris RA, McInerney G, Murrell B, Aoun M, Bäckdahl L, Holmdahl R, Pekalski M, Wedell A, Engvall M, Wredenberg A, Karlsson Hedestam GB, Castro Dopico X, Rorbach J. Antigen receptor stimulation induces purifying selection against pathogenic mitochondrial tRNA mutations. JCI Insight 2023; 8:e167656. [PMID: 37681412 PMCID: PMC10544217 DOI: 10.1172/jci.insight.167656] [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: 12/06/2022] [Accepted: 07/27/2023] [Indexed: 09/09/2023] Open
Abstract
Pathogenic mutations in mitochondrial (mt) tRNA genes that compromise oxidative phosphorylation (OXPHOS) exhibit heteroplasmy and cause a range of multisyndromic conditions. Although mitochondrial disease patients are known to suffer from abnormal immune responses, how heteroplasmic mtDNA mutations affect the immune system at the molecular level is largely unknown. Here, in mice carrying pathogenic C5024T in mt-tRNAAla and in patients with mitochondrial encephalomyopathy, lactic acidosis, stroke-like episodes (MELAS) syndrome carrying A3243G in mt-tRNALeu, we found memory T and B cells to have lower pathogenic mtDNA mutation burdens than their antigen-inexperienced naive counterparts, including after vaccination. Pathogenic burden reduction was less pronounced in myeloid compared with lymphoid lineages, despite C5024T compromising macrophage OXPHOS capacity. Rapid dilution of the C5024T mutation in T and B cell cultures could be induced by antigen receptor-triggered proliferation and was accelerated by metabolic stress conditions. Furthermore, we found C5024T to dysregulate CD8+ T cell metabolic remodeling and IFN-γ production after activation. Together, our data illustrate that the generation of memory lymphocytes shapes the mtDNA landscape, wherein pathogenic variants dysregulate the immune response.
Collapse
Affiliation(s)
- Jingdian Zhang
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Camilla Koolmeister
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Jinming Han
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Roberta Filograna
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Leo Hanke
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Àdori
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Daniel J. Sheward
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Sina Teifel
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shreekara Gopalakrishna
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Qiuya Shao
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Yong Liu
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Keying Zhu
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Robert A. Harris
- Applied Immunology and Immunotherapy, Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Gerald McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ben Murrell
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mike Aoun
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Liselotte Bäckdahl
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Rikard Holmdahl
- Division of Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Marcin Pekalski
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Wedell
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Engvall
- Center for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Wredenberg
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
- Center for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden
| | | | - Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Joanna Rorbach
- Department of Medical Biochemistry and Biophysics, and
- Max Planck Institute Biology of Ageing-Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
31
|
Bied M, Ho WW, Ginhoux F, Blériot C. Roles of macrophages in tumor development: a spatiotemporal perspective. Cell Mol Immunol 2023; 20:983-992. [PMID: 37429944 PMCID: PMC10468537 DOI: 10.1038/s41423-023-01061-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/16/2023] [Indexed: 07/12/2023] Open
Abstract
Macrophages are critical regulators of tissue homeostasis but are also abundant in the tumor microenvironment (TME). In both primary tumors and metastases, such tumor-associated macrophages (TAMs) seem to support tumor development. While we know that TAMs are the dominant immune cells in the TME, their vast heterogeneity and associated functions are only just being unraveled. In this review, we outline the various known TAM populations found thus far and delineate their specialized roles associated with the main stages of cancer progression. We discuss how macrophages may prime the premetastatic niche to enable the growth of a metastasis and then how subsequent metastasis-associated macrophages can support secondary tumor growth. Finally, we speculate on the challenges that remain to be overcome in TAM research.
Collapse
Affiliation(s)
- Mathilde Bied
- Institut Gustave Roussy, INSERM U1015, Villejuif, France
| | - William W Ho
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore
| | - Florent Ginhoux
- Institut Gustave Roussy, INSERM U1015, Villejuif, France.
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A∗STAR), Singapore, Singapore.
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China.
- Translational Immunology Institute, SingHealth Duke-NUS, Singapore, Singapore.
| | - Camille Blériot
- Institut Gustave Roussy, INSERM U1015, Villejuif, France.
- Institut Necker des Enfants Malades, INSERM, CNRS, Université Paris Cité, Paris, France.
| |
Collapse
|
32
|
De Simone G, Soldani C, Morabito A, Franceschini B, Ferlan F, Costa G, Pastorelli R, Donadon M, Brunelli L. Implication of metabolism in the polarization of tumor-associated-macrophages: the mass spectrometry-based point of view. Front Immunol 2023; 14:1193235. [PMID: 37503340 PMCID: PMC10368868 DOI: 10.3389/fimmu.2023.1193235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/22/2023] [Indexed: 07/29/2023] Open
Abstract
Tumor-associated macrophages (TAMs) represent one of the main tumor-infiltrating immune cell types and are generally categorized into either of two functionally contrasting subtypes, namely classical activated M1 macrophages and alternatively activated M2 macrophages. TAMs showed different activation states that can be represent by the two extremes of the complex profile of macrophages biology, the M1-like phenotype (pro-inflammatory activity) and the M2-like phenotype (anti-inflammatory activity). Based on the tumor type, and grades, TAMs can acquire different functions and properties; usually, the M1-like phenotype is typical of early tumor stages and is associated to an anti-tumor activity, while M2-like phenotype has a pro-inflammatory activity and is related to a poor patients' prognosis. The classification of macrophages into M1/M2 groups based on well-defined stimuli does not model the infinitely more complex tissue milieu where macrophages (potentially of different origin) would be exposed to multiple signals in different sequential order. This review aims to summarize the recent mass spectrometry-based (MS-based) metabolomics findings about the modifications of metabolism in TAMs polarization in different tumors. The published data shows that MS-based metabolomics is a promising tool to help better understanding TAMs metabolic phenotypes, although it is still poorly applied for TAMs metabolism. The knowledge of key metabolic alterations in TAMs is an essential step for discovering TAMs polarization novel biomarkers and developing novel therapeutic approaches targeting TAM metabolism to repolarize TAMs towards their anti-tumor phenotype.
Collapse
Affiliation(s)
- Giulia De Simone
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biotechnologies and Biosciences, Università degli Studi Milano Bicocca, Milan, Italy
| | - Cristiana Soldani
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Aurelia Morabito
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Barbara Franceschini
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Fabrizio Ferlan
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Guido Costa
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Roberta Pastorelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Matteo Donadon
- Hepatobiliary Immunopathology Laboratory, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy
- Department of General Surgery, University Maggiore Hospital, Novara, Italy
| | - Laura Brunelli
- Laboratory of Metabolites and Proteins in Translational Research, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| |
Collapse
|
33
|
Zhang Z, Zhu H, Dang P, Wang J, Chang W, Wang X, Alghamdi N, Lu A, Zang Y, Wu W, Wang Y, Zhang Y, Cao S, Zhang C. FLUXestimator: a webserver for predicting metabolic flux and variations using transcriptomics data. Nucleic Acids Res 2023; 51:W180-W190. [PMID: 37216602 PMCID: PMC10320190 DOI: 10.1093/nar/gkad444] [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: 03/28/2023] [Revised: 04/29/2023] [Accepted: 05/11/2023] [Indexed: 05/24/2023] Open
Abstract
Quantitative assessment of single cell fluxome is critical for understanding the metabolic heterogeneity in diseases. Unfortunately, laboratory-based single cell fluxomics is currently impractical, and the current computational tools for flux estimation are not designed for single cell-level prediction. Given the well-established link between transcriptomic and metabolomic profiles, leveraging single cell transcriptomics data to predict single cell fluxome is not only feasible but also an urgent task. In this study, we present FLUXestimator, an online platform for predicting metabolic fluxome and variations using single cell or general transcriptomics data of large sample-size. The FLUXestimator webserver implements a recently developed unsupervised approach called single cell flux estimation analysis (scFEA), which uses a new neural network architecture to estimate reaction rates from transcriptomics data. To the best of our knowledge, FLUXestimator is the first web-based tool dedicated to predicting cell-/sample-wise metabolic flux and metabolite variations using transcriptomics data of human, mouse and 15 other common experimental organisms. The FLUXestimator webserver is available at http://scFLUX.org/, and stand-alone tools for local use are available at https://github.com/changwn/scFEA. Our tool provides a new avenue for studying metabolic heterogeneity in diseases and has the potential to facilitate the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Zixuan Zhang
- College of Software, College of Computer Science and Technology, Jilin University, Changchun 130012, China
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Haiqi Zhu
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Computer Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Pengtao Dang
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Electric Computer Engineering, Purdue University, Indianapolis, IN 46202, USA
| | - Jia Wang
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Computer Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Wennan Chang
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Xiao Wang
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Computer Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Norah Alghamdi
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alex Lu
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yong Zang
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wenzhuo Wu
- Department of Industrial Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Yijie Wang
- Department of Computer Sciences, Indiana University, Bloomington, IN 47405, USA
| | - Yu Zhang
- College of Software, College of Computer Science and Technology, Jilin University, Changchun 130012, China
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sha Cao
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics and Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
34
|
Wu H, Huang H, Zhao Y. Interplay between metabolic reprogramming and post-translational modifications: from glycolysis to lactylation. Front Immunol 2023; 14:1211221. [PMID: 37457701 PMCID: PMC10338923 DOI: 10.3389/fimmu.2023.1211221] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
Cellular metabolism plays a critical role in determining the fate and function of cells. Metabolic reprogramming and its byproducts have a complex impact on cellular activities. In quiescent T cells, oxidative phosphorylation (OXPHOS) is the primary pathway for survival. However, upon antigen activation, T cells undergo rapid metabolic reprogramming, characterized by an elevation in both glycolysis and OXPHOS. While both pathways are induced, the balance predominantly shifts towards glycolysis, enabling T cells to rapidly proliferate and enhance their functionality, representing the most distinctive signature during activation. Metabolic processes generate various small molecules resulting from enzyme-catalyzed reactions, which also modulate protein function and exert regulatory control. Notably, recent studies have revealed the direct modification of histones, known as lactylation, by lactate derived from glycolysis. This lactylation process influences gene transcription and adds a novel variable to the regulation of gene expression. Protein lactylation has been identified as an essential mechanism by which lactate exerts its diverse functions, contributing to crucial biological processes such as uterine remodeling, tumor proliferation, neural system regulation, and metabolic regulation. This review focuses on the metabolic reprogramming of T cells, explores the interplay between lactate and the immune system, highlights the impact of lactylation on cellular function, and elucidates the intersection of metabolic reprogramming and epigenetics.
Collapse
Affiliation(s)
- Hengwei Wu
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, People's Government of Zhejiang Province, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - He Huang
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, People's Government of Zhejiang Province, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Yanmin Zhao
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Institute of Hematology, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, People's Government of Zhejiang Province, Hangzhou, Zhejiang, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Nabhan M, Egan D, Kreileder M, Zhernovkov V, Timosenko E, Slidel T, Dovedi S, Glennon K, Brennan D, Kolch W. Deciphering the tumour immune microenvironment cell by cell. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 18:100383. [PMID: 37234284 PMCID: PMC10206805 DOI: 10.1016/j.iotech.2023.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have rejuvenated therapeutic approaches in oncology. Although responses tend to be durable, response rates vary in many cancer types. Thus, the identification and validation of predictive biomarkers is a key clinical priority, the answer to which is likely to lie in the tumour microenvironment (TME). A wealth of data demonstrates the huge impact of the TME on ICI response and resistance. However, these data also reveal the complexity of the TME composition including the spatiotemporal interactions between different cell types and their dynamic changes in response to ICIs. Here, we briefly review some of the modalities that sculpt the TME, in particular the metabolic milieu, hypoxia and the role of cancer-associated fibroblasts. We then discuss recent approaches to dissect the TME with a focus on single-cell RNA sequencing, spatial transcriptomics and spatial proteomics. We also discuss some of the clinically relevant findings these multi-modal analyses have yielded.
Collapse
Affiliation(s)
- M. Nabhan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - D. Egan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - M. Kreileder
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - V. Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
| | - E. Timosenko
- ICC, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, , UK
| | - T. Slidel
- Oncology Data Science, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - S. Dovedi
- ICC, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, , UK
| | - K. Glennon
- UCD Gynaecological Oncology Group, UCD School of Medicine Mater Misericordiae University Hospital, Dublin, Ireland
| | - D. Brennan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
- UCD Gynaecological Oncology Group, UCD School of Medicine Mater Misericordiae University Hospital, Dublin, Ireland
| | - W. Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Ireland
| |
Collapse
|
36
|
Chaudagar K, Hieromnimon HM, Kelley A, Labadie B, Shafran J, Rameshbabu S, Drovetsky C, Bynoe K, Solanki A, Markiewicz E, Fan X, Loda M, Patnaik A. Suppression of tumor cell lactate-generating signaling pathways eradicates murine PTEN/p53-deficient aggressive-variant prostate cancer via macrophage phagocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.540590. [PMID: 37292972 PMCID: PMC10245812 DOI: 10.1101/2023.05.23.540590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Purpose PTEN loss-of-function/PI3K pathway hyperactivation occurs in ∼50% of metastatic, castrate-resistant prostate cancer patients, resulting in poor therapeutic outcomes and resistance to immune checkpoint inhibitors across multiple malignancies. Our prior studies in prostate-specific PTEN/p53-deleted genetically engineered mice (Pb-Cre;PTEN fl/fl Trp53 fl/fl GEM) with aggressive-variant prostate cancer (AVPC) demonstrated feedback Wnt/β-catenin signaling activation in 40% mice resistant to androgen deprivation therapy (ADT)/PI3K inhibitor (PI3Ki)/PD-1 antibody (aPD-1) combination, resulting in restoration of lactate cross-talk between tumor-cells and tumor-associated macrophages (TAM), histone lactylation (H3K18lac) and phagocytic suppression within TAM. Here, we targeted immunometabolic mechanism(s) of resistance to ADT/PI3Ki/aPD-1 combination, with the goal of durable tumor control in PTEN/p53-deficient PC. Experimental design Pb-Cre;PTEN fl/fl Trp53 fl/fl GEM were treated with either ADT (degarelix), PI3Ki (copanlisib), aPD-1, MEK inhibitor (trametinib) or Porcupine inhibitor (LGK 974) as single agents or their combinations. MRI was used to monitor tumor kinetics and immune/proteomic profiling/ ex vivo co-culture mechanistic studies were performed on prostate tumors or established GEM-derived cell lines. Results We tested whether Wnt/β-catenin pathway inhibition with LGK 974 addition to degarelix/copanlisib/aPD-1 therapy enhances tumor control in GEM, and observed de novo resistance due to feedback activation of MEK signaling. Based on our observation that degarelix/aPD-1 treatment resulted in partial inhibition of MEK signaling, we substituted trametinib for degarelix/aPD-1 treatment, and observed a durable tumor growth control of PI3Ki/MEKi/PORCNi in 100% mice via H3K18lac suppression and complete TAM activation within TME. Conclusions Abrogation of lactate-mediated cross-talk between cancer cells and TAM results in durable ADT-independent tumor control in PTEN/p53-deficient AVPC, and warrants further investigation in clinical trials. STATEMENT OF TRANSLATIONAL RELEVANCE PTEN loss-of-function occurs in ∼50% of mCRPC patients, and associated with poor prognosis, and immune checkpoint inhibitor resistance across multiple malignancies. Our prior studies have demonstrated that ADT/PI3Ki/PD-1 triplet combination therapy controls PTEN/p53-deficient PC in 60% of mice via enhancement of TAM phagocytosis. Here, we discovered that resistance to ADT/PI3K/PD-1 therapy occurred via restoration of lactate production via feedback Wnt/MEK signaling following treatment with PI3Ki, resulting in inhibition of TAM phagocytosis. Critically, co-targeting of PI3K/MEK/Wnt signaling pathways using an intermittent dosing schedule of corresponding targeted agents resulted in complete tumor control and significantly prolonged survival without significant long-term toxicity. Collectively, our findings provide "proof-of-concept" that targeting lactate as a macrophage phagocytic checkpoint controls growth of murine PTEN/p53-deficient PC and warrant further investigation in AVPC clinical trials.
Collapse
Affiliation(s)
- Kiranj Chaudagar
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Hanna M. Hieromnimon
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Anne Kelley
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Brian Labadie
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jordan Shafran
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Srikrishnan Rameshbabu
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Catherine Drovetsky
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Kaela Bynoe
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL, USA
| | | | - Xiaobing Fan
- Department of Radiology, University of Chicago, Chicago IL, USA
| | - Massimo Loda
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Akash Patnaik
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
37
|
Chaudagar K, Hieromnimon HM, Khurana R, Labadie B, Hirz T, Mei S, Hasan R, Shafran J, Kelley A, Apostolov E, Al-Eryani G, Harvey K, Rameshbabu S, Loyd M, Bynoe K, Drovetsky C, Solanki A, Markiewicz E, Zamora M, Fan X, Schürer S, Swarbrick A, Sykes DB, Patnaik A. Reversal of Lactate and PD-1-mediated Macrophage Immunosuppression Controls Growth of PTEN/p53-deficient Prostate Cancer. Clin Cancer Res 2023; 29:1952-1968. [PMID: 36862086 PMCID: PMC10192075 DOI: 10.1158/1078-0432.ccr-22-3350] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/28/2022] [Accepted: 02/24/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE Phosphatase and tensin homolog (PTEN) loss of function occurs in approximately 50% of patients with metastatic castrate-resistant prostate cancer (mCRPC), and is associated with poor prognosis and responsiveness to standard-of-care therapies and immune checkpoint inhibitors. While PTEN loss of function hyperactivates PI3K signaling, combinatorial PI3K/AKT pathway and androgen deprivation therapy (ADT) has demonstrated limited anticancer efficacy in clinical trials. Here, we aimed to elucidate mechanism(s) of resistance to ADT/PI3K-AKT axis blockade, and to develop rational combinatorial strategies to effectively treat this molecular subset of mCRPC. EXPERIMENTAL DESIGN Prostate-specific PTEN/p53-deficient genetically engineered mice (GEM) with established 150-200 mm3 tumors, as assessed by ultrasound, were treated with either ADT (degarelix), PI3K inhibitor (copanlisib), or anti-PD-1 antibody (aPD-1), as single agents or their combinations, and tumors were monitored by MRI and harvested for immune, transcriptomic, and proteomic profiling, or ex vivo co-culture studies. Single-cell RNA sequencing on human mCRPC samples was performed using 10X Genomics platform. RESULTS Coclinical trials in PTEN/p53-deficient GEM revealed that recruitment of PD-1-expressing tumor-associated macrophages (TAM) thwarts ADT/PI3Ki combination-induced tumor control. The addition of aPD-1 to ADT/PI3Ki combination led to TAM-dependent approximately 3-fold increase in anticancer responses. Mechanistically, decreased lactate production from PI3Ki-treated tumor cells suppressed histone lactylation within TAM, resulting in their anticancer phagocytic activation, which was augmented by ADT/aPD-1 treatment and abrogated by feedback activation of Wnt/β-catenin pathway. Single-cell RNA-sequencing analysis in mCRPC patient biopsy samples revealed a direct correlation between high glycolytic activity and TAM phagocytosis suppression. CONCLUSIONS Immunometabolic strategies that reverse lactate and PD-1-mediated TAM immunosuppression, in combination with ADT, warrant further investigation in patients with PTEN-deficient mCRPC.
Collapse
Affiliation(s)
- Kiranj Chaudagar
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Hanna M. Hieromnimon
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Rimpi Khurana
- Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Brian Labadie
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Taghreed Hirz
- Center for Regenerative Medicine, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Shenglin Mei
- Center for Regenerative Medicine, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Raisa Hasan
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Jordan Shafran
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Anne Kelley
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Eva Apostolov
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Ghamdan Al-Eryani
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Kate Harvey
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Srikrishnan Rameshbabu
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Mayme Loyd
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Kaela Bynoe
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Catherine Drovetsky
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Ani Solanki
- Animal Resource Center, University of Chicago, Chicago, IL, USA
| | | | - Marta Zamora
- Department of Radiology, University of Chicago, Chicago IL, USA
| | - Xiaobing Fan
- Department of Radiology, University of Chicago, Chicago IL, USA
| | - Stephan Schürer
- Department of Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alex Swarbrick
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- St Vincent’s Clinical School, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - David B. Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Akash Patnaik
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL, USA
| |
Collapse
|
38
|
Mortazavi Farsani SS, Verma V. Lactate mediated metabolic crosstalk between cancer and immune cells and its therapeutic implications. Front Oncol 2023; 13:1175532. [PMID: 37234972 PMCID: PMC10206240 DOI: 10.3389/fonc.2023.1175532] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Metabolism is central to energy generation and cell signaling in all life forms. Cancer cells rely heavily on glucose metabolism wherein glucose is primarily converted to lactate even in adequate oxygen conditions, a process famously known as "the Warburg effect." In addition to cancer cells, Warburg effect was found to be operational in other cell types, including actively proliferating immune cells. According to current dogma, pyruvate is the end product of glycolysis that is converted into lactate in normal cells, particularly under hypoxic conditions. However, several recent observations suggest that the final product of glycolysis may be lactate, which is produced irrespective of oxygen concentrations. Traditionally, glucose-derived lactate can have three fates: it can be used as a fuel in the TCA cycle or lipid synthesis; it can be converted back into pyruvate in the cytosol that feeds into the mitochondrial TCA; or, at very high concentrations, accumulated lactate in the cytosol may be released from cells that act as an oncometabolite. In immune cells as well, glucose-derived lactate seems to play a major role in metabolism and cell signaling. However, immune cells are much more sensitive to lactate concentrations, as higher lactate levels have been found to inhibit immune cell function. Thus, tumor cell-derived lactate may serve as a major player in deciding the response and resistance to immune cell-directed therapies. In the current review, we will provide a comprehensive overview of the glycolytic process in eukaryotic cells with a special focus on the fate of pyruvate and lactate in tumor and immune cells. We will also review the evidence supporting the idea that lactate, not pyruvate, is the end product of glycolysis. In addition, we will discuss the impact of glucose-lactate-mediated cross-talk between tumor and immune cells on the therapeutic outcomes after immunotherapy.
Collapse
Affiliation(s)
- Seyedeh Sahar Mortazavi Farsani
- Section of Cancer Immunotherapy and Immune Metabolism, The Hormel Institute, University of Minnesota, Austin, MN, United States
| | - Vivek Verma
- Section of Cancer Immunotherapy and Immune Metabolism, The Hormel Institute, University of Minnesota, Austin, MN, United States
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
39
|
Mouton AJ, do Carmo JM, da Silva AA, Omoto ACM, Hall JE. Targeting immunometabolism during cardiorenal injury: roles of conventional and alternative macrophage metabolic fuels. Front Physiol 2023; 14:1139296. [PMID: 37234412 PMCID: PMC10208225 DOI: 10.3389/fphys.2023.1139296] [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: 01/06/2023] [Accepted: 04/14/2023] [Indexed: 05/28/2023] Open
Abstract
Macrophages play critical roles in mediating and resolving tissue injury as well as tissue remodeling during cardiorenal disease. Altered immunometabolism, particularly macrophage metabolism, is a critical underlying mechanism of immune dysfunction and inflammation, particularly in individuals with underlying metabolic abnormalities. In this review, we discuss the critical roles of macrophages in cardiac and renal injury and disease. We also highlight the roles of macrophage metabolism and discuss metabolic abnormalities, such as obesity and diabetes, which may impair normal macrophage metabolism and thus predispose individuals to cardiorenal inflammation and injury. As the roles of macrophage glucose and fatty acid metabolism have been extensively discussed elsewhere, we focus on the roles of alternative fuels, such as lactate and ketones, which play underappreciated roles during cardiac and renal injury and heavily influence macrophage phenotypes.
Collapse
Affiliation(s)
- Alan J. Mouton
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Jussara M. do Carmo
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Alexandre A. da Silva
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - Ana C. M. Omoto
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| | - John E. Hall
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, United States
- Mississippi Center for Obesity Research, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
40
|
Abstract
Over the last decade, immunometabolism has emerged as a novel interdisciplinary field of research and yielded significant fundamental insights into the regulation of immune responses. Multiple classical approaches to interrogate immunometabolism, including bulk metabolic profiling and analysis of metabolic regulator expression, paved the way to appreciating the physiological complexity of immunometabolic regulation in vivo. Studying immunometabolism at the systems level raised the need to transition towards the next-generation technology for metabolic profiling and analysis. Spatially resolved metabolic imaging and computational algorithms for multi-modal data integration are new approaches to connecting metabolism and immunity. In this review, we discuss recent studies that highlight the complex physiological interplay between immune responses and metabolism and give an overview of technological developments that bear the promise of capturing this complexity most directly and comprehensively.
Collapse
Affiliation(s)
- Denis A Mogilenko
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Current affiliation: Department of Medicine, Department of Pathology, Microbiology, and Immunology, and Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, USA;
| | - Alexey Sergushichev
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA; ,
| |
Collapse
|
41
|
Kabat AM, Pearce EL, Pearce EJ. Metabolism in type 2 immune responses. Immunity 2023; 56:723-741. [PMID: 37044062 PMCID: PMC10938369 DOI: 10.1016/j.immuni.2023.03.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/11/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023]
Abstract
The immune response is tailored to the environment in which it takes place. Immune cells sense and adapt to changes in their surroundings, and it is now appreciated that in addition to cytokines made by stromal and epithelial cells, metabolic cues provide key adaptation signals. Changes in immune cell activation states are linked to changes in cellular metabolism that support function. Furthermore, metabolites themselves can signal between as well as within cells. Here, we discuss recent progress in our understanding of how metabolic regulation relates to type 2 immunity firstly by considering specifics of metabolism within type 2 immune cells and secondly by stressing how type 2 immune cells are integrated more broadly into the metabolism of the organism as a whole.
Collapse
Affiliation(s)
- Agnieszka M Kabat
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Erika L Pearce
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA
| | - Edward J Pearce
- Bloomberg Kimmel Institute, and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21287, USA.
| |
Collapse
|
42
|
Chaib M, Tanveer UA, Makowski L. Myeloid cells in the era of cancer immunotherapy: Top 3 unanswered questions. Pharmacol Ther 2023; 244:108370. [PMID: 36871784 PMCID: PMC10798582 DOI: 10.1016/j.pharmthera.2023.108370] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/09/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Myeloid cells are increasingly being recognized as central players orchestrating or suppressing antitumor immune responses. With the advent of high-resolution analytical methods such as single-cell technologies, we now appreciate the heterogeneity and complexity of the myeloid compartment in the context of cancer. Because of their highly plastic nature, targeting myeloid cells has shown promising results either as a monotherapy or in combination with immunotherapy in preclinical models and cancer patients. However, the complexity of myeloid cell cellular crosstalk and molecular networks contributes to our poor understanding of the different myeloid cell subsets in tumorigenesis, which makes targeting myeloid cells challenging. Here, we summarize varied myeloid cell subsets and their contribution to tumor progression with a main focus on mononuclear phagocytes. The top three unanswered questions challenging the field of myeloid cells and cancer in the era of cancer immunotherapy are addressed. Through these questions, we discuss how myeloid cell origin and identity influence their function and disease outcomes. Different therapeutic strategies used to target myeloid cells in cancer are also addressed. Finally, the durability of myeloid cell targeting is interrogated by examining the complexity of resultant compensatory cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ubaid A Tanveer
- Division of Hematology Oncology, Department of Medicine, College of Medicine, USA; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Liza Makowski
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Division of Hematology Oncology, Department of Medicine, College of Medicine, USA; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
43
|
Abstract
Few metabolites can claim a more central and versatile role in cell metabolism than acetyl coenzyme A (acetyl-CoA). Acetyl-CoA is produced during nutrient catabolism to fuel the tricarboxylic acid cycle and is the essential building block for fatty acid and isoprenoid biosynthesis. It also functions as a signalling metabolite as the substrate for lysine acetylation reactions, enabling the modulation of protein functions in response to acetyl-CoA availability. Recent years have seen exciting advances in our understanding of acetyl-CoA metabolism in normal physiology and in cancer, buoyed by new mouse models, in vivo stable-isotope tracing approaches and improved methods for measuring acetyl-CoA, including in specific subcellular compartments. Efforts to target acetyl-CoA metabolic enzymes are also advancing, with one therapeutic agent targeting acetyl-CoA synthesis receiving approval from the US Food and Drug Administration. In this Review, we give an overview of the regulation and cancer relevance of major metabolic pathways in which acetyl-CoA participates. We further discuss recent advances in understanding acetyl-CoA metabolism in normal tissues and tumours and the potential for targeting these pathways therapeutically. We conclude with a commentary on emerging nodes of acetyl-CoA metabolism that may impact cancer biology.
Collapse
Affiliation(s)
- David A Guertin
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, USA.
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
44
|
Lactate induces tumor-associated macrophage polarization independent of mitochondrial pyruvate carrier-mediated metabolism. Int J Biol Macromol 2023; 237:123810. [PMID: 36868333 DOI: 10.1016/j.ijbiomac.2023.123810] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/02/2022] [Accepted: 02/18/2023] [Indexed: 03/05/2023]
Abstract
Tumor cell-derived lactate has been recognized as the key driver of polarization in tumor-associated macrophages (TAMs). Intratumoral lactate can be transported into macrophages to fuel the TCA cycle, which is mediated by mitochondrial pyruvate carrier (MPC). At the heart of intracellular metabolism, MPC-mediated transport has been investigated in studies which suggested its role and importance in the process of TAMs polarization. However, previous studies relied on pharmacological inhibition instead of genetic approaches to evaluate the role of MPC in TAMs polarization. Here, we demonstrated that genetic depletion of MPC blocks the entry of lactate into mitochondria in macrophages. However, MPC-mediated metabolism was dispensable for IL-4/lactate-induced macrophages polarization as well as tumor growth. In addition, MPC depletion had no impact on hypoxia-inducible factor 1α (HIF-1α) stabilization and histone lactylation, both of which are required for TAMs polarization. Our study suggests that lactate itself, rather than its downstream metabolites, is responsible for TAMs polarization.
Collapse
|
45
|
Dias AS, Almeida CR, Helguero LA, Duarte IF. Metabolic Reprogramming of Breast Tumor-Educated Macrophages Revealed by NMR Metabolomics. Cancers (Basel) 2023; 15:cancers15041211. [PMID: 36831553 PMCID: PMC9954003 DOI: 10.3390/cancers15041211] [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: 12/15/2022] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023] Open
Abstract
The metabolic crosstalk between tumor cells and tumor-associated macrophages (TAMs) has emerged as a critical contributor to tumor development and progression. In breast cancer (BC), the abundance of immune-suppressive TAMs positively correlates with poor prognosis. However, little is known about how TAMs reprogram their metabolism in the BC microenvironment. In this work, we have assessed the metabolic and phenotypic impact of incubating THP-1-derived macrophages in conditioned media (CM) from two BC cell lines cultured in normoxia/hypoxia: MDA-MB-231 cells (highly metastatic, triple-negative BC), and MCF-7 cells (less aggressive, luminal BC). The resulting tumor-educated macrophages (TEM) displayed prominent differences in their metabolic activity and composition, compared to control cells (M0), as assessed by exo- and endometabolomics. In particular, TEM turned to the utilization of extracellular pyruvate, alanine, and branched chain keto acids (BCKA), while exhibiting alterations in metabolites associated with several intracellular pathways, including polyamines catabolism (MDA-TEM), collagen degradation (mainly MCF-TEM), adenosine accumulation (mainly MDA-TEM) and lipid metabolism. Interestingly, following a second-stage incubation in fresh RPMI medium, TEM still displayed several metabolic differences compared to M0, indicating persistent reprogramming. Overall, this work provided new insights into the metabolic plasticity of TEM, revealing potentially important nutritional exchanges and immunoregulatory metabolites in the BC TME.
Collapse
Affiliation(s)
- Ana S. Dias
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- iBiMED—Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Catarina R. Almeida
- iBiMED—Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Luisa A. Helguero
- iBiMED—Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Iola F. Duarte
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
- Correspondence: ; Tel.: +351-234-401-418
| |
Collapse
|
46
|
Dubey S, Ghosh S, Goswami D, Ghatak D, De R. Immunometabolic attributes and mitochondria-associated signaling of Tumor-Associated Macrophages in tumor microenvironment modulate cancer progression. Biochem Pharmacol 2023; 208:115369. [PMID: 36481347 DOI: 10.1016/j.bcp.2022.115369] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Macrophages are specialized immune cells, which have the capacity to phagocytize and destroy the target cells, including tumor cells. Some macrophages, however on their way to devour the cancer cells undergo a change due to a complex set of signaling pathways. They are induced to change into a polarized state known as M2. The M2 macrophages help in metastasis, tumor suppression, and angiogenesis. The macrophage which gets associated with this TME, are referred to as tumor-associated macrophages (TAMs). TAMS undergo a metabolic reprogramming toward oxidative metabolism for bioenergetic purposes (OXPHOS), fatty acid oxidation (FAO), decreased glycolysis, decreased metabolism via the PPP, and upregulation of arginase 1 (ARG1) which triggers immunosuppressive pro-tumor signaling in the tumor microenvironment (TME) in which mitochondria plays an instrumental role. Reports have suggested that a complex series of interactions and exchange of materials, such as cytokines, metabolic intermediates and sometimes even transfer of mitochondria take place between TAMS and other TME components most importantly cancer cells that reprogram their metabolism to encourage cell growth, division, epithelial to mesenchymal transition, that ultimately play an important role in tumor progression. This review will try to focus on the crosstalk between the TAMs with several other components of TME, what instrumental role mitochondria play in that and also try to explore some of the therapeutic options available in cancer patients.
Collapse
Affiliation(s)
- Srijan Dubey
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India
| | - Sayak Ghosh
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India
| | - Debosmita Goswami
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India
| | - Debapriya Ghatak
- Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700032, India
| | - Rudranil De
- Amity Institute of Biotechnology, Amity University, Kolkata, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Newtown, Kolkata 700135, West Bengal, India.
| |
Collapse
|
47
|
Knudsen-Clark AM, Cazarin J, Altman BJ. Do macrophages follow the beat of circadian rhythm in TIME (Tumor Immune Microenvironment)? F1000Res 2023; 12:101. [PMID: 37469718 PMCID: PMC10352629 DOI: 10.12688/f1000research.129863.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 07/21/2023] Open
Abstract
Advances in cancer research have made clear the critical role of the immune response in clearing tumors. This breakthrough in scientific understanding was heralded by the success of immune checkpoint blockade (ICB) therapies such as anti-programmed cell death protein 1 (PD-1)/ programmed death-ligand 1 (PD-L1) and anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), as well as the success of chimeric antigen receptor (CAR) T cells in treating liquid tumors. Thus, much effort has been made to further understand the role of the immune response in tumor progression, and how we may target it to treat cancer. Macrophages are a component of the tumor immune microenvironment (TIME) that can promote tumor growth both indirectly, by suppressing T cell responses necessary for tumor killing, as well as directly, through deposition of extracellular matrix and promotion of angiogenesis. Thus, understanding regulation of macrophages within the tumor microenvironment (TME) is key to targeting them for immunotherapy. However, circadian rhythms (24-hour cycles) are a fundamental aspect of macrophage biology that have yet to be investigated for their role in macrophage-mediated suppression of the anti-tumor immune response Circadian rhythms regulate macrophage-mediated immune responses through time-of-day-dependent regulation of macrophage function. A better understanding of the circadian biology of macrophages in the context of the TME may allow us to exploit synergy between existing and upcoming treatments and circadian regulation of immunity.
Collapse
Affiliation(s)
- Amelia M. Knudsen-Clark
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14620, USA
| | - Juliana Cazarin
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14620, USA
| | - Brian J. Altman
- Department of Biomedical Genetics, University of Rochester School of Medicine and Dentistry, Rochester, NY, 14620, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY, 14620, USA
| |
Collapse
|
48
|
Hallmarks of Cancer Affected by the MIF Cytokine Family. Cancers (Basel) 2023; 15:cancers15020395. [PMID: 36672343 PMCID: PMC9856758 DOI: 10.3390/cancers15020395] [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: 11/25/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
New diagnostic methods and treatments have significantly decreased the mortality rates of cancer patients, but further improvements are warranted based on the identification of novel tumor-promoting molecules that can serve as therapeutic targets. The macrophage migration inhibitory factor (MIF) family of cytokines, comprising MIF and DDT (also known as MIF2), are overexpressed in almost all cancer types, and their high expressions are related to a worse prognosis for the patients. MIF is involved in 9 of the 10 hallmarks of cancer, and its inhibition by antibodies, nanobodies, or small synthetic molecules has shown promising results. Even though DDT is also proposed to be involved in several of the hallmarks of cancer, the available information about its pro-tumoral role and mechanism of action is more limited. Here, we provide an overview of the involvement of both MIF and DDT in cancer, and we propose that blocking both cytokines is needed to obtain the maximum anti-tumor response.
Collapse
|
49
|
Khan F, Pang L, Dunterman M, Lesniak MS, Heimberger AB, Chen P. Macrophages and microglia in glioblastoma: heterogeneity, plasticity, and therapy. J Clin Invest 2023; 133:163446. [PMID: 36594466 PMCID: PMC9797335 DOI: 10.1172/jci163446] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive tumor in the central nervous system and contains a highly immunosuppressive tumor microenvironment (TME). Tumor-associated macrophages and microglia (TAMs) are a dominant population of immune cells in the GBM TME that contribute to most GBM hallmarks, including immunosuppression. The understanding of TAMs in GBM has been limited by the lack of powerful tools to characterize them. However, recent progress on single-cell technologies offers an opportunity to precisely characterize TAMs at the single-cell level and identify new TAM subpopulations with specific tumor-modulatory functions in GBM. In this Review, we discuss TAM heterogeneity and plasticity in the TME and summarize current TAM-targeted therapeutic potential in GBM. We anticipate that the use of single-cell technologies followed by functional studies will accelerate the development of novel and effective TAM-targeted therapeutics for GBM patients.
Collapse
|
50
|
Wei J, Yu W, Chen J, Huang G, Zhang L, Chen Z, Hu M, Gong X, Du H. Single-cell and spatial analyses reveal the association between gene expression of glutamine synthetase with the immunosuppressive phenotype of APOE+CTSZ+TAM in cancers. Mol Oncol 2023; 17:611-628. [PMID: 36587392 PMCID: PMC10061288 DOI: 10.1002/1878-0261.13373] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/28/2022] [Accepted: 12/30/2022] [Indexed: 01/02/2023] Open
Abstract
An immunosuppressive state is regulated by various factors in the tumor microenvironment (TME), including, but not limited to, metabolic plasticity of immunosuppressive cells and cytokines secreted by these cells. We used single-cell RNA-sequencing (scRNA-seq) data and applied single-cell flux estimation analysis to characterize the link between metabolism and cellular function within the hypoxic TME of colorectal (CRC) and lung cancer. In terms of metabolic heterogeneity, we found myeloid cells potentially inclined to accumulate glutamine but tumor cells inclined to accumulate glutamate. In particular, we uncovered a tumor-associated macrophage (TAM) subpopulation, APOE+CTSZ+TAM, that was present in high proportions in tumor samples and exhibited immunosuppressive characteristics through upregulating the expression of anti-inflammatory genes. The proportion of APOE+CTSZ+TAM and regulatory T cells (Treg) were positively correlated across CRC scRNA-seq samples. APOE+CTSZ+TAM potentially interacted with Treg via CXCL16-CCR6 signals, as seen by ligand-receptor interactions analysis. Notably, glutamate-to-glutamine metabolic flux score and glutamine synthetase (GLUL) expression were uniquely higher in APOE+CTSZ+TAM, compared with other cell types within the TME. GLUL expression in macrophages was positively correlated with anti-inflammatory score and was higher in high-grade and invasive tumor samples. Moreover, spatial transcriptome and multiplex immunofluorescence staining of samples showed that APOE+CTSZ+TAM and Treg potentially colocalized in the tissue sections from CRC clinical samples. These results highlight the specific role and metabolic characteristic of the APOE+CTSZ+TAM subpopulation and provide a new perspective for macrophage subcluster-targeted therapeutic interventions or metabolic checkpoint-based cancer therapies.
Collapse
Affiliation(s)
- Jinfen Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Wenqi Yu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Juanzhi Chen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guanda Huang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Lingjie Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China.,Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zixi Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Meiling Hu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Xiaocheng Gong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Hongli Du
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| |
Collapse
|