101
|
Mirzaei R, Sabokroo N, Ahmadyousefi Y, Motamedi H, Karampoor S. Immunometabolism in biofilm infection: lessons from cancer. Mol Med 2022; 28:10. [PMID: 35093033 PMCID: PMC8800364 DOI: 10.1186/s10020-022-00435-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Biofilm is a community of bacteria embedded in an extracellular matrix, which can colonize different human cells and tissues and subvert the host immune reactions by preventing immune detection and polarizing the immune reactions towards an anti-inflammatory state, promoting the persistence of biofilm-embedded bacteria in the host. MAIN BODY OF THE MANUSCRIPT It is now well established that the function of immune cells is ultimately mediated by cellular metabolism. The immune cells are stimulated to regulate their immune functions upon sensing danger signals. Recent studies have determined that immune cells often display distinct metabolic alterations that impair their immune responses when triggered. Such metabolic reprogramming and its physiological implications are well established in cancer situations. In bacterial infections, immuno-metabolic evaluations have primarily focused on macrophages and neutrophils in the planktonic growth mode. CONCLUSION Based on differences in inflammatory reactions of macrophages and neutrophils in planktonic- versus biofilm-associated bacterial infections, studies must also consider the metabolic functions of immune cells against biofilm infections. The profound characterization of the metabolic and immune cell reactions could offer exciting novel targets for antibiofilm therapy.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Niloofar Sabokroo
- Department of Microbiology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Motamedi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
102
|
van Genugten EAJ, Weijers JAM, Heskamp S, Kneilling M, van den Heuvel MM, Piet B, Bussink J, Hendriks LEL, Aarntzen EHJG. Imaging the Rewired Metabolism in Lung Cancer in Relation to Immune Therapy. Front Oncol 2022; 11:786089. [PMID: 35070990 PMCID: PMC8779734 DOI: 10.3389/fonc.2021.786089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic reprogramming is recognized as one of the hallmarks of cancer. Alterations in the micro-environmental metabolic characteristics are recognized as important tools for cancer cells to interact with the resident and infiltrating T-cells within this tumor microenvironment. Cancer-induced metabolic changes in the micro-environment also affect treatment outcomes. In particular, immune therapy efficacy might be blunted because of somatic mutation-driven metabolic determinants of lung cancer such as acidity and oxygenation status. Based on these observations, new onco-immunological treatment strategies increasingly include drugs that interfere with metabolic pathways that consequently affect the composition of the lung cancer tumor microenvironment (TME). Positron emission tomography (PET) imaging has developed a wide array of tracers targeting metabolic pathways, originally intended to improve cancer detection and staging. Paralleling the developments in understanding metabolic reprogramming in cancer cells, as well as its effects on stromal, immune, and endothelial cells, a wave of studies with additional imaging tracers has been published. These tracers are yet underexploited in the perspective of immune therapy. In this review, we provide an overview of currently available PET tracers for clinical studies and discuss their potential roles in the development of effective immune therapeutic strategies, with a focus on lung cancer. We report on ongoing efforts that include PET/CT to understand the outcomes of interactions between cancer cells and T-cells in the lung cancer microenvironment, and we identify areas of research which are yet unchartered. Thereby, we aim to provide a starting point for molecular imaging driven studies to understand and exploit metabolic features of lung cancer to optimize immune therapy.
Collapse
Affiliation(s)
- Evelien A J van Genugten
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Jetty A M Weijers
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Sandra Heskamp
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| | - Manfred Kneilling
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center, Eberhard Karls University, Tuebingen, Germany.,Department of Dermatology, Eberhard Karls University, Tuebingen, Germany
| | | | - Berber Piet
- Department of Respiratory Diseases, Radboudumc, Nijmegen, Netherlands
| | - Johan Bussink
- Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboudumc, Netherlands
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Centre (UMC), Maastricht, Netherlands
| | - Erik H J G Aarntzen
- Department of Medical Imaging, Radboud University Medical Centre (Radboudumc), Nijmegen, Netherlands
| |
Collapse
|
103
|
Abstract
As cancers progress, they produce a local environment that acts to redirect, paralyze, exhaust, or otherwise evade immune detection and destruction. The tumor microenvironment (TME) has long been characterized as a metabolic desert, depleted of essential nutrients such as glucose, oxygen, and amino acids, that starves infiltrating immune cells and renders them dysfunctional. While not incorrect, this perspective is only half the picture. The TME is not a metabolic vacuum, only consuming essential nutrients and never producing by-products. Rather, the by-products of depleted nutrients, "toxic" metabolites in the TME such as lactic acid, kynurenine, ROS, and adenosine, play an important role in shaping immune cell function and cannot be overlooked in cancer immunotherapy. Moreover, while the metabolic landscape is distinct, it is not unique, as these toxic metabolites are encountered in non-tumor tissues, where they evolutionarily shape immune cells and their response. In this Review, we discuss how depletion of essential nutrients and production of toxic metabolites shape the immune response within the TME and how toxic metabolites can be targeted to improve current cancer immunotherapies.
Collapse
Affiliation(s)
- McLane J. Watson
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Tumor Microenvironment Center, Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Greg M. Delgoffe
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Tumor Microenvironment Center, Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
104
|
Sun R, Kim AH. The multifaceted mechanisms of malignant glioblastoma progression and clinical implications. Cancer Metastasis Rev 2022; 41:871-898. [PMID: 35920986 PMCID: PMC9758111 DOI: 10.1007/s10555-022-10051-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023]
Abstract
With the application of high throughput sequencing technologies at single-cell resolution, studies of the tumor microenvironment in glioblastoma, one of the most aggressive and invasive of all cancers, have revealed immense cellular and tissue heterogeneity. A unique extracellular scaffold system adapts to and supports progressive infiltration and migration of tumor cells, which is characterized by altered composition, effector delivery, and mechanical properties. The spatiotemporal interactions between malignant and immune cells generate an immunosuppressive microenvironment, contributing to the failure of effective anti-tumor immune attack. Among the heterogeneous tumor cell subpopulations of glioblastoma, glioma stem cells (GSCs), which exhibit tumorigenic properties and strong invasive capacity, are critical for tumor growth and are believed to contribute to therapeutic resistance and tumor recurrence. Here we discuss the role of extracellular matrix and immune cell populations, major components of the tumor ecosystem in glioblastoma, as well as signaling pathways that regulate GSC maintenance and invasion. We also highlight emerging advances in therapeutic targeting of these components.
Collapse
Affiliation(s)
- Rui Sun
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Albert H. Kim
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO 63110 USA ,The Brain Tumor Center, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
105
|
Zhang M, Zhou Y, Xie Z, Luo S, Zhou Z, Huang J, Zhao B. New Developments in T Cell Immunometabolism and Therapeutic Implications for Type 1 Diabetes. Front Endocrinol (Lausanne) 2022; 13:914136. [PMID: 35757405 PMCID: PMC9226440 DOI: 10.3389/fendo.2022.914136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 05/09/2022] [Indexed: 11/23/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease mediated by T cells and is becoming a serious public health threat. Despite the increasing incidence rate of T1D worldwide, our understanding of why T1D develops and how T cells lose their self-tolerance in this process remain limited. Recent advances in immunometabolism have shown that cellular metabolism plays a fundamental role in shaping T cell responses. T cell activation and proliferation are supported by metabolic reprogramming to meet the increased energy and biomass demand, and deregulation in immune metabolism can lead to autoimmune disorders. Specific metabolic pathways and factors have been investigated to rectify known deficiencies in several autoimmune diseases, including T1D. Most therapeutic strategies have concentrated on aerobic glycolysis to limit T cell responses, whereas glycolysis is the main metabolic pathway for T cell activation and proliferation. The use of metabolic inhibitors, especially glycolysis inhibitors may largely leave T cell function intact but primarily target those autoreactive T cells with hyperactivated metabolism. In this review, we provide an overview of metabolic reprogramming used by T cells, summarize the recent findings of key metabolic pathways and regulators modulating T cell homeostasis, differentiation, and function in the context of T1D, and discuss the opportunities for metabolic intervention to be employed to suppress autoreactive T cells and limit the progression of β-cell destruction.
Collapse
Affiliation(s)
- Mengdi Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yanyan Zhou
- Department of Critical Care Medicine, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shuoming Luo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiaqi Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bin Zhao, ; ; Jiaqi Huang, ;
| | - Bin Zhao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Bin Zhao, ; ; Jiaqi Huang, ;
| |
Collapse
|
106
|
Aoki T, Nishida N, Kudo M. Current Perspectives on the Immunosuppressive Niche and Role of Fibrosis in Hepatocellular Carcinoma and the Development of Antitumor Immunity. J Histochem Cytochem 2022; 70:53-81. [PMID: 34751050 PMCID: PMC8721576 DOI: 10.1369/00221554211056853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Immune checkpoint inhibitors have become the mainstay of treatment for hepatocellular carcinoma (HCC). However, they are ineffective in some cases. Previous studies have reported that genetic alterations in oncogenic pathways such as Wnt/β-catenin are the important triggers in HCC for primary refractoriness. T-cell exhaustion has been reported in various tumors and is likely to play a prominent role in the emergence of HCC due to chronic inflammation and cirrhosis-associated immune dysfunction. Immunosuppressive cells including regulatory T-cells and tumor-associated macrophages infiltrating the tumor are associated with hyperprogressive disease in the early stages of immune checkpoint inhibitor treatment. In addition, stellate cells and tumor-associated fibroblasts create an abundant desmoplastic environment by producing extracellular matrix. This strongly contributes to epithelial to mesenchymal transition via signaling activities including transforming growth factor beta, Wnt/β-catenin, and Hippo pathway. The abundant desmoplastic environment has been demonstrated in pancreatic ductal adenocarcinoma and cholangiocarcinoma to suppress cytotoxic T-cell infiltration, PD-L1 expression, and neoantigen expression, resulting in a highly immunosuppressive niche. It is possible that a similar immunosuppressive environment is created in HCC with advanced fibrosis in the background liver. Although sufficient understanding is required for the establishment of immune therapies of HCC, further investigations are still required in this field.
Collapse
Affiliation(s)
- Tomoko Aoki
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| | - Naoshi Nishida
- Naoshi Nishida, Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, 377-2 Ohno-higashi, Osaka-Sayama 589-8511, Japan. E-mail:
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University, Osaka-Sayama, Japan
| |
Collapse
|
107
|
Importance of T, NK, CAR T and CAR NK Cell Metabolic Fitness for Effective Anti-Cancer Therapy: A Continuous Learning Process Allowing the Optimization of T, NK and CAR-Based Anti-Cancer Therapies. Cancers (Basel) 2021; 14:cancers14010183. [PMID: 35008348 PMCID: PMC8782435 DOI: 10.3390/cancers14010183] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/18/2021] [Accepted: 12/29/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Cancer treatments are evolving at a very rapid pace. Some of the most novel anti-cancer medicines under development rely on the modification of immune cells in order to transform them into potent tumor-killing cells. However, the tumor microenvironment (TME) is competing for nutrients with these harnessed immune cells and therefore paralyzes their metabolic effective and active anti-cancer activities. Here we describe strategies to overcome these hurdles imposed on immune cell activity, which lead to therapeutic approaches to enhance metabolic fitness of the patient’s immune system with the objective to improve their anti-cancer capacity. Abstract Chimeric antigen receptor (CAR) T and CAR NK cell therapies opened new avenues for cancer treatment. Although original successes of CAR T and CAR NK cells for the treatment of hematological malignancies were extraordinary, several obstacles have since been revealed, in particular their use for the treatment of solid cancers. The tumor microenvironment (TME) is competing for nutrients with T and NK cells and their CAR-expressing counterparts, paralyzing their metabolic effective and active states. Consequently, this can lead to alterations in their anti-tumoral capacity and persistence in vivo. High glucose uptake and the depletion of key amino acids by the TME can deprive T and NK cells of energy and building blocks, which turns them into a state of anergy, where they are unable to exert cytotoxic activity against cancer cells. This is especially true in the context of an immune-suppressive TME. In order to re-invigorate the T, NK, CAR T and CAR NK cell-mediated antitumor response, the field is now attempting to understand how metabolic pathways might change T and NK responses and functions, as well as those from their CAR-expressing partners. This revealed ways to metabolically rewire these cells by using metabolic enhancers or optimizing pre-infusion in vitro cultures of these cells. Importantly, next-generation CAR T and CAR NK products might include in the future the necessary metabolic requirements by improving their design, manufacturing process and other parameters. This will allow the overcoming of current limitations due to their interaction with the suppressive TME. In a clinical setting, this might improve their anti-cancer effector activity in synergy with immunotherapies. In this review, we discuss how the tumor cells and TME interfere with T and NK cell metabolic requirements. This may potentially lead to therapeutic approaches that enhance the metabolic fitness of CAR T and CAR NK cells, with the objective to improve their anti-cancer capacity.
Collapse
|
108
|
Wan M, Ding Y, Li Z, Wang X, Xu M. Metabolic manipulation of the tumour immune microenvironment. Immunology 2021; 165:290-300. [PMID: 34962655 DOI: 10.1111/imm.13444] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 11/26/2022] Open
Abstract
In the past few years, the evolution of immunotherapy has resulted in a shift in cancer treatment models. However, with immunosuppressive effects of the tumour microenvironment continue to limit advances in tumour immunotherapy. The tumour microenvironment induces metabolic reprogramming in cancer cells, which results in competition for nutrients between tumour cells and host immunocytes. Metabolic and waste products originating in tumour cells can influence the activation and effector properties of immunocytes in numerous ways and ultimately promote the survival and propagation of tumour cells. In this paper, we discuss metabolic reprogramming in tumour cells and the influence of metabolite byproducts on the immune microenvironment, providing novel insights into tumour immunotherapy.
Collapse
Affiliation(s)
- Mengtian Wan
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Yuzhu Ding
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Zheng Li
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Xu Wang
- Department of Radiation Oncology, Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, Jiangsu Province, China
| |
Collapse
|
109
|
Krapić M, Kavazović I, Wensveen FM. Immunological Mechanisms of Sickness Behavior in Viral Infection. Viruses 2021; 13:v13112245. [PMID: 34835051 PMCID: PMC8624889 DOI: 10.3390/v13112245] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/04/2021] [Indexed: 12/11/2022] Open
Abstract
Sickness behavior is the common denominator for a plethora of changes in normal behavioral routines and systemic metabolism during an infection. Typical symptoms include temperature, muscle weakness, and loss of appetite. Whereas we experience these changes as a pathology, in fact they are a carefully orchestrated response mediated by the immune system. Its purpose is to optimize immune cell functionality against pathogens whilst minimizing viral replication in infected cells. Sickness behavior is controlled at several levels, most notably by the central nervous system, but also by other organs that mediate systemic homeostasis, such as the liver and adipose tissue. Nevertheless, the changes mediated by these organs are ultimately initiated by immune cells, usually through local or systemic secretion of cytokines. The nature of infection determines which cytokine profile is induced by immune cells and therefore which sickness behavior ensues. In context of infection, sickness behavior is typically beneficial. However, inappropriate activation of the immune system may induce adverse aspects of sickness behavior. For example, tissue stress caused by obesity may result in chronic activation of the immune system, leading to lasting changes in systemic metabolism. Concurrently, metabolic disease prevents induction of appropriate sickness behavior following viral infection, thus impairing the normal immune response. In this article, we will revisit recent literature that elucidates both the benefits and the negative aspects of sickness behavior in context of viral infection.
Collapse
|
110
|
Selection and stability validation of reference gene candidates for transcriptional analysis in Rousettus aegyptiacus. Sci Rep 2021; 11:21662. [PMID: 34737406 PMCID: PMC8568961 DOI: 10.1038/s41598-021-01260-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 10/18/2021] [Indexed: 11/08/2022] Open
Abstract
Bats are the only mammals capable of powered flight and their body temperature can reach up to 42 °C during flight. Additionally, bats display robust type I IFN interferon (IFN-I) responses and some species constitutively express IFN-α. Reference genes with stable expression under temperature oscillations and IFN-I release are therefore critical for normalization of quantitative reverse-transcription polymerase chain reaction (qRT-PCR) data in bats. The expression stability of reference genes in Rousettus aegyptiacus remains elusive, although this species is frequently used in the infection research. We selected ACTB, EEF1A1, GAPDH and PGK1 as candidate reference genes and evaluated their expression stability in various tissues and cells from this model bat species upon IFN-I treatment at 35 °C, 37 °C and 40 °C by qRT-PCR. We employed two statistical algorithms, BestKeeper and NormFinder, and found that EEF1A1 exhibited the highest expression stability under all tested conditions. ACTB and GAPDH displayed unstable expression upon temperature change and IFN-I treatment, respectively. By normalizing to EEF1A1, we uncovered that GAPDH expression was significantly induced by IFN-I in R. aegyptiacus. Our study identifies EEF1A1 as the most suitable reference gene for qRT-PCR studies upon temperature changes and IFN-I treatment and unveils the induction of GAPDH expression by IFN-I in R. aegyptiacus. These findings are pertinent to other bat species and may be relevant for non-volant mammals that show physiological fluctuations of core body temperature.
Collapse
|
111
|
Matias MI, Yong CS, Foroushani A, Goldsmith C, Mongellaz C, Sezgin E, Levental KR, Talebi A, Perrault J, Rivière A, Dehairs J, Delos O, Bertand-Michel J, Portais JC, Wong M, Marie JC, Kelekar A, Kinet S, Zimmermann VS, Levental I, Yvan-Charvet L, Swinnen JV, Muljo SA, Hernandez-Vargas H, Tardito S, Taylor N, Dardalhon V. Regulatory T cell differentiation is controlled by αKG-induced alterations in mitochondrial metabolism and lipid homeostasis. Cell Rep 2021; 37:109911. [PMID: 34731632 PMCID: PMC10167917 DOI: 10.1016/j.celrep.2021.109911] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 08/18/2021] [Accepted: 10/08/2021] [Indexed: 12/15/2022] Open
Abstract
Suppressive regulatory T cell (Treg) differentiation is controlled by diverse immunometabolic signaling pathways and intracellular metabolites. Here we show that cell-permeable α-ketoglutarate (αKG) alters the DNA methylation profile of naive CD4 T cells activated under Treg polarizing conditions, markedly attenuating FoxP3+ Treg differentiation and increasing inflammatory cytokines. Adoptive transfer of these T cells into tumor-bearing mice results in enhanced tumor infiltration, decreased FoxP3 expression, and delayed tumor growth. Mechanistically, αKG leads to an energetic state that is reprogrammed toward a mitochondrial metabolism, with increased oxidative phosphorylation and expression of mitochondrial complex enzymes. Furthermore, carbons from ectopic αKG are directly utilized in the generation of fatty acids, associated with lipidome remodeling and increased triacylglyceride stores. Notably, inhibition of either mitochondrial complex II or DGAT2-mediated triacylglyceride synthesis restores Treg differentiation and decreases the αKG-induced inflammatory phenotype. Thus, we identify a crosstalk between αKG, mitochondrial metabolism and triacylglyceride synthesis that controls Treg fate.
Collapse
MESH Headings
- Animals
- Cell Differentiation/drug effects
- Cells, Cultured
- Cytokines/genetics
- Cytokines/metabolism
- Diacylglycerol O-Acyltransferase/metabolism
- Energy Metabolism/drug effects
- Fibrosarcoma/genetics
- Fibrosarcoma/immunology
- Fibrosarcoma/metabolism
- Fibrosarcoma/therapy
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Homeostasis
- Humans
- Immunotherapy, Adoptive
- Ketoglutaric Acids/pharmacology
- Lipid Metabolism/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/drug effects
- Mitochondria/genetics
- Mitochondria/metabolism
- Phenotype
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Signal Transduction
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Mice
Collapse
Affiliation(s)
- Maria I Matias
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Carmen S Yong
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France; Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Amir Foroushani
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Chloe Goldsmith
- Cancer Research Center of Lyon, University Lyon 1, Inserm/ CNRS, Labex DEVweCAN, Lyon France
| | - Cédric Mongellaz
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Erdinc Sezgin
- Science for Life Laboratory, Department of Women's and Children's Health, Karolinska Institute, Solna, Sweden
| | - Kandice R Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Ali Talebi
- Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, Leuven, Belgium
| | - Julie Perrault
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Anais Rivière
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, Leuven, Belgium
| | - Océane Delos
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France; I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Justine Bertand-Michel
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France; I2MC, Université de Toulouse, Inserm, Toulouse, France
| | - Jean-Charles Portais
- MetaboHUB-MetaToul, National Infrastructure of Metabolomics and Fluxomics, Toulouse, France
| | - Madeline Wong
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Julien C Marie
- Cancer Research Center of Lyon, University Lyon 1, Inserm/ CNRS, Labex DEVweCAN, Lyon France
| | - Ameeta Kelekar
- Department of Laboratory Medicine and Pathology, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Valérie S Zimmermann
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France
| | - Ilya Levental
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | | | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, Leuven, Belgium
| | - Stefan A Muljo
- Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, USA
| | - Hector Hernandez-Vargas
- Cancer Research Center of Lyon, University Lyon 1, Inserm/ CNRS, Labex DEVweCAN, Lyon France
| | - Saverio Tardito
- Cancer Research UK, Beatson Institute, Glasgow, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France; Pediatric Oncology Branch, NCI, CCR, NIH, Bethesda, MD, USA.
| | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, Univ Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
112
|
Tran DT, Sundararaj K, Atkinson C, Nadig SN. T-cell Immunometabolism: Therapeutic Implications in Organ Transplantation. Transplantation 2021; 105:e191-e201. [PMID: 33795597 PMCID: PMC8464628 DOI: 10.1097/tp.0000000000003767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although solid-organ transplantation has evolved steadily with many breakthroughs in the past 110 y, many problems remain to be addressed, and advanced therapeutic strategies need to be considered. T-cell immunometabolism is a rapidly advancing field that has gathered much attention recently, providing ample mechanistic insight from which many novel therapeutic approaches have been developed. Applications from the field include antitumor and antimicrobial therapies, as well as for reversing graft-versus-host disease and autoimmune diseases. However, the immunometabolism of T cells remains underexplored in solid-organ transplantation. In this review, we will highlight key findings from hallmark studies centered around various metabolic modes preferred by different T-cell subtypes (categorized into naive, effector, regulatory, and memory T cells), including glycolysis, glutaminolysis, oxidative phosphorylation, fatty acid synthesis, and oxidation. This review will discuss the underlying cellular signaling components that affect these processes, including the transcription factors myelocytomatosis oncogene, hypoxia-inducible factor 1-alpha, estrogen-related receptor alpha, and sterol regulatory element-binding proteins, along with the mechanistic target of rapamycin and adenosine monophosphate-activated protein kinase signaling. We will also explore potential therapeutic strategies targeting these pathways, as applied to the potential for tolerance induction in solid-organ transplantation.
Collapse
Affiliation(s)
- Danh T. Tran
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
| | - Kamala Sundararaj
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Carl Atkinson
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Satish N. Nadig
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC
- Department of Surgery, Division of Transplant Surgery, Lee Patterson Allen Transplant Immunobiology Laboratory, Medical University of South Carolina, Charleston, SC
- South Carolina Investigators in Transplantation, Department of Surgery, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
113
|
Emerging Bone Marrow Microenvironment-Driven Mechanisms of Drug Resistance in Acute Myeloid Leukemia: Tangle or Chance? Cancers (Basel) 2021; 13:cancers13215319. [PMID: 34771483 PMCID: PMC8582363 DOI: 10.3390/cancers13215319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Despite high rates of remission obtained with conventional chemotherapy, the persistence of leukemic cells after treatments, eventually exiting in disease relapse, remains the main challenge in acute myeloid leukemia (AML). Increasing evidence indicates that, besides AML cell mutations, stromal and immune cells, as leukemic microenvironment components, may protect AML cells from therapies. Here, we will recapitulate emerging bone marrow (BM) microenvironment-dependent mechanisms of therapy resistance. The understanding of these processes will help find new drug combinations and conceive novel and more effective treatments. Abstract Acute myeloid leukemia (AML) has been considered for a long time exclusively driven by critical mutations in hematopoietic stem cells. Recently, the contribution of further players, such as stromal and immune bone marrow (BM) microenvironment components, to AML onset and progression has been pointed out. In particular, mesenchymal stromal cells (MSCs) steadily remodel the leukemic niche, not only favoring leukemic cell growth and development but also tuning their responsiveness to treatments. The list of mechanisms driven by MSCs to promote a leukemia drug-resistant phenotype has progressively expanded. Moreover, the relative proportion and the activation status of immune cells in the BM leukemic microenvironment may vary by influencing their reactivity against leukemic cells. In that, the capacity of the stroma to re-program immune cells, thus promoting and/or hampering therapeutic efficacy, is emerging as a crucial aspect in AML biology, adding an extra layer of complexity. Current treatments for AML have mainly focused on eradicating leukemia cells, with little consideration for the leukemia-damaged BM niche. Increasing evidence on the contribution of stromal and immune cells in response to therapy underscores the need to hold the mutual interplay, which takes place in the BM. A careful dissection of these interactions will help provide novel applications for drugs already under experimentation and open a wide array of opportunities for new drug discovery.
Collapse
|
114
|
Ross SH, Rollings CM, Cantrell DA. Quantitative Analyses Reveal How Hypoxia Reconfigures the Proteome of Primary Cytotoxic T Lymphocytes. Front Immunol 2021; 12:712402. [PMID: 34603285 PMCID: PMC8484760 DOI: 10.3389/fimmu.2021.712402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/26/2021] [Indexed: 12/18/2022] Open
Abstract
Metabolic and nutrient-sensing pathways play an important role in controlling the efficacy of effector T cells. Oxygen is a critical regulator of cellular metabolism. However, during immune responses T cells must function in oxygen-deficient, or hypoxic, environments. Here, we used high resolution mass spectrometry to investigate how the proteome of primary murine CD8+ cytotoxic T lymphocytes (CTLs) is reconfigured in response to hypoxia in vitro. We identified and quantified over 7,600 proteins and discovered that hypoxia increased the abundance of a selected number of proteins in CTLs. This included glucose transporters, metabolic enzymes, transcription factors, cytolytic effector molecules, checkpoint receptors and adhesion molecules. While some of these proteins may augment the effector functions of CTLs, others may limit their cytotoxicity. Moreover, we determined that hypoxia could inhibit IL-2-induced proliferation cues and antigen-induced pro-inflammatory cytokine production in CTLs. These data provide a comprehensive resource for understanding the magnitude of the CTL response to hypoxia and emphasise the importance of oxygen-sensing pathways for controlling CD8+ T cells. Additionally, this study provides new understanding about how hypoxia may promote the effector function of CTLs, while contributing to their dysfunction in some contexts.
Collapse
Affiliation(s)
- Sarah H Ross
- Immunology Programme, The Babraham Institute, Cambridge, United Kingdom.,Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Christina M Rollings
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Doreen A Cantrell
- Division of Cell Signalling and Immunology, School of Life Sciences, University of Dundee, Dundee, United Kingdom
| |
Collapse
|
115
|
Holthaus L, Sharma V, Brandt D, Ziegler AG, Jastroch M, Bonifacio E. Functional and metabolic fitness of human CD4 + T lymphocytes during metabolic stress. Life Sci Alliance 2021; 4:4/12/e202101013. [PMID: 34580176 PMCID: PMC8500231 DOI: 10.26508/lsa.202101013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
Human T-cell activation, expansion, and effector function is grossly impaired in conditions that combine glucose deprivation and mild mitochondrial stress. Human CD4+ T cells are essential mediators of immune responses. By altering the mitochondrial and metabolic states, we defined metabolic requirements of human CD4+ T cells for in vitro activation, expansion, and effector function. T-cell activation and proliferation were reduced by inhibiting oxidative phosphorylation, whereas early cytokine production was maintained by either OXPHOS or glycolytic activity. Glucose deprivation in the presence of mild mitochondrial stress markedly reduced all three T-cell functions, contrasting the exposure to resveratrol, an antioxidant and sirtuin-1 activator, which specifically inhibited cytokine production and T-cell proliferation, but not T-cell activation. Conditions that inhibited T-cell activation were associated with the down-regulation of 2′,5′-oligoadenylate synthetase genes via interferon response pathways. Our findings indicate that T-cell function is grossly impaired by stressors combined with nutrient deprivation, suggesting that correcting nutrient availability, metabolic stress, and/or the function of T cells in these conditions will improve the efficacy of T-cell–based therapies.
Collapse
Affiliation(s)
- Lisa Holthaus
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Virag Sharma
- German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital and Faculty of Medicine of TU Dresden, Dresden, Germany
| | - Daniel Brandt
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Forschergruppe Diabetes e.V. at Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Martin Jastroch
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Ezio Bonifacio
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany .,German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany.,Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Paul Langerhans Institute Dresden of the Helmholtz Center Munich at the University Hospital and Faculty of Medicine of TU Dresden, Dresden, Germany
| |
Collapse
|
116
|
Kareva I, Luddy KA, O’Farrelly C, Gatenby RA, Brown JS. Predator-Prey in Tumor-Immune Interactions: A Wrong Model or Just an Incomplete One? Front Immunol 2021; 12:668221. [PMID: 34531851 PMCID: PMC8438324 DOI: 10.3389/fimmu.2021.668221] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/05/2021] [Indexed: 01/05/2023] Open
Abstract
Tumor-immune interactions are often framed as predator-prey. This imperfect analogy describes how immune cells (the predators) hunt and kill immunogenic tumor cells (the prey). It allows for evaluation of tumor cell populations that change over time during immunoediting and it also considers how the immune system changes in response to these alterations. However, two aspects of predator-prey type models are not typically observed in immuno-oncology. The first concerns the conversion of prey killed into predator biomass. In standard predator-prey models, the predator relies on the prey for nutrients, while in the tumor microenvironment the predator and prey compete for resources (e.g. glucose). The second concerns oscillatory dynamics. Standard predator-prey models can show a perpetual cycling in both prey and predator population sizes, while in oncology we see increases in tumor volume and decreases in infiltrating immune cell populations. Here we discuss the applicability of predator-prey models in the context of cancer immunology and evaluate possible causes for discrepancies. Key processes include "safety in numbers", resource availability, time delays, interference competition, and immunoediting. Finally, we propose a way forward to reconcile differences between model predictions and empirical observations. The immune system is not just predator-prey. Like natural food webs, the immune-tumor community of cell types forms an immune-web of different and identifiable interactions.
Collapse
Affiliation(s)
- Irina Kareva
- EMD Serono, Merck KGaA, Billerica, MA, United States
| | - Kimberly A. Luddy
- Department of Cancer Physiology, H. Lee Moffitt Cancer Center, Tampa, FL, United States
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Cliona O’Farrelly
- School of Biochemistry and Immunology, Trinity College Dublin, Dublin, Ireland
| | - Robert A. Gatenby
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Joel S. Brown
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
117
|
Peng Z, Zhang Y, Ma X, Zhou M, Wu S, Song Z, Yuan Y, Chen Y, Li Y, Wang G, Huang F, Qiao Y, Xia B, Liu W, Liu J, Zhang X, He X, Pan T, Xu H, Zhang H. Brd4 Regulates the Homeostasis of CD8 + T-Lymphocytes and Their Proliferation in Response to Antigen Stimulation. Front Immunol 2021; 12:728082. [PMID: 34512660 PMCID: PMC8427756 DOI: 10.3389/fimmu.2021.728082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 08/05/2021] [Indexed: 12/24/2022] Open
Abstract
CD8+ T cells are major components of adaptive immunity and confer robust protective cellular immunity, which requires adequate T-cell numbers, targeted migration, and efficient T-cell proliferation. Altered CD8+ T-cell homeostasis and impaired proliferation result in dysfunctional immune response to infection or tumorigenesis. However, intrinsic factors controlling CD8+ T-cell homeostasis and immunity remain largely elusive. Here, we demonstrate the prominent role of Brd4 on CD8+ T cell homeostasis and immune response. By upregulating Myc and GLUT1 expression, Brd4 facilitates glucose uptake and energy production in mitochondria, subsequently supporting naïve CD8+ T-cell survival. Besides, Brd4 promotes the trafficking of naïve CD8+ T cells partially through maintaining the expression of homing receptors (CD62L and LFA-1). Furthermore, Brd4 is required for CD8+ T cell response to antigen stimulation, as Brd4 deficiency leads to a severe defect in clonal expansion and terminal differentiation by decreasing glycolysis. Importantly, as JQ1, a pan-BRD inhibitor, severely dampens CD8+ T-cell immune response, its usage as an anti-tumor agent or latency-reversing agent for human immunodeficiency virus type I (HIV-1) should be more cautious. Collectively, our study identifies a previously-unexpected role of Brd4 in the metabolic regulation of CD8+ T cell-mediated immune surveillance and also provides a potential immunomodulation target.
Collapse
Affiliation(s)
- Zhilin Peng
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yiwen Zhang
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiancai Ma
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mo Zhou
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shiyu Wu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zheng Song
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yaochang Yuan
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yingshi Chen
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuzhuang Li
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guanwen Wang
- Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Feng Huang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Yidan Qiao
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Baijing Xia
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Weiwei Liu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jun Liu
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xu Zhang
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xin He
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ting Pan
- Center for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Hanshi Xu
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Zhang
- Key Laboratory of Tropical Disease Control of Ministry of Education, Institute of Human Virology, Guangdong Engineering Research Center for Antimicrobial Agent and Immunotechnology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
118
|
Khurana P, Burudpakdee C, Grupp SA, Beier UH, Barrett DM, Bassiri H. Distinct Bioenergetic Features of Human Invariant Natural Killer T Cells Enable Retained Functions in Nutrient-Deprived States. Front Immunol 2021; 12:700374. [PMID: 34434191 PMCID: PMC8380770 DOI: 10.3389/fimmu.2021.700374] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/20/2021] [Indexed: 12/19/2022] Open
Abstract
Invariant natural killer T (iNKT) cells comprise a unique subset of lymphocytes that are primed for activation and possess innate NK-like functional features. Currently, iNKT cell-based immunotherapies remain in early clinical stages, and little is known about the ability of these cells to survive and retain effector functions within the solid tumor microenvironment (TME) long-term. In conventional T cells (TCONV), cellular metabolism is linked to effector functions and their ability to adapt to the nutrient-poor TME. In contrast, the bioenergetic requirements of iNKT cells - particularly those of human iNKT cells - at baseline and upon stimulation are not well understood; neither is how these requirements affect effector functions such as production of cytokines and cytolytic proteins. We find that unlike TCONV, human iNKT cells are not dependent upon glucose or glutamine for these effector functions upon stimulation with anti-CD3 and anti-CD28. Additionally, transcriptional profiling revealed that stimulated human iNKT cells are less glycolytic than TCONV and display higher expression of fatty acid oxidation (FAO) and adenosine monophosphate-activated protein kinase (AMPK) pathway genes. Furthermore, stimulated iNKT cells displayed higher mitochondrial mass and membrane potential relative to TCONV. Real-time Seahorse metabolic flux analysis revealed that stimulated human iNKT cells utilize fatty acids as substrates for oxidation more than stimulated TCONV. Together, our data suggest that human iNKT cells possess different bioenergetic requirements from TCONV and display a more oxidative metabolic program relative to effector TCONV. Importantly, iNKT cell-based immunotherapeutic strategies could co-opt such unique features of iNKT cells to improve their efficacy and longevity of anti-tumor responses.
Collapse
Affiliation(s)
- Priya Khurana
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Chakkapong Burudpakdee
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Stephan A. Grupp
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Cell and Therapy Transplant Section, Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Ulf H. Beier
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Nephrology, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Janssen Research and Development, Spring House, PA, United States
| | | | - Hamid Bassiri
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
119
|
Sheldon RD, Ma EH, DeCamp LM, Williams KS, Jones RG. Interrogating in vivo T-cell metabolism in mice using stable isotope labeling metabolomics and rapid cell sorting. Nat Protoc 2021; 16:4494-4521. [PMID: 34349284 DOI: 10.1038/s41596-021-00586-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/31/2021] [Indexed: 11/10/2022]
Abstract
T cells are integral players in the adaptive immune system that readily adapt their metabolism to meet their energetic and biosynthetic needs. A major hurdle to understand physiologic T-cell metabolism has been the differences between in vitro cell culture conditions and the complex in vivo milieu. To address this, we have developed a protocol that merges traditional immunology infection models with whole-body metabolite infusion and mass-spectrometry-based metabolomic profiling to assess T-cell metabolism in vivo. In this protocol, pathogen-infected mice are infused via the tail vein with an isotopically labeled metabolite (2-6 h), followed by rapid magnetic bead isolation to purify T-cell populations (<1 h) and then stable isotope labeling analysis conducted by mass spectrometry (~1-2 d). This procedure enables researchers to evaluate metabolic substrate utilization into central carbon metabolic pathways (i.e., glycolysis and the tricarboxylic acid cycle) by specific T-cell subpopulations in the context of physiological immune responses in vivo.
Collapse
Affiliation(s)
- Ryan D Sheldon
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA.,Metabolomics and Bioenergetics Core Facility, Van Andel Institute, Grand Rapids, MI, USA
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Lisa M DeCamp
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Kelsey S Williams
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Institute, Grand Rapids, MI, USA.
| |
Collapse
|
120
|
Ma G, Li C, Zhang Z, Liang Y, Liang Z, Chen Y, Wang L, Li D, Zeng M, Shan W, Niu H. Targeted Glucose or Glutamine Metabolic Therapy Combined With PD-1/PD-L1 Checkpoint Blockade Immunotherapy for the Treatment of Tumors - Mechanisms and Strategies. Front Oncol 2021; 11:697894. [PMID: 34327138 PMCID: PMC8314994 DOI: 10.3389/fonc.2021.697894] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy, especially PD-1/PD-L1 checkpoint blockade immunotherapy, has led tumor therapy into a new era. However, the vast majority of patients do not benefit from immunotherapy. One possible reason for this lack of response is that the association between tumors, immune cells and metabolic reprogramming in the tumor microenvironment affect tumor immune escape. Generally, the limited amount of metabolites in the tumor microenvironment leads to nutritional competition between tumors and immune cells. Metabolism regulates tumor cell expression of PD-L1, and the PD-1/PD-L1 immune checkpoint regulates the metabolism of tumor and T cells, which suggests that targeted tumor metabolism may have a synergistic therapeutic effect together with immunotherapy. However, the targeting of different metabolic pathways in different tumors may have different effects on tumor immune escape. Herein, we discuss the influence of glucose metabolism and glutamine metabolism on tumor immune escape and describe the theoretical basis for strategies targeting glucose or glutamine metabolism in combination with PD-1/PD-L1 checkpoint blockade immunotherapy.
Collapse
Affiliation(s)
- Guofeng Ma
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chun Li
- Department of Pharmacy, Central Hospital of Shengli Oil Field, Dongying, China
| | - Zhilei Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ye Liang
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhijuan Liang
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuanbin Chen
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liping Wang
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Li
- Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Manqin Zeng
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenhong Shan
- Department of Nephrology, Qingdao Central Hospital, The Second Clinical Medical College of Qingdao University, Qingdao, China
| | - Haitao Niu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.,Key Laboratory, Department of Urology and Andrology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
121
|
Slaats J, Dieteren CE, Wagena E, Wolf L, Raaijmakers TK, van der Laak JA, Figdor CG, Weigelin B, Friedl P. Metabolic Screening of Cytotoxic T-cell Effector Function Reveals the Role of CRAC Channels in Regulating Lethal Hit Delivery. Cancer Immunol Res 2021; 9:926-938. [PMID: 34226201 DOI: 10.1158/2326-6066.cir-20-0741] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/24/2021] [Accepted: 04/30/2021] [Indexed: 11/16/2022]
Abstract
Cytotoxic T lymphocytes (CTL) mediate cytotoxicity toward tumor cells by multistep cell-cell interactions. However, the tumor microenvironment can metabolically perturb local CTL effector function. CTL activity is typically studied in two-dimensional (2D) liquid coculture, which is limited in recapitulating the mechanisms and efficacy of the multistep CTL effector response. We here developed a microscopy-based, automated three-dimensional (3D) interface coculture model suitable for medium-throughput screening to delineate the steps and CTL effector mechanisms affected by microenvironmental perturbation. CTL effector function was compromised by deregulated redox homeostasis, deficient mitochondrial respiration, as well as dysfunctional Ca2+ release-activated Ca2+ (CRAC) channels. Perturbation of CRAC channel function dampened calcium influx into CTLs, delayed CTL degranulation, and lowered the frequency of sublethal hits (i.e., additive cytotoxicity) delivered to the target cell. Thus, CRAC channel activity controls both individual contact efficacy and CTL cooperativity required for serial killing of target cells. The multistep analysis of CTL effector responses in 3D coculture will facilitate the identification of immune-suppressive mechanisms and guide the rational design of targeted intervention strategies to restore CTL effector function.
Collapse
Affiliation(s)
- Jeroen Slaats
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cindy E Dieteren
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands.,Protinhi Therapeutics, Noviotech Campus, Nijmegen, the Netherlands
| | - Esther Wagena
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Louis Wolf
- Microscopic Imaging Center, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tonke K Raaijmakers
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands.,Radiotherapy and OncoImmunology Laboratory, Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen A van der Laak
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bettina Weigelin
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands.,Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tübingen, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies," University of Tübingen, Tübingen, Germany
| | - Peter Friedl
- Department of Cell Biology, RIMLS, Radboud University Medical Center, Nijmegen, the Netherlands. .,Department of Genitourinary Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Cancer Genomics Center, Utrecht, the Netherlands
| |
Collapse
|
122
|
Russell BL, Sooklal SA, Malindisa ST, Daka LJ, Ntwasa M. The Tumor Microenvironment Factors That Promote Resistance to Immune Checkpoint Blockade Therapy. Front Oncol 2021; 11:641428. [PMID: 34268109 PMCID: PMC8276693 DOI: 10.3389/fonc.2021.641428] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/16/2021] [Indexed: 12/13/2022] Open
Abstract
Through genetic and epigenetic alterations, cancer cells present the immune system with a diversity of antigens or neoantigens, which the organism must distinguish from self. The immune system responds to neoantigens by activating naïve T cells, which mount an anticancer cytotoxic response. T cell activation begins when the T cell receptor (TCR) interacts with the antigen, which is displayed by the major histocompatibility complex (MHC) on antigen-presenting cells (APCs). Subsequently, accessory stimulatory or inhibitory molecules transduce a secondary signal in concert with the TCR/antigen mediated stimulus. These molecules serve to modulate the activation signal's strength at the immune synapse. Therefore, the activation signal's optimum amplitude is maintained by a balance between the costimulatory and inhibitory signals. This system comprises the so-called immune checkpoints such as the programmed cell death (PD-1) and Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and is crucial for the maintenance of self-tolerance. Cancers often evade the intrinsic anti-tumor activity present in normal physiology primarily by the downregulation of T cell activation. The blockade of the immune checkpoint inhibitors using specific monoclonal antibodies has emerged as a potentially powerful anticancer therapy strategy. Several drugs have been approved mainly for solid tumors. However, it has emerged that there are innate and acquired mechanisms by which resistance is developed against these therapies. Some of these are tumor-intrinsic mechanisms, while others are tumor-extrinsic whereby the microenvironment may have innate or acquired resistance to checkpoint inhibitors. This review article will examine mechanisms by which resistance is mounted against immune checkpoint inhibitors focussing on anti-CTL4-A and anti-PD-1/PD-Ll since drugs targeting these checkpoints are the most developed.
Collapse
Affiliation(s)
- Bonnie L. Russell
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
- Innovation Hub, Buboo (Pty) Ltd, Pretoria, South Africa
| | - Selisha A. Sooklal
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
| | - Sibusiso T. Malindisa
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
| | | | - Monde Ntwasa
- Department of Life & Consumer Sciences, University of South Africa, Johannesburg, South Africa
| |
Collapse
|
123
|
Braun MY. The Natural History of T Cell Metabolism. Int J Mol Sci 2021; 22:ijms22136779. [PMID: 34202553 PMCID: PMC8269353 DOI: 10.3390/ijms22136779] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
The cells of the immune system, particularly the T lymphocytes, have two main features that distinguish them from the cells of other tissues. They proliferate after activation and have the ability to move in tissues and organs. These characteristics compel them to develop metabolic plasticity in order to fulfil their immune function. This review focuses on the different known mechanisms that allow T cells to adapt their metabolism to the real-life circumstances they operate in, whether it is to exit quiescence, to differentiate into effector cells, or to participate in immune memory formation. Some of the metabolic adaptations to environmental variations that T cells are likely to undergo in their immune monitoring function are also discussed.
Collapse
Affiliation(s)
- Michel Y Braun
- Institute for Medical Immunology (IMI), Faculty of Medicine, Université Libre de Bruxelles (ULB), 6041 Gosselies, Belgium
| |
Collapse
|
124
|
Amitrano AM, Berry BJ, Lim K, Kim KD, Waugh RE, Wojtovich AP, Kim M. Optical Control of CD8 + T Cell Metabolism and Effector Functions. Front Immunol 2021; 12:666231. [PMID: 34149701 PMCID: PMC8209468 DOI: 10.3389/fimmu.2021.666231] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/12/2021] [Indexed: 11/17/2022] Open
Abstract
Although cancer immunotherapy is effective against hematological malignancies, it is less effective against solid tumors due in part to significant metabolic challenges present in the tumor microenvironment (TME), where infiltrated CD8+ T cells face fierce competition with cancer cells for limited nutrients. Strong metabolic suppression in the TME is often associated with impaired T cell recruitment to the tumor site and hyporesponsive effector function via T cell exhaustion. Increasing evidence suggests that mitochondria play a key role in CD8+ T cell activation, effector function, and persistence in tumors. In this study, we showed that there was an increase in overall mitochondrial function, including mitochondrial mass and membrane potential, during both mouse and human CD8+ T cell activation. CD8+ T cell mitochondrial membrane potential was closely correlated with granzyme B and IFN-γ production, demonstrating the significance of mitochondria in effector T cell function. Additionally, activated CD8+ T cells that migrate on ICAM-1 and CXCL12 consumed significantly more oxygen than stationary CD8+ T cells. Inhibition of mitochondrial respiration decreased the velocity of CD8+ T cell migration, indicating the importance of mitochondrial metabolism in CD8+ T cell migration. Remote optical stimulation of CD8+ T cells that express our newly developed "OptoMito-On" successfully enhanced mitochondrial ATP production and improved overall CD8+ T cell migration and effector function. Our study provides new insight into the effect of the mitochondrial membrane potential on CD8+ T cell effector function and demonstrates the development of a novel optogenetic technique to remotely control T cell metabolism and effector function at the target tumor site with outstanding specificity and temporospatial resolution.
Collapse
Affiliation(s)
- Andrea M. Amitrano
- Department of Pathology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Brandon J. Berry
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, United States
| | - Kihong Lim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Kyun-Do Kim
- Center for Convergent Research of Emerging Virus Infection, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Richard E. Waugh
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, United States
| | - Andrew P. Wojtovich
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, United States
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
125
|
Bahadoran A, Bezavada L, Smallwood HS. Fueling influenza and the immune response: Implications for metabolic reprogramming during influenza infection and immunometabolism. Immunol Rev 2021; 295:140-166. [PMID: 32320072 DOI: 10.1111/imr.12851] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/19/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
Recent studies support the notion that glycolysis and oxidative phosphorylation are rheostats in immune cells whose bioenergetics have functional outputs in terms of their biology. Specific intrinsic and extrinsic molecular factors function as molecular potentiometers to adjust and control glycolytic to respiratory power output. In many cases, these potentiometers are used by influenza viruses and immune cells to support pathogenesis and the host immune response, respectively. Influenza virus infects the respiratory tract, providing a specific environmental niche, while immune cells encounter variable nutrient concentrations as they migrate in response to infection. Immune cell subsets have distinct metabolic programs that adjust to meet energetic and biosynthetic requirements to support effector functions, differentiation, and longevity in their ever-changing microenvironments. This review details how influenza coopts the host cell for metabolic reprogramming and describes the overlap of these regulatory controls in immune cells whose function and fate are dictated by metabolism. These details are contextualized with emerging evidence of the consequences of influenza-induced changes in metabolic homeostasis on disease progression.
Collapse
Affiliation(s)
- Azadeh Bahadoran
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Lavanya Bezavada
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Heather S Smallwood
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
126
|
Dastmalchi F, Deleyrolle LP, Karachi A, Mitchell DA, Rahman M. Metabolomics Monitoring of Treatment Response to Brain Tumor Immunotherapy. Front Oncol 2021; 11:691246. [PMID: 34150663 PMCID: PMC8209463 DOI: 10.3389/fonc.2021.691246] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has revolutionized care for many solid tissue malignancies, and is being investigated for efficacy in the treatment of malignant brain tumors. Identifying a non-invasive monitoring technique such as metabolomics monitoring to predict patient response to immunotherapy has the potential to simplify treatment decision-making and to ensure therapy is tailored based on early patient response. Metabolomic analysis of peripheral immune response is feasible due to large metabolic shifts that immune cells undergo when activated. The utility of this approach is under investigation. In this review, we discuss the metabolic changes induced during activation of an immune response, and the role of metabolic profiling to monitor immune responses in the context of immunotherapy for malignant brain tumors. This review provides original insights into how metabolomics monitoring could have an important impact in the field of tumor immunotherapy if achievable.
Collapse
Affiliation(s)
- Farhad Dastmalchi
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, United States
| | - Loic P Deleyrolle
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, United States
| | - Aida Karachi
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, United States
| | - Duane A Mitchell
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, United States
| | - Maryam Rahman
- Department of Neurosurgery, Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida, Gainesville, FL, United States
| |
Collapse
|
127
|
Metabolic Interplay between the Immune System and Melanoma Cells: Therapeutic Implications. Biomedicines 2021; 9:biomedicines9060607. [PMID: 34073463 PMCID: PMC8227307 DOI: 10.3390/biomedicines9060607] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Malignant melanoma represents the most fatal skin cancer due to its aggressive biological behavior and high metastatic potential. Treatment strategies for advanced disease have dramatically changed over the last years due to the introduction of BRAF/MEK inhibitors and immunotherapy. However, many patients either display primary (i.e., innate) or eventually develop secondary (i.e., acquired) resistance to systemic treatments. Treatment resistance depends on multiple mechanisms driven by a set of rewiring processes, which involve cancer metabolism, epigenetic, gene expression, and interactions within the tumor microenvironment. Prognostic and predictive biomarkers are needed to guide patients’ selection and treatment decisions. Indeed, there are no recognized clinical or biological characteristics that identify which patients will benefit more from available treatments, but several biomarkers have been studied with promising preliminary results. In this review, we will summarize novel tumor metabolic pathways and tumor-host metabolic crosstalk mechanisms leading to melanoma progression and drug resistance, with an overview on their translational potential as novel therapeutic targets.
Collapse
|
128
|
Traba J, Sack MN, Waldmann TA, Anton OM. Immunometabolism at the Nexus of Cancer Therapeutic Efficacy and Resistance. Front Immunol 2021; 12:657293. [PMID: 34079545 PMCID: PMC8166297 DOI: 10.3389/fimmu.2021.657293] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/29/2021] [Indexed: 12/22/2022] Open
Abstract
Constitutive activity of the immune surveillance system detects and kills cancerous cells, although many cancers have developed strategies to avoid detection and to resist their destruction. Cancer immunotherapy entails the manipulation of components of the endogenous immune system as targeted approaches to control and destroy cancer cells. Since one of the major limitations for the antitumor activity of immune cells is the immunosuppressive tumor microenvironment (TME), boosting the immune system to overcome the inhibition provided by the TME is a critical component of oncotherapeutics. In this article, we discuss the main effects of the TME on the metabolism and function of immune cells, and review emerging strategies to potentiate immune cell metabolism to promote antitumor effects either as monotherapeutics or in combination with conventional chemotherapy to optimize cancer management.
Collapse
Affiliation(s)
- Javier Traba
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Michael N. Sack
- Cardiovascular Branch, National Heart, Lung and Blood Institute, NIH, Bethesda, MD, United States
| | - Thomas A. Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| | - Olga M. Anton
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, United States
| |
Collapse
|
129
|
Certo M, Elkafrawy H, Pucino V, Cucchi D, Cheung KC, Mauro C. Endothelial cell and T-cell crosstalk: Targeting metabolism as a therapeutic approach in chronic inflammation. Br J Pharmacol 2021; 178:2041-2059. [PMID: 31999357 PMCID: PMC8246814 DOI: 10.1111/bph.15002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
The role of metabolic reprogramming in the coordination of the immune response has gained increasing consideration in recent years. Indeed, it has become clear that changes in the metabolic status of immune cells can alter their functional properties. During inflammation, T cells need to generate sufficient energy and biomolecules to support growth, proliferation, and effector functions. Therefore, T cells need to rearrange their metabolism to meet these demands. A similar metabolic reprogramming has been described in endothelial cells, which have the ability to interact with and modulate the function of immune cells. In this overview, we will discuss recent insights in the complex crosstalk between endothelial cells and T cells as well as their metabolic reprogramming following activation. We highlight key components of this metabolic switch that can lead to the development of new therapeutics against chronic inflammatory disorders. LINKED ARTICLES: This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc.
Collapse
Affiliation(s)
- Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Hagar Elkafrawy
- Medical Biochemistry and Molecular Biology Department, Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Valentina Pucino
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Danilo Cucchi
- Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Kenneth C.P. Cheung
- School of Life SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Cardiovascular Sciences, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Metabolism and Systems Research, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
130
|
Metabolic Modulation of Immunity: A New Concept in Cancer Immunotherapy. Cell Rep 2021; 32:107848. [PMID: 32640218 DOI: 10.1016/j.celrep.2020.107848] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/22/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022] Open
Abstract
Immunotherapy shifted the paradigm of cancer treatment. The clinical approval of immune checkpoint blockade and adoptive cell transfer led to considerable success in several tumor types. However, for a significant number of patients, these therapies have proven ineffective. Growing evidence shows that the metabolic requirements of immune cells in the tumor microenvironment (TME) greatly influence the success of immunotherapy. It is well established that the TME influences energy consumption and metabolic reprogramming of immune cells, often inducing them to become tolerogenic and inefficient in cancer cell eradication. Increasing nutrient availability using pharmacological modulators of metabolism or antibodies targeting specific immune receptors are strategies that support energetic rewiring of immune cells and boost their anti-tumor capacity. In this review, we describe the metabolic features of the diverse immune cell types in the context of the TME and discuss how these immunomodulatory strategies could synergize with immunotherapy to circumvent its current limitations.
Collapse
|
131
|
Abstract
Recent evidence supports the notion that mitochondrial metabolism is necessary for T cell activation, proliferation, and function. Mitochondrial metabolism supports T cell anabolism by providing key metabolites for macromolecule synthesis and generating metabolites for T cell function. In this review, we focus on how mitochondrial metabolism controls conventional and regulatory T cell fates and function.
Collapse
Affiliation(s)
- Elizabeth M Steinert
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Karthik Vasan
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Navdeep S Chandel
- Department of Medicine, Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| |
Collapse
|
132
|
Rangel Rivera GO, Knochelmann HM, Dwyer CJ, Smith AS, Wyatt MM, Rivera-Reyes AM, Thaxton JE, Paulos CM. Fundamentals of T Cell Metabolism and Strategies to Enhance Cancer Immunotherapy. Front Immunol 2021; 12:645242. [PMID: 33815400 PMCID: PMC8014042 DOI: 10.3389/fimmu.2021.645242] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/01/2021] [Indexed: 01/11/2023] Open
Abstract
Emerging reports show that metabolic pathways can be targeted to enhance T cell-mediated immunity to tumors. Yet, tumors consume key metabolites in the host to survive, thus robbing T cells of these nutrients to function and thrive. T cells are often deprived of basic building blocks for energy in the tumor, including glucose and amino acids needed to proliferate or produce cytotoxic molecules against tumors. Immunosuppressive molecules in the host further compromise the lytic capacity of T cells. Moreover, checkpoint receptors inhibit T cell responses by impairing their bioenergetic potential within tumors. In this review, we discuss the fundamental metabolic pathways involved in T cell activation, differentiation and response against tumors. We then address ways to target metabolic pathways to improve the next generation of immunotherapies for cancer patients.
Collapse
Affiliation(s)
- Guillermo O Rangel Rivera
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Hannah M Knochelmann
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Connor J Dwyer
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Aubrey S Smith
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Surgery, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Megan M Wyatt
- Department of Surgery, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Amalia M Rivera-Reyes
- Department of Surgery, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| | - Jessica E Thaxton
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Department of Orthopaedics and Physical Medicine, Medical University of South Carolina, Charleston, SC, United States
| | - Chrystal M Paulos
- Department of Surgery, Emory University, Atlanta, GA, United States.,Department of Microbiology and Immunology, Emory University, Atlanta, GA, United States
| |
Collapse
|
133
|
Mangal JL, Handlos JL, Esrafili A, Inamdar S, Mcmillian S, Wankhede M, Gottardi R, Acharya AP. Engineering Metabolism of Chimeric Antigen Receptor (CAR) Cells for Developing Efficient Immunotherapies. Cancers (Basel) 2021; 13:1123. [PMID: 33807867 PMCID: PMC7962004 DOI: 10.3390/cancers13051123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/23/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell-based therapies have shown tremendous advancement in clinical and pre-clinical studies for the treatment of hematological malignancies, such as the refractory of pre-B cell acute lymphoblastic leukemia (B-ALL), chronic lymphocytic leukemia (CLL), and large B cell lymphoma (LBCL). However, CAR T cell therapy for solid tumors has not been successful clinically. Although, some research efforts, such as combining CARs with immune checkpoint inhibitor-based therapy, have been used to expand the application of CAR T cells for the treatment of solid tumors. Importantly, further understanding of the coordination of nutrient and energy supplies needed for CAR T cell expansion and function, especially in the tumor microenvironment (TME), is greatly needed. In addition to CAR T cells, there is great interest in utilizing other types of CAR immune cells, such as CAR NK and CAR macrophages that can infiltrate solid tumors. However, the metabolic competition in the TME between cancer cells and immune cells remains a challenge. Bioengineering technologies, such as metabolic engineering, can make a substantial contribution when developing CAR cells to have an ability to overcome nutrient-paucity in the solid TME. This review introduces technologies that have been used to generate metabolically fit CAR-immune cells as a treatment for hematological malignancies and solid tumors, and briefly discusses the challenges to treat solid tumors with CAR-immune cells.
Collapse
Affiliation(s)
- Joslyn L. Mangal
- Biological Design Graduate Program, School for Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA;
| | - Jamie L. Handlos
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; (J.L.H.); (A.E.); (S.I.); (S.M.); (M.W.)
| | - Arezoo Esrafili
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; (J.L.H.); (A.E.); (S.I.); (S.M.); (M.W.)
| | - Sahil Inamdar
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; (J.L.H.); (A.E.); (S.I.); (S.M.); (M.W.)
| | - Sidnee Mcmillian
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; (J.L.H.); (A.E.); (S.I.); (S.M.); (M.W.)
| | - Mamta Wankhede
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; (J.L.H.); (A.E.); (S.I.); (S.M.); (M.W.)
| | - Riccardo Gottardi
- Department of Pediatrics, Division of Pulmonary Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA;
- Fondazione Ri.MED, 90133 Palermo, Italy
| | - Abhinav P. Acharya
- Biological Design Graduate Program, School for Biological and Health Systems Engineering, Arizona State University, Tempe, AZ 85281, USA;
- Department of Chemical Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA; (J.L.H.); (A.E.); (S.I.); (S.M.); (M.W.)
- Department of Materials Science and Engineering, School for the Engineering of Matter, Transport, and Energy, Arizona State University, Tempe, AZ 85281, USA
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Tempe, AZ 85281, USA
| |
Collapse
|
134
|
Manzo T, Prentice BM, Anderson KG, Raman A, Schalck A, Codreanu GS, Nava Lauson CB, Tiberti S, Raimondi A, Jones MA, Reyzer M, Bates BM, Spraggins JM, Patterson NH, McLean JA, Rai K, Tacchetti C, Tucci S, Wargo JA, Rodighiero S, Clise-Dwyer K, Sherrod SD, Kim M, Navin NE, Caprioli RM, Greenberg PD, Draetta G, Nezi L. Accumulation of long-chain fatty acids in the tumor microenvironment drives dysfunction in intrapancreatic CD8+ T cells. J Exp Med 2021; 217:151833. [PMID: 32491160 PMCID: PMC7398173 DOI: 10.1084/jem.20191920] [Citation(s) in RCA: 195] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/14/2020] [Accepted: 03/19/2020] [Indexed: 12/13/2022] Open
Abstract
CD8+ T cells are master effectors of antitumor immunity, and their presence at tumor sites correlates with favorable outcomes. However, metabolic constraints imposed by the tumor microenvironment (TME) can dampen their ability to control tumor progression. We describe lipid accumulation in the TME areas of pancreatic ductal adenocarcinoma (PDA) populated by CD8+ T cells infiltrating both murine and human tumors. In this lipid-rich but otherwise nutrient-poor TME, access to using lipid metabolism becomes particularly valuable for sustaining cell functions. Here, we found that intrapancreatic CD8+ T cells progressively accumulate specific long-chain fatty acids (LCFAs), which, rather than provide a fuel source, impair their mitochondrial function and trigger major transcriptional reprogramming of pathways involved in lipid metabolism, with the subsequent reduction of fatty acid catabolism. In particular, intrapancreatic CD8+ T cells specifically exhibit down-regulation of the very-long-chain acyl-CoA dehydrogenase (VLCAD) enzyme, which exacerbates accumulation of LCFAs and very-long-chain fatty acids (VLCFAs) that mediate lipotoxicity. Metabolic reprogramming of tumor-specific T cells through enforced expression of ACADVL enabled enhanced intratumoral T cell survival and persistence in an engineered mouse model of PDA, overcoming one of the major hurdles to immunotherapy for PDA.
Collapse
Affiliation(s)
- Teresa Manzo
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Boone M Prentice
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Kristin G Anderson
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Ayush Raman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aislyn Schalck
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Carina B Nava Lauson
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Silvia Tiberti
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Andrea Raimondi
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Marissa A Jones
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Michelle Reyzer
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Breanna M Bates
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Jeffrey M Spraggins
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Nathan H Patterson
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - John A McLean
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Carlo Tacchetti
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, San Raffaele Vita-Salute University, Milano, Italy
| | - Sara Tucci
- Laboratory of Clinical Biochemistry and Metabolism Center for Pediatrics and Adolescent Medicine, University of Freiburg, Freiburg, Germany
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Simona Rodighiero
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy
| | - Karen Clise-Dwyer
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Stacy D Sherrod
- Center for Innovative Technology, Vanderbilt University, Nashville, TN
| | - Michael Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nicholas E Navin
- Department of Genetics and Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard M Caprioli
- Department of Biochemistry, Mass Spectrometry Research Center, Department of Chemistry, Department of Pharmacology and Medicine, Vanderbilt University, Nashville, TN
| | - Philip D Greenberg
- Clinical Research Division and Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA.,Departments of Medicine/Oncology and Immunology, University of Washington School of Medicine, Seattle, WA
| | - Giulio Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Luigi Nezi
- Department of Experimental Oncology, IRCCS European Institute of Oncology, Milano, Italy.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
135
|
El Sayed R, Haibe Y, Amhaz G, Bouferraa Y, Shamseddine A. Metabolic Factors Affecting Tumor Immunogenicity: What Is Happening at the Cellular Level? Int J Mol Sci 2021; 22:2142. [PMID: 33670011 PMCID: PMC7927105 DOI: 10.3390/ijms22042142] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/07/2021] [Accepted: 02/11/2021] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy has changed the treatment paradigm in multiple solid and hematologic malignancies. However, response remains limited in a significant number of cases, with tumors developing innate or acquired resistance to checkpoint inhibition. Certain "hot" or "immune-sensitive" tumors become "cold" or "immune-resistant", with resultant tumor growth and disease progression. Multiple factors are at play both at the cellular and host levels. The tumor microenvironment (TME) contributes the most to immune-resistance, with nutrient deficiency, hypoxia, acidity and different secreted inflammatory markers, all contributing to modulation of immune-metabolism and reprogramming of immune cells towards pro- or anti-inflammatory phenotypes. Both the tumor and surrounding immune cells require high amounts of glucose, amino acids and fatty acids to fulfill their energy demands. Thus, both compete over one pool of nutrients that falls short on needs, obliging cells to resort to alternative adaptive metabolic mechanisms that take part in shaping their inflammatory phenotypes. Aerobic or anaerobic glycolysis, oxidative phosphorylation, tryptophan catabolism, glutaminolysis, fatty acid synthesis or fatty acid oxidation, etc. are all mechanisms that contribute to immune modulation. Different pathways are triggered leading to genetic and epigenetic modulation with consequent reprogramming of immune cells such as T-cells (effector, memory or regulatory), tumor-associated macrophages (TAMs) (M1 or M2), natural killers (NK) cells (active or senescent), and dendritic cells (DC) (effector or tolerogenic), etc. Even host factors such as inflammatory conditions, obesity, caloric deficit, gender, infections, microbiota and smoking status, may be as well contributory to immune modulation, anti-tumor immunity and response to immune checkpoint inhibition. Given the complex and delicate metabolic networks within the tumor microenvironment controlling immune response, targeting key metabolic modulators may represent a valid therapeutic option to be combined with checkpoint inhibitors in an attempt to regain immune function.
Collapse
Affiliation(s)
- Rola El Sayed
- Global Health Institute, American University of Beirut, Beirut 11-0236, Lebanon;
| | - Yolla Haibe
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| | - Ghid Amhaz
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| | - Youssef Bouferraa
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| | - Ali Shamseddine
- Division of Hematology/Oncology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut 11-0236, Lebanon; (Y.H.); (G.A.); (Y.B.)
| |
Collapse
|
136
|
Briceño P, Rivas-Yañez E, Rosemblatt MV, Parra-Tello B, Farías P, Vargas L, Simon V, Cárdenas C, Lladser A, Salazar-Onfray F, Elorza AA, Rosemblatt M, Bono MR, Sauma D. CD73 Ectonucleotidase Restrains CD8+ T Cell Metabolic Fitness and Anti-tumoral Activity. Front Cell Dev Biol 2021; 9:638037. [PMID: 33681221 PMCID: PMC7930398 DOI: 10.3389/fcell.2021.638037] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 01/20/2021] [Indexed: 12/19/2022] Open
Abstract
CD39 and CD73 are ectoenzymes that dephosphorylate ATP into its metabolites; ADP, AMP, and adenosine, and thus are considered instrumental in the development of immunosuppressive microenvironments. We have previously shown that within the CD8+ T cell population, naïve and memory cells express the CD73 ectonucleotidase, while terminally differentiated effector cells are devoid of this enzyme. This evidence suggests that adenosine might exert an autocrine effect on CD8+ T cells during T cell differentiation. To study the possible role of CD73 and adenosine during this process, we compared the expression of the adenosinergic signaling components, the phenotype, and the functional properties between CD73-deficient and WT CD8+ T cells. Upon activation, we observed an upregulation of CD73 expression in CD8+ T cells along with an upregulation of the adenosine A2A receptor. Interestingly, when we differentiated CD8+ T cells to Tc1 cells in vitro, we observed that these cells produce adenosine and that CD73-deficient cells present a higher cytotoxic potential evidenced by an increase in IFN-γ, TNF-α, and granzyme B production. Moreover, CD73-deficient cells presented a increased glucose uptake and higher mitochondrial respiration, indicating that this ectonucleotidase restrict the mitochondrial capacity in CD8+ T cells. In agreement, when adoptively transferred, antigen-specific CD73-deficient CD8+ T cells were more effective in reducing the tumor burden in B16.OVA melanoma-bearing mice and presented lower levels of exhaustion markers than wild type cells. All these data suggest an autocrine effect of CD73-mediated adenosine production, limiting differentiation and cytotoxic T cells' metabolic fitness.
Collapse
Affiliation(s)
- Pedro Briceño
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Elizabeth Rivas-Yañez
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Mariana V Rosemblatt
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile
| | - Brian Parra-Tello
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Paula Farías
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
| | - Leonardo Vargas
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Valeska Simon
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - César Cárdenas
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
| | - Alvaro Lladser
- Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile.,Fundacion Ciencia & Vida, Santiago, Chile
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Alvaro A Elorza
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Mario Rosemblatt
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Facultad de Medicina y Ciencia, Universidad San Sebastian, Santiago, Chile.,Fundacion Ciencia & Vida, Santiago, Chile
| | - María Rosa Bono
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Daniela Sauma
- Departamento de Biologia, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| |
Collapse
|
137
|
Lopes N, McIntyre C, Martin S, Raverdeau M, Sumaria N, Kohlgruber AC, Fiala GJ, Agudelo LZ, Dyck L, Kane H, Douglas A, Cunningham S, Prendeville H, Loftus R, Carmody C, Pierre P, Kellis M, Brenner M, Argüello RJ, Silva-Santos B, Pennington DJ, Lynch L. Distinct metabolic programs established in the thymus control effector functions of γδ T cell subsets in tumor microenvironments. Nat Immunol 2021; 22:179-192. [PMID: 33462452 PMCID: PMC7610600 DOI: 10.1038/s41590-020-00848-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 11/24/2020] [Indexed: 01/29/2023]
Abstract
Metabolic programming controls immune cell lineages and functions, but little is known about γδ T cell metabolism. Here, we found that γδ T cell subsets making either interferon-γ (IFN-γ) or interleukin (IL)-17 have intrinsically distinct metabolic requirements. Whereas IFN-γ+ γδ T cells were almost exclusively dependent on glycolysis, IL-17+ γδ T cells strongly engaged oxidative metabolism, with increased mitochondrial mass and activity. These distinct metabolic signatures were surprisingly imprinted early during thymic development and were stably maintained in the periphery and within tumors. Moreover, pro-tumoral IL-17+ γδ T cells selectively showed high lipid uptake and intracellular lipid storage and were expanded in obesity and in tumors of obese mice. Conversely, glucose supplementation enhanced the antitumor functions of IFN-γ+ γδ T cells and reduced tumor growth upon adoptive transfer. These findings have important implications for the differentiation of effector γδ T cells and their manipulation in cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Cell Line, Tumor
- Cell Lineage
- Colonic Neoplasms/immunology
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Energy Metabolism
- Female
- Glucose/metabolism
- Glycolysis
- Humans
- Immunotherapy, Adoptive
- Interferon-gamma/metabolism
- Interleukin-17/metabolism
- Lipid Metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/transplantation
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitochondria/metabolism
- Obesity/immunology
- Obesity/metabolism
- Organ Culture Techniques
- Phenotype
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Signal Transduction
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/transplantation
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Tumor Burden
- Tumor Microenvironment
- Mice
Collapse
Affiliation(s)
- Noella Lopes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Claire McIntyre
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stefania Martin
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Mathilde Raverdeau
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Nital Sumaria
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | | | - Gina J Fiala
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Leandro Z Agudelo
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Lydia Dyck
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Harry Kane
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Aaron Douglas
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Stephen Cunningham
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Hannah Prendeville
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Roisin Loftus
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Colleen Carmody
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Philippe Pierre
- Aix Marseille Université, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
- Institute for Research in Biomedicine (iBiMED) and Ilidio Pinho Foundation, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Manolis Kellis
- MIT Computer Science and Artificial Intelligence Laboratory, Cambridge, MA, USA
| | - Michael Brenner
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rafael J Argüello
- Aix Marseille Université, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Daniel J Pennington
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK.
| | - Lydia Lynch
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
138
|
Zhao S, Peralta RM, Avina-Ochoa N, Delgoffe GM, Kaech SM. Metabolic regulation of T cells in the tumor microenvironment by nutrient availability and diet. Semin Immunol 2021; 52:101485. [PMID: 34462190 PMCID: PMC8545851 DOI: 10.1016/j.smim.2021.101485] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 08/12/2021] [Indexed: 12/11/2022]
Abstract
Recent advances in immunotherapies such as immune checkpoint blockade (ICB) and chimeric antigen receptor T cells (CAR-T) for the treatment of cancer have generated excitement over their ability to yield durable, and potentially curative, responses in a multitude of cancers. These findings have established that the immune system is capable of eliminating tumors and led us to a better, albeit still incomplete, understanding of the mechanisms by which tumors interact with and evade destruction by the immune system. Given the central role of T cells in immunotherapy, elucidating the cell intrinsic and extrinsic factors that govern T cell function in tumors will facilitate the development of immunotherapies that establish durable responses in a greater number of patients. One such factor is metabolism, a set of fundamental cellular processes that not only sustains cell survival and proliferation, but also serves as a means for cells to interpret their local environment. Nutrient sensing is critical for T cells that must infiltrate into a metabolically challenging tumor microenvironment and expand under these harsh conditions to eliminate cancerous cells. Here we introduce T cell exhaustion with respect to cellular metabolism, followed by a discussion of nutrient availability at the tumor and organismal level in relation to T cell metabolism and function to provide rationale for the study and targeting of metabolism in anti-tumor immune responses.
Collapse
Affiliation(s)
- Steven Zhao
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ronal M Peralta
- Tumor Microenvironment Center, Department of Immunology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, USA
| | - Natalia Avina-Ochoa
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Greg M Delgoffe
- Tumor Microenvironment Center, Department of Immunology, UPMC Hillman Cancer Center and University of Pittsburgh, Pittsburgh, PA, USA.
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
139
|
Yu T, Dong T, Eyvani H, Fang Y, Wang X, Zhang X, Lu X. Metabolic interventions: A new insight into the cancer immunotherapy. Arch Biochem Biophys 2021; 697:108659. [PMID: 33144083 PMCID: PMC8638212 DOI: 10.1016/j.abb.2020.108659] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/15/2020] [Accepted: 10/29/2020] [Indexed: 12/13/2022]
Abstract
Metabolic reprogramming confers cancer cells plasticity and viability under harsh conditions. Such active alterations lead to cell metabolic dependency, which can be exploited as an attractive target in development of effective antitumor therapies. Similar to cancer cells, activated T cells also execute global metabolic reprogramming for their proliferation and effector functions when recruited to the tumor microenvironment (TME). However, the high metabolic activity of rapidly proliferating cancer cells can compete for nutrients with immune cells in the TME, and consequently, suppressing their anti-tumor functions. Thus, therapeutic strategies could aim to restore T cell metabolism and anti-tumor responses in the TME by targeting the metabolic dependence of cancer cells. In this review, we highlight current research progress on metabolic reprogramming and the interplay between cancer cells and immune cells. We also discuss potential therapeutic intervention strategies for targeting metabolic pathways to improve cancer immunotherapy efficacy.
Collapse
Affiliation(s)
- Tao Yu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tianhan Dong
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Haniyeh Eyvani
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yuanzhang Fang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiyu Wang
- Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xinna Zhang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA; Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
140
|
Hematopoietic versus Solid Cancers and T Cell Dysfunction: Looking for Similarities and Distinctions. Cancers (Basel) 2021; 13:cancers13020284. [PMID: 33466674 PMCID: PMC7828769 DOI: 10.3390/cancers13020284] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Dysfunction of the immune T cell compartment occurs in many hematopoietic as well as solid cancers and hampers successful application of new immunotherapeutic approaches. A complete understanding of T cell dysfunction might improve the outcome of such therapies, but an overview in the various cancers is still lacking. We aim to map areas of similarities and differences in solid versus hematopoietic malignancies, providing a high-level rather than a detailed perspective on T cell dysfunction in those tumors. Abstract Cancer cells escape, suppress and exploit the host immune system to sustain themselves, and the tumor microenvironment (TME) actively dampens T cell function by various mechanisms. Over the last years, new immunotherapeutic approaches, such as adoptive chimeric antigen receptor (CAR) T cell therapy and immune checkpoint inhibitors, have been successfully applied for refractory malignancies that could only be treated in a palliative manner previously. Engaging the anti-tumor activity of the immune system, including CAR T cell therapy to target the CD19 B cell antigen, proved to be effective in acute lymphocytic leukemia. In low-grade hematopoietic B cell malignancies, such as chronic lymphocytic leukemia, clinical outcomes have been tempered by cancer-induced T cell dysfunction characterized in part by a state of metabolic lethargy. In multiple myeloma, novel antigens such as BCMA and CD38 are being explored for CAR T cells. In solid cancers, T cell-based immunotherapies have been applied successfully to melanoma and lung cancers, whereas application in e.g., breast cancer lags behind and is modestly effective as yet. The main hurdles for CAR T cell immunotherapy in solid tumors are the lack of suitable antigens, anatomical inaccessibility, and T cell anergy due to immunosuppressive TME. Given the wide range of success and failure of immunotherapies in various cancer types, it is crucial to comprehend the underlying similarities and distinctions in T cell dysfunction. Hence, this review aims at comparing selected, distinct B cell-derived versus solid cancer types and at describing means by which malignant cells and TME might dampen T cell anti-tumor activity, with special focus on immunometabolism. Drawing a meaningful parallel between the efficacy of immunotherapy and the extent of T cell dysfunction will shed light on areas where we can improve immune function to battle cancer.
Collapse
|
141
|
Jung JG, Le A. Metabolism of Immune Cells in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1311:173-185. [PMID: 34014543 DOI: 10.1007/978-3-030-65768-0_13] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The tumor microenvironment (TME) is a complex biological structure surrounding tumor cells and includes blood vessels, immune cells, fibroblasts, adipocytes, and extracellular matrix (ECM) [1, 2]. These heterogeneous surrounding structures provide nutrients, metabolites, and signaling molecules to provide a cancer-friendly environment. The metabolic interplay between immune cells and cancer cells in the TME is a key feature not only for understanding tumor biology but also for discovering cancer cells' vulnerability. As cancer immunotherapy to treat cancer patients and the use of metabolomics technologies become more and more common [3], the importance of the interplay between cancer cells and immune cells in the TME is emerging with respect to not only cell-to-cell interactions but also metabolic pathways. This interaction between immune cells and cancer cells is a complex and dynamic process in which immune cells act as a determinant factor of cancer cells' fate and vice versa. In this chapter, we provide an overview of the metabolic interplay between immune cells and cancer cells and discuss the therapeutic opportunities as a result of this interplay in order to define targets for cancer treatment. It is important to understand and identify therapeutic targets that interrupt this cancerpromoting relationship between cancer cells and the surrounding immune cells, allowing for maximum efficacy of immune checkpoint inhibitors as well as other genetic and cellular therapies.
Collapse
Affiliation(s)
- Jin G Jung
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anne Le
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD, USA.
| |
Collapse
|
142
|
Poorebrahim M, Melief J, Pico de Coaña Y, L Wickström S, Cid-Arregui A, Kiessling R. Counteracting CAR T cell dysfunction. Oncogene 2021; 40:421-435. [PMID: 33168929 PMCID: PMC7808935 DOI: 10.1038/s41388-020-01501-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 02/08/2023]
Abstract
In spite of high rates of complete remission following chimeric antigen receptor (CAR) T cell therapy, the efficacy of this approach is limited by generation of dysfunctional CAR T cells in vivo, conceivably induced by immunosuppressive tumor microenvironment (TME) and excessive antigen exposure. Exhaustion and senescence are two critical dysfunctional states that impose a pivotal hurdle for successful CAR T cell therapies. Recently, modified CAR T cells with an "exhaustion-resistant" phenotype have shown superior antitumor functions and prolonged lifespan. In addition, several studies have indicated the feasibility of senescence delay in CAR T cells. Here, we review the latest reports regarding blockade of CAR T cell exhaustion and senescence with a particular focus on the exhaustion-inducing pathways. Subsequently, we describe what potential these latest insights offer for boosting the potency of adoptive cell transfer (ACT) therapies involving CAR T cells. Furthermore, we discuss how induction of costimulation, cytokine exposure, and TME modulation can impact on CAR T cell efficacy and persistence, while potential safety issues associated with reinvigorated CAR T cells will also be addressed.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden. .,Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Jeroen Melief
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yago Pico de Coaña
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stina L Wickström
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Angel Cid-Arregui
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rolf Kiessling
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
143
|
Elia I, Haigis MC. Metabolites and the tumour microenvironment: from cellular mechanisms to systemic metabolism. Nat Metab 2021; 3:21-32. [PMID: 33398194 PMCID: PMC8097259 DOI: 10.1038/s42255-020-00317-z] [Citation(s) in RCA: 351] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 10/29/2020] [Indexed: 02/07/2023]
Abstract
Metabolic transformation is a hallmark of cancer and a critical target for cancer therapy. Cancer metabolism and behaviour are regulated by cell-intrinsic factors as well as metabolite availability in the tumour microenvironment (TME). This metabolic niche within the TME is shaped by four tiers of regulation: (1) intrinsic tumour cell metabolism, (2) interactions between cancer cells and non-cancerous cells, (3) tumour location and heterogeneity and (4) whole-body metabolic homeostasis. Here, we define these modes of metabolic regulation and review how distinct cell types contribute to the metabolite composition of the TME. Finally, we connect these insights to understand how each of these tiers offers unique therapeutic potential to modulate the metabolic profile and function of all cells inhabiting the TME.
Collapse
Affiliation(s)
- Ilaria Elia
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Ludwig Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Ludwig Cancer Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
144
|
Zhang Y, Lin A, Li Y, Ding W, Meng H, Luo P, Zhang J. Age and Mutations as Predictors of the Response to Immunotherapy in Head and Neck Squamous Cell Cancer. Front Cell Dev Biol 2020; 8:608969. [PMID: 33363171 PMCID: PMC7755718 DOI: 10.3389/fcell.2020.608969] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/19/2020] [Indexed: 12/17/2022] Open
Abstract
The immunosuppressive tumor microenvironment plays an essential role in the treatment of head and neck squamous cell carcinoma (HNSC). Compared to traditional chemoradiotherapy, immune checkpoint inhibitors (ICIs) have become increasingly important in HNSC therapy. Prior studies linked the efficacy of ICIs to PD-L1, microsatellite instability (MSI), HPV infection, tumor mutation burden (TMB), and tumor lymphocyte infiltration in patients with HNSC, but further verification is needed. Additional predictors are needed to recognize HNSC patients with a good response to ICIs. We collected the clinical information and mutation data of HNSC patients from Memorial Sloan Kettering Cancer Center (MSKCC) and The Cancer Genome Atlas (TCGA) databases to generate two clinical cohorts. The MSKCC cohort was used to recognize predictors related to the efficacy of ICIs, and the TCGA cohort was used to further examine the immune microenvironment features and signaling pathways that are significantly enriched in the subgroups of predictors. Multivariate Cox regression analysis indicated that age (HR = 0.50, p = 0.014) and ARID1A (HR = 0.13, p = 0.048), PIK3CA (HR = 0.45, p = 0.021), and TP53 (HR = 1.82, p = 0.035) mutations were potential predictors for ICI efficacy in HNSC patients. Age > 65 years and ARID1A or PIK3CA mutations correlated with good overall survival (OS). TP53 mutant-type (MT) patients experienced a worse prognosis than TP53 wild-type (WT) patients. The subgroups associated with a good prognosis (age > 65 years, ARID1A-MT, and PIK3CA-MT) universally had a high TMB and increased expression of immune checkpoint molecules. Although TP53-MT was associated with a high TMB, the expression of most immune checkpoint molecules and immune-related genes was lower in TP53-MT patients than TP53-WT patients, which may reflect low immunogenicity. Pathways related to the immunosuppressive tumor microenvironment were mostly enriched in the subgroups associated with a poor prognosis (age ≤ 65 years, low TMB, ARID1A-WT, PIK3CA-WT, and TP53-MT). In conclusion, the factors age > 65 years, PIK3CA-MT, and ARID1A-MT predicted favorable efficacy for ICI treatment in HNSC patients, and TP53 mutation was a negative predictor.
Collapse
Affiliation(s)
- Yueming Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yonghe Li
- Department of Otolaryngology-Head and Neck Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Weimin Ding
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hui Meng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
145
|
Elebo N, Fru P, Omoshoro-Jones J, Candy GP, Nweke EE. Role of different immune cells and metabolic pathways in modulating the immune response in pancreatic cancer (Review). Mol Med Rep 2020; 22:4981-4991. [PMID: 33174057 PMCID: PMC7646946 DOI: 10.3892/mmr.2020.11622] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is an aggressive cancer, making it a leading cause of cancer‑related deaths. It is characteristically resistant to treatment, which results in low survival rates. In pancreatic cancer, immune cells undergo transitions that can inhibit or promote their functions, enabling treatment resistance and tumor progression. These transitions can be fostered by metabolic pathways that are dysregulated during tumorigenesis. The present review aimed to summarize the different immune cells and their roles in pancreatic cancer. The review also highlighted the individual metabolic pathways in pancreatic cancer and how they enable transitions in immune cells. Finally, the potential of targeting metabolic pathways for effective therapeutic strategies was considered.
Collapse
Affiliation(s)
- Nnenna Elebo
- Department of Surgery, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, Gauteng 2193, South Africa
| | - Pascaline Fru
- Department of Surgery, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, Gauteng 2193, South Africa
| | - Jones Omoshoro-Jones
- Department of Surgery, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, Gauteng 2193, South Africa
| | - Geoffrey Patrick Candy
- Department of Surgery, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, Gauteng 2193, South Africa
| | - Ekene Emmanuel Nweke
- Department of Surgery, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, Gauteng 2193, South Africa
| |
Collapse
|
146
|
Lim J, Murthy A. Targeting Autophagy to Treat Cancer: Challenges and Opportunities. Front Pharmacol 2020; 11:590344. [PMID: 33381037 PMCID: PMC7768823 DOI: 10.3389/fphar.2020.590344] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
Autophagy is a catabolic process that targets its cargo for lysosomal degradation. In addition to its function in maintaining tissue homeostasis, autophagy is recognized to play a context-dependent role in cancer. Autophagy may inhibit tumor initiation under specific contexts; however, a growing body of evidence supports a pro-tumorigenic role of this pathway in established disease. In this setting, autophagy drives treatment resistance, metabolic changes, and immunosuppression both in a tumor-intrinsic and extrinsic manner. This observation has prompted renewed interest in targeting autophagy for cancer therapy. Novel genetic models have proven especially insightful, revealing unique and overlapping roles of individual autophagy-related genes in tumor progression. Despite identification of pharmacologically actionable nodes in the pathway, fundamental challenges still exist for successful therapeutic inhibition of autophagy. Here we summarize the current understanding of autophagy as a driver of resistance against targeted and immuno-therapies and highlight knowledge gaps that, if addressed, may provide meaningful advances in the treatment of cancer.
Collapse
Affiliation(s)
| | - Aditya Murthy
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA, United States
| |
Collapse
|
147
|
Galyean S, Sawant D, Shin AC. Immunometabolism, Micronutrients, and Bariatric Surgery: The Use of Transcriptomics and Microbiota-Targeted Therapies. Mediators Inflamm 2020; 2020:8862034. [PMID: 33281501 PMCID: PMC7685844 DOI: 10.1155/2020/8862034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/29/2020] [Accepted: 10/31/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Obesity is associated with the gut microbiota and decreased micronutrient status. Bariatric surgery is a recommended therapy for obesity. It can positively affect the composition of the gut bacteria but also disrupt absorption of nutrients. Low levels of micronutrients can affect metabolic processes, like glycolysis, TCA cycle, and oxidative phosphorylation, that are associated with the immune system also known as immunometabolism. METHODS MEDLINE, PUBMED, and Google Scholar were searched. Articles involving gut microbiome, micronutrient deficiency, gut-targeted therapies, transcriptome analysis, micronutrient supplementation, and bariatric surgery were included. RESULTS Studies show that micronutrients play a pivotal role in the intestinal immune system and regulating immunometabolism. Research demonstrates that gut-targeting therapies may improve the microbiome health for bariatric surgery populations. There is limited research that examines the role of micronutrients in modulating the gut microbiota among the bariatric surgery population. CONCLUSIONS Investigations are needed to understand the influence that micronutrient deficiencies have on the gut, particularly immunometabolism. Nutritional transcriptomics shows great potential in providing this type of analysis to develop gut-modulating therapies as well as more personalized nutrition recommendations for bariatric surgery patients.
Collapse
Affiliation(s)
- Shannon Galyean
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Dhanashree Sawant
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Andrew C. Shin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| |
Collapse
|
148
|
Sanchez J, Jackson I, Flaherty KR, Muliaditan T, Schurich A. Divergent Impact of Glucose Availability on Human Virus-Specific and Generically Activated CD8 T Cells. Metabolites 2020; 10:metabo10110461. [PMID: 33202938 PMCID: PMC7696163 DOI: 10.3390/metabo10110461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/27/2022] Open
Abstract
Upon activation T cells engage glucose metabolism to fuel the costly effector functions needed for a robust immune response. Consequently, the availability of glucose can impact on T cell function. The glucose concentrations used in conventional culture media and common metabolic assays are often artificially high, representing hyperglycaemic levels rarely present in vivo. We show here that reducing glucose concentration to physiological levels in culture differentially impacted on virus-specific compared to generically activated human CD8 T cell responses. In virus-specific T cells, limiting glucose availability significantly reduced the frequency of effector-cytokine producing T cells, but promoted the upregulation of CD69 and CD103 associated with an increased capacity for tissue retention. In contrast the functionality of generically activated T cells was largely unaffected and these showed reduced differentiation towards a residency phenotype. Furthermore, T cells being cultured at physiological glucose concentrations were more susceptible to viral infection. This setting resulted in significantly improved lentiviral transduction rates of primary cells. Our data suggest that CD8 T cells are exquisitely adapted to their niche and provide a reminder of the need to better mimic physiological conditions to study the complex nature of the human CD8 T cell immune response.
Collapse
Affiliation(s)
- Jenifer Sanchez
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy’s Hospital, King’s College London, London SE1 9RT, UK; (J.S.); (K.R.F.)
| | - Ian Jackson
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Guy’s Hospital, King’s College London, London SE1 9RT, UK;
| | - Katie R. Flaherty
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy’s Hospital, King’s College London, London SE1 9RT, UK; (J.S.); (K.R.F.)
| | - Tamara Muliaditan
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK;
- Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, David de Wiedgebouw, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Anna Schurich
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy’s Hospital, King’s College London, London SE1 9RT, UK; (J.S.); (K.R.F.)
- Correspondence:
| |
Collapse
|
149
|
Iranparast S, Tayebi S, Ahmadpour F, Yousefi B. Tumor-Induced Metabolism and T Cells Located in Tumor Environment. Curr Cancer Drug Targets 2020; 20:741-756. [PMID: 32691710 DOI: 10.2174/1568009620666200720010647] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 12/14/2022]
Abstract
Several subtypes of T cells are located in a tumor environment, each of which supplies their energy using different metabolic mechanisms. Since the cancer cells require high levels of glucose, the conditions of food poverty in the tumor environment can cause inactivation of immune cells, especially the T-effector cells, due to the need for glucose in the early stages of these cells activity. Different signaling pathways, such as PI3K-AKt-mTOR, MAPK, HIF-1α, etc., are activated or inactivated by the amount and type of energy source or oxygen levels that determine the fate of T cells in a cancerous environment. This review describes the metabolites in the tumor environment and their effects on the function of T cells. It also explains the signaling pathway of T cells in the tumor and normal conditions, due to the level of access to available metabolites and subtypes of T cells in the tumor environment.
Collapse
Affiliation(s)
- Sara Iranparast
- Department of Immunology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran,Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Sanaz Tayebi
- Department of Immunology, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fatemeh Ahmadpour
- Department of Biochemistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
150
|
The Role of Metabolic Enzymes in the Regulation of Inflammation. Metabolites 2020; 10:metabo10110426. [PMID: 33114536 PMCID: PMC7693344 DOI: 10.3390/metabo10110426] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 10/19/2020] [Accepted: 10/19/2020] [Indexed: 12/26/2022] Open
Abstract
Immune cells undergo dramatic metabolic reprogramming in response to external stimuli. These metabolic pathways, long considered as simple housekeeping functions, are increasingly understood to critically regulate the immune response, determining the activation, differentiation, and downstream effector functions of both lymphoid and myeloid cells. Within the complex metabolic networks associated with immune activation, several enzymes play key roles in regulating inflammation and represent potential therapeutic targets in human disease. In some cases, these enzymes control flux through pathways required to meet specific energetic or metabolic demands of the immune response. In other cases, key enzymes control the concentrations of immunoactive metabolites with direct roles in signaling. Finally, and perhaps most interestingly, several metabolic enzymes have evolved moonlighting functions, with roles in the immune response that are entirely independent of their conventional enzyme activities. Here, we review key metabolic enzymes that critically regulate inflammation, highlighting mechanistic insights and opportunities for clinical intervention.
Collapse
|