1
|
Alim L, Adityan S, Chen R, Neilson T, Coleborn E, Wilkinson AN, He Y, Irgam G, Bhavsar C, Lourie R, Rogers R, Cabraal N, Jagasia N, Chetty N, Perrin L, Hooper JD, Steptoe R, Wu SY. Antigen presentation potential is variable among human ovarian tumour and syngeneic murine models and dictates pre-clinical outcomes of immunotherapy. Biomed Pharmacother 2025; 187:118141. [PMID: 40347847 DOI: 10.1016/j.biopha.2025.118141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
High grade serous ovarian carcinoma (HGSC) is a fatal gynaecological malignancy with limited therapeutic options. Immunotherapies targeting MHC-I-dependent antigen presentation offer potential. Currently, the antigen presentation machinery (APM) of widely used syngeneic murine HGSC models remains poorly characterised, limiting translational relevance. Here, we systematically evaluate APM gene expression in syngeneic murine and patient samples. Tap1 and Psmb8 were identified as critical APM markers, deficient in murine models and strongly correlating with MHC-I expression. Hierarchical clustering correlation analysis using these markers revealed that ID8-p53⁻/⁻BRCA1⁻/⁻ was the most strongly correlated model and aligned with the largest patient subset. Moreover, ID8-ip1 correlated to the smaller second patient subset strongly. The low MHC-I expressing IG10 model was unique clustering alongside patient derived LP28 tumour and not fitting either patient subset. In vivo test of a novel combination immune therapy consisting of Flt3L, Poly(I:C), and paclitaxel therapy demonstrated significantly reduced tumour burden in high APM models (p53⁻/⁻BRCA1⁻/⁻, ID8-ip1; p < 0.01), but not IG10. Furthermore, high expressing MHC-I models were linked to enhanced DC expansion, CD8⁺ T-cell infiltration, and effector differentiation (131 % increase in ID8-ip1), alongside improved CD8⁺ T-cell activation and CD86⁺ B-cell co-stimulation. These findings establish MHC-I as a predictive biomarker for immunotherapy response and underscore the need for APM-enhancing strategies in antigen-poor tumours. By bridging murine models to human APM heterogeneity, this work provides a framework for optimising preclinical immunotherapy evaluation and patient stratification, advancing tailored therapeutic approaches for HGSC.
Collapse
Affiliation(s)
- Louisa Alim
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Siddharth Adityan
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Trent Neilson
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elaina Coleborn
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew N Wilkinson
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yaowu He
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Gowri Irgam
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rohan Lourie
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Rebecca Rogers
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Nimithri Cabraal
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Nisha Jagasia
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Naven Chetty
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Lewis Perrin
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - John D Hooper
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland 4102, Australia; Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia
| | - Raymond Steptoe
- Frazer Institute, University of Queensland, Brisbane, Australia
| | - Sherry Y Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
2
|
Juhász P, Méhes G. Tumor Hypoxia: How Conventional Histology Is Reshaped in Breast Carcinoma. Int J Mol Sci 2025; 26:4423. [PMID: 40362659 PMCID: PMC12072647 DOI: 10.3390/ijms26094423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Intratumoral hypoxia is common in any form of malignancy initializing focal necrosis or tumor cell adaptation. Hypoxia inducible factor-1-driven reprogramming favors the loss of tumor cell proliferation (quiescence) and partial cellular reversion, induces stemness and/or mesenchymal-like features in the exposed tumor areas. The characteristic hypoxia-driven tumor cell phenotype is principally directed to reduce energy consumption and to enhance survival, but the gained features also contribute to growth advantage and induce the reorganization of the microenvironment and protective mechanisms against external stress. The hypoxia-induced phenotypic changes are at least in part reflected by conventional morphology in breast carcinoma. Intratumoral variability of classical morphological signs, such as the growth pattern, the histological grade, cell proliferation, necrosis, microcalcification, angiogenesis, and the immune cell infiltration is also related with the co-existence of hypoxic areas. Thus, a deeper understanding of hypoxia-activated mechanisms is required. The current paper aims to summarize the major tissue factors involved in the response to hypoxia and their potential contribution to the breast carcinoma phenotype.
Collapse
Affiliation(s)
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen, Hungary;
| |
Collapse
|
3
|
Jeng LB, Shih FY, Liao YW, Shyu WC, Teng CF. Hypoxic tumor cell line lysate-pulsed dendritic cell vaccine exhibits better therapeutic effects on hepatocellular carcinoma. Br J Cancer 2025; 132:837-848. [PMID: 40050434 PMCID: PMC12041587 DOI: 10.1038/s41416-025-02975-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/03/2025] [Accepted: 02/26/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Dendritic cell (DC) vaccine is a promising immunotherapy for hepatocellular carcinoma (HCC) via triggering antigen-specific anti-tumor immunity. Hypoxia contributes to higher level and broader spectrum of antigen expression in tumor cells. METHODS This study aims to compare immunological activity and therapeutic efficacy between hypoxic and normoxic HCC cell line lysate-pulsed DC vaccines. RESULTS The results showed that hypoxic HCC cell line lysate-pulsed DC vaccines exhibited a stronger activity in producing interleukin-12 and promoting T cell proliferation and cytotoxicity in vitro. In HCC mice, hypoxic HCC cell line lysate-pulsed DC vaccines displayed a better efficacy in improving survival time and tumor volume and inducing intratumoral cytotoxic T cell infiltration and activation as well as tumor cell apoptosis. Adenylate kinase 4-derived antigens were important for hypoxic HCC cell line lysate-pulsed DC vaccine-elicited T cell killing. CONCLUSIONS In conclusion, this study demonstrated hypoxic HCC cell line lysate-pulsed DC vaccine as a potential therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, ROC
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan, ROC
- Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan, ROC
- School of Medicine, China Medical University, Taichung, Taiwan, ROC
| | - Fu-Ying Shih
- Ph.D. Program for Biotech Pharmaceutical Industry, China Medical University, Taichung, Taiwan, ROC
| | - Yu-Wen Liao
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
- Translational Medicine Research Center, China Medical University Hospital, Taichung, 404, Taiwan, ROC
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan, ROC
- Department of Occupational Therapy, Asia University, Taichung, Taiwan, ROC
| | - Chiao-Fang Teng
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan, ROC.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC.
- Master Program for Cancer Biology and Drug Discovery, China Medical University, Taichung, Taiwan, ROC.
| |
Collapse
|
4
|
Halpin-Veszeleiova K, Mallouh MP, Williamson LM, Apro AC, Botticello-Romero NR, Bahr C, Shin M, Ward KM, Rosenberg L, Ritov VB, Sitkovsky MV, Jackson EK, Spiess BD, Hatfield SM. Oxygen-carrying nanoemulsions and respiratory hyperoxia eliminate tumor hypoxia-induced immunosuppression. JCI Insight 2025; 10:e174675. [PMID: 40125552 PMCID: PMC11949039 DOI: 10.1172/jci.insight.174675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025] Open
Abstract
Hypoxia/hypoxia-inducible factor 1α-driven immunosuppressive transcription and cAMP-elevating signaling through A2A adenosine receptors (A2ARs) represent a major tumor-protecting pathway that enables immune evasion. Recent promising clinical outcomes due to the blockade of the adenosine-generating enzyme CD73 and A2AR in patients refractory to all other therapies have confirmed the importance of targeting hypoxia-adenosinergic signaling. We report a feasible approach to target the upstream stage of hypoxia-adenosinergic immunosuppression using an oxygen-carrying nanoemulsion (perfluorocarbon blood substitute). We show that oxygenation agent therapy (a) eliminates tumor hypoxia, (b) improves efficacy of endogenously developed and adoptively transferred T cells, and thereby (c) promotes regression of tumors in different anatomical locations. We show that both T cells and NK cells avoid hypoxic tumor areas and that reversal of hypoxia by oxygenation agent therapy increases intratumoral infiltration of activated T cells and NK cells due to reprogramming of the tumor microenvironment (TME). Thus, repurposing oxygenation agents in combination with supplemental oxygen may improve current cancer immunotherapies by preventing hypoxia-adenosinergic suppression, promoting immune cell infiltration and enhancing effector responses. These data also suggest that pretreating patients with oxygenation agent therapy may reprogram the TME from immunosuppressive to immune-permissive prior to adoptive cell therapy, or other forms of immunotherapy.
Collapse
Affiliation(s)
- Katarina Halpin-Veszeleiova
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Michael P. Mallouh
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
- Department of Surgery, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Lucy M. Williamson
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Ashley C. Apro
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Nuria R. Botticello-Romero
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Camille Bahr
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Maureen Shin
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Kelly M. Ward
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Laura Rosenberg
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Vladimir B. Ritov
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Michail V. Sitkovsky
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
| | - Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bruce D. Spiess
- Department of Anesthesiology, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Stephen M. Hatfield
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouve College of Health Sciences, Northeastern University, Boston, Massachusetts, USA
- Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Estephan H, Tailor A, Parker R, Kreamer M, Papandreou I, Campo L, Easton A, Moon EJ, Denko NC, Ternette N, Hammond EM, Giaccia AJ. Hypoxia promotes tumor immune evasion by suppressing MHC-I expression and antigen presentation. EMBO J 2025; 44:903-922. [PMID: 39753950 PMCID: PMC11790895 DOI: 10.1038/s44318-024-00319-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/29/2024] [Accepted: 10/14/2024] [Indexed: 02/05/2025] Open
Abstract
Hypoxia is a common feature of solid tumors that has previously been linked to resistance to radiotherapy and chemotherapy, and more recently to immunotherapy. In particular, hypoxic tumors exclude T cells and inhibit their activity, suggesting that tumor cells acquire a mechanism to evade T-cell recognition and killing. Our analysis of hypoxic tumors indicates that hypoxia downregulates the expression of MHC class I and its bound peptides (i.e., the immunopeptidome). Hypoxia decreases MHC-I expression in an oxygen-dependent manner, via activation of autophagy through the PERK arm of the unfolded protein response. Using an immunopeptidomics-based LC-MS approach, we find a significant reduction of presented antigens under hypoxia. Inhibition of autophagy under hypoxia enhances antigen presentation. In experimental tumors, reducing mitochondrial metabolism through a respiratory complex-I inhibitor increases tumor oxygenation, as well as MHC-I levels and the immunopeptidome. These data explain the molecular basis of tumor immune evasion in hypoxic conditions, and have implications for future therapeutic interventions targeting hypoxia-induced alterations in antigen presentation.
Collapse
Affiliation(s)
- Hala Estephan
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Arun Tailor
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, OX37BN, UK
| | - Robert Parker
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, OX37BN, UK
| | - McKenzie Kreamer
- Department of Radiation Oncology, OSU Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, USA
| | - Ioanna Papandreou
- Department of Radiation Oncology, OSU Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, USA
| | - Leticia Campo
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Alistair Easton
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Eui Jung Moon
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Nicholas C Denko
- Department of Radiation Oncology, OSU Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, USA
| | - Nicola Ternette
- Centre for Immuno-Oncology, Nuffield Department of Medicine, University of Oxford, Oxford, OX37BN, UK
| | - Ester M Hammond
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK
| | - Amato J Giaccia
- Department of Oncology, The University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
6
|
Song CW, Kim H, Kim MS, Park HJ, Paek SH, Terezakis S, Cho LC. Role of HIF-1α in the Responses of Tumors to Radiotherapy and Chemotherapy. Cancer Res Treat 2025; 57:1-10. [PMID: 38853541 PMCID: PMC11729307 DOI: 10.4143/crt.2024.255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
Tumor microenvironment is intrinsically hypoxic with abundant hypoxia-inducible factors-1α (HIF-1α), a primary regulator of the cellular response to hypoxia and various stresses imposed on the tumor cells. HIF-1α increases radioresistance and chemoresistance by reducing DNA damage, increasing repair of DNA damage, enhancing glycolysis that increases antioxidant capacity of tumors cells, and promoting angiogenesis. In addition, HIF-1α markedly enhances drug efflux, leading to multidrug resistance. Radiotherapy and certain chemotherapy drugs evoke profound anti-tumor immunity by inducing immunologic cell death that release tumor-associated antigens together with numerous pro-immunological factors, leading to priming of cytotoxic CD8+ T cells and enhancing the cytotoxicity of macrophages and natural killer cells. Radiotherapy and chemotherapy of tumors significantly increase HIF-1α activity in tumor cells. Unfortunately, HIF-1α effectively promotes various immune suppressive pathways including secretion of immune suppressive cytokines, activation of myeloid-derived suppressor cells, activation of regulatory T cells, inhibition of T cells priming and activity, and upregulation of immune checkpoints. Consequently, the anti-tumor immunity elevated by radiotherapy and chemotherapy is counterbalanced or masked by the potent immune suppression promoted by HIF-1α. Effective inhibition of HIF-1α may significantly increase the efficacy of radiotherapy and chemotherapy by increasing radiosensitivity and chemosensitivity of tumor cells and also by upregulating anti-tumor immunity.
Collapse
Affiliation(s)
- Chang W Song
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Hyunkyung Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Mi-Sook Kim
- Department of Radiation Oncology, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Heon J Park
- Department of Microbiology, College of Medicine, Inha University, Incheon, Korea
| | - Sun-Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul, Korea
| | - Stephanie Terezakis
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - L Chinsoo Cho
- Department of Radiation Oncology, University of Minnesota Medical School, Minneapolis, MN, USA
| |
Collapse
|
7
|
Ward MB, Jones AB, Krenciute G. Therapeutic advantage of combinatorial chimeric antigen receptor T cell and chemotherapies. Pharmacol Rev 2025; 77:100011. [PMID: 39952691 DOI: 10.1124/pharmrev.124.001070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/28/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapies have transformed outcomes for many patients with hematological malignancies. However, some patients do not respond to CAR T cell treatment, and adapting CAR T cells for treatment of solid and brain tumors has been met with many challenges, including a hostile tumor microenvironment and poor CAR T cell persistence. Thus, it is unlikely that CAR T cell therapy alone will be sufficient for consistent, complete tumor clearance across patients with cancer. Combinatorial therapies of CAR T cells and chemotherapeutics are a promising approach for overcoming this because chemotherapeutics could augment CAR T cells for improved antitumor activity or work in tandem with CAR T cells to clear tumors. Herein, we review efforts toward achieving successful CAR T cell and chemical drug combination therapies. We focus on combination therapies with approved chemotherapeutics because these will be more easily translated to the clinic but also review nonapproved chemotherapeutics and drug screens designed to reveal promising new CAR T cell and chemical drug combinations. Overall, this review highlights the promise of CAR T cell and chemotherapy combinations with a specific focus on how combinatorial therapy overcomes challenges faced by either monotherapy and supports the potential of this therapeutic strategy to improve outcomes for patients with cancer. SIGNIFICANCE STATEMENT: Improving currently available CAR T cell products via combinatorial therapy with chemotherapeutics has the potential to drastically expand the types of cancers and number of patients that could benefit from these therapies when neither alone has been sufficient to achieve tumor clearance. Herein, we provide a thorough review of the current efforts toward studying CAR T and chemotherapy combinatorial therapies and offer perspectives on optimal ways to identify new and effective combinations moving forward.
Collapse
Affiliation(s)
- Meghan B Ward
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Amber B Jones
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, Tennessee.
| |
Collapse
|
8
|
Geng S, Zhu L, Wang Y, Liu Q, Yu C, Shi S, Yu S. Co-Colorectal cancer stem cells employ the FADS1/DDA axis to evade NK cell-mediated immunosuppression after co-cultured with NK cells under hypoxia. Int Immunopharmacol 2024; 143:113535. [PMID: 39488917 DOI: 10.1016/j.intimp.2024.113535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 10/19/2024] [Accepted: 10/27/2024] [Indexed: 11/05/2024]
Abstract
Colorectal cancer (CRC) ranks as China's second most common cancer and fifth top cancer death cause. The study highlights the role of Natural Killer (NK) cells in targeting cancer stem cells (CSCs) that evade immune responses in CRC. Colorectal cancer stem cells (CCSCs) were stem from HT-29 cells and co-cultured with NK cells under normoxic or hypoxic conditions. The impact of this co-culture was evaluated using CCK8 assays for NK cell viability, ELISA for cytokine level changes, and flow cytometry for assessing NK cell apoptosis and activation. Comprehensive metabolomic and transcriptomic analyses were also performed to identify key genes and metabolites involved in the interaction between CCSCs and NK cells Co-culture of CCSCs with NK cells under hypoxia reduced NK cytotoxicity, increased NK apoptosis, and altered cytokine secretion by decreasing IFN-γ and TNF-α levels while increasing IL-6. Transcriptomic and metabolomic analysis identified 4 genes (FADS1, ALDH3A2, GCSH, MTCL1) and 3 metabolites (glyoxylic acid, spermine, DDA) as significant. Interfering with FADS1 counteracted the suppression of IFN-γ and TNF-α induced by CSC cells. Curiously, this inhibition caused by si-FADS1 could be neutralized by the addition of exogenous DDA. Co-culturing with NK cells notably increased spermine levels. Exogenous spermine resulted in a significant reduction in HT-29 cell death rates at 32 µM, 64 µM, and 128 µM, compared to NK cells without spermine. Our research explored CCSCs employed the FADS1/DDA axis to evade NK cell-mediated immunosuppression after co-cultured with NK cells under hypoxia.
Collapse
Affiliation(s)
- Shan Geng
- Central Laboratory of the People's Hospital of Dazu, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China
| | - Lei Zhu
- Department of General Surgery, The First People's Hospital of Kunming, 650034 Kunming, Yunnan Province, China
| | - Yanping Wang
- Central Laboratory of the People's Hospital of Dazu, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China
| | - Qiang Liu
- Department of General Surgery, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China
| | - Caiyu Yu
- Department of Hernia Surgery, Qujing No.1 Hospital, 655099 Qujing, Yunnan Province, China
| | - Shan Shi
- Office of Hospital, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China.
| | - Shaohong Yu
- Department of General Surgery, The Affiliated Dazu Hospital of Chongqing Medical University, 402360 Chongqing, China.
| |
Collapse
|
9
|
Buzaglo GBB, Telles GD, Araújo RB, Junior GDS, Ruberti OM, Ferreira MLV, Derchain SFM, Vechin FC, Conceição MS. The Therapeutic Potential of Physical Exercise in Cancer: The Role of Chemokines. Int J Mol Sci 2024; 25:13740. [PMID: 39769501 PMCID: PMC11678861 DOI: 10.3390/ijms252413740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 01/11/2025] Open
Abstract
The global increase in cancer cases and mortality has been associated with inflammatory processes, in which chemokines play crucial roles. These molecules, a subfamily of cytokines, are essential for the migration, adhesion, interaction, and positioning of immune cells throughout the body. Chemokines primarily originate in response to pathogenic stimuli and inflammatory cytokines. They are expressed by lymphocytes in the bloodstream and are divided into four classes (CC, CXC, XC, and CX3C), playing multifaceted roles in the tumor environment (TME). In the TME, chemokines regulate immune behavior by recruiting cells such as tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), which promote tumor survival. Additionally, they directly influence tumor behavior, promoting pathological angiogenesis, invasion, and metastasis. On the other hand, chemokines can also induce antitumor responses by mobilizing CD8+ T cells and natural killer (NK) cells to the tumor, reducing pro-inflammatory chemokines and enhancing essential antitumor responses. Given the complex interaction between chemokines, the immune system, angiogenic factors, and metastasis, it becomes evident how important it is to target these pathways in therapeutic interventions to counteract cancer progression. In this context, physical exercise emerges as a promising strategy due to its role modulating the expression of anti-inflammatory chemokines and enhancing the antitumor response. Aerobic and resistance exercises have been associated with a beneficial inflammatory profile in cancer, increased infiltration of CD8+ T cells in the TME, and improvement of intratumoral vasculature. This creates an environment less favorable to tumor growth and supports the circulation of antitumor immune cells and chemokines. Therefore, understanding the impact of exercise on the expression of chemokines can provide valuable insights for therapeutic interventions in cancer treatment and prevention.
Collapse
Affiliation(s)
- Glenda B. B. Buzaglo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Guilherme D. Telles
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Rafaela B. Araújo
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Gilmar D. S. Junior
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Olivia M. Ruberti
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Marina L. V. Ferreira
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| | - Sophie F. M. Derchain
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, University of Campinas, Campinas, Sao Paulo 13083-881, Brazil;
| | - Felipe C. Vechin
- School of Physical Education and Sport, University of Sao Paulo, Sao Paulo 05508-030, Brazil; (G.D.T.); (F.C.V.)
| | - Miguel S. Conceição
- Health Sciences Postgraduate Program, São Francisco University, Av. São Francisco de Assis, 218, Bragança Paulista, Sao Paulo 12916-900, Brazil; (G.B.B.B.); (R.B.A.); (G.D.S.J.); (O.M.R.); (M.L.V.F.)
| |
Collapse
|
10
|
Rachunek-Medved K, Krauß S, Daigeler A, Adams C, Eckert F, Ganser K, Gonzalez-Menendez I, Quintanilla-Martinez L, Kolbenschlag J. Acute remote ischemic conditioning enhances (CD3+)- but not (FoxP3+)-T-cell invasion in the tumor center and increases IL 17 and TNF-alpha expression in a murine melanoma model. Front Immunol 2024; 15:1501885. [PMID: 39650654 PMCID: PMC11621216 DOI: 10.3389/fimmu.2024.1501885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction Hypoxia can drive tumor progression, suppress anti-tumor immunity, and reduce the effectiveness of radiotherapy and chemotherapy. This study aimed to assess the impact of remote ischemic conditioning (RIC) on tumor oxygenation (sO2) and the anti-tumor immune response. Material and methods Fourteen B16-Ova tumor-bearing C57BL/6N mice received six 5-minute RIC cycles, while another fourteen underwent anesthesia only. Pimonidazole was administered 1.5 hours before sacrifice. Blood flow, sO2, and hemoglobin levels were measured in the non-ischemic hind limb and tumor. Tumor hypoxia was assessed using pimonidazole and CA IX immunohistochemistry, and T cell infiltration by CD3 and FoxP3 staining. Serum levels of 23 cytokines were analyzed using a multiplex immunoassay. Results Isoflurane anesthesia caused a slight intraindividual increase in blood flow (p = 0.07) and sO2 (p = 0.06) of the hind limb and a sole increase in tumor sO2 (p = 0.035), whereas RIC improved sO2 of the tumor in relation to the hind limb (p=0.03). The median of the tumor oxygen saturation reached 51.4% in the control group and 62.7% in the RIC group (p = 0.09), exhibiting a slight tendency towards better oxygenation in the RIC group. Pimonidazole (p=0.24) and CA IX hypoxia score (p=0.48) did not reveal statistically significant differences between the two groups. In RIC-treated tumors, the number of CD3 (p=0.006), but not FoxP3- positive cells (p = 0.84), in the tumor core was significantly higher compared to the control group. In the RIC group, the mean fluorescence intensity (MFI) of IL-17 was significantly higher (p=0.035), and TNF-α was trend-wise higher (p=0.063) compared to the control group. Conclusion Both isoflurane anesthesia and RIC have an impact on microcirculation. The application of RIC counteracted some of the effects of isoflurane, primarily in healthy tissue, and led to a significant improvement in relative tumor tissue oxygenation compared to the non-ischemic hind limb. RIC selectively enhanced immune infiltration within the tumor center, probably by previously activated tumor infiltrating T cells, while having no significant impact on T-regulatory cells. RIC appears to impact the cytokine profile, as indicated by elevated MFIs of TNF-α and IL-17.
Collapse
Affiliation(s)
- Katarzyna Rachunek-Medved
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Sabrina Krauß
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Adrien Daigeler
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| | - Constantin Adams
- Department of Paediatrics, University Hospital Tuebingen, Tuebingen, Germany
| | - Franziska Eckert
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Katrin Ganser
- Department of Radiation Oncology, University of Tuebingen, Tuebingen, Germany
| | - Irene Gonzalez-Menendez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany
| | - Leticia Quintanilla-Martinez
- Institute of Pathology and Neuropathology, Comprehensive Cancer Center, University Hospital Tuebingen, Eberhard Karls University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany
| | - Jonas Kolbenschlag
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center, Eberhard Karls University of Tuebingen, Tuebingen, Germany
| |
Collapse
|
11
|
Mastrogiovanni M, Donnadieu E, Pathak R, Di Bartolo V. Subverting Attachment to Prevent Attacking: Alteration of Effector Immune Cell Migration and Adhesion as a Key Mechanism of Tumor Immune Evasion. BIOLOGY 2024; 13:860. [PMID: 39596815 PMCID: PMC11591779 DOI: 10.3390/biology13110860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/29/2024]
Abstract
Cell adhesion regulates specific migratory patterns, location, communication with other cells, physical interactions with the extracellular matrix, and the establishment of effector programs. Proper immune control of cancer strongly depends on all these events occurring in a highly accurate spatiotemporal sequence. In response to cancer-associated inflammatory signals, effector immune cells navigating the bloodstream shift from their patrolling exploratory migration mode to establish adhesive interactions with vascular endothelial cells. This interaction enables them to extravasate through the blood vessel walls and access the cancer site. Further adhesive interactions within the tumor microenvironment (TME) are crucial for coordinating their distribution in situ and for mounting an effective anti-tumor immune response. In this review, we examine how alterations of adhesion cues in the tumor context favor tumor escape by affecting effector immune cell infiltration and trafficking within the TME. We discuss the mechanisms by which tumors directly modulate immune cell adhesion and migration patterns to affect anti-tumor immunity and favor tumor evasion. We also explore indirect immune escape mechanisms that involve modifications of TME characteristics, such as vascularization, immunogenicity, and structural topography. Finally, we highlight the significance of these aspects in designing more effective drug treatments and cellular immunotherapies.
Collapse
Affiliation(s)
- Marta Mastrogiovanni
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Gottesman Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Emmanuel Donnadieu
- Equipe Labellisée Ligue Contre le Cancer, CNRS, INSERM, Institut Cochin, Université Paris Cité, F-75014 Paris, France;
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Vincenzo Di Bartolo
- Immunoregulation Unit, Institut Pasteur, Université Paris Cité, F-75015 Paris, France;
| |
Collapse
|
12
|
Taghizadeh-Hesary F, Ghadyani M, Kashanchi F, Behnam B. Exploring TSGA10 Function: A Crosstalk or Controlling Mechanism in the Signaling Pathway of Carcinogenesis? Cancers (Basel) 2024; 16:3044. [PMID: 39272902 PMCID: PMC11393850 DOI: 10.3390/cancers16173044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/18/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Cancer-specific antigens have been a significant area of focus in cancer treatment since their discovery in the mid-twentieth century. Cancer germline antigens are a class of antigens specifically overexpressed in germline tissues and cancer cells. Among these, TSGA10 (testis-specific gene antigen 10) is of great interest because of its crucial impact on cancer progression. Early studies explored TSGA10 expression in a variety of cancer types. More recent studies revealed that TSGA10 can suppress tumor progression by blocking cancer cell metabolism, angiogenesis, and metastasis. An open question regarding the TSGA10 is why cancer cells must express a protein that prevents their progression. To answer this question, we conducted a comprehensive review to engage the TSGA10 in the context of the current understanding of "malignant transformation". This review demonstrated that TSGA10 expression level in cancer cells depends on the cancer stage across malignant transformation. In addition, we evaluated how TSGA10 expression can prevent the "cancer hallmarks". Given this information, TSGA10 can be of great interest in developing effective targeted anti-cancer therapies.
Collapse
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Mobina Ghadyani
- Chester Medical School, University of Chester, Chester CH2 1BR, UK
| | - Fatah Kashanchi
- Laboratory of Molecular Virology, George Mason University, Manassas, VA 20110, USA
| | - Babak Behnam
- Avicenna Biotech Research, Germantown, MD 20871, USA
| |
Collapse
|
13
|
Mpekris F, Panagi M, Charalambous A, Voutouri C, Stylianopoulos T. Modulating cancer mechanopathology to restore vascular function and enhance immunotherapy. Cell Rep Med 2024; 5:101626. [PMID: 38944037 PMCID: PMC11293360 DOI: 10.1016/j.xcrm.2024.101626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/12/2024] [Accepted: 06/07/2024] [Indexed: 07/01/2024]
Abstract
Solid tumor pathology, characterized by abnormalities in the tumor microenvironment (TME), challenges therapeutic effectiveness. Mechanical factors, including increased tumor stiffness and accumulation of intratumoral forces, can determine the success of cancer treatments, defining the tumor's "mechanopathology" profile. These abnormalities cause extensive vascular compression, leading to hypoperfusion and hypoxia. Hypoperfusion hinders drug delivery, while hypoxia creates an unfavorable TME, promoting tumor progression through immunosuppression, heightened metastatic potential, drug resistance, and chaotic angiogenesis. Strategies targeting TME mechanopathology, such as vascular and stroma normalization, hold promise in enhancing cancer therapies with some already advancing to the clinic. Normalization can be achieved using anti-angiogenic agents, mechanotherapeutics, immune checkpoint inhibitors, engineered bacterial therapeutics, metronomic nanomedicine, and ultrasound sonopermeation. Here, we review the methods developed to rectify tumor mechanopathology, which have even led to cures in preclinical models, and discuss their bench-to-bedside translation, including the derivation of biomarkers from tumor mechanopathology for personalized therapy.
Collapse
Affiliation(s)
- Fotios Mpekris
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Myrofora Panagi
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Antonia Charalambous
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
14
|
Uher O, Hadrava Vanova K, Taïeb D, Calsina B, Robledo M, Clifton-Bligh R, Pacak K. The Immune Landscape of Pheochromocytoma and Paraganglioma: Current Advances and Perspectives. Endocr Rev 2024; 45:521-552. [PMID: 38377172 PMCID: PMC11244254 DOI: 10.1210/endrev/bnae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/19/2023] [Accepted: 02/02/2024] [Indexed: 02/22/2024]
Abstract
Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors derived from neural crest cells from adrenal medullary chromaffin tissues and extra-adrenal paraganglia, respectively. Although the current treatment for PPGLs is surgery, optimal treatment options for advanced and metastatic cases have been limited. Hence, understanding the role of the immune system in PPGL tumorigenesis can provide essential knowledge for the development of better therapeutic and tumor management strategies, especially for those with advanced and metastatic PPGLs. The first part of this review outlines the fundamental principles of the immune system and tumor microenvironment, and their role in cancer immunoediting, particularly emphasizing PPGLs. We focus on how the unique pathophysiology of PPGLs, such as their high molecular, biochemical, and imaging heterogeneity and production of several oncometabolites, creates a tumor-specific microenvironment and immunologically "cold" tumors. Thereafter, we discuss recently published studies related to the reclustering of PPGLs based on their immune signature. The second part of this review discusses future perspectives in PPGL management, including immunodiagnostic and promising immunotherapeutic approaches for converting "cold" tumors into immunologically active or "hot" tumors known for their better immunotherapy response and patient outcomes. Special emphasis is placed on potent immune-related imaging strategies and immune signatures that could be used for the reclassification, prognostication, and management of these tumors to improve patient care and prognosis. Furthermore, we introduce currently available immunotherapies and their possible combinations with other available therapies as an emerging treatment for PPGLs that targets hostile tumor environments.
Collapse
Affiliation(s)
- Ondrej Uher
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - Katerina Hadrava Vanova
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| | - David Taïeb
- Department of Nuclear Medicine, CHU de La Timone, Marseille 13005, France
| | - Bruna Calsina
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Familiar Cancer Clinical Unit, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Mercedes Robledo
- Hereditary Endocrine Cancer Group, Human Cancer Genetics Program, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Institute of Health Carlos III (ISCIII), Madrid 28029, Spain
| | - Roderick Clifton-Bligh
- Department of Endocrinology, Royal North Shore Hospital, Sydney 2065, NSW, Australia
- Cancer Genetics Laboratory, Kolling Institute, University of Sydney, Sydney 2065, NSW, Australia
| | - Karel Pacak
- Section of Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1109, USA
| |
Collapse
|
15
|
De Velasco MA, Kura Y, Fujita K, Uemura H. Moving toward improved immune checkpoint immunotherapy for advanced prostate cancer. Int J Urol 2024; 31:307-324. [PMID: 38167824 DOI: 10.1111/iju.15378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 12/10/2023] [Indexed: 01/05/2024]
Abstract
Human prostate cancer is a heterogenous malignancy that responds poorly to immunotherapy targeting immune checkpoints. The immunosuppressive tumor microenvironment that is typical of human prostate cancer has been the main obstacle to these treatments. The effectiveness of these therapies is also hindered by acquired resistance, leading to slow progress in prostate cancer immunotherapy. Results from the highly anticipated late-stage clinical trials of PD-1/PD-L1 immune checkpoint blockade in patients with advanced prostate cancer have highlighted some of the obstacles to immunotherapy. Despite the setbacks, there is much that has been learned about the mechanisms that drive resistance, and new strategies are being developed and tested. Here, we review the status of immune checkpoint blockade and the immunosuppressive tumor microenvironment and discuss factors contributing to innate and adaptive resistance to immune checkpoint blockade within the context of prostate cancer. We then examine current strategies aiming to overcome these challenges as well as prospects.
Collapse
Affiliation(s)
- Marco A De Velasco
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Yurie Kura
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Kazutoshi Fujita
- Department of Urology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osakasayama, Japan
| |
Collapse
|
16
|
Fan P, Zhang N, Candi E, Agostini M, Piacentini M, Shi Y, Huang Y, Melino G. Alleviating hypoxia to improve cancer immunotherapy. Oncogene 2023; 42:3591-3604. [PMID: 37884747 DOI: 10.1038/s41388-023-02869-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/07/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023]
Abstract
Tumor hypoxia resulting from abnormal and dysfunctional tumor vascular network poses a substantial obstacle to immunotherapy. In fact, hypoxia creates an immunosuppressive tumor microenvironment (TME) through promoting angiogenesis, metabolic reprogramming, extracellular matrix remodeling, epithelial-mesenchymal transition (EMT), p53 inactivation, and immune evasion. Vascular normalization, a strategy aimed at restoring the structure and function of tumor blood vessels, has been shown to improve oxygen delivery and reverse hypoxia-induced signaling pathways, thus alleviates hypoxia and potentiates cancer immunotherapy. In this review, we discuss the mechanisms of tumor tissue hypoxia and its impacts on immune cells and cancer immunotherapy, as well as the approaches to induce tumor vascular normalization. We also summarize the evidence supporting the use of vascular normalization in combination with cancer immunotherapy, and highlight the challenges and future directions of this overlooked important field. By targeting the fundamental problem of tumor hypoxia, vascular normalization proposes a promising strategy to enhance the efficacy of cancer immunotherapy and improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Peng Fan
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
- National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 215123, Suzhou, China
| | - Naidong Zhang
- National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 215123, Suzhou, China
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Mauro Piacentini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, 215123, Suzhou, China.
| | - Yuhui Huang
- National Clinical Research Center for Hematologic Diseases, Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, 215123, Suzhou, China.
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
17
|
Wada A, Hirohashi Y, Kutomi G, Murata K, Iwabuchi S, Mizue Y, Murai A, Kyuno D, Shima H, Minowa T, Sasaki K, Kubo T, Kanaseki T, Tsukahara T, Nakatsugawa M, Hashimoto S, Osanai M, Torigoe T, Takemasa I. Eribulin is an immune potentiator in breast cancer that upregulates human leukocyte antigen class I expression via the induction of NOD-like receptor family CARD domain-containing 5. Cancer Sci 2023; 114:4511-4520. [PMID: 37991442 PMCID: PMC10728009 DOI: 10.1111/cas.15986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/10/2023] [Accepted: 09/18/2023] [Indexed: 11/23/2023] Open
Abstract
Eribulin inhibits microtubule polymerization and improves the overall survival of patients with recurrent metastatic breast cancer. A subgroup analysis revealed a low neutrophil to lymphocyte ratio (NLR) (<3) to be a prognostic factor of eribulin treatment. We thus hypothesized that eribulin might be related to the immune response for breast cancer cells and we analyzed the effects of eribulin on the immune system. Immunohistochemical staining revealed that human leukocyte antigen (HLA) class I expression was increased in clinical samples after eribulin treatment. In vitro assays revealed that eribulin treatment increased HLA class I expression in breast cancer line cells. RNA-sequencing demonstrated that eribulin treatment increased the expression of the NOD-like family CARD domain-containing 5 (NLRC5), a master regulator of HLA class I expression. Eribulin treatment increased the NY-ESO-1-specific T-cell receptor (TCR) transduced T (TCR-T) cell response for New York oesophageal squamous cell carcinoma 1 (NY-ESO-1) overexpressed breast cancer cells. The eribulin and TCR-T combined therapy model revealed that eribulin and immunotherapy using TCR-T cells has a synergistic effect. In summary, eribulin increases the expression of HLA class 1 via HLA class 1 transactivatior NLRC5 and eribulin combination with immunotherapy can be effective for the treatment of breast cancer.
Collapse
Affiliation(s)
- Asaka Wada
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Yoshihiko Hirohashi
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Goro Kutomi
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Kenji Murata
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Sadahiro Iwabuchi
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Yuka Mizue
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Aiko Murai
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Daisuke Kyuno
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Hiroaki Shima
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| | - Tomoyuki Minowa
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Kenta Sasaki
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Terufumi Kubo
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Takayuki Kanaseki
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Tomohide Tsukahara
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Munehide Nakatsugawa
- Department of PathologyTokyo Medical University Hachioji Medical CenterTokyoJapan
| | - Shinichi Hashimoto
- Department of Molecular PathophysiologyInstitute of Advanced Medicine, Wakayama Medical UniversityWakayamaJapan
| | - Makoto Osanai
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Toshihiko Torigoe
- Department of PathologySapporo Medical University School of MedicineSapporoJapan
| | - Ichiro Takemasa
- Department of SurgerySapporo Medical University School of MedicineSapporoJapan
| |
Collapse
|
18
|
Malviya M, Aretz Z, Molvi Z, Lee J, Pierre S, Wallisch P, Dao T, Scheinberg DA. Challenges and solutions for therapeutic TCR-based agents. Immunol Rev 2023; 320:58-82. [PMID: 37455333 PMCID: PMC11141734 DOI: 10.1111/imr.13233] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 06/18/2023] [Indexed: 07/18/2023]
Abstract
Recent development of methods to discover and engineer therapeutic T-cell receptors (TCRs) or antibody mimics of TCRs, and to understand their immunology and pharmacology, lag two decades behind therapeutic antibodies. Yet we have every expectation that TCR-based agents will be similarly important contributors to the treatment of a variety of medical conditions, especially cancers. TCR engineered cells, soluble TCRs and their derivatives, TCR-mimic antibodies, and TCR-based CAR T cells promise the possibility of highly specific drugs that can expand the scope of immunologic agents to recognize intracellular targets, including mutated proteins and undruggable transcription factors, not accessible by traditional antibodies. Hurdles exist regarding discovery, specificity, pharmacokinetics, and best modality of use that will need to be overcome before the full potential of TCR-based agents is achieved. HLA restriction may limit each agent to patient subpopulations and off-target reactivities remain important barriers to widespread development and use of these new agents. In this review we discuss the unique opportunities for these new classes of drugs, describe their unique antigenic targets, compare them to traditional antibody therapeutics and CAR T cells, and review the various obstacles that must be overcome before full application of these drugs can be realized.
Collapse
Affiliation(s)
- Manish Malviya
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Zita Aretz
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Physiology, Biophysics & Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| | - Zaki Molvi
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Physiology, Biophysics & Systems Biology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| | - Jayop Lee
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - Stephanie Pierre
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Tri-Institutional Medical Scientist Program, 1300 York Avenue, New York, NY 10021
| | - Patrick Wallisch
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| | - Tao Dao
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065
- Pharmacology Program, Weill Cornell Graduate School of Medical Sciences, 1300 York Avenue, New York, NY 10021
| |
Collapse
|
19
|
Galassi C, Klapp V, Formenti SC, Demaria S, Galluzzi L. Immunologically relevant effects of radiation therapy on the tumor microenvironment. Essays Biochem 2023; 67:979-989. [PMID: 37199227 PMCID: PMC10543618 DOI: 10.1042/ebc20220248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023]
Abstract
Focal radiation therapy (RT) has been successfully employed to clinically manage multiple types of cancer for more than a century. Besides being preferentially cytotoxic for malignant cells over their nontransformed counterparts, RT elicits numerous microenvironmental alterations that appear to factor into its therapeutic efficacy. Here, we briefly discuss immunostimulatory and immunosuppressive microenvironmental changes elicited by RT and their impact on tumor recognition by the host immune system.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Vanessa Klapp
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Silvia C. Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| |
Collapse
|
20
|
Bandopadhyay S, Patranabis S. Mechanisms of HIF-driven immunosuppression in tumour microenvironment. J Egypt Natl Canc Inst 2023; 35:27. [PMID: 37646847 DOI: 10.1186/s43046-023-00186-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/12/2023] [Indexed: 09/01/2023] Open
Abstract
Hypoxia arises due to insufficient oxygen delivery to rapidly proliferating tumour cells that outpace the available blood supply. It is a characteristic feature of most solid tumour microenvironments and plays a critical role in regulating anti-tumour immunity, enhancing tumoral heterogeneity, and promoting therapeutic resistance and poor clinical outcomes. Hypoxia-inducible factors (HIFs) are the major hypoxia-responsive transcription factors that are activated under low oxygenation conditions and have been identified to drive multifunctional roles in tumour immune evasion. The HIF signalling network serves as an attractive target for targeted therapeutic approaches. This review aims to provide a comprehensive overview of the most crucial mechanisms by which HIF controls the expression of immunosuppressive molecules and immune checkpoints, disrupts cancer immunogenicity, and induces immunotherapeutic resistance.
Collapse
Affiliation(s)
| | - Somi Patranabis
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, India.
| |
Collapse
|
21
|
Joseph JP, Gugulothu SB, Nandi D, Chatterjee K. Mechanical Properties Affect Primary T Cell Activation in 3D Bioprinted Hydrogels. ACS Macro Lett 2023; 12:1085-1093. [PMID: 37466277 DOI: 10.1021/acsmacrolett.3c00271] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
T cells play a critical role in the adaptive immune response of the body, especially against intracellular pathogens and cancer. In vitro, T cell activation studies typically employ planar (two-dimensional, 2D) culture systems that do not mimic native cell-to-extracellular matrix (ECM) interactions, which influence activation. The goal of this work was to study T cell responses in a cell line (EL4) and primary mouse T cells in three-dimensional (3D) bioprinted matrices of varied stiffness. Cell-laden hydrogels were 3D bioprinted from gelatin methacryloyl (GelMA) using a digital light processing (DLP)-based 3D bioprinter operated with visible light (405 nm). Mechanical characterization revealed that the hydrogels had pathophysiologically relevant stiffnesses for a lymph node-mimetic tissue construct. EL4, a mouse T cell lymphoma line, or primary mouse T cells were 3D bioprinted and activated using a combination of 10 ng/mL of phorbol myristate acetate (PMA) and 0.1 μM of ionomycin. Cellular responses revealed differences between 2D and 3D cultures and that the biomechanical properties of the 3D bioprinted hydrogel influence T cell activation. Cellular responses of the 2D and 3D cultures in a soft matrix (19.83 ± 2.36 kPa) were comparable; however, they differed in a stiff matrix (52.95 ± 1.36 kPa). The fraction of viable EL4 cells was 1.3-fold higher in the soft matrix than in the stiff matrix. Furthermore, primary mouse T cells activated with PMA and ionomycin showed 1.35-fold higher viable cells in the soft matrix than in the stiff matrix. T cells bioprinted in a soft matrix and a stiff matrix released 7.4-fold and 5.9-fold higher amounts of interleukin-2 (IL-2) than 2D cultured cells, respectively. Overall, the study demonstrates the changes in the response of T cells in 3D bioprinted scaffolds toward engineering an ex vivo lymphoid tissue-mimetic system that can faithfully recapitulate T cell activation and unravel pathophysiological characteristics of T cells in infectious biology, autoimmunity, and cancers.
Collapse
Affiliation(s)
- Joel P Joseph
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru - 560012, India
| | | | - Dipankar Nandi
- Department of Biochemistry, Indian Institute of Science, Bengaluru - 560012, India
| | - Kaushik Chatterjee
- Centre for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru - 560012, India
- Department of Materials Engineering, Indian Institute of Science, Bengaluru - 560012, India
| |
Collapse
|
22
|
Sari G, Rock KL. Tumor immune evasion through loss of MHC class-I antigen presentation. Curr Opin Immunol 2023; 83:102329. [PMID: 37130455 PMCID: PMC10524158 DOI: 10.1016/j.coi.2023.102329] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 05/04/2023]
Abstract
CD8 T cells recognize cancers when they detect antigenic peptides presented on a tumor's surface MHC-I molecules. Since MHC-I antigen presentation is not essential for cell growth or survival, many cancers inactivate this pathway, and thereby escape control by CD8 T cells. Such immune evasion allows cancers to progress and also become resistant to CD8 T- cell-based immunotherapies, such as checkpoint blockade. Here, we review recent findings about the various different mechanisms that cancers use to impair antigen presentation, the consequence of such changes, and, in some cases, the potential to reverse these defects.
Collapse
Affiliation(s)
- Gulce Sari
- University of Massachusetts Medical School, Department of Pathology, Worcester, MA, USA
| | - Kenneth L Rock
- University of Massachusetts Medical School, Department of Pathology, Worcester, MA, USA.
| |
Collapse
|
23
|
Wang SF, Tseng LM, Lee HC. Role of mitochondrial alterations in human cancer progression and cancer immunity. J Biomed Sci 2023; 30:61. [PMID: 37525297 PMCID: PMC10392014 DOI: 10.1186/s12929-023-00956-w] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/11/2023] [Indexed: 08/02/2023] Open
Abstract
Dysregulating cellular metabolism is one of the emerging cancer hallmarks. Mitochondria are essential organelles responsible for numerous physiologic processes, such as energy production, cellular metabolism, apoptosis, and calcium and redox homeostasis. Although the "Warburg effect," in which cancer cells prefer aerobic glycolysis even under normal oxygen circumstances, was proposed a century ago, how mitochondrial dysfunction contributes to cancer progression is still unclear. This review discusses recent progress in the alterations of mitochondrial DNA (mtDNA) and mitochondrial dynamics in cancer malignant progression. Moreover, we integrate the possible regulatory mechanism of mitochondrial dysfunction-mediated mitochondrial retrograde signaling pathways, including mitochondrion-derived molecules (reactive oxygen species, calcium, oncometabolites, and mtDNA) and mitochondrial stress response pathways (mitochondrial unfolded protein response and integrated stress response) in cancer progression and provide the possible therapeutic targets. Furthermore, we discuss recent findings on the role of mitochondria in the immune regulatory function of immune cells and reveal the impact of the tumor microenvironment and metabolism remodeling on cancer immunity. Targeting the mitochondria and metabolism might improve cancer immunotherapy. These findings suggest that targeting mitochondrial retrograde signaling in cancer malignancy and modulating metabolism and mitochondria in cancer immunity might be promising treatment strategies for cancer patients and provide precise and personalized medicine against cancer.
Collapse
Affiliation(s)
- Sheng-Fan Wang
- Department of Pharmacy, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist., Taipei, 112, Taiwan
- School of Pharmacy, Taipei Medical University, No. 250, Wuxing St., Xinyi Dist., Taipei, 110, Taiwan
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei, 112, Taiwan
| | - Ling-Ming Tseng
- Division of General Surgery, Department of Surgery, Comprehensive Breast Health Center, Taipei Veterans General Hospital, No. 201, Sec. 2, Shipai Rd., Beitou Dist., Taipei, 112, Taiwan
- Department of Surgery, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei, 112, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, College of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei, 112, Taiwan.
- Department of Pharmacy, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei, 112, Taiwan.
| |
Collapse
|
24
|
Benoit A, Vogin G, Duhem C, Berchem G, Janji B. Lighting Up the Fire in the Microenvironment of Cold Tumors: A Major Challenge to Improve Cancer Immunotherapy. Cells 2023; 12:1787. [PMID: 37443821 PMCID: PMC10341162 DOI: 10.3390/cells12131787] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Immunotherapy includes immune checkpoint inhibitors (ICI) such as antibodies targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) or the programmed cell death protein/programmed death ligand 1 (PD-1/PD-L1) axis. Experimental and clinical evidence show that immunotherapy based on immune checkpoint inhibitors (ICI) provides long-term survival benefits to cancer patients in whom other conventional therapies have failed. However, only a minority of patients show high clinical benefits via the use of ICI alone. One of the major factors limiting the clinical benefits to ICI can be attributed to the lack of immune cell infiltration within the tumor microenvironment. Such tumors are classified as "cold/warm" or an immune "desert"; those displaying significant infiltration are considered "hot" or inflamed. This review will provide a brief summary of different tumor properties contributing to the establishment of cold tumors and describe major strategies that could reprogram non-inflamed cold tumors into inflamed hot tumors. More particularly, we will describe how targeting hypoxia can induce metabolic reprogramming that results in improving and extending the benefit of ICI.
Collapse
Affiliation(s)
- Alice Benoit
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg; (A.B.); (G.B.)
| | - Guillaume Vogin
- Centre National de Radiothérapie François Baclesse, L-4005 Esch-sur-Alzette, Luxembourg;
- Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Université de Lorraine—UMR 7365, 54505 Vandoeuvre-lès-Nancy, France
| | - Caroline Duhem
- Department of Hemato-Oncology, Centre Hospitalier du Luxembourg, L-1210 Luxembourg, Luxembourg;
| | - Guy Berchem
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg; (A.B.); (G.B.)
- Department of Hemato-Oncology, Centre Hospitalier du Luxembourg, L-1210 Luxembourg, Luxembourg;
- Faculty of Science, Technology and Medicine, University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Bassam Janji
- Tumor Immunotherapy and Microenvironment (TIME) Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg; (A.B.); (G.B.)
| |
Collapse
|
25
|
Purushothaman AK, Nelson EJR. Role of innate immunity and systemic inflammation in cystic fibrosis disease progression. Heliyon 2023; 9:e17553. [PMID: 37449112 PMCID: PMC10336457 DOI: 10.1016/j.heliyon.2023.e17553] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023] Open
Abstract
Pathophysiological manifestations of cystic fibrosis (CF) result from a functional defect in the cystic fibrosis transmembrane conductance regulator (CFTR) paving way for mucus obstruction and pathogen colonization. The role of CFTR in modulating immune cell function and vascular integrity, irrespective of mucus thickening, in determining the host cell response to pathogens/allergens and causing systemic inflammation is least appreciated. Since CFTR plays a key role in the conductance of anions like Cl-, loss of CFTR function could affect various basic cellular processes, such as cellular homeostasis, lysosome acidification, and redox balance. CFTR aids in endotoxin tolerance by regulating Toll-like receptor-mediated signaling resulting in uncontrolled activation of innate immune cells. Although leukocytes of CF patients are hyperactivated, they exhibit compromised phagosome activity thus favouring the orchestration of sepsis from defective pathogen clearance. This review will emphasize the importance of innate immunity and systemic inflammatory response in the development of CF and other CFTR-associated pathologies.
Collapse
Affiliation(s)
- Anand Kumar Purushothaman
- Gene Therapy Laboratory, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Everette Jacob Remington Nelson
- Gene Therapy Laboratory, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
26
|
Galluzzi L, Aryankalayil MJ, Coleman CN, Formenti SC. Emerging evidence for adapting radiotherapy to immunotherapy. Nat Rev Clin Oncol 2023:10.1038/s41571-023-00782-x. [PMID: 37280366 DOI: 10.1038/s41571-023-00782-x] [Citation(s) in RCA: 134] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2023] [Indexed: 06/08/2023]
Abstract
Immunotherapy has revolutionized the clinical management of many malignancies but is infrequently associated with durable objective responses when used as a standalone treatment approach, calling for the development of combinatorial regimens with superior efficacy and acceptable toxicity. Radiotherapy, the most commonly used oncological treatment, has attracted considerable attention as a combination partner for immunotherapy owing to its well-known and predictable safety profile, widespread clinical availability, and potential for immunostimulatory effects. However, numerous randomized clinical trials investigating radiotherapy-immunotherapy combinations have failed to demonstrate a therapeutic benefit compared with either modality alone. Such a lack of interaction might reflect suboptimal study design, choice of end points and/or administration of radiotherapy according to standard schedules and target volumes. Indeed, radiotherapy has empirically evolved towards radiation doses and fields that enable maximal cancer cell killing with manageable toxicity to healthy tissues, without much consideration of potential radiation-induced immunostimulatory effects. Herein, we propose the concept that successful radiotherapy-immunotherapy combinations might require modifications of standard radiotherapy regimens and target volumes to optimally sustain immune fitness and enhance the antitumour immune response in support of meaningful clinical benefits.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Molykutty J Aryankalayil
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - C Norman Coleman
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
27
|
Bai R, Cui J. Mitochondrial immune regulation and anti-tumor immunotherapy strategies targeting mitochondria. Cancer Lett 2023; 564:216223. [PMID: 37172686 DOI: 10.1016/j.canlet.2023.216223] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/25/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Cancer cells adapt to increasing energy and biosynthetic demands by reprogramming their metabolic pathways. Mitochondria are important organelles for the metabolic reprogramming of tumor cells. In addition to supplying energy, they play crucial roles in the survival, immune evasion, tumor progression, and treatment resistance of the hypoxic tumor microenvironment (TME) in cancer cells. With the development of the life sciences, scientists have gained an in-depth understanding of immunity, metabolism, and cancer, and numerous studies have emphasized that mitochondria are essential for tumor immune escape and the regulation of immune cell metabolism and activation. Moreover, recent evidence suggests that targeting the mitochondria-related pathway with anticancer drugs can initiate the killing of cancer cells by increasing the ability of cancer cells to be recognized by immune cells, tumor antigen presentation ability, and the anti-tumor function of immune cells. This review discusses the effects of mitochondrial morphology and function on the phenotype and function of immune cells under normal and TME conditions, the effects of mitochondrial changes in tumors and microenvironments on tumor immune escape and immune cell function, and finally focuses on the recent research progress and future challenges of novel anti-tumor immunotherapy strategies targeting mitochondria.
Collapse
Affiliation(s)
- Rilan Bai
- Cancer Center, the First Hospital of Jilin University, Changchun, 130021, China
| | - Jiuwei Cui
- Cancer Center, the First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
28
|
Finisguerra V, Dvorakova T, Formenti M, Van Meerbeeck P, Mignion L, Gallez B, Van den Eynde BJ. Metformin improves cancer immunotherapy by directly rescuing tumor-infiltrating CD8 T lymphocytes from hypoxia-induced immunosuppression. J Immunother Cancer 2023; 11:jitc-2022-005719. [PMID: 37147018 PMCID: PMC10163559 DOI: 10.1136/jitc-2022-005719] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2023] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND Despite their revolutionary success in cancer treatment over the last decades, immunotherapies encounter limitations in certain tumor types and patients. The efficacy of immunotherapies depends on tumor antigen-specific CD8 T-cell viability and functionality within the immunosuppressive tumor microenvironment, where oxygen levels are often low. Hypoxia can reduce CD8 T-cell fitness in several ways and CD8 T cells are mostly excluded from hypoxic tumor regions. Given the challenges to achieve durable reduction of hypoxia in the clinic, ameliorating CD8 T-cell survival and effector function in hypoxic condition could improve tumor response to immunotherapies. METHODS Activated CD8 T cells were exposed to hypoxia and metformin and analyzed by fluorescence-activated cell sorting for cell proliferation, apoptosis and phenotype. In vivo, metformin was administered to mice bearing hypoxic tumors and receiving either adoptive cell therapy with tumor-specific CD8 T cells, or immune checkpoint inhibitors; tumor growth was followed over time and CD8 T-cell infiltration, survival and localization in normoxic or hypoxic tumor regions were assessed by flow cytometry and immunofluorescence. Tumor oxygenation and hypoxia were measured by electron paramagnetic resonance and pimonidazole staining, respectively. RESULTS We found that the antidiabetic drug metformin directly improved CD8 T-cell fitness in hypoxia, both in vitro and in vivo. Metformin rescued murine and human CD8 T cells from hypoxia-induced apoptosis and increased their proliferation and cytokine production, while blunting the upregulation of programmed cell death protein 1 and lymphocyte-activation gene 3. This appeared to result from a reduced production of reactive oxygen species, due to the inhibition of mitochondrial complex I. Differently from what others reported, metformin did not reduce tumor hypoxia, but rather increased CD8 T-cell infiltration and survival in hypoxic tumor areas, and synergized with cyclophosphamide to enhance tumor response to adoptive cell therapy or immune checkpoint blockade in different tumor models. CONCLUSIONS This study describes a novel mechanism of action of metformin and presents a promising strategy to achieve immune rejection in hypoxic and immunosuppressive tumors, which would otherwise be resistant to immunotherapy.
Collapse
Affiliation(s)
- Veronica Finisguerra
- de Duve Institute, UCLouvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, de Duve Institute, Brussels, Belgium
- Walloon Excellence in Life Science and Biotechnology (WELBIO), WEL Research Institute, Brussels, Belgium
| | - Tereza Dvorakova
- de Duve Institute, UCLouvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, de Duve Institute, Brussels, Belgium
- Walloon Excellence in Life Science and Biotechnology (WELBIO), WEL Research Institute, Brussels, Belgium
| | - Matteo Formenti
- de Duve Institute, UCLouvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, de Duve Institute, Brussels, Belgium
- Walloon Excellence in Life Science and Biotechnology (WELBIO), WEL Research Institute, Brussels, Belgium
| | | | - Lionel Mignion
- Biomedical Magnetic Resonance (REMA) Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance (REMA) Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Benoit J Van den Eynde
- de Duve Institute, UCLouvain, Brussels, Belgium
- Ludwig Institute for Cancer Research, de Duve Institute, Brussels, Belgium
- Walloon Excellence in Life Science and Biotechnology (WELBIO), WEL Research Institute, Brussels, Belgium
- Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, UK
| |
Collapse
|
29
|
Bi J, Witt E, Voltarelli VA, Feig VR, Venkatachalam V, Boyce H, McGovern M, Gutierrez WR, Rytlewski JD, Bowman KR, Rhodes AC, Cook AN, Muller BN, Smith MG, Ramos AR, Panchal H, Dodd RD, Henry MD, Mailloux A, Traverso G, Otterbein LE, Byrne JD. Low-Cost, High-Pressure-Synthesized Oxygen-Entrapping Materials to Improve Treatment of Solid Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205995. [PMID: 36727291 PMCID: PMC10074083 DOI: 10.1002/advs.202205995] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/30/2022] [Indexed: 05/10/2023]
Abstract
Tumor hypoxia drives resistance to many cancer therapies, including radiotherapy and chemotherapy. Methods that increase tumor oxygen pressures, such as hyperbaric oxygen therapy and microbubble infusion, are utilized to improve the responses to current standard-of-care therapies. However, key obstacles remain, in particular delivery of oxygen at the appropriate dose and with optimal pharmacokinetics. Toward overcoming these hurdles, gas-entrapping materials (GeMs) that are capable of tunable oxygen release are formulated. It is shown that injection or implantation of these materials into tumors can mitigate tumor hypoxia by delivering oxygen locally and that these GeMs enhance responsiveness to radiation and chemotherapy in multiple tumor types. This paper also demonstrates, by comparing an oxygen (O2 )-GeM to a sham GeM, that the former generates an antitumorigenic and immunogenic tumor microenvironment in malignant peripheral nerve sheath tumors. Collectively the results indicate that the use of O2 -GeMs is promising as an adjunctive strategy for the treatment of solid tumors.
Collapse
|
30
|
Zhuang Y, Liu K, He Q, Gu X, Jiang C, Wu J. Hypoxia signaling in cancer: Implications for therapeutic interventions. MedComm (Beijing) 2023; 4:e203. [PMID: 36703877 PMCID: PMC9870816 DOI: 10.1002/mco2.203] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/14/2022] [Accepted: 12/18/2022] [Indexed: 01/25/2023] Open
Abstract
Hypoxia is a persistent physiological feature of many different solid tumors and a key driver of malignancy, and in recent years, it has been recognized as an important target for cancer therapy. Hypoxia occurs in the majority of solid tumors due to a poor vascular oxygen supply that is not sufficient to meet the needs of rapidly proliferating cancer cells. A hypoxic tumor microenvironment (TME) can reduce the effectiveness of other tumor therapies, such as radiotherapy, chemotherapy, and immunotherapy. In this review, we discuss the critical role of hypoxia in tumor development, including tumor metabolism, tumor immunity, and tumor angiogenesis. The treatment methods for hypoxic TME are summarized, including hypoxia-targeted therapy and improving oxygenation by alleviating tumor hypoxia itself. Hyperoxia therapy can be used to improve tissue oxygen partial pressure and relieve tumor hypoxia. We focus on the underlying mechanisms of hyperoxia and their impact on current cancer therapies and discuss the prospects of hyperoxia therapy in cancer treatment.
Collapse
Affiliation(s)
- Yan Zhuang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Kua Liu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Qinyu He
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
| | - Xiaosong Gu
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing UniversityJiangsu Key Laboratory of Molecular MedicineMedicineMedical School of Nanjing UniversityNanjing UniversityNanjingChina
- Microecological, Regenerative and Microfabrication Technical Platform for Biomedicine and Tissue EngineeringJinan Microecological Biomedicine Shandong LaboratoryJinan CityChina
| |
Collapse
|
31
|
Abou Khouzam R, Sharda M, Rao SP, Kyerewah-Kersi SM, Zeinelabdin NA, Mahmood AS, Nawafleh H, Khan MS, Venkatesh GH, Chouaib S. Chronic hypoxia is associated with transcriptomic reprogramming and increased genomic instability in cancer cells. Front Cell Dev Biol 2023; 11:1095419. [PMID: 36968212 PMCID: PMC10033758 DOI: 10.3389/fcell.2023.1095419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
Hypoxia afflicts the microenvironment of solid tumors fueling malignancy. We investigated the impact of long hypoxia exposure on transcriptional remodeling, tumor mutational burden (TMB), and genomic instability of cancer cells that were grouped based on their inherent sensitivity or resistance to hypoxia. A hypoxia score was used as a metric to distinguish between the most hypoxia-sensitive (hypoxia high (HH)), and most resistant (hypoxia low (HL)) cancer cells. By applying whole exome sequencing and microarray analysis, we showed that the HH group was indeed more sensitive to hypoxia, having significantly higher TMB (p = 0.03) and copy number losses (p = 0.03), as well as a trend of higher transcriptional response. Globally cells adapted by decreasing expression of genes involved in metabolism, proliferation, and protein maturation, and increasing alternative splicing. They accumulated mutations, especially frameshift insertions, and harbored increased copy number alterations, indicating increased genomic instability. Cells showing highest TMB simultaneously experienced a significant downregulation of DNA replication and repair and chromosomal maintenance pathways. A sixteen-gene common response to chronic hypoxia was put forth, including genes regulating angiogenesis and proliferation. Our findings show that chronic hypoxia enables survival of tumor cells by metabolic reprogramming, modulating proliferation, and increasing genomic instability. They additionally highlight key adaptive pathways that can potentially be targeted to prevent cancer cells residing in chronically hypoxic tumor areas from thriving.
Collapse
Affiliation(s)
- Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Mohak Sharda
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, Karnataka, India
- School of Life Science, The University of Trans-Disciplinary Health Sciences & Technology (TDU), Bangalore, India
| | - Shyama Prasad Rao
- Center for Bioinformatics, NITTE deemed to be University, Mangaluru, India
| | | | - Nagwa Ahmed Zeinelabdin
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Ayda Shah Mahmood
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Husam Nawafleh
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Munazza Samar Khan
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Goutham Hassan Venkatesh
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- INSERM UMR 1186, Integrative Tumor Immunology and Cancer Immunotherapy, Gustave Roussy, EPHE, Faculty De médecine University Paris-Sud, University Paris-Saclay, Villejuif, France
- *Correspondence: Salem Chouaib, ,
| |
Collapse
|
32
|
Mirjolet C, Baude J, Galluzzi L. Dual impact of radiation therapy on tumor-targeting immune responses. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 378:xiii-xxiv. [PMID: 37438022 DOI: 10.1016/s1937-6448(23)00114-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Affiliation(s)
- Céline Mirjolet
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France; TIReCS Team, UMR INSERM 1231, Dijon, France.
| | - Jérémy Baude
- Radiation Oncology Department, Preclinical Radiation Therapy and Radiobiology Unit, GF Leclerc Centre, Unicancer, Dijon, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
33
|
Shayan S, Arashkia A, Azadmanesh K. Modifying oncolytic virotherapy to overcome the barrier of the hypoxic tumor microenvironment. Where do we stand? Cancer Cell Int 2022; 22:370. [PMID: 36424577 PMCID: PMC9686061 DOI: 10.1186/s12935-022-02774-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022] Open
Abstract
Viruses are completely dependent on host cell machinery for their reproduction. As a result, factors that influence the state of cells, such as signaling pathways and gene expression, could determine the outcome of viral pathogenicity. One of the important factors influencing cells or the outcome of viral infection is the level of oxygen. Recently, oncolytic virotherapy has attracted attention as a promising approach to improving cancer treatment. However, it was shown that tumor cells are mostly less oxygenated compared with their normal counterparts, which might affect the outcome of oncolytic virotherapy. Therefore, knowing how oncolytic viruses could cope with stressful environments, particularly hypoxic environments, might be essential for improving oncolytic virotherapy.
Collapse
Affiliation(s)
- Sara Shayan
- grid.420169.80000 0000 9562 2611Department of Molecular Virology, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran, Iran
| | - Arash Arashkia
- grid.420169.80000 0000 9562 2611Department of Molecular Virology, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran, Iran
| | - Kayhan Azadmanesh
- grid.420169.80000 0000 9562 2611Department of Molecular Virology, Pasteur Institute of Iran, No. 69, Pasteur Ave, Tehran, Iran
| |
Collapse
|
34
|
Mohan N, Wellach K, Özerdem C, Veits N, Förster JD, Foehr S, Bonsack M, Riemer AB. Effects of hypoxia on antigen presentation and T cell-based immune recognition of HPV16-transformed cells. Front Immunol 2022; 13:918528. [PMID: 36341354 PMCID: PMC9634485 DOI: 10.3389/fimmu.2022.918528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 09/26/2022] [Indexed: 02/27/2025] Open
Abstract
Attempts to develop a therapeutic vaccine against human papillomavirus (HPV)-induced malignancies have mostly not been clinically successful to date. One reason may be the hypoxic microenvironment present in most tumors, including cervical cancer. Hypoxia dysregulates the levels of human leukocyte antigen (HLA) class I molecules in different tumor entities, impacts the function of cytotoxic T cells, and leads to decreased protein levels of the oncoproteins E6 and E7 in HPV-transformed cells. Therefore, we investigated the effect of hypoxia on the presentation of HPV16 E6- and E7-derived epitopes in cervical cancer cells and its effect on epitope-specific T cell cytotoxicity. Hypoxia induced downregulation of E7 protein levels in all analyzed cell lines, as assessed by Western blotting. However, contrary to previous reports, no perturbation of antigen processing and presentation machinery (APM) components and HLA-A2 surface expression upon hypoxia treatment was detected by mass spectrometry and flow cytometry, respectively. Cytotoxicity assays performed in hypoxic conditions showed differential effects on the specific killing of HPV16-positive cervical cancer cells by epitope-specific CD8+ T cell lines in a donor- and peptide-specific manner. Effects of hypoxia on the expression of PD-L1 were ruled out by flow cytometry analysis. Altogether, our results under hypoxia show a decreased expression of E6 and E7, but an intact APM, and epitope- and donor-dependent effects on T cell cytotoxicity towards HPV16-positive target cells. This suggests that successful immunotherapies can be developed for hypoxic HPV-induced cervical cancer, with careful choice of target epitopes, and ideally in combination with hypoxia-alleviating measures.
Collapse
Affiliation(s)
- Nitya Mohan
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Kathrin Wellach
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Ceren Özerdem
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Nisha Veits
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Jonas D. Förster
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Sophia Foehr
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Maria Bonsack
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Angelika B. Riemer
- Immunotherapy and Immunoprevention, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Molecular Vaccine Design, German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
35
|
Taghizadeh-Hesary F, Akbari H, Bahadori M, Behnam B. Targeted Anti-Mitochondrial Therapy: The Future of Oncology. Genes (Basel) 2022; 13:1728. [PMID: 36292613 PMCID: PMC9602426 DOI: 10.3390/genes13101728] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/16/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
Like living organisms, cancer cells require energy to survive and interact with their environment. Mitochondria are the main organelles for energy production and cellular metabolism. Recently, investigators demonstrated that cancer cells can hijack mitochondria from immune cells. This behavior sheds light on a pivotal piece in the cancer puzzle, the dependence on the normal cells. This article illustrates the benefits of new functional mitochondria for cancer cells that urge them to hijack mitochondria. It describes how functional mitochondria help cancer cells' survival in the harsh tumor microenvironment, immune evasion, progression, and treatment resistance. Recent evidence has put forward the pivotal role of mitochondria in the metabolism of cancer stem cells (CSCs), the tumor components responsible for cancer recurrence and metastasis. This theory highlights the mitochondria in cancer biology and explains how targeting mitochondria may improve oncological outcomes.
Collapse
Affiliation(s)
- Farzad Taghizadeh-Hesary
- ENT and Head and Neck Research Center and Department, The Five Senses Health Institute, School of Medicine, Iran University of Medical Sciences, Tehran 1445613131, Iran
- Department of Radiation Oncology, Iran University of Medical Sciences, Tehran 1445613131, Iran
| | - Hassan Akbari
- Department of Pathology, Shahid Beheshti University of Medical Sciences, Tehran P.O. Box 4739-19395, Iran
- Traditional Medicine School, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Moslem Bahadori
- Faculty of Medicine, Tehran University of Medical Sciences, Tehran P.O. Box 14155-6559, Iran
| | - Babak Behnam
- Department of Regulatory Affairs, Amarex Clinical Research, Germantown, MD 20874, USA
| |
Collapse
|
36
|
Wei C, Ma Y, Wang F, Liao Y, Chen Y, Zhao B, Zhao Q, Wang D, Tang D. Igniting Hope for Tumor Immunotherapy: Promoting the "Hot and Cold" Tumor Transition. Clin Med Insights Oncol 2022; 16:11795549221120708. [PMID: 36147198 PMCID: PMC9486259 DOI: 10.1177/11795549221120708] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/05/2022] [Indexed: 12/02/2022] Open
Abstract
The discovery of immune checkpoint inhibitors (ICIs) has ushered a new era for immunotherapy against malignant tumors through the killing effects of cytotoxic T lymphocytes in the tumor microenvironment (TME), resulting in long-lasting tumor suppression and regression. Nevertheless, given that ICIs are highly dependent on T cells in the TME and that most tumors lack T-cell infiltration, promoting the conversion of such immunosuppressive "cold" tumors to "hot" tumors is currently a key challenge in tumor immunotherapy. Herein, we systematically outlined the mechanisms underlying the formation of the immunosuppressive TME in cold tumors, including the role of immunosuppressive cells, impaired antigen presentation, transforming growth factor-β, STAT3 signaling, adenosine, and interferon-γ signaling. Moreover, therapeutic strategies for promoting cold tumors to hot tumors with adequate T-cell infiltration were also discussed. Finally, the prospects of therapeutic tools such as oncolytic viruses, nanoparticles, and photothermal therapy in restoring immune activity in cold tumors were thoroughly reviewed.
Collapse
Affiliation(s)
- Chen Wei
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Yichao Ma
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Fei Wang
- Clinical Medical College, Dalian
Medical University, Dalian, China
| | - Yiqun Liao
- Clinical Medical College, Dalian
Medical University, Dalian, China
| | - Yuji Chen
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Bin Zhao
- Clinical Medical College, Dalian
Medical University, Dalian, China
| | - Qi Zhao
- Clinical Medical College, Yangzhou
University, Yangzhou, China
| | - Daorong Wang
- Department of General Surgery,
Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s
Hospital, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery,
Institute of General Surgery, Clinical Medical College, Northern Jiangsu People’s
Hospital, Yangzhou University, Yangzhou, China
| |
Collapse
|
37
|
Melia F, Udomjarumanee P, Zinovkin D, Arghiani N, Pranjol MZI. Pro-tumorigenic role of type 2 diabetes-induced cellular senescence in colorectal cancer. Front Oncol 2022; 12:975644. [PMID: 36059680 PMCID: PMC9434004 DOI: 10.3389/fonc.2022.975644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related mortality worldwide. The disease still remains incurable and highly lethal in the advanced stage, representing a global health concern. Therefore, it is essential to understand the causes and risk factors leading to its development. Because age-related cellular senescence and type 2 diabetes (T2D) have been recognised as risk factors for CRC development, the recent finding that type 2 diabetic patients present an elevated circulating volume of senescent cells raises the question whether type 2 diabetes facilitates the process of CRC tumorigenesis by inducing premature cell senescence. In this review, we will discuss the mechanisms according to which T2D induces cellular senescence and the role of type 2 diabetes-induced cellular senescence in the pathogenesis and progression of colorectal cancer. Lastly, we will explore the current therapeutic approaches and challenges in targeting senescence.
Collapse
Affiliation(s)
- Francesco Melia
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Palita Udomjarumanee
- Department of Immunology and Inflammation, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Dmitry Zinovkin
- Department of Pathology, Gomel State Medical University, Gomel, Belarus
| | - Nahid Arghiani
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- *Correspondence: Nahid Arghiani, ; Md Zahidul Islam Pranjol,
| | - Md Zahidul Islam Pranjol
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- *Correspondence: Nahid Arghiani, ; Md Zahidul Islam Pranjol,
| |
Collapse
|
38
|
HIF-1α Inhibition Improves Anti-Tumor Immunity and Promotes the Efficacy of Stereotactic Ablative Radiotherapy (SABR). Cancers (Basel) 2022; 14:cancers14133273. [PMID: 35805044 PMCID: PMC9265101 DOI: 10.3390/cancers14133273] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 01/10/2023] Open
Abstract
Simple Summary Stereotactic ablative radiotherapy (SABR), which irradiates tumors with high-dose radiation per fraction, promotes anti-tumor immunity by stimulating various immune processes. SABR also induces vascular damage and obstructs blood flow, thereby increasing tumor hypoxia and upregulation of hypoxia-inducible factors HIF-1α and HIF-2α, master transcription factors for the cellular response to hypoxia. HIF-1α and HIF-2α are key players in the upregulation of immune suppression in hypoxia. Therefore, the radiation-induced increase in anti-tumor immunity is masked by the HIF-mediated immune suppression. Pre-clinical experiments show that inhibition of HIF-1α effectively prevents immune suppression and improves anti-tumor immunity. A combination of HIF-1α inhibitors with immunotherapy with checkpoint blocking antibodies may represent a novel approach to boost anti-tumor immunity and enhance the efficacy of SABR. Abstract High-dose hypofractionated radiation such as SABR (stereotactic ablative radiotherapy) evokes an anti-tumor immune response by promoting a series of immune-stimulating processes, including the release of tumor-specific antigens from damaged tumor cells and the final effector phase of immune-mediated lysis of target tumor cells. High-dose hypofractionated radiation also causes vascular damage in tumors, thereby increasing tumor hypoxia and upregulation of hypoxia-inducible factors HIF-1α and HIF-2α, the master transcription factors for the cellular response to hypoxia. HIF-1α and HIF-2α are critical factors in the upregulation of immune suppression and are the master regulators of immune evasion of tumors. Consequently, SABR-induced increase in anti-tumor immunity is counterbalanced by the increase in immune suppression mediated by HIFα. Inhibition of HIF-1α with small molecules such as metformin downregulates immunosuppressive pathways, including the expression of immune checkpoints, and it improves or restores the anti-tumor immunity stimulated by irradiation. Combinations of HIFα inhibitors, particularly HIF-1α inhibitors, with immune checkpoint blocking antibodies may represent a novel approach to boost the overall anti-tumor immune profile in patients and thus enhance outcomes after SABR.
Collapse
|
39
|
Salman S, Meyers DJ, Wicks EE, Lee SN, Datan E, Thomas AM, Anders NM, Hwang Y, Lyu Y, Yang Y, Jackson W, Dordai D, Rudek MA, Semenza GL. HIF inhibitor 32-134D eradicates murine hepatocellular carcinoma in combination with anti-PD1 therapy. J Clin Invest 2022; 132:156774. [PMID: 35499076 PMCID: PMC9057582 DOI: 10.1172/jci156774] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/01/2022] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer mortality worldwide and available therapies, including immunotherapies, are ineffective for many patients. HCC is characterized by intratumoral hypoxia, and increased expression of hypoxia-inducible factor 1α (HIF-1α) in diagnostic biopsies is associated with patient mortality. Here we report the development of 32-134D, a low-molecular-weight compound that effectively inhibits gene expression mediated by HIF-1 and HIF-2 in HCC cells, and blocks human and mouse HCC tumor growth. In immunocompetent mice bearing Hepa1-6 HCC tumors, addition of 32-134D to anti-PD1 therapy increased the rate of tumor eradication from 25% to 67%. Treated mice showed no changes in appearance, behavior, body weight, hemoglobin, or hematocrit. Compound 32-134D altered the expression of a large battery of genes encoding proteins that mediate angiogenesis, glycolytic metabolism, and responses to innate and adaptive immunity. This altered gene expression led to significant changes in the tumor immune microenvironment, including a decreased percentage of tumor-associated macrophages and myeloid-derived suppressor cells, which mediate immune evasion, and an increased percentage of CD8+ T cells and natural killer cells, which mediate antitumor immunity. Taken together, these preclinical findings suggest that combining 32-134D with immune checkpoint blockade may represent a breakthrough therapy for HCC.
Collapse
Affiliation(s)
- Shaima Salman
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- McKusick-Nathans Department of Genetic Medicine
| | | | | | - Sophia N. Lee
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
| | - Emmanuel Datan
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- McKusick-Nathans Department of Genetic Medicine
| | - Aline M. Thomas
- Institute for Cell Engineering
- Department of Radiology and Radiological Science
| | - Nicole M. Anders
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center
| | - Yousang Hwang
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- Department of Pharmacology and Molecular Sciences
| | - Yajing Lyu
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- McKusick-Nathans Department of Genetic Medicine
| | - Yongkang Yang
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center
| | - Walter Jackson
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- McKusick-Nathans Department of Genetic Medicine
| | - Dominic Dordai
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- McKusick-Nathans Department of Genetic Medicine
| | - Michelle A. Rudek
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Gregg L. Semenza
- Armstrong Oxygen Biology Research Center
- Institute for Cell Engineering
- McKusick-Nathans Department of Genetic Medicine
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center
| |
Collapse
|
40
|
Corgnac S, Damei I, Gros G, Caidi A, Terry S, Chouaib S, Deloger M, Mami-Chouaib F. Cancer stem-like cells evade CD8 +CD103 + tumor-resident memory T (T RM) lymphocytes by initiating an epithelial-to-mesenchymal transition program in a human lung tumor model. J Immunother Cancer 2022; 10:jitc-2022-004527. [PMID: 35418483 PMCID: PMC9014106 DOI: 10.1136/jitc-2022-004527] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2022] [Indexed: 02/06/2023] Open
Abstract
Background Cancer stem cells (CSC) define a population of rare malignant cells endowed with ‘stemness’ properties, such as self-renewing, multipotency and tumorigenicity. They are responsible for tumor initiation and progression, and could be associated with resistance to immunotherapies by negatively regulating antitumor immune response and acquiring molecular features enabling escape from CD8 T-cell immunity. However, the immunological hallmarks of human lung CSC and their potential interactions with resident memory T (TRM) cells within the tumor microenvironment have not been investigated. Methods We generated a non-small cell lung cancer model, including CSC line and clones, and autologous CD8+CD103+ TRM and CD8+CD103− non-TRM clones, to dissect out immune properties of CSC and their susceptibility to specific T-cell-mediated cytotoxic activity. Results Unlike their parental tumor cells, lung CSC are characterized by the initiation of an epithelial-to-mesenchymal transition program defined by upregulation of the SNAIL1 transcription factor and downregulation of phosphorylated-GSK-3β and cell surface E-cadherin. Acquisition of a CSC profile results in partial resistance to TRM-cell-mediated cytotoxicity, which correlates with decreased surface expression of the CD103 ligand E-cadherin and human leukocyte antigen-A2-neoepitope complexes. On the other hand, CSC gained expression of intercellular adhesion molecule (ICAM)-1 and thereby sensitivity to leukocyte function-associated antigen (LFA)-1-dependent non-TRM-cell-mediated killing. Cytotoxicity is inhibited by anti-ICAM-1 and anti-major histocompatibility complex class I neutralizing antibodies further emphasizing the role of LFA-1/ICAM-1 interaction in T-cell receptor-dependent lytic function. Conclusion Our data support the rational design of immunotherapeutic strategies targeting CSC to optimize their responsiveness to local CD8+CD103+ TRM cells for more efficient anticancer treatments.
Collapse
Affiliation(s)
- Stéphanie Corgnac
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Isabelle Damei
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Gwendoline Gros
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Aziza Caidi
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,Gustave Roussy, Plateforme de Bioinformatique, Université Paris-Saclay, INSERM US23, CNRS UMS 3655, Villejuif, France
| | - Stéphane Terry
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| | - Salem Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France.,Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, UAE
| | - Marc Deloger
- Gustave Roussy, Plateforme de Bioinformatique, Université Paris-Saclay, INSERM US23, CNRS UMS 3655, Villejuif, France
| | - Fathia Mami-Chouaib
- INSERM UMR 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Fac. de Médecine - Univ. Paris-Sud, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
41
|
Koukourakis IM, Giatromanolaki A, Mitrakas A, Koukourakis MI. Loss of HLA-class-I expression in non-small-cell lung cancer: Association with prognosis and anaerobic metabolism. Cell Immunol 2022; 373:104495. [DOI: 10.1016/j.cellimm.2022.104495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/03/2022] [Accepted: 02/04/2022] [Indexed: 01/21/2023]
|
42
|
Lappano R, Todd LA, Stanic M, Cai Q, Maggiolini M, Marincola F, Pietrobon V. Multifaceted Interplay between Hormones, Growth Factors and Hypoxia in the Tumor Microenvironment. Cancers (Basel) 2022; 14:539. [PMID: 35158804 PMCID: PMC8833523 DOI: 10.3390/cancers14030539] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Hormones and growth factors (GFs) are signaling molecules implicated in the regulation of a variety of cellular processes. They play important roles in both healthy and tumor cells, where they function by binding to specific receptors on target cells and activating downstream signaling cascades. The stages of tumor progression are influenced by hormones and GF signaling. Hypoxia, a hallmark of cancer progression, contributes to tumor plasticity and heterogeneity. Most solid tumors contain a hypoxic core due to rapid cellular proliferation that outgrows the blood supply. In these circumstances, hypoxia-inducible factors (HIFs) play a central role in the adaptation of tumor cells to their new environment, dramatically reshaping their transcriptional profile. HIF signaling is modulated by a variety of factors including hormones and GFs, which activate signaling pathways that enhance tumor growth and metastatic potential and impair responses to therapy. In this review, we summarize the role of hormones and GFs during cancer onset and progression with a particular focus on hypoxia and the interplay with HIF proteins. We also discuss how hypoxia influences the efficacy of cancer immunotherapy, considering that a hypoxic environment may act as a determinant of the immune-excluded phenotype and a major hindrance to the success of adoptive cell therapies.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Lauren A. Todd
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Mia Stanic
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Qi Cai
- Kite Pharma Inc., Santa Monica, CA 90404, USA; (Q.C.); (F.M.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | | | | |
Collapse
|
43
|
Maggs L, Sadagopan A, Moghaddam AS, Ferrone S. HLA class I antigen processing machinery defects in antitumor immunity and immunotherapy. Trends Cancer 2021; 7:1089-1101. [PMID: 34489208 PMCID: PMC8651070 DOI: 10.1016/j.trecan.2021.07.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 07/28/2021] [Accepted: 07/30/2021] [Indexed: 12/14/2022]
Abstract
Human leukocyte antigen (HLA) class I antigen-processing machinery (APM) plays a crucial role in the synthesis and expression of HLA class I tumor antigen-derived peptide complexes; the latter mediate the recognition and elimination of malignant cells by cognate T cells. Defects in HLA class I APM component expression and/or function are frequently found in cancer cells, providing them with an immune escape mechanism that has relevance in the clinical course of the disease and in the response to T-cell-based immunotherapy. The majority of HLA class I APM defects (>75%) are caused by epigenetic mechanisms or dysregulated signaling and therefore can be corrected by strategies that counteract the underlying mechanisms. Their application in oncology is likely to improve responses to T-cell-based immunotherapies, including checkpoint inhibition.
Collapse
Affiliation(s)
- Luke Maggs
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Ananthan Sadagopan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ali Sanjari Moghaddam
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Soldano Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
44
|
Therapeutic targeting of the hypoxic tumour microenvironment. Nat Rev Clin Oncol 2021; 18:751-772. [PMID: 34326502 DOI: 10.1038/s41571-021-00539-4] [Citation(s) in RCA: 246] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2021] [Indexed: 02/07/2023]
Abstract
Hypoxia is prevalent in human tumours and contributes to microenvironments that shape cancer evolution and adversely affect therapeutic outcomes. Historically, two different tumour microenvironment (TME) research communities have been discernible. One has focused on physicochemical gradients of oxygen, pH and nutrients in the tumour interstitium, motivated in part by the barrier that hypoxia poses to effective radiotherapy. The other has focused on cellular interactions involving tumour and non-tumour cells within the TME. Over the past decade, strong links have been established between these two themes, providing new insights into fundamental aspects of tumour biology and presenting new strategies for addressing the effects of hypoxia and other microenvironmental features that arise from the inefficient microvascular system in solid tumours. This Review provides a perspective on advances at the interface between these two aspects of the TME, with a focus on translational therapeutic opportunities relating to the elimination and/or exploitation of tumour hypoxia.
Collapse
|
45
|
Bouleftour W, Rowinski E, Louati S, Sotton S, Wozny AS, Moreno-Acosta P, Mery B, Rodriguez-Lafrasse C, Magne N. A Review of the Role of Hypoxia in Radioresistance in Cancer Therapy. Med Sci Monit 2021; 27:e934116. [PMID: 34728593 PMCID: PMC8573967 DOI: 10.12659/msm.934116] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hypoxia involves neoplastic cells. Unlike normal tissue, solid tumors are composed of aberrant vasculature, leading to a hypoxic microenvironment. Hypoxia is also known to be involved in both metastasis initiation and therapy resistance. Radiotherapy is the appropriate treatment in about half of all cancers, but loco-regional control failure and a disease recurrence often occur due to clinical radioresistance. Hypoxia induces radioresistance through a number of molecular pathways, and numerous strategies have been developed to overcome this. Nevertheless, these strategies have resulted in disappointing results, including adverse effects and limited efficacy. Additional clinical studies are needed to achieve a better understanding of the complex hypoxia pathways. This review presents an update on the mechanisms of hypoxia in radioresistance in solid tumors and the potential therapeutic solutions.
Collapse
Affiliation(s)
- Wafa Bouleftour
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Elise Rowinski
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Safa Louati
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| | - Sandrine Sotton
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Anne-Sophie Wozny
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| | - Pablo Moreno-Acosta
- Research Group in Cancer Biology, National Cancer Institute, Bogotá, Colombia
| | - Benoite Mery
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France
| | - Claire Rodriguez-Lafrasse
- Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| | - Nicolas Magne
- Radiotherapy Department, Lucien Neuwirth Cancerology Institute, Saint Priest en Jarez, France.,Université Lyon 1, Faculté de Médecine-Lyon-Sud, Oullins, France.,Laboratoire de Radiobiologie Cellulaire et Moléculaire, CNRS UMR 5822, Institut de Physique Nucléaire de Lyon, IPNL, Villeurbanne, France.,Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
46
|
Link B, Torres Crigna A, Hölzel M, Giordano FA, Golubnitschaja O. Abscopal Effects in Metastatic Cancer: Is a Predictive Approach Possible to Improve Individual Outcomes? J Clin Med 2021; 10:5124. [PMID: 34768644 PMCID: PMC8584726 DOI: 10.3390/jcm10215124] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Patients with metastatic cancers often require radiotherapy (RT) as a palliative therapy for cancer pain. RT can, however, also induce systemic antitumor effects outside of the irradiated field (abscopal effects) in various cancer entities. The occurrence of the abscopal effect is associated with a specific immunological activation in response to RT-induced cell death, which is mainly seen under concomitant immune checkpoint blockade. Even if the number of reported apscopal effects has increased since the introduction of immune checkpoint inhibition, its occurrence is still considered rare and unpredictable. The cases reported so far may nevertheless allow for identifying first biomarkers and clinical patterns. We here review biomarkers that may be helpful to predict the occurrence of abscopal effects and hence to optimize therapy for patients with metastatic cancers.
Collapse
Affiliation(s)
- Barbara Link
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany; (B.L.); (A.T.C.); (F.A.G.)
| | - Adriana Torres Crigna
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany; (B.L.); (A.T.C.); (F.A.G.)
| | - Michael Hölzel
- Institute of Experimental Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany;
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany; (B.L.); (A.T.C.); (F.A.G.)
| | - Olga Golubnitschaja
- Predictive, Preventive, Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| |
Collapse
|
47
|
Hassan Venkatesh G, Abou Khouzam R, Shaaban Moustafa Elsayed W, Ahmed Zeinelabdin N, Terry S, Chouaib S. Tumor hypoxia: an important regulator of tumor progression or a potential modulator of tumor immunogenicity? Oncoimmunology 2021; 10:1974233. [PMID: 34595058 PMCID: PMC8477925 DOI: 10.1080/2162402x.2021.1974233] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Affiliation(s)
- Goutham Hassan Venkatesh
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Raefa Abou Khouzam
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | | | - Nagwa Ahmed Zeinelabdin
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Stéphane Terry
- Inserm Umr 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, Villejuif, France
| | - Salem Chouaib
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
- Inserm Umr 1186, Integrative Tumor Immunology and Immunotherapy, Gustave Roussy, Faculty of Medicine, University Paris-Saclay, Villejuif, France
| |
Collapse
|
48
|
Kim H, Shim BY, Lee SJ, Lee JY, Lee HJ, Kim IH. Loss of Von Hippel-Lindau ( VHL) Tumor Suppressor Gene Function: VHL-HIF Pathway and Advances in Treatments for Metastatic Renal Cell Carcinoma (RCC). Int J Mol Sci 2021; 22:9795. [PMID: 34575959 PMCID: PMC8470481 DOI: 10.3390/ijms22189795] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/02/2021] [Accepted: 09/07/2021] [Indexed: 12/18/2022] Open
Abstract
Renal cell carcinoma (RCC) is a malignancy of the kidney originating from the tubular epithelium. Inactivation of the von Hippel-Lindau tumor-suppressor gene (VHL) is found in most clear cell renal cell carcinomas (ccRCCs). The VHL-HIF-VEGF/VEGFR pathway, which involves the von Hippel-Lindau tumor suppressor protein (VHL), hypoxia-inducible factor (HIF), vascular endothelial growth factor (VEGF), and its receptor (VEGFR), is a well-studied therapeutic target for metastatic ccRCC. Therefore, over the past decade, anti-angiogenic agents targeting VEGFR have served as the standard treatment for metastatic RCC. Recently, based on the immunomodulatory effect of anti-VEGFR therapy, anti-angiogenic agents and immune checkpoint inhibitor combination strategies have also emerged as therapeutic strategies. These advances were made possible by the improved understanding of the VHL-HIF pathway. In this review, we summarize the historical evolution of ccRCC treatments, with a focus on the involvement of the VHL-HIF pathway.
Collapse
Affiliation(s)
- Hyunho Kim
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent’s Hospital, The Catholic University of Korea, 93 Jungbu-daero, Paldal-gu, Suwon 16247, Korea; (H.K.); (B.Y.S.)
| | - Byoung Yong Shim
- Division of Medical Oncology, Department of Internal Medicine, St. Vincent’s Hospital, The Catholic University of Korea, 93 Jungbu-daero, Paldal-gu, Suwon 16247, Korea; (H.K.); (B.Y.S.)
| | - Seung-Ju Lee
- Department of Urology, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, 93 Jungbu-daero, Paldal-gu, Suwon 16247, Korea;
| | - Ji Youl Lee
- Department of Urology Cancer Center, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea;
| | - Hyo-Jin Lee
- Department of Internal Medicine, Chungnam National University School of Medicine, 266 Munhwa-ro, Jung-gu, Daejeon 35015, Korea;
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea
| |
Collapse
|
49
|
Colombani T, Eggermont LJ, Hatfield SM, Rogers ZJ, Rezaeeyazdi M, Memic A, Sitkovsky MV, Bencherif SA. Oxygen-Generating Cryogels Restore T Cell Mediated Cytotoxicity in Hypoxic Tumors. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2102234. [PMID: 37745940 PMCID: PMC10516343 DOI: 10.1002/adfm.202102234] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Indexed: 09/26/2023]
Abstract
Solid tumors are protected from antitumor immune responses due to their hypoxic microenvironments. Weakening hypoxia-driven immunosuppression by hyperoxic breathing of 60% oxygen has shown to be effective in unleashing antitumor immune cells against solid tumors. However, efficacy of systemic oxygenation is limited against solid tumors outside of lungs and has been associated with unwanted side effects. As a result, it is essential to develop targeted oxygenation alternatives to weaken tumor hypoxia as novel approaches to restore immune responses against cancer. Herein, we report on injectable oxygen-generating cryogels (O2-cryogels) to reverse tumor-induced hypoxia. These macroporous biomaterials were designed to locally deliver oxygen, inhibit the expression of hypoxia-inducible genes in hypoxic melanoma cells, and reduce the accumulation of immunosuppressive extracellular adenosine. Our data show that O2-cryogels enhance T cell-mediated secretion of cytotoxic proteins, restoring the killing ability of tumor-specific CTLs, both in vitro and in vivo. In summary, O2-cryogels provide a unique and safe platform to supply oxygen as a co-adjuvant in hypoxic tumors and have the potential to improve cancer immunotherapies.
Collapse
Affiliation(s)
- Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Loek J. Eggermont
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | - Stephen M. Hatfield
- New England Inflammation and Tissue Protection Institute, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Zachary J. Rogers
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| | | | - Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah, Makkah 21589, Saudi Arabia
| | - Michail V. Sitkovsky
- New England Inflammation and Tissue Protection Institute, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Biomechanics and Bioengineering (BMBI), UTC CNRS UMR 7338, University of Technology of Compiègne, Sorbonne University, 60203 Compiègne, France
| |
Collapse
|
50
|
Hazini A, Fisher K, Seymour L. Deregulation of HLA-I in cancer and its central importance for immunotherapy. J Immunother Cancer 2021; 9:e002899. [PMID: 34353849 PMCID: PMC8344275 DOI: 10.1136/jitc-2021-002899] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 12/28/2022] Open
Abstract
It is now well accepted that many tumors undergo a process of clonal selection which means that tumor antigens arising at various stages of tumor progression are likely to be represented in just a subset of tumor cells. This process is thought to be driven by constant immunosurveillance which applies selective pressure by eliminating tumor cells expressing antigens that are recognized by T cells. It is becoming increasingly clear that the same selective pressure may also select for tumor cells that evade immune detection by acquiring deficiencies in their human leucocyte antigen (HLA) presentation pathways, allowing important tumor antigens to persist within cells undetected by the immune system. Deficiencies in antigen presentation pathway can arise by a variety of mechanisms, including genetic and epigenetic changes, and functional antigen presentation is a hard phenomenon to assess using our standard analytical techniques. Nevertheless, it is likely to have profound clinical significance and could well define whether an individual patient will respond to a particular type of therapy or not. In this review we consider the mechanisms by which HLA function may be lost in clinical disease, we assess the implications for current immunotherapy approaches using checkpoint inhibitors and examine the prognostic impact of HLA loss demonstrated in clinical trials so far. Finally, we propose strategies that might be explored for possible patient stratification.
Collapse
Affiliation(s)
- Ahmet Hazini
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, UK
| | - Kerry Fisher
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, UK
| | - Len Seymour
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, UK
| |
Collapse
|