1
|
Chen B, Yang Y, Wang X, Yang W, Lu Y, Wang D, Zhuo E, Tang Y, Su J, Tang G, Shao S, Gu K. mRNA vaccine development and applications: A special focus on tumors (Review). Int J Oncol 2024; 65:81. [PMID: 38994758 PMCID: PMC11251742 DOI: 10.3892/ijo.2024.5669] [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: 01/14/2024] [Accepted: 05/20/2024] [Indexed: 07/13/2024] Open
Abstract
Cancer is characterized by unlimited proliferation and metastasis, and traditional therapeutic strategies usually result in the acquisition of drug resistance, thus highlighting the need for more personalized treatment. mRNA vaccines transfer the gene sequences of exogenous target antigens into human cells through transcription and translation to stimulate the body to produce specific immune responses against the encoded proteins, so as to enable the body to obtain immune protection against said antigens; this approach may be adopted for personalized cancer therapy. Since the recent coronavirus pandemic, the development of mRNA vaccines has seen substantial progress and widespread adoption. In the present review, the development of mRNA vaccines, their mechanisms of action, factors influencing their function and the current clinical applications of the vaccine are discussed. A focus is placed on the application of mRNA vaccines in cancer, with the aim of highlighting unique advances and the remaining challenges of this novel and promising therapeutic approach.
Collapse
Affiliation(s)
- Bangjie Chen
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yipin Yang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Xinyi Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Wenzhi Yang
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - You Lu
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Daoyue Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Enba Zhuo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Yanchao Tang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Junhong Su
- Department of Rehabilitation, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Guozheng Tang
- Department of Orthopedics, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui 237008, P.R. China
| | - Song Shao
- Department of Orthopedics, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui 237008, P.R. China
| | - Kangsheng Gu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
2
|
Barboy O, Bercovich A, Li H, Eyal-Lubling Y, Yalin A, Shapir Itai Y, Abadie K, Zada M, David E, Shlomi-Loubaton S, Katzenelenbogen Y, Jaitin DA, Gur C, Yofe I, Feferman T, Cohen M, Dahan R, Newell EW, Lifshitz A, Tanay A, Amit I. Modeling T cell temporal response to cancer immunotherapy rationalizes development of combinatorial treatment protocols. NATURE CANCER 2024; 5:742-759. [PMID: 38429414 DOI: 10.1038/s43018-024-00734-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Accepted: 01/19/2024] [Indexed: 03/03/2024]
Abstract
Successful immunotherapy relies on triggering complex responses involving T cell dynamics in tumors and the periphery. Characterizing these responses remains challenging using static human single-cell atlases or mouse models. To address this, we developed a framework for in vivo tracking of tumor-specific CD8+ T cells over time and at single-cell resolution. Our tools facilitate the modeling of gene program dynamics in the tumor microenvironment (TME) and the tumor-draining lymph node (tdLN). Using this approach, we characterize two modes of anti-programmed cell death protein 1 (PD-1) activity, decoupling induced differentiation of tumor-specific activated precursor cells from conventional type 1 dendritic cell (cDC1)-dependent proliferation and recruitment to the TME. We demonstrate that combining anti-PD-1 therapy with anti-4-1BB agonist enhances the recruitment and proliferation of activated precursors, resulting in tumor control. These data suggest that effective response to anti-PD-1 therapy is dependent on sufficient influx of activated precursor CD8+ cells to the TME and highlight the importance of understanding system-level dynamics in optimizing immunotherapies.
Collapse
Affiliation(s)
- Oren Barboy
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Akhiad Bercovich
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hanjie Li
- Department of Synthetic Immunology, Shenzhen Institutes of Advanced Technology, Shenzhen, China
| | - Yaniv Eyal-Lubling
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Adam Yalin
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yuval Shapir Itai
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Kathleen Abadie
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Mor Zada
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eyal David
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shir Shlomi-Loubaton
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Diego Adhemar Jaitin
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Chamutal Gur
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
- The Hebrew University, Jerusalem, Israel
| | - Ido Yofe
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Tali Feferman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Cohen
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Rony Dahan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Evan W Newell
- Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | - Aviezer Lifshitz
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Amos Tanay
- Department of Computer Science and Applied Mathematics and Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
3
|
Wang M, Yu F, Zhang Y, Li P. Novel insights into Notch signaling in tumor immunity: potential targets for cancer immunotherapy. Front Immunol 2024; 15:1352484. [PMID: 38444855 PMCID: PMC10912471 DOI: 10.3389/fimmu.2024.1352484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 02/06/2024] [Indexed: 03/07/2024] Open
Abstract
Notch signaling pathway is a highly conserved system of cell-to-cell communication that participates in various biological processes, such as stem cell maintenance, cell fate decision, cell proliferation and death during homeostasis and development. Dysregulation of Notch signaling has been associated with many aspects of cancer biology, such as maintenance of cancer stem-like cells (CSCs), cancer cell metabolism, angiogenesis and tumor immunity. Particularly, Notch signaling can regulate antitumor or pro-tumor immune cells within the tumor microenvironment (TME). Currently, Notch signaling has drawn significant attention in the therapeutic development of cancer treatment. In this review, we focus on the role of Notch signaling pathway in remodeling tumor immune microenvironment. We describe the impact of Notch signaling on the efficacy of cancer immunotherapies. Furthermore, we summarize the results of relevant preclinical and clinical trials of Notch-targeted therapeutics and discuss the challenges in their clinical application in cancer therapy. An improved understanding of the involvement of Notch signaling in tumor immunity will open the door to new options in cancer immunotherapy treatment.
Collapse
Affiliation(s)
- Man Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | | | | | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Jiang Q, Chen H, Zhou S, Zhu T, Liu W, Wu H, Zhang Y, Liu F, Sun Y. Ubiquilin-4 induces immune escape in gastric cancer by activating the notch signaling pathway. Cell Oncol (Dordr) 2024; 47:303-319. [PMID: 37702916 DOI: 10.1007/s13402-023-00869-8] [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] [Accepted: 08/25/2023] [Indexed: 09/14/2023] Open
Abstract
PURPOSE We aimed to investigate the role of ubiquilin-4 in predicting the immunotherapy response in gastric cancer. METHODS Retrospective RNA-sequencing and immunohistochemical analysis were performed for patients with gastric cancer who received programmed death-1 blockade therapy after recurrence. Multiplex immunohistochemistry identified immune cell types in gastric cancer tissues. We used immunocompetent 615 mice and immunodeficient nude mice to perform tumorigenic experiments. RESULTS Ubiquilin-4 expression was significantly higher in responders (p < 0.05, false discovery rate > 2.5) and showed slight superiority over programmed death ligand 1 in predicting programmed death-1 inhibitor therapy response (area under the curve: 87.08 vs. 72.50). Ubiquilin-4-high patients exhibited increased CD4+ and CD8+ T cells, T follicular helper cells, monocytes, and macrophages. Ubiquilin-4-overexpressed mouse forestomach carcinoma cells showed significantly enhanced growth in immunocompetent mice but not in immunodeficient mice. Upregulation or downregulation of ubiquilin-4 synergistically affected programmed death ligand 1 at the protein and messenger RNA levels. Functional enrichment analysis revealed significant enrichment of the Notch, JAK-STAT, and WNT signaling pathways in ubiquilin-4-high gastric cancers. Ubiquilin-4 promoted Numb degaration, activating the Notch signaling pathway and upregulating programmed death ligand 1. CONCLUSIONS Ubiquilin-4 may contribute to immune escape in gastric cancer by upregulating programmed death ligand 1 expression in tumor cells through Notch signaling activation. Thus, ubiquilin-4 could serve as a predictive marker for programmed death ligand 1 inhibitor therapy response in gastric cancer.
Collapse
Affiliation(s)
- Quan Jiang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Retroperitoneal Tumor and Soft Tissue Sarcoma Surgery, Fudan University, Shanghai, China
| | - Hao Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shixin Zhou
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Zhu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Retroperitoneal Tumor and Soft Tissue Sarcoma Surgery, Fudan University, Shanghai, China
| | - Wenshuai Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yong Zhang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Retroperitoneal Tumor and Soft Tissue Sarcoma Surgery, Fudan University, Shanghai, China.
| | - Fenglin Liu
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Gastric Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Huntington KE, Louie AD, Srinivasan PR, Schorl C, Lu S, Silverberg D, Newhouse D, Wu Z, Zhou L, Borden BA, Giles FJ, Dooner M, Carneiro BA, El-Deiry WS. GSK-3 Inhibitor Elraglusib Enhances Tumor-Infiltrating Immune Cell Activation in Tumor Biopsies and Synergizes with Anti-PD-L1 in a Murine Model of Colorectal Cancer. Int J Mol Sci 2023; 24:10870. [PMID: 37446056 PMCID: PMC10342141 DOI: 10.3390/ijms241310870] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 06/13/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that has been implicated in numerous oncogenic processes. GSK-3 inhibitor elraglusib (9-ING-41) has shown promising preclinical and clinical antitumor activity across multiple tumor types. Despite promising early-phase clinical trial results, there have been limited efforts to characterize the potential immunomodulatory properties of elraglusib. We report that elraglusib promotes immune cell-mediated tumor cell killing of microsatellite stable colorectal cancer (CRC) cells. Mechanistically, elraglusib sensitized CRC cells to immune-mediated cytotoxicity and enhanced immune cell effector function. Using western blots, we found that elraglusib decreased CRC cell expression of NF-κB p65 and several survival proteins. Using microarrays, we discovered that elraglusib upregulated the expression of proapoptotic and antiproliferative genes and downregulated the expression of cell proliferation, cell cycle progression, metastasis, TGFβ signaling, and anti-apoptotic genes in CRC cells. Elraglusib reduced CRC cell production of immunosuppressive molecules such as VEGF, GDF-15, and sPD-L1. Elraglusib increased immune cell IFN-γ secretion, which upregulated CRC cell gasdermin B expression to potentially enhance pyroptosis. Elraglusib enhanced immune effector function resulting in augmented granzyme B, IFN-γ, TNF-α, and TRAIL production. Using a syngeneic, immunocompetent murine model of microsatellite stable CRC, we evaluated elraglusib as a single agent or combined with immune checkpoint blockade (anti-PD-1/L1) and observed improved survival in the elraglusib and anti-PD-L1 group. Murine responders had increased tumor-infiltrating T cells, augmented granzyme B expression, and fewer regulatory T cells. Murine responders had reduced immunosuppressive (VEGF, VEGFR2) and elevated immunostimulatory (GM-CSF, IL-12p70) cytokine plasma concentrations. To determine the clinical significance, we then utilized elraglusib-treated patient plasma samples and found that reduced VEGF and BAFF and elevated IL-1 beta, CCL22, and CCL4 concentrations correlated with improved survival. Using paired tumor biopsies, we found that tumor-infiltrating immune cells had a reduced expression of inhibitory immune checkpoints (VISTA, PD-1, PD-L2) and an elevated expression of T-cell activation markers (CTLA-4, OX40L) after elraglusib treatment. These results address a significant gap in knowledge concerning the immunomodulatory mechanisms of GSK-3 inhibitor elraglusib, provide a rationale for the clinical evaluation of elraglusib in combination with immune checkpoint blockade, and are expected to have an impact on additional tumor types, besides CRC.
Collapse
Affiliation(s)
- Kelsey E. Huntington
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Lifespan Health System, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Pathobiology Graduate Program, Brown University, Providence, RI 02903, USA
| | - Anna D. Louie
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Lifespan Health System, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Department of Surgery, Lifespan Health System, Providence, RI 02903, USA
| | - Praveen R. Srinivasan
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Lifespan Health System, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Christoph Schorl
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Genomics Core Facility, Brown University, Providence, RI 02903, USA
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02903, USA
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Lifespan Health System, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - David Silverberg
- Molecular Pathology Core Facility, Brown University, Providence, RI 02903, USA
| | | | - Zhijin Wu
- Department of Biostatistics, Brown University, Providence, RI 02903, USA
| | - Lanlan Zhou
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Lifespan Health System, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Brittany A. Borden
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | | | - Mark Dooner
- Division of Hematology/Oncology, Department of Medicine, Lifespan Health System, Providence, RI 02903, USA
| | - Benedito A. Carneiro
- The Joint Program in Cancer Biology, Lifespan Health System, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Division of Hematology/Oncology, Department of Medicine, Lifespan Health System, Providence, RI 02903, USA
| | - Wafik S. El-Deiry
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI 02903, USA
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Brown University, Providence, RI 02903, USA
- The Joint Program in Cancer Biology, Lifespan Health System, Brown University, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Pathobiology Graduate Program, Brown University, Providence, RI 02903, USA
- Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
- Division of Hematology/Oncology, Department of Medicine, Lifespan Health System, Providence, RI 02903, USA
| |
Collapse
|
6
|
Tran TT, Caulfield J, Zhang L, Schoenfeld D, Djureinovic D, Chiang VL, Oria V, Weiss SA, Olino K, Jilaveanu LB, Kluger HM. Lenvatinib or anti-VEGF in combination with anti-PD-1 differentially augments antitumor activity in melanoma. JCI Insight 2023; 8:e157347. [PMID: 36821392 PMCID: PMC10132152 DOI: 10.1172/jci.insight.157347] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/22/2023] [Indexed: 02/24/2023] Open
Abstract
Targeting tumor-associated blood vessels to increase immune infiltration may enhance treatment effectiveness, yet limited data exist regarding anti-angiogenesis effects on the tumor microenvironment (TME). We hypothesized that dual targeting of angiogenesis with immune checkpoints would improve both intracranial and extracranial disease. We used subcutaneous and left ventricle melanoma models to evaluate anti-PD-1/anti-VEGF and anti-PD-1/lenvatinib (pan-VEGFR inhibitor) combinations. Cytokine/chemokine profiling and flow cytometry were performed to assess signaling and immune-infiltrating populations. An in vitro blood-brain barrier (BBB) model was utilized to study intracranial treatment effects on endothelial integrity and leukocyte transmigration. Anti-PD-1 with either anti-VEGF or lenvatinib improved survival and decreased tumor growth in systemic melanoma murine models; treatment increased Th1 cytokine/chemokine signaling. Lenvatinib decreased tumor-associated macrophages but increased plasmacytoid DCs early in treatment; this effect was not evident with anti-VEGF. Both lenvatinib and anti-VEGF resulted in decreased intratumoral blood vessels. Although anti-VEGF promoted endothelial stabilization in an in vitro BBB model, while lenvatinib did not, both regimens enabled leukocyte transmigration. The combined targeting of PD-1 and VEGF or its receptors promotes enhanced melanoma antitumor activity, yet their effects on the TME are quite different. These studies provide insights into dual anti-PD-1 and anti-angiogenesis combinations.
Collapse
Affiliation(s)
- Thuy T. Tran
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - Jasmine Caulfield
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - Lin Zhang
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - David Schoenfeld
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - Dijana Djureinovic
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - Veronica L. Chiang
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
- Yale School of Medicine, Department of Neurosurgery, New Haven, Connecticut, USA
| | - Victor Oria
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - Sarah A. Weiss
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - Kelly Olino
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
- Yale School of Medicine, Department of Surgery, New Haven, Connecticut, USA
| | - Lucia B. Jilaveanu
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| | - Harriet M. Kluger
- Yale School of Medicine and Yale Cancer Center, New Haven, Connecticut, USA
| |
Collapse
|
7
|
Bafaloukos D, Gazouli I, Koutserimpas C, Samonis G. Evolution and Progress of mRNA Vaccines in the Treatment of Melanoma: Future Prospects. Vaccines (Basel) 2023; 11:vaccines11030636. [PMID: 36992220 DOI: 10.3390/vaccines11030636] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/01/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
mRNA vaccines encoding tumor antigens may be able to sensitize the immune system of the host against cancer cells, enhancing antigen presentation and immune response. Since the breakout of the COVID19 pandemic, interest in mRNA vaccines has been accelerating, as vaccination against the virus served as a measure to limit disease spread. Given that immunotherapy has been the cornerstone of melanoma treatment over the last several decades, further innate immunity enhancement by targeted mRNA vaccines could be the next pivotal achievement in melanoma treatment. Preclinical data coming from murine cancer models have already provided evidence of mRNA vaccines’ ability to induce host immune responses against cancer. Moreover, specific immune responses have been observed in melanoma patients receiving mRNA vaccines, while the recent KEYNOTE-942 trial may establish the incorporation of the mRNA-4157/V940 vaccine into the melanoma treatment algorithm, in combination with immune checkpoint inhibition. As the existing data are further tested and reviewed, investigators are already gaining enthusiasm about this novel, promising pathway in cancer therapy.
Collapse
Affiliation(s)
- Dimitrios Bafaloukos
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, 18547 Attica, Greece
| | - Ioanna Gazouli
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, 18547 Attica, Greece
| | - Christos Koutserimpas
- Department of Orthopaedics and Traumatology, "251" Hellenic Air Force General Hospital of Athens, 11525 Athens, Greece
| | - George Samonis
- First Department of Medical Oncology, "Metropolitan" Hospital, Neon Faliron, 18547 Attica, Greece
- Department of Medicine, University of Crete, 71500 Heraklion, Greece
| |
Collapse
|
8
|
Heng Z, Zhao C, Gao Y. Comparison of urine proteomes from tumor-bearing mice with those from tumor-resected mice. PeerJ 2023; 11:e14737. [PMID: 36718454 PMCID: PMC9884041 DOI: 10.7717/peerj.14737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 12/22/2022] [Indexed: 01/26/2023] Open
Abstract
Objective This study aimed to address on the most important concern of surgeons-whether to completely resect tumor. Urine can indicate early changes associated with physiological or pathophysiological processes. Based on these ideas, we conducted experiments to explore changes in the urine proteome between tumor-bearing mice and tumor-resected mice. Method The tumor-bearing mouse model was established with MC38 mouse colon cancer cells, and the mice were divided into the control group, tumor-resected group, and tumor-bearing group. Urine was collected 7 and 30 days after tumor resection. Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) was used to identify the urine proteome, which was analyzed for differentially expressed proteins and functional annotation. Results (1) Seven days after tumor resection, 20 differentially expressed proteins distinguished the tumor-resected group and the tumor-bearing group. The identified biological processes included circadian rhythm, Notch signaling pathway, leukocyte cell-cell adhesion, and heterophilic cell-cell adhesion via plasma membrane cell adhesion molecules. (2) Thirty days after tumor resection, 33 differentially expressed proteins distinguished the tumor-resected group and the tumor-bearing group. The identified biological processes included cell adhesion; complement activation, the alternative pathway; the immune system process; and angiogenesis. (3) The difference in the urine proteome between the tumor-resected group and the healthy control group was smaller 30 days after tumor resection. Conclusion Changes in the urinary proteome can reflect the complete resection of MC38 tumors.
Collapse
Affiliation(s)
- Ziqi Heng
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, China
| | - Chenyang Zhao
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, China
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, College of Life Sciences, Beijing Normal University, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing, China
| |
Collapse
|
9
|
Denis M, Mathé D, Micoud M, Choffour PA, Grasselly C, Matera EL, Dumontet C. Impact of mouse model tumor implantation site on acquired resistance to anti-PD-1 immune checkpoint therapy. Front Immunol 2023; 13:1011943. [PMID: 36703964 PMCID: PMC9872099 DOI: 10.3389/fimmu.2022.1011943] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/20/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction The use of tumor subcutaneous (SC) implantations rather than orthotopic sites is likely to induce a significant bias, in particular, in the field of immunotherapy. Methods In this study, we developed and characterized MC38 models, implanted subcutaneously and orthotopically, which were either sensitive or rendered resistant to anti-PD1 therapy. We characterized the tumor immune infiltrate by flow cytometry at baseline and after treatment. Results and Discussion Our results demonstrate several differences between SC and orthotopic models at basal state, which tend to become similar after therapy. These results emphasize the need to take into account tumor implantation sites when performing preclinical studies with immunotherapeutic agents.
Collapse
Affiliation(s)
- Morgane Denis
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France,R&D Department, Antineo, Lyon, France
| | | | - Manon Micoud
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | | | - Chloé Grasselly
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Eva-Laure Matera
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Charles Dumontet
- Univ Lyon, Université Claude Bernard Lyon, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France,Hematology Department, Hospices Civils de Lyon, Lyon, France,*Correspondence: Charles Dumontet,
| |
Collapse
|
10
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 174:xv-xxv. [PMID: 37039770 DOI: 10.1016/s0091-679x(23)00036-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
11
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2023; 180:xv-xxv. [PMID: 37890936 DOI: 10.1016/s0091-679x(23)00166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023]
Affiliation(s)
- Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Jeffrey Kraynak
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Ariel E Marciscano
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - 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
|
12
|
Co-Targeting Luminal B Breast Cancer with S-Adenosylmethionine and Immune Checkpoint Inhibitor Reduces Primary Tumor Growth and Progression, and Metastasis to Lungs and Bone. Cancers (Basel) 2022; 15:cancers15010048. [PMID: 36612044 PMCID: PMC9818024 DOI: 10.3390/cancers15010048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BCa) is the most prevalent cancer in females and has a high rate of mortality, especially due to increased metastasis to skeletal and non-skeletal sites. Despite the marked clinical accomplishment of immune checkpoint inhibitor (CPI) therapy in patients with several cancers, it has had limited success in luminal subtypes of BCa. Accordingly, recent efforts have focused on combination therapy with CPI, including epigenetic modulators, to increase response rates of CPI in luminal BCa. We have previously shown that S-adenosylmethionine (SAM), the ubiquitous methyl donor, has strong anti-cancer effects in various cancers, including all subtypes of BCa. In the current study, we took a novel approach and examined the effect of CPI alone and in combination with SAM on tumor growth and metastasis in a syngeneic mouse model of luminal B BCa. We showed that SAM decreases cell proliferation, colony-formation (survival), and invasion of luminal B BCa cell lines (Eo771, R221A) in vitro. In in vivo studies, in Eo771 tumor-bearing mice, either SAM or anti-PD-1 antibody treatment alone significantly reduced tumor growth and progression, while the SAM+anti-PD-1 combination treatment had the highest anti-cancer efficacy of all groups. The SAM+anti-PD-1 combination reduced the percentage of animals with lung metastasis, as well as total metastatic lesion area, compared to control. Additionally, the SAM+anti-PD-1 combination significantly reduced the skeletal lesion area and protected tibial integrity to a greater extent than the monotherapies in an Eo771 bone metastasis model. Transcriptome analysis of Eo771 primary tumors revealed significant downregulation of pro-metastatic genes, including Matrix metalloproteinases (MMPs) and related pathways. On the other hand, CD8+ T cell infiltration, CD8+ T cell cytotoxicity (elevated granzymes), and immunostimulatory genes and pathways were significantly upregulated by the combination treatment. The results presented point to a combination of SAM with CPI as a possible treatment for luminal B BCa that should be tested in clinical studies.
Collapse
|
13
|
Tong L, Jiménez-Cortegana C, Tay AHM, Wickström S, Galluzzi L, Lundqvist A. NK cells and solid tumors: therapeutic potential and persisting obstacles. Mol Cancer 2022; 21:206. [PMID: 36319998 PMCID: PMC9623927 DOI: 10.1186/s12943-022-01672-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/10/2022] [Accepted: 09/24/2022] [Indexed: 11/05/2022] Open
Abstract
Natural killer (NK) cells, which are innate lymphocytes endowed with potent cytotoxic activity, have recently attracted attention as potential anticancer therapeutics. While NK cells mediate encouraging responses in patients with leukemia, the therapeutic effects of NK cell infusion in patients with solid tumors are limited. Preclinical and clinical data suggest that the efficacy of NK cell infusion against solid malignancies is hampered by several factors including inadequate tumor infiltration and persistence/activation in the tumor microenvironment (TME). A number of metabolic features of the TME including hypoxia as well as elevated levels of adenosine, reactive oxygen species, and prostaglandins negatively affect NK cell activity. Moreover, cancer-associated fibroblasts, tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells actively suppress NK cell-dependent anticancer immunity. Here, we review the metabolic and cellular barriers that inhibit NK cells in solid neoplasms as we discuss potential strategies to circumvent such obstacles towards superior therapeutic activity.
Collapse
Affiliation(s)
- Le Tong
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - Carlos Jiménez-Cortegana
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Department of Medical Biochemistry, Molecular Biology and Immunology, Faculty of Medicine, University of Seville, Seville, Spain
| | - Apple H M Tay
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Biological Science, Nanyang Technological University, Singapore, Singapore
| | - Stina Wickström
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | - 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.
| | - Andreas Lundqvist
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
14
|
Souza-Fonseca Guimaraes F, Rossi GR, Dagley LF, Foroutan M, McCulloch TR, Yousef J, Park HY, Gunter JH, Beavis PA, Lin CY, Hediyeh-Zadeh S, Camilleri T, Davis MJ, Huntington ND. TGF-β and CIS inhibition overcomes NK cell suppression to restore anti-tumor immunity. Cancer Immunol Res 2022; 10:1047-1054. [PMID: 35759796 DOI: 10.1158/2326-6066.cir-21-1052] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/21/2022] [Accepted: 06/23/2022] [Indexed: 11/16/2022]
Abstract
Antibodies targeting "immune checkpoints" have revolutionized cancer therapy by reactivating tumor-resident cytotoxic lymphocytes, primarily CD8+ T cells. Interest in targeting analogous pathways in other cytotoxic lymphocytes is growing. Natural killer (NK) cells are key to cancer immunosurveillance by eradicating metastases and driving solid tumor inflammation. NK cell anti-tumor function is dependent on the cytokine interleukin (IL)-15. Ablation of the IL-15 signaling inhibitor CIS (Cish) enhances NK cell anti-tumor immunity by increasing NK cell metabolism and persistence within the tumor microenvironment (TME). The TME has also been shown to impair NK cell fitness via the production of immunosuppressive TGF-β, a suppression which occurs even in the presence of high IL-15 signaling. Here, we identified an unexpected interaction between CIS and the TGF-β signaling pathway in NK cells. Independently, Cish- and Tgfbr2-deficient NK cells are both hyper-responsive to IL-15 and hypo-responsive to TGF-β, with dramatically enhanced anti-tumor immunity. Remarkably, when both these immunosuppressive genes are simultaneously deleted in NK cells, mice are largely resistant to tumor development, suggesting that combining suppression of these two pathways might represent a novel therapeutic strategy to enhance innate anti-cancer immunity.
Collapse
Affiliation(s)
| | | | - Laura F Dagley
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | | | | | - Jumana Yousef
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| | | | | | - Paul A Beavis
- Peter MacCallum Cancer Research Centre, Melbourne, Victoria, Australia
| | - Cheng-Yu Lin
- University of Queensland, Woolloongabba, QLD, Australia
| | | | | | - Melissa J Davis
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | | |
Collapse
|
15
|
Gkountakos A, Simbolo M, Bariani E, Scarpa A, Luchini C. Undifferentiated Sarcomatoid Carcinoma of the Pancreas: From Histology and Molecular Pathology to Precision Oncology. Int J Mol Sci 2022; 23:1283. [PMID: 35163206 PMCID: PMC8835772 DOI: 10.3390/ijms23031283] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/22/2022] [Accepted: 01/22/2022] [Indexed: 12/12/2022] Open
Abstract
Undifferentiated sarcomatoid carcinoma of the pancreas (SCP) is a rare and aggressive subtype of pancreatic cancer. Histologically, SCP is a poorly differentiated tumor characterized by the lack of glandular differentiation and the presence of mesenchymal-like, spindle-shaped tumor cells. Due to its rarity, only sporadic cases have been reported, while its molecular characterization has not been sufficiently described. Surgical resection with curative intent is the gold-standard of SCP management, but this strategy is possible only in a small proportion of cases due to SCP early metastasization. Although SCP is generally associated with a poor prognosis, some clinical cases amenable to surgical resection and followed by adjuvant chemotherapy have demonstrated a remarkably long survival. Preliminary molecular insights on the SCP molecular landscape have demonstrated the recurrent presence of KRAS and TP53 mutations, highlighting genetic similarities with conventional pancreatic ductal adenocarcinoma (PDAC). Although the use of immunotherapy in PDAC remains an unmet challenge, recent insights indicated a potentially significant role of the PD-L1/Notch3 axis in SCP, opening new horizons for immunotherapy in this cancer subtype. In this review, we described the most important clinic-pathologic features of SCP, with a specific focus on their molecular landscape and the potential targets for precision oncology.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
| | - Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| | - Elena Bariani
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| | - Aldo Scarpa
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| | - Claudio Luchini
- ARC-NET Applied Research on Cancer Center, University of Verona, 37134 Verona, Italy; (A.G.); (A.S.)
- Department of Diagnostics and Public Health, Section of Pathology, University of Verona, 37134 Verona, Italy; (M.S.); (E.B.)
| |
Collapse
|
16
|
Sato A, Kraynak J, Marciscano AE, Galluzzi L. Radiation therapy: An old dog learning new tricks. Methods Cell Biol 2022; 172:xiii-xxiii. [PMID: 36064230 PMCID: PMC10087864 DOI: 10.1016/s0091-679x(22)00139-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
17
|
Silvestris N, Argentiero A, Brunetti O, Sonnessa M, Colonna F, Delcuratolo S, Luchini C, Scarpa A, Lonardi S, Nappo F, Fassan M, Solimando AG, Fucci L, Saponaro C. PD-L1 and Notch as novel biomarkers in pancreatic sarcomatoid carcinoma: a pilot study. Expert Opin Ther Targets 2021; 25:1007-1016. [PMID: 34846251 DOI: 10.1080/14728222.2021.2011859] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 11/24/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND The improved immunological understanding revealed the tumor microenvironment as an appealing driver to restore the immune response against cancer cells resulting in a paradigm shift in the oncology field. However, the complexity of the tumor milieu suggests the role of several pathways linking in immunomodulation mechanisms. Pancreatic cancer represents a model of the intricate relationship between malignant cells and their surrounding neighborhood. RESEARCH DESIGN AND METHODS In this study, we analyzed, retrospectively, six cases of rare pancreatic sarcomatoid carcinoma (PSC) and evaluated the expression of PD-L1 and Notch, aiming to explore new attributes in immunophenotype. RESULTS PD-L1 CPS ≥ 1was common in PSCs (83%) with half samples expressing PD-L1 CPS ≥ 50. Notch1 and Notch3 demonstrated a high range of expression. A direct significant correlation between PD-L1 and Notch3 overexpression (r = 0.7; p = 0.036) has been observed. Immunofluorescence studies revealed a co-localization of Notch3 and PD-L1 when both proteins were over-expressed within cytoplasmic or membranous compartments of the same cells. CONCLUSIONS Our data identify a unique biological characterization of this rare pancreatic histotype. These findings provide a rationale for future studies evaluating the potential crosstalk between PD-L1/PD-1 axis and Notch pathways and prompting the development of novel therapeutics strategy.
Collapse
Affiliation(s)
- Nicola Silvestris
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Antonella Argentiero
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Oronzo Brunetti
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Margherita Sonnessa
- Functional Biomorphology Laboratory, Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Fulvia Colonna
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Sabina Delcuratolo
- Clinical Trial Office IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Claudio Luchini
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Enets Center of Excellence of Verona, Verona, Italy
| | - Aldo Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
- Enets Center of Excellence of Verona, Verona, Italy
- Arc-net Applied Research on Cancer Centre, University of Verona, Verona, Italy
| | - Sara Lonardi
- Early Phase Clinical Trial Unit, Department of Oncology, Veneto Institute of Oncology Iov-irccs, Padua, Italy
- Medical Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology Iov - Irccs, Padua, Italy
| | - Floriana Nappo
- Early Phase Clinical Trial Unit, Department of Oncology, Veneto Institute of Oncology Iov-irccs, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Matteo Fassan
- Department of Medicine (Dimed), Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
- Veneto Institute of Oncology Iov - Irccs, Padua, Italy
| | - Antonio Giovanni Solimando
- Medical Oncology Unit - IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Livia Fucci
- Pathology Department, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| | - Concetta Saponaro
- Functional Biomorphology Laboratory, Functional Biomorphology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo Ii" of Bari, Bari, Italy
| |
Collapse
|
18
|
Immunosuppressive Effects of Myeloid-Derived Suppressor Cells in Cancer and Immunotherapy. Cells 2021; 10:cells10051170. [PMID: 34065010 PMCID: PMC8150533 DOI: 10.3390/cells10051170] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/01/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
The primary function of myeloid cells is to protect the host from infections. However, during cancer progression or states of chronic inflammation, these cells develop into myeloid-derived suppressor cells (MDSCs) that play a prominent role in suppressing anti-tumor immunity. Overcoming the suppressive effects of MDSCs is a major hurdle in cancer immunotherapy. Therefore, understanding the mechanisms by which MDSCs promote tumor growth is essential for improving current immunotherapies and developing new ones. This review explores mechanisms by which MDSCs suppress T-cell immunity and how this impacts the efficacy of commonly used immunotherapies.
Collapse
|