1
|
Mivehchi H, Eskandari-Yaghbastlo A, Ghazanfarpour M, Ziaei S, Mesgari H, Faghihinia F, Zokaei Ashtiani N, Afjadi MN. Microenvironment-based immunotherapy in oral cancer: a comprehensive review. Med Oncol 2025; 42:140. [PMID: 40153139 DOI: 10.1007/s12032-025-02694-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
Oral cancer, a prevalent form of head and neck malignancy, accounts for 4% of global cancer cases. The most common type, oral squamous cell carcinoma (OSCC), has a survival rate of about 50%. Even though emerging molecular therapies show promise for managing oral cancer, current treatments like surgery, radiotherapy, and chemotherapy have significant side effects. In addition, the complex tumor microenvironment (TME), involving the extracellular matrix (ECM) and cells like fibroblasts and stromal cells like immune cells, promotes tumor growth and inhibits immune responses, complicating treatment. Nonetheless, immunotherapy is crucial in cancer treatment, especially in oral cancers. Indeed, its effectiveness lies in targeting immune checkpoints such as PD-1 and CTLA-4 inhibitors, as well as monoclonal antibodies like pembrolizumab and cetuximab, adoptive cell transfer methods (including CAR-T cell therapy), cytokine therapy such as IL-2, and tumor vaccines. Thus, these interventions collectively regulate tumor proliferation and metastasis by targeting the TME through autocrine-paracrine signaling pathways. Immunotherapy indeed aims to stimulate the immune system, leveraging both innate and adaptive immunity to counteract cancer cell signals and promote tumor destruction. This review will explore how the TME controls tumor proliferation and metastasis via autocrine-paracrine signaling pathways. It will then detail the effectiveness of immunotherapy in oral cancers, focusing on immune checkpoints, targeted monoclonal antibodies, adoptive cell transfer, cytokine therapy, and tumor vaccines.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - SeyedMehdi Ziaei
- Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Hung SK, Lee MS, Chiou WY, Liu DW, Yu CC, Chen LC, Lin RI, Chew CH, Hsu FC, Yang HJ, Chan MWY, Lin HY. Epigenetic modification in radiotherapy and immunotherapy for cancers. Tzu Chi Med J 2024; 36:396-406. [PMID: 39421493 PMCID: PMC11483092 DOI: 10.4103/tcmj.tcmj_3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/20/2024] [Accepted: 06/18/2024] [Indexed: 10/19/2024] Open
Abstract
Radiotherapy (RT) is one of the primary treatment modalities in managing cancer patients. Recently, combined RT and immunotherapy (IT) (i.e., radio-IT [RIT]) have been aggressively investigated in managing cancer patients. However, several issues in conducting RIT are challenging, such as incorporating advanced irradiation techniques, predictive/prognostic biomarkers, and other treatment modalities. Several clinical efforts and novel biomarkers have been introduced and developed to solve these challenges. For example, stereotactic radiosurgery/stereotactic radiotherapy, stereotactic body radiotherapy/stereotactic ablative body radiotherapy, and FLASH-RT have been applied for delivering precise irradiation to lung and liver tumors in conjunction with IT. Besides, several novel IT agents and incorporations of other therapies, such as targeted and thermal therapies, have been further investigated. The present study reviewed the emerging challenges of RIT in modern oncology. We also evaluated clinical practice, bench research, and multimodality treatments. In addition to several clinically applicable biomarkers, we emphasize the roles of advanced irradiation techniques and epigenetic modification as predictive/prognostic biomarkers and potential therapeutic targets. For example, 6(m) A-based epigenetic agents demonstrate the potential to enhance the treatment effects of RIT. However, further prospective randomized trials should be conducted to confirm their roles.
Collapse
Affiliation(s)
- Shih-Kai Hung
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Moon-Sing Lee
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Yen Chiou
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Dai-Wei Liu
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chih-Chia Yu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Liang-Cheng Chen
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ru-Inn Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chia-Hui Chew
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Feng-Chun Hsu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Hsuan-Ju Yang
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Michael W. Y. Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Hon-Yi Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
3
|
Tsarovsky N, Felder M, Heck M, Slowinski J, Rasmussen K, VandenHeuvel S, Zaborek J, Morris ZS, Erbe AK, Sondel PM, Rakhmilevich AL. Cyclophosphamide augments the efficacy of in situ vaccination in a mouse melanoma model. Front Oncol 2023; 13:1200436. [PMID: 37746303 PMCID: PMC10516537 DOI: 10.3389/fonc.2023.1200436] [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: 04/04/2023] [Accepted: 08/21/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction We have previously shown that an intratumoral (IT) injection of the hu14.18-IL2 immunocytokine (IC), an anti-GD2 antibody linked to interleukin 2, can serve as an in situ vaccine and synergize with local radiotherapy (RT) to induce T cell-mediated antitumor effects. We hypothesized that cyclophosphamide (CY), a chemotherapeutic agent capable of depleting T regulatory cells (Tregs), would augment in situ vaccination. GD2+ B78 mouse melanoma cells were injected intradermally in syngeneic C57BL/6 mice. Methods Treatments with RT (12Gy) and/or CY (100 mg/kg i.p.) started when tumors reached 100-300 mm3 (day 0 of treatment), followed by five daily injections of IT-IC (25 mcg) on days 5-9. Tumor growth and survival were followed. In addition, tumors were analyzed by flow cytometry. Results Similar to RT, CY enhanced the antitumor effect of IC. The strongest antitumor effect was achieved when CY, RT and IC were combined, as compared to combinations of IC+RT or IC+CY. Flow cytometric analyses showed that the combined treatment with CY, RT and IC decreased Tregs and increased the ratio of CD8+ cells/Tregs within the tumors. Moreover, in mice bearing two separate tumors, the combination of RT and IT-IC delivered to one tumor, together with systemic CY, led to a systemic antitumor effect detected as shrinkage of the tumor not treated directly with RT and IT-IC. Cured mice developed immunological memory as they were able to reject B78 tumor rechallenge. Conclusion Taken together, these preclinical results show that CY can augment the antitumor efficacy of IT- IC, given alone or in combination with local RT, suggesting potential benefit in clinical testing of these combinations.
Collapse
Affiliation(s)
- Noah Tsarovsky
- Department of Human Oncology, Madison, WI, United States
| | - Mildred Felder
- Department of Human Oncology, Madison, WI, United States
| | - Mackenzie Heck
- Department of Human Oncology, Madison, WI, United States
| | | | | | | | - Jen Zaborek
- Department of Biostatistics and Medical Informatics, Madison, WI, United States
| | - Zachary S. Morris
- Department of Human Oncology, Madison, WI, United States
- Paul P. Carbone Comprehensive Cancer Center, Madison, WI, United States
| | - Amy K. Erbe
- Department of Human Oncology, Madison, WI, United States
- Paul P. Carbone Comprehensive Cancer Center, Madison, WI, United States
| | - Paul M. Sondel
- Department of Human Oncology, Madison, WI, United States
- Paul P. Carbone Comprehensive Cancer Center, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin, Madison, WI, United States
| | - Alexander L. Rakhmilevich
- Department of Human Oncology, Madison, WI, United States
- Paul P. Carbone Comprehensive Cancer Center, Madison, WI, United States
| |
Collapse
|
4
|
Affolter A, Liebel K, Tengler L, Seiz E, Tiedtke M, Azhakesan A, Schütz J, Theodoraki MN, Kern J, Ruder AM, Fleckenstein J, Weis CA, Bieback K, Kramer B, Lammert A, Scherl C, Rotter N, Ludwig S. Modulation of PD‑L1 expression by standard therapy in head and neck cancer cell lines and exosomes. Int J Oncol 2023; 63:102. [PMID: 37503786 PMCID: PMC10552694 DOI: 10.3892/ijo.2023.5550] [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: 11/25/2022] [Accepted: 05/15/2023] [Indexed: 07/29/2023] Open
Abstract
Although checkpoint inhibitors (CPI) have recently extended the treatment options and improved clinical response of advanced stage head and neck squamous cell carcinoma (HNSCC), treatment success remains unpredictable. Programmed cell death ligand‑1 (PD‑L1) is a key player in immunotherapy. Tumor cells, and exosomes derived therefrom, are carriers of PD‑L1 and efficiently suppress immune responses. The aim of the present study was to analyze the influence of established therapies on PD‑L1 expression of HNSCC cell lines and their exosomes. The HNSCC cell lines, UM‑SCC‑11B, UM‑SCC‑14C and UM‑SCC‑22C were treated with fractionated radiotherapy (RT; 5x2 Gy), cisplatin (CT) and cetuximab (Cetux) as monotherapy, or combined therapy, chemoradiotherapy (CRT; RT and CT) or radioimmunotherapy (RT and Cetux). The expression of PD‑L1 and phosphorylated (p)ERK1/2 as a mediator of radioresistance were assessed using western blotting, immunohistochemistry and an ex vivo vital tissue culture model. Additionally, exosomes were isolated from concentrated supernatants of the (un‑)treated HNSCC cell lines by size exclusion chromatography. Exosomal protein expression levels of PD‑L1 were detected using western blotting and semi‑quantitative levels were calculated. The functional impact of exosomes from the (un‑)treated HNSCC cell lines on the proliferation (MTS assay) and apoptosis (Caspase 3/7 assay) of the untreated HNSCC cell lines were measured and compared. The HNSCC cell lines UM‑SCC‑11B and UM‑SCC‑22B showed strong expression of pERK1/2 and PD‑L1, respectively. RT upregulated the PD‑L1 expression in UM‑SCC‑11B and UM‑SCC‑14C and in exosomes from all three cell lines. CT alone induced PD‑L1 expression in all cell lines. CRT induced the expression of PD‑L1 in all HNSCC cell lines and exosomes from UM‑SCC‑14C and UM‑SCC‑22B. The data indicated a potential co‑regulation of PD‑L1 and activated ERK1/2, most evident in UM‑SCC‑14C. Exosomes from irradiated UM‑SCC‑14C cells protected the unirradiated cells from apoptosis by Caspase 3/7 downregulation. The present study suggested a tumor cell‑mediated regulation of PD‑L1 upon platinum‑based CRT in HNSCC and in exosomes. A co‑regulation of PD‑L1 and MAPK signaling response was hypothesized.
Collapse
Affiliation(s)
- Annette Affolter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Kai Liebel
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Luisa Tengler
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Elena Seiz
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Moritz Tiedtke
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Alexya Azhakesan
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Julia Schütz
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Marie-Nicole Theodoraki
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Ulm, D-89075 Ulm
| | - Johann Kern
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Arne M. Ruder
- Department of Radiation Oncology, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
- Department of Radiation Oncology, University Hospital Heidelberg, D-69120 Heidelberg
| | - Jens Fleckenstein
- Department of Radiation Oncology, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Cleo-Aron Weis
- Department of Pathology, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, Mannheim
- Department of Pathology, Heidelberg University Hospital
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, D-68167 Mannheim, Germany
| | - Benedikt Kramer
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Anne Lammert
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Claudia Scherl
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Nicole Rotter
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| | - Sonja Ludwig
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Mannheim, Medical Faculty Mannheim of Heidelberg University, D-68167 Mannheim
| |
Collapse
|
5
|
Zhang W, Kong X, Ai B, Wang Z, Wang X, Wang N, Zheng S, Fang Y, Wang J. Research Progresses in Immunological Checkpoint Inhibitors for Breast Cancer Immunotherapy. Front Oncol 2021; 11:582664. [PMID: 34631507 PMCID: PMC8495193 DOI: 10.3389/fonc.2021.582664] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 08/12/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor immune escape refers to the phenomenon in which tumor cells escape the recognition and attack of the body’s immune system through various mechanisms so that they can survive and proliferate in vivo. The imbalance of immune checkpoint protein expression is the primary mechanism for breast cancer to achieve immune escape. Cytotoxic T lymphocyte antigen 4 (CTLA4) and programmed cell death protein 1 (PD-1)/programmed cell death protein-ligand 1 (PD-L1) are critical immune checkpoints for breast cancer. Immune checkpoint inhibitors block the checkpoint and relieve its inhibition effect on immune cells, reactivate T-cells and destroy cancer cells and restore the body’s ability to resist tumors. At present, immunological checkpoint inhibitors have made significant progress in breast cancer immunotherapy, and it is expected to become a new treatment for breast cancer.
Collapse
Affiliation(s)
- Wenxiang Zhang
- Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Bolun Ai
- Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Xiangyu Wang
- Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Nianchang Wang
- Department of Cancer Prevention, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Shan Zheng
- Department of Pathology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Yi Fang
- Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| | - Jing Wang
- Department of Breast Surgical Oncology, China National Cancer Center/Cancer Hospital, Chinese Academy of Medical and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Varayathu H, Sarathy V, Thomas BE, Mufti SS, Naik R. Combination Strategies to Augment Immune Check Point Inhibitors Efficacy - Implications for Translational Research. Front Oncol 2021; 11:559161. [PMID: 34123767 PMCID: PMC8193928 DOI: 10.3389/fonc.2021.559161] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 04/30/2021] [Indexed: 12/22/2022] Open
Abstract
Immune checkpoint inhibitor therapy has revolutionized the field of cancer immunotherapy. Even though it has shown a durable response in some solid tumors, several patients do not respond to these agents, irrespective of predictive biomarker (PD-L1, MSI, TMB) status. Multiple preclinical, as well as early-phase clinical studies are ongoing for combining immune checkpoint inhibitors with anti-cancer and/or non-anti-cancer drugs for beneficial therapeutic interactions. In this review, we discuss the mechanistic basis behind the combination of immune checkpoint inhibitors with other drugs currently being studied in early phase clinical studies including conventional chemotherapy drugs, metronomic chemotherapy, thalidomide and its derivatives, epigenetic therapy, targeted therapy, inhibitors of DNA damage repair, other small molecule inhibitors, anti-tumor antibodies hormonal therapy, multiple checkpoint Inhibitors, microbiome therapeutics, oncolytic viruses, radiotherapy, drugs targeting myeloid-derived suppressor cells, drugs targeting Tregs, drugs targeting renin-angiotensin system, drugs targeting the autonomic nervous system, metformin, etc. We also highlight how translational research strategies can help better understand the true therapeutic potential of such combinations.
Collapse
Affiliation(s)
- Hrishi Varayathu
- Department of Translational Medicine and Therapeutics, HealthCare Global Enterprises Limited, Bangalore, India
| | - Vinu Sarathy
- Department of Medical Oncology, HealthCare Global Enterprises Limited, Bangalore, India
| | - Beulah Elsa Thomas
- Department of Clinical Pharmacology, HealthCare Global Enterprises Limited, Bangalore, India
| | - Suhail Sayeed Mufti
- Department of Translational Medicine and Therapeutics, HealthCare Global Enterprises Limited, Bangalore, India
| | - Radheshyam Naik
- Department of Medical Oncology, HealthCare Global Enterprises Limited, Bangalore, India
| |
Collapse
|
7
|
Merlano MC, Abbona A, Paccagnella M, Falletta A, Granetto C, Ricci V, Fea E, Denaro N, Ruatta F, Merlotti A, Bertetto O, Crosetto N, Galizia D, Basiricò M, Gammaitoni L, Sangiolo D, Aglietta M, Garrone O. Cytokine Profiling of End Stage Cancer Patients Treated with Immunotherapy. Vaccines (Basel) 2021; 9:vaccines9030235. [PMID: 33800511 PMCID: PMC7999072 DOI: 10.3390/vaccines9030235] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 01/22/2023] Open
Abstract
Published data suggest that immunotherapy plays a role even in patients with very advanced tumours. We investigated the immune profile of end-stage cancer patients treated with immunotherapy to identify changes induced by treatment. Breast, colon, renal and prostate cancer patients were eligible. Treatment consisted of metronomic cyclophosphamide, low-dose interleukin-2 (IL-2) and a single radiation shot. A panel of 16 cytokines was assessed using automated ELISA before treatment (T0), after radiation (RT; T1), at cycle 2 (T2) and at disease progression (TPD). Receiving operating characteristic (ROC) analysis was used to identify cytokine cut-off related to overall survival (OS). Principal component analysis (PCA) was used to identify the immune profile correlating better with OS and progression-free survival. Twenty-three patients were enrolled. High IL-2, low IL-8 and CCL-2 correlated with OS. The PCA identified a cluster of patients, with high IL-2, IL-12 and IFN-γ levels at T0 having longer PFS and OS. In all cohorts, IL-2 and IL-5 increased from T0 to T2; a higher CCL-4 level compared to T2 and a higher IL-8 level compared to T0 were found at TPD. The progressive increase of the IL-10 level during treatment negatively correlated with OS. Our data suggested that baseline cytokine levels may predict patients’ outcome and that the treatment may affect their kinetic even in end-stage patients. Cytokine profiling of end-stage patients might offer a tool for medical decisions (EUDRACT: 2016-000578-39).
Collapse
Affiliation(s)
- Marco Carlo Merlano
- Experimental Cell Therapy Lab, Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy; (L.G.); (D.S.); (M.A.)
- Correspondence: or ; Tel.: +39-0171-616342
| | - Andrea Abbona
- Translational Oncology, ARCO Foundation, 12100 Cuneo, Italy; (A.A.); (M.P.); (A.F.); (O.G.)
| | - Matteo Paccagnella
- Translational Oncology, ARCO Foundation, 12100 Cuneo, Italy; (A.A.); (M.P.); (A.F.); (O.G.)
| | - Antonella Falletta
- Translational Oncology, ARCO Foundation, 12100 Cuneo, Italy; (A.A.); (M.P.); (A.F.); (O.G.)
| | - Cristina Granetto
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (C.G.); (V.R.); (E.F.); (N.D.); (F.R.)
| | - Vincenzo Ricci
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (C.G.); (V.R.); (E.F.); (N.D.); (F.R.)
| | - Elena Fea
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (C.G.); (V.R.); (E.F.); (N.D.); (F.R.)
| | - Nerina Denaro
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (C.G.); (V.R.); (E.F.); (N.D.); (F.R.)
| | - Fiorella Ruatta
- Department of Medical Oncology, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy; (C.G.); (V.R.); (E.F.); (N.D.); (F.R.)
| | - Anna Merlotti
- Department of Radiotherapy, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy;
| | - Oscar Bertetto
- Rete Oncologica del Piemonte e Della Valle d’Aosta, 10125 Turin, Italy;
| | - Nicola Crosetto
- Department of Medical Biochemistry and Biophysiscs, Karolinska Institute, 17177 Stockholm, Sweden;
| | - Danilo Galizia
- Multidisciplinary Oncology Outpatient Clinic, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy;
| | - Marco Basiricò
- Department of Oncology, University of Turin, 10043 Turin, Italy;
| | - Loretta Gammaitoni
- Experimental Cell Therapy Lab, Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy; (L.G.); (D.S.); (M.A.)
| | - Dario Sangiolo
- Experimental Cell Therapy Lab, Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy; (L.G.); (D.S.); (M.A.)
- Department of Oncology, University of Turin, 10043 Turin, Italy;
| | - Massimo Aglietta
- Experimental Cell Therapy Lab, Department of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy; (L.G.); (D.S.); (M.A.)
- Department of Oncology, University of Turin, 10043 Turin, Italy;
| | - Ornella Garrone
- Translational Oncology, ARCO Foundation, 12100 Cuneo, Italy; (A.A.); (M.P.); (A.F.); (O.G.)
- Breast Unit, S. Croce e Carle Teaching Hospital, 12100 Cuneo, Italy
| |
Collapse
|
8
|
Kok VC. Current Understanding of the Mechanisms Underlying Immune Evasion From PD-1/PD-L1 Immune Checkpoint Blockade in Head and Neck Cancer. Front Oncol 2020; 10:268. [PMID: 32185135 PMCID: PMC7058818 DOI: 10.3389/fonc.2020.00268] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/17/2020] [Indexed: 02/05/2023] Open
Abstract
Starting in 2014, large phase III clinical trials began to disclose the study results of using programmed death (PD)-1 immune checkpoint inhibitors (ICIs) (pembrolizumab, nivolumab) and PD-ligand (L)1 (atezolizumab, durvalumab, avelumab) ICIs immunotherapy in patients with advanced head and neck squamous cell carcinoma (HNSCC). In the recurrent and metastatic (R/M), cisplatin-refractory setting, nivolumab achieved a 2.2-fold increase of the median 1-year overall survival as compared with investigators' choice of salvage chemotherapy (36.0 vs. 16.6%). A paradigm shift to the winning regimen, pembrolizumab combined with platinum and infusional fluorouracil, has outperformed the past gold standard of cetuximab-based platinum and fluorouracil combination in terms of overall survival (median, 13.6 vs. 10.1 mo) when administered as the first-line treatment for R/M HNSCC. Nevertheless, many patients still did not respond to the PD-1/PD-L1 checkpoint inhibitor treatment, indicating innate, adapted, or quickly acquired resistance to the immunotherapy. The mechanisms of resistance to ICIs targeting the PD-1/PD-L1 signaling pathway in the context of HNSCC are the focus of this review. The past 5 years have seen improved understanding of the mechanisms underlying checkpoint inhibition resistance in tumor cells, such as: tumor cell adaption with malfunction of the antigen-presenting machinery via class I human leukocyte antigen (HLA), reintroduction of cyclin D-cyclin-dependent kinase (CDK) 4 complex to cell cycles, enrichment of CD44+ cancer stem-like cells, or development of inactivating mutation in IKZF1 gene; impairment of T-cell functions and proliferation through mutations in the interferon-γ-regulating genes, suppression of the stimulator of interferon genes (STING) pathway, or resulted from constitutional nutritional iron deficiency state; metabolic reprogramming by cancer cells with changes in metabolites such as GTP cyclohydrolase 1, tetrahydrobiopterin, kynurenine, indoleamine 2,3-dioxygenase, and arginase 1; defective dendritic cells, CD-69 sufficient state; and the upregulation or activation of the alternative immune checkpoints, including lymphocyte activation gene-3 (LAG3), T-cell immunoglobulin and ITIM domain (TIGIT)/CD155 pathway, T-cell immunoglobulin mucin-3 (TIM-3), and V domain-containing Ig suppressor of T-cell activation (VISTA). Several potential biomarkers or biosignatures, which could predict the response or resistance to the PD-1/PD-L1 checkpoint immunotherapy, are also discussed.
Collapse
Affiliation(s)
- Victor C. Kok
- Department of Medical Oncology, Kuang Tien General Hospital Cancer Center, Taichung, Taiwan
- Department of Bioinformatics and Medical Engineering, Asia University Taiwan, Taichung, Taiwan
| |
Collapse
|
9
|
Bailly C, Thuru X, Quesnel B. Combined cytotoxic chemotherapy and immunotherapy of cancer: modern times. NAR Cancer 2020; 2:zcaa002. [PMID: 34316682 PMCID: PMC8209987 DOI: 10.1093/narcan/zcaa002] [Citation(s) in RCA: 145] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Monoclonal antibodies targeting programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) immune checkpoints have improved the treatments of cancers. However, not all patients equally benefit from immunotherapy. The use of cytotoxic drugs is practically inevitable to treat advanced cancers and metastases. The repertoire of cytotoxics includes 80 products that principally target nucleic acids or the microtubule network in rapidly proliferating tumor cells. Paradoxically, many of these compounds tend to become essential to promote the activity of immunotherapy and to offer a sustained therapeutic effect. We have analyzed each cytotoxic drug with respect to effect on expression and function of PD-(L)1. The major cytotoxic drugs—carboplatin, cisplatin, cytarabine, dacarbazine, docetaxel, doxorubicin, ecteinascidin, etoposide, fluorouracil, gemcitabine, irinotecan, oxaliplatin, paclitaxel and pemetrexed—all have the capacity to upregulate PD-L1 expression on cancer cells (via the generation of danger signals) and to promote antitumor immunogenicity, via activation of cytotoxic T lymphocytes, maturation of antigen-presenting cells, depletion of immunosuppressive regulatory T cells and/or expansion of myeloid-derived suppressor cells. The use of ‘immunocompatible’ cytotoxic drugs combined with anti-PD-(L)1 antibodies is a modern approach, not only for increasing the direct killing of cancer cells, but also as a strategy to minimize the activation of immunosuppressive and cancer cell prosurvival program responses.
Collapse
Affiliation(s)
| | - Xavier Thuru
- Centre de Recherche Jean-Pierre Aubert, INSERM, University of Lille, UMR-S 1172, CHU Lille, 59045 Lille, France
| | - Bruno Quesnel
- Centre de Recherche Jean-Pierre Aubert, INSERM, University of Lille, UMR-S 1172, CHU Lille, 59045 Lille, France
| |
Collapse
|
10
|
Cristina V, Herrera-Gómez RG, Szturz P, Espeli V, Siano M. Immunotherapies and Future Combination Strategies for Head and Neck Squamous Cell Carcinoma. Int J Mol Sci 2019; 20:E5399. [PMID: 31671550 PMCID: PMC6862353 DOI: 10.3390/ijms20215399] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 10/26/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is often diagnosed at an advanced stage and has a dismal prognosis. Nearly 10 years after the approval of cetuximab, anti-PD1/PD-L1 checkpoint inhibitors are the first drugs that have shown any survival benefit for the treatment on platinum-refractory recurrent/metastatic (R/M) HNSCC. Furthermore, checkpoint inhibitors are better tolerated than chemotherapy. The state of the art in the treatment of R/M HNSCC is changing, thanks to improved results for checkpoint inhibitors. Results for these treatments are also awaited in curative settings and for locally advanced HNSCC. Unfortunately, the response rate of immunotherapy is low. Therefore, the identification of predictive biomarkers of response and resistance to anti-PD1/PD-L1 is a key point for better selecting patients that would benefit the most from immunotherapy. Furthermore, the combination of checkpoint inhibitors with various agents is being currently evaluated to improve the response rate, prolong response duration, and even increase the chances for a cure. In this review, we summarize the most important results regarding immune targeting agents for HNSCC, predictive biomarkers for resistance to immune therapies, and future perspectives.
Collapse
Affiliation(s)
- Valerie Cristina
- Oncology Department, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland.
| | | | - Petr Szturz
- Oncology Department, Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland.
| | - Vittoria Espeli
- Oncology Department, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland.
| | - Marco Siano
- Interdisciplinary Cancer Service-SIC, Hôpital Riviera-Chablais, 1847 Rennaz, Switzerland.
- Faculty of Medicine, University and Unive rsity Hospital of Zurich, 8032 Zurich, Switzerland.
| |
Collapse
|
11
|
Longo V, Brunetti O, Azzariti A, Galetta D, Nardulli P, Leonetti F, Silvestris N. Strategies to Improve Cancer Immune Checkpoint Inhibitors Efficacy, Other Than Abscopal Effect: A Systematic Review. Cancers (Basel) 2019; 11:cancers11040539. [PMID: 30991686 PMCID: PMC6521062 DOI: 10.3390/cancers11040539] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/27/2022] Open
Abstract
Despite that the impact of immune checkpoint inhibitors on malignancies treatment is unprecedented, a lack of response to these molecules is observed in several cases. Differently from melanoma and non-small cell lung cancer, where the use of immune checkpoint inhibitors results in a high efficacy, the response rate in other tumors, such as gastrointestinal cancers, breast cancer, sarcomas, and part of genitourinary cancers remains low. The first strategy evaluated to improve the response rate to immune checkpoint inhibitors is the use of predictive factors for the response such as PD-L1 expression, tumor mutational burden, and clinical features. In addition to the identification of the patients with a higher expression of immune checkpoint molecules, another approach currently under intensive investigation is the use of therapeutics in a combinatory manner with immune checkpoint inhibitors in order to obtain an enhancement of efficacy through the modification of the tumor immune microenvironment. In addition to the abscopal effect induced by radiotherapy, a lot of studies are evaluating several drugs able to improve the response rate to immune checkpoint inhibitors, including microbiota modifiers, drugs targeting co-inhibitory receptors, anti-angiogenic therapeutics, small molecules, and oncolytic viruses. In view of the rapid and extensive development of this research field, we conducted a systematic review of the literature identifying which of these drugs are closer to achieving validation in the clinical practice.
Collapse
Affiliation(s)
- Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124 Bari, Italy.
| | - Oronzo Brunetti
- Medical Oncology Unit, Hospital of Barletta, Viale Ippocrate, 15, 70051 Barletta, Italy.
| | - Amalia Azzariti
- Experimental Pharmacology Laboratory, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124 Bari, Italy.
| | - Domenico Galetta
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124 Bari, Italy.
| | - Patrizia Nardulli
- Pharmacy Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124 Bari, Italy.
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze del Farmaco, University of Bari, Piazza Umberto I, 1, 70121 Bari, Italy.
| | - Nicola Silvestris
- Scientific Guidance, IRCCS Istituto Tumori "Giovanni Paolo II", Viale Orazio Flacco, 65, 70124 Bari, Italy.
| |
Collapse
|
12
|
Shinde A, Novak J, Freeman ML, Glaser S, Amini A. Induction of the Abscopal Effect with Immunotherapy and Palliative Radiation in Metastatic Head and Neck Squamous Cell Carcinoma: A Case Report and Review of the Literature. Cureus 2019; 11:e4201. [PMID: 31114720 PMCID: PMC6505722 DOI: 10.7759/cureus.4201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The induction of the abscopal effect using immunotherapy and radiation is under investigation through case reports and institutional studies. We describe a case of the abscopal effect with a combination of ipilimumab, nivolumab, and palliative radiation, in a patient with metastatic head and neck squamous cell carcinoma (mHNSCC).
Collapse
Affiliation(s)
- Ashwin Shinde
- Radiation Oncology, City of Hope National Medical Center, Duarte, USA
| | - Jennifer Novak
- Radiation Oncology, City of Hope National Medical Center, Duarte, USA
| | | | - Scott Glaser
- Radiation Oncology, City of Hope National Medical Center, Duarte, USA
| | - Arya Amini
- Radiation Oncology, City of Hope National Medical Center, Duarte, USA
| |
Collapse
|