1
|
Saeidpour Masouleh S, Nasiri K, Ostovar Ravari A, Saligheh Rad M, Kiani K, Sharifi Sultani A, Nejati ST, Nabi Afjadi M. Advances and challenges in CAR-T cell therapy for head and neck squamous cell carcinoma. Biomark Res 2025; 13:69. [PMID: 40312353 PMCID: PMC12044960 DOI: 10.1186/s40364-025-00783-1] [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: 02/08/2025] [Accepted: 04/24/2025] [Indexed: 05/03/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) remains among the most aggressive malignancies with limited treatment options, especially in recurrent and metastatic cases. Despite advances in surgery, radiotherapy, chemotherapy, and immune checkpoint inhibitors, survival rates remain suboptimal due to tumor heterogeneity, immune evasion, and treatment resistance. In recent years, Chimeric Antigen Receptor (CAR) T-cell therapy has revolutionized hematologic cancer treatment by genetically modifying T cells to target tumor-specific antigens like CD19, CD70, BCMA, EGFR, and HER2, leading to high remission rates. Its success is attributed to precise antigen recognition, sustained immune response, and long-term immunological memory, though challenges like cytokine release syndrome and antigen loss remain. Notably, its translation to solid tumors, including HNSCC, faces significant challenges, such as tumor microenvironment (TME)-induced immunosuppression, antigen heterogeneity, and limited CAR T-cell infiltration. To address these barriers, several tumor-associated antigens (TAAs), including EGFR, HER2 (ErbB2), B7-H3, CD44v6, CD70, CD98, and MUC1, have been identified as potential CAR T-cell targets in HNSCC. Moreover, innovative approaches, such as dual-targeted CAR T-cells, armored CARs, and CRISPR-engineered modifications, aim to enhance efficacy and overcome resistance. Notably, combination therapies integrating CAR T-cells with immune checkpoint inhibitors (e.g., PD-1/CTLA-4 blockade) and TGF-β-resistant CAR T designs are being explored to improve therapeutic outcomes. This review aimed to elucidate the current landscape of CAR T-cell therapy in HNSCC, by exploring its mechanisms, targeted antigens, challenges, emerging strategies, and future therapeutic potential.
Collapse
Affiliation(s)
| | - Kamyar Nasiri
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Ava Ostovar Ravari
- Faculty of Dentistry, Haybusak University of Medical Sciences, Yerevan, Armenia
| | - Mona Saligheh Rad
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | - Kiarash Kiani
- Faculty of Dentistry, Islamic Azad University of Medical Sciences, Tehran, Iran
| | | | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
2
|
Hu C, Liu M, Li Y, Zhao Y, Sharma A, Liu H, Schmidt-Wolf IGH. Recent advances and future perspectives of CAR-T cell therapy in head and neck cancer. Front Immunol 2023; 14:1213716. [PMID: 37457699 PMCID: PMC10346844 DOI: 10.3389/fimmu.2023.1213716] [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: 04/28/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023] Open
Abstract
Head and neck cancer (HNC) ranks as the sixth most prevalent type of cancer globally and accounts for about 4% of all types of cancer. Among all HNC, most are head and neck squamous cell carcinoma (HNSCC) with clinical therapies that include surgery, radiation therapy, chemotherapy, immunotherapy, targeted therapy, and multimodal treatments. In recent years, chimeric antigen receptor (CAR)-T cell immunotherapy has significantly transformed the therapeutic approaches for leukemia and lymphoma and has garnered increased attention as a potential treatment for a wide range of cancers. However, CAR-T immunotherapy in solid tumors, especially HNSCCs, lags significantly behind due to the paucity of tumor-specific antigens, high levels of tumor heterogeneity, immunosuppressive tumor microenvironment, the risk of treatment-related toxicities and off-target adverse events in HNSCCs. The objective of this review is to explore the advancement of CAR-T cell therapy in the treatment of HNSCCs. We aim to outline the targeted antigens in HNSCCs, highlight the challenges and potential solutions, and discuss the relevant combination therapies. Our review presents a comprehensive overview of the recent developments in CAR-T cell therapy for HNSCCs, and provides valuable insights into future research avenues.
Collapse
Affiliation(s)
- Chunmei Hu
- Department of Otolaryngology-Head and Neck Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Liu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yutao Li
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| | - Yi Zhao
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Haotian Liu
- Department of Otolaryngology-Head and Neck Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ingo G. H. Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, Bonn, Germany
| |
Collapse
|
3
|
Papa S, Adami A, Metoudi M, Beatson R, George MS, Achkova D, Williams E, Arif S, Reid F, Elstad M, Beckley-Hoelscher N, Douri A, Delord M, Lyne M, Shivapatham D, Fisher C, Hope A, Gooljar S, Mitra A, Gomm L, Morton C, Henley-Smith R, Thavaraj S, Santambrogio A, Andoniadou C, Allen S, Gibson V, Cook GJR, Parente-Pereira AC, Davies DM, Farzaneh F, Schurich A, Guerrero-Urbano T, Jeannon JP, Spicer J, Maher J. Intratumoral pan-ErbB targeted CAR-T for head and neck squamous cell carcinoma: interim analysis of the T4 immunotherapy study. J Immunother Cancer 2023; 11:e007162. [PMID: 37321663 PMCID: PMC10277526 DOI: 10.1136/jitc-2023-007162] [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: 05/26/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Locally advanced/recurrent head and neck squamous cell carcinoma (HNSCC) is associated with significant morbidity and mortality. To target upregulated ErbB dimer expression in this cancer, we developed an autologous CD28-based chimeric antigen receptor T-cell (CAR-T) approach named T4 immunotherapy. Patient-derived T-cells are engineered by retroviral transduction to coexpress a panErbB-specific CAR called T1E28ζ and an IL-4-responsive chimeric cytokine receptor, 4αβ, which allows IL-4-mediated enrichment of transduced cells during manufacture. These cells elicit preclinical antitumor activity against HNSCC and other carcinomas. In this trial, we used intratumoral delivery to mitigate significant clinical risk of on-target off-tumor toxicity owing to low-level ErbB expression in healthy tissues. METHODS We undertook a phase 1 dose-escalation 3+3 trial of intratumoral T4 immunotherapy in HNSCC (NCT01818323). CAR T-cell batches were manufactured from 40 to 130 mL of whole blood using a 2-week semiclosed process. A single CAR T-cell treatment, formulated as a fresh product in 1-4 mL of medium, was injected into one or more target lesions. Dose of CAR T-cells was escalated in 5 cohorts from 1×107-1×109 T4+ T-cells, administered without prior lymphodepletion. RESULTS Despite baseline lymphopenia in most enrolled subjects, the target cell dose was successfully manufactured in all cases, yielding up to 7.5 billion T-cells (67.5±11.8% transduced), without any batch failures. Treatment-related adverse events were all grade 2 or less, with no dose-limiting toxicities (Common Terminology Criteria for Adverse Events V.4.0). Frequent treatment-related adverse events were tumor swelling, pain, pyrexias, chills, and fatigue. There was no evidence of leakage of T4+ T-cells into the circulation following intratumoral delivery, and injection of radiolabeled cells demonstrated intratumoral persistence. Despite rapid progression at trial entry, stabilization of disease (Response Evaluation Criteria in Solid Tumors V.1.1) was observed in 9 of 15 subjects (60%) at 6 weeks post-CAR T-cell administration. Subsequent treatment with pembrolizumab and T-VEC oncolytic virus achieved a rapid complete clinical response in one subject, which was durable for over 3 years. Median overall survival was greater than for historical controls. Disease stabilization was associated with the administration of an immunophenotypically fitter, less exhausted, T4 CAR T-cell product. CONCLUSIONS These data demonstrate the safe intratumoral administration of T4 immunotherapy in advanced HNSCC.
Collapse
Affiliation(s)
- Sophie Papa
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Antonella Adami
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Michael Metoudi
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Richard Beatson
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Molly Sarah George
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Daniela Achkova
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Evangelia Williams
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Sefina Arif
- Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Fiona Reid
- School of Life Course & Population Sciences, King's College London, London, UK
| | - Maria Elstad
- School of Life Course & Population Sciences, King's College London, London, UK
| | - Nicholas Beckley-Hoelscher
- Department of Biostatistics and Health Informatics, Institute of Psychiatry Psychology & Neuroscience, King's College London, London, UK
| | - Abdel Douri
- School of Life Course & Population Sciences, King's College London, London, UK
| | - Marc Delord
- School of Life Course & Population Sciences, King's College London, London, UK
| | - Mike Lyne
- Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Dharshene Shivapatham
- Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Christopher Fisher
- Good Manufacturing Practice Unit, Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Andrew Hope
- Good Manufacturing Practice Unit, Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Sakina Gooljar
- Good Manufacturing Practice Unit, Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Arindam Mitra
- Good Manufacturing Practice Unit, Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Linda Gomm
- Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Cienne Morton
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Guy's and St Thomas' Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| | - Rhonda Henley-Smith
- Head and Neck Pathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Selvam Thavaraj
- Head and Neck Pathology, Guy's and St Thomas' NHS Foundation Trust, London, UK
- Faculty of Dentistry, Oral and Craniofacial Sciences, Guy's Hospital, King's College London, London, UK
| | - Alice Santambrogio
- Faculty of Dentistry, Oral and Craniofacial Sciences, Guy's Hospital, King's College London, London, UK
| | - Cynthia Andoniadou
- Faculty of Dentistry, Oral and Craniofacial Sciences, Guy's Hospital, King's College London, London, UK
| | - Sarah Allen
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Victoria Gibson
- Department of Nuclear Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Gary J R Cook
- London School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | | | - David M Davies
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Farzin Farzaneh
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Anna Schurich
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Teresa Guerrero-Urbano
- Department of Head and Neck Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Jean-Pierre Jeannon
- Department of Head and Neck Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - James Spicer
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - John Maher
- School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
- Department of Immunology, Eastbourne Hospital, Eastbourne, UK
- Leucid Bio Ltd, London, London, UK
| |
Collapse
|
4
|
Ramírez-Chacón A, Betriu-Méndez S, Bartoló-Ibars A, González A, Martí M, Juan M. Ligand-based CAR-T cell: Different strategies to drive T cells in future new treatments. Front Immunol 2022; 13:932559. [PMID: 36172370 PMCID: PMC9511026 DOI: 10.3389/fimmu.2022.932559] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-based therapies are presented as innovative treatments for multiple malignancies. Despite their clinical success, there is scientific evidence of the limitations of these therapies mainly due to immunogenicity issues, toxicities associated with the infusion of the product, and relapses of the tumor. As a result, novel approaches are appearing aiming to solve and/or mitigate the harmful effects of CAR-T therapies. These include strategies based on the use of ligands as binding moieties or ligand-based CAR-T cells. Several proposals are currently under development, with some undergoing clinical trials to assess their potential benefits. In addition to these, therapies such as chimeric autoantibody receptor (CAAR), B-cell receptor antigen for reverse targeting (BAR), and even chimeric human leukocyte antigen (HLA) antibody receptor (CHAR) have emerged, benefiting from the advantages of antigenic ligands as antibody-binding motifs. This review focuses on the potential role that ligands can play in current and future antitumor treatments and in other types of diseases, such as autoimmune diseases or problems associated with transplantation.
Collapse
Affiliation(s)
- Alejandro Ramírez-Chacón
- Immunology Unit, Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Laboratory of Cellular Immunology, Institute of Biotechnology and Biomedicine (IBB), Cerdanyola del Vallès, Spain
| | - Sergi Betriu-Méndez
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
| | - Ariadna Bartoló-Ibars
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
| | - Azucena González
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
- Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mercè Martí
- Immunology Unit, Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Laboratory of Cellular Immunology, Institute of Biotechnology and Biomedicine (IBB), Cerdanyola del Vallès, Spain
| | - Manel Juan
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
- Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- *Correspondence: Manel Juan,
| |
Collapse
|
5
|
Deshet-Unger N, Horn G, Rawet-Slobodkin M, Waks T, Laskov I, Michaan N, Raz Y, Bar V, Zundelevich A, Aharon S, Turovsky L, Mallel G, Salpeter S, Neev G, Hollander KS, Katz BZ, Grisaru D, Globerson Levin A. Comparing Intraperitoneal and Intravenous Personalized ErbB2CAR-T for the Treatment of Epithelial Ovarian Cancer. Biomedicines 2022; 10:biomedicines10092216. [PMID: 36140319 PMCID: PMC9496506 DOI: 10.3390/biomedicines10092216] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 12/24/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most common type of epithelial ovarian cancer. The majority of cases are diagnosed at advanced stages, when intraperitoneal (IP) spread has already occurred. Despite significant surgical and chemotherapeutic advances in HGSOC treatment over the past decades, survival rates with HGSOC have only modestly improved. Chimeric antigen receptor (CAR)-T cells enable T cells to directly bind to tumor-associated antigens in a major histocompatibility complex-independent manner, thereby inducing tumor rejection. While CAR-T cell therapy shows great promise in hematological malignancies, its use in solid tumors is limited. Therefore, innovative approaches are needed to increase the specificity of CAR-modified T cells against solid tumors. The aim of this study was to assess the efficacy and safety of intraperitoneal (IP) versus intravenous (IV) CAR-T cell therapy in the treatment of HGSOC. We constructed a CAR that targets the ErbB2/HER2 protein (ErbB2CAR), which is overexpressed in HGSOC, and evaluated the functionality of ErbB2CAR on ovarian cancer cell lines (OVCAR8, SKOV3, and NAR). Our findings show that an IP injection of ErbB2CAR-T cells to tumor-bearing mice led to disease remission and increased survival compared to the IV route. Moreover, we found that IP-injected ErbB2CART cells circulate to a lesser extent, making them safer for non-tumor tissues than IV-injected cells. Further supporting our findings, we show that the effect of ErbB2CAR-T cells on primary HGSOC tumors is correlated with ErbB2 expression. Together, these data demonstrate the advantages of an IP administration of CAR-T cells over IV administration, offering not only a safer strategy but also the potential for counteracting the effect of ErbB2CAR in HGSOC. Significance: IP-injected ErbB2CAR-T cells led to disease remission and increased survival compared to the IV route. These findings demonstrate the advantages of IP administration, offering a safe treatment strategy with the potential for counteracting the effect of ErbB2CAR in HGSOC.
Collapse
Affiliation(s)
- Naamit Deshet-Unger
- Immunology and Advanced CAR-T Therapy, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
| | - Galit Horn
- Immunology and Advanced CAR-T Therapy, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
| | - Moran Rawet-Slobodkin
- Immunology and Advanced CAR-T Therapy, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
| | - Tova Waks
- Immunology and Advanced CAR-T Therapy, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
- Department of Immunology, The Weizmann Institute, Rehovot 7610001, Israel
| | - Ido Laskov
- Department of Gynecologic Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Nadav Michaan
- Department of Gynecologic Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Yael Raz
- Department of Gynecologic Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Vered Bar
- cResponce Company, Rehovot 7670102, Israel
| | | | | | | | | | | | - Guy Neev
- cResponce Company, Rehovot 7670102, Israel
| | - Kenneth Samuel Hollander
- Immunology and Advanced CAR-T Therapy, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
| | - Ben-Zion Katz
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- The Hematology Laboratory, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6997801, Israel
| | - Dan Grisaru
- Department of Gynecologic Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Anat Globerson Levin
- Immunology and Advanced CAR-T Therapy, Tel-Aviv Sourasky Medical Center, Tel-Aviv 6423906, Israel
- Dotan Center for Advanced Therapies, Tel-Aviv Sourasky Medical Center, Tel-Aviv University, Tel-Aviv 6423906, Israel
- Correspondence: ; Tel.: +972-3-6972503
| |
Collapse
|
6
|
Cavalieri S, Filippini DM, Ottini A, Bergamini C, Resteghini C, Colombo E, Lombardo R, Nuzzolese I, Alfieri S, Licitra L, Locati LD. Immunotherapy in head and neck squamous cell carcinoma and rare head and neck malignancies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:522-542. [PMID: 36046116 PMCID: PMC9400733 DOI: 10.37349/etat.2021.00062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/29/2021] [Indexed: 11/19/2022] Open
Abstract
The dismal prognosis of recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) prompted recent advances in the field of therapeutic approaches beyond cytotoxic cancer therapy. In recent years, the deeper and increasing knowledge on the genomic landscape and the upcoming new data on immunotherapy enacted by HNSCCs have led to successful therapeutic targeting of the immune system. Immune checkpoint inhibitors (ICIs) have changed state of the art in R/M patients and could have a potential role even in early disease. The purpose of this work is to summarize the role of immunotherapy for R/M HNSCC in clinical practice, with insights about future perspectives. Updated immunotherapy results in other R/M head and neck cancers such as thyroid, salivary glands, nasopharynx, sinonasal cancers, and nuclear protein in testis (NUT) are presented.
Collapse
Affiliation(s)
- Stefano Cavalieri
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Daria Maria Filippini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Arianna Ottini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Cristiana Bergamini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Carlo Resteghini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Elena Colombo
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Roberta Lombardo
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Imperia Nuzzolese
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Salvatore Alfieri
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Lisa Licitra
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Laura D. Locati
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| |
Collapse
|
7
|
Short Review on Advances in Hydrogel-Based Drug Delivery Strategies for Cancer Immunotherapy. Tissue Eng Regen Med 2021; 19:263-280. [PMID: 34596839 DOI: 10.1007/s13770-021-00369-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy has become the new paradigm of cancer treatment. The introduction and discovery of various therapeutic agents have also accelerated the application of immunotherapy in clinical trials. However, despite the significant potency and demonstrated advantages of cancer immunotherapy, its clinical application to patients faces several safety and efficacy issues, including autoimmune reactions, cytokine release syndrome, and vascular leak syndrome-related issues. In addressing these problems, biomaterials traditionally used for tissue engineering and drug delivery are attracting attention. Among them, hydrogels can be easily injected into tumors with drugs, and they can minimize side effects by retaining immune therapeutics at the tumor site for a long time. This article reviews the status of functional hydrogels for effective cancer immunotherapy. First, we describe the basic mechanisms of cancer immunotherapy and the advantages of using hydrogels to apply these mechanisms. Next, we summarize recent advances in the development of functional hydrogels designed to locally release various immunotherapeutic agents, including cytokines, cancer immune vaccines, immune checkpoint inhibitors, and chimeric antigen receptor-T cells. Finally, we briefly discuss the current problems and possible prospects of hydrogels for effective cancer immunotherapy.
Collapse
|
8
|
Glover M, Avraamides S, Maher J. How Can We Engineer CAR T Cells to Overcome Resistance? Biologics 2021; 15:175-198. [PMID: 34040345 PMCID: PMC8141613 DOI: 10.2147/btt.s252568] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved unrivalled success in the treatment of B cell and plasma cell malignancies, with five CAR T cell products now approved by the US Food and Drug Administration (FDA). However, CAR T cell therapies for solid tumours have not been nearly as successful, owing to several additional challenges. Here, we discuss mechanisms of tumour resistance in CAR T cell therapy and the emerging strategies that are under development to engineer CAR T cells to overcome resistance.
Collapse
Affiliation(s)
- Maya Glover
- Leucid Bio Ltd., Guy's Hospital, London, SE1 9RT, UK
| | - Stephanie Avraamides
- King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, SE1 9RT, UK
| | - John Maher
- Leucid Bio Ltd., Guy's Hospital, London, SE1 9RT, UK.,King's College London, School of Cancer and Pharmaceutical Sciences, Guy's Hospital, London, SE1 9RT, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, SE5 9RS, UK.,Department of Immunology, Eastbourne Hospital, Eastbourne, East Sussex, BN21 2UD, UK
| |
Collapse
|
9
|
Guo F, Cui J. CAR-T in solid tumors: Blazing a new trail through the brambles. Life Sci 2020; 260:118300. [DOI: 10.1016/j.lfs.2020.118300] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/31/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
|
10
|
Benard E, Casey NP, Inderberg EM, Wälchli S. SJI 2020 special issue: A catalogue of Ovarian Cancer targets for CAR therapy. Scand J Immunol 2020; 92:e12917. [PMID: 32557659 DOI: 10.1111/sji.12917] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/09/2020] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Ovarian Cancer (OC) is currently difficult to cure, mainly due to its late detection and the advanced state of the disease at the time of diagnosis. Therefore, conventional treatments such as debulking surgery and combination chemotherapy are rarely able to control progression of the tumour, and relapses are frequent. Alternative therapies are currently being evaluated, including immunotherapy and advanced T cell-based therapy. In the present review, we will focus on a description of those Chimeric Antigen Receptors (CARs) that have been validated in the laboratory or are being tested in the clinic. Numerous target antigens have been defined due to the identification of OC biomarkers, and many are being used as CAR targets. We provide an exhaustive list of these constructs and their current status. Despite being innovative and efficient, the OC-specific CARs face a barrier to their clinical efficacy: the tumour microenvironment (TME). Indeed, effector cells expressing CARs have been shown to be severely inhibited, rendering the CAR T cells useless once at the tumour site. Herein, we give a thorough description of the highly immunosuppressive OC TME and present recent studies and innovations that have enabled CAR T cells to counteract this negative environment and to destroy tumours.
Collapse
Affiliation(s)
- Emmanuelle Benard
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Nicholas P Casey
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Else Marit Inderberg
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section for Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
11
|
Editor's Pick: Tumour-Associated Hypoxia: Can We Give Chimeric Antigen Receptor T Cells More Breathing Space? EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR)-engineered T cells has encountered important limitations in the transition of their use from liquid to solid tumours. Success is dependent upon T-cell trafficking to, and functional persistence within, tumours that often present a metabolically and immunologically hostile microenvironment. Moreover, CAR targets that are tumour specific are extremely scarce. To address these issues, several strategies have been proposed to improve both tumour selectivity and safety. One approach involves the engineering of CAR-T cells that only deploy their effector function at tumour sites. Conceptually, a solution for this exploits the oxygen-limited nature of advanced tumour deposits through the engineering of CAR that are exclusively expressed or activated under conditions of profound hypoxia. T cells have a complex inter-relationship with oxygen, which also needs to be factored into the refinement of these technologies. Ideally, oxygen-sensing CAR should only function when oxygen tension is below 2%, as is commonly the case in solid tumours but rare in healthy tissue. Successful advancement of such technologies presents opportunities for solid tumour immunotherapy because it should broaden the target repertoire that may safely be exploited in this context.
Collapse
|
12
|
The Landscape of CAR-T Cell Clinical Trials against Solid Tumors-A Comprehensive Overview. Cancers (Basel) 2020; 12:cancers12092567. [PMID: 32916883 PMCID: PMC7563774 DOI: 10.3390/cancers12092567] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/03/2020] [Accepted: 09/04/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Certain immune cells, namely T cells, of cancer patients can be genetically manipulated to express so-called chimeric antigen receptors (CARs), which enables these cells to kill the tumor cells after recognition by the receptor. This therapy is very successful in the treatment of hematologic tumors such as lymphoma or leukemia. However, tumors growing as a solid mass are less susceptible to this kind of treatment. This review summarizes known data of all clinical trials using this therapy against solid tumors that are registered at clinicaltrials.gov. Abstract CAR-T cells showed great potential in the treatment of patients with hematologic tumors. However, the clinical efficacy of CAR-T cells against solid tumors lags behind. To obtain a comprehensive overview of the landscape of CAR-T cell clinical trials against this type of cancer, this review summarizes all the 196 studies registered at clinicaltrials.gov. Special focus is on: (1) geographical distribution; (2) targeted organs, tumor entities, and antigens; (3) CAR transfer methods, CAR formats, and extra features introduced into the T cells; and (4) patient pretreatments, injection sites, and safety measurements. Finally, the few data on clinical outcome are reported. The last assessment of clinicaltrials.gov for the data summarized in this paper was on 4 August 2020.
Collapse
|
13
|
Abdou P, Wang Z, Chen Q, Chan A, Zhou DR, Gunadhi V, Gu Z. Advances in engineering local drug delivery systems for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1632. [PMID: 32255276 PMCID: PMC7725287 DOI: 10.1002/wnan.1632] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aims to leverage the immune system to suppress the growth of tumors and to inhibit metastasis. The recent promising clinical outcomes associated with cancer immunotherapy have prompted research and development efforts towards enhancing the efficacy of immune checkpoint blockade, cancer vaccines, cytokine therapy, and adoptive T cell therapy. Advancements in biomaterials, nanomedicine, and micro-/nano-technology have facilitated the development of enhanced local delivery systems for cancer immunotherapy, which can enhance treatment efficacy while minimizing toxicity. Furthermore, locally administered cancer therapies that combine immunotherapy with chemotherapy, radiotherapy, or phototherapy have the potential to achieve synergistic antitumor effects. Herein, the latest studies on local delivery systems for cancer immunotherapy are surveyed, with an emphasis on the therapeutic benefits associated with the design of biomaterials and nanomedicines. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peter Abdou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Road, Suzhou, 215123, Jiangsu, PR China
| | - Amanda Chan
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Daojia R. Zhou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Vivienne Gunadhi
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
14
|
Gershovich PM, Karabelskii AV, Ulitin AB, Ivanov RA. The Role of Checkpoint Inhibitors and Cytokines in Adoptive Cell-Based Cancer Immunotherapy with Genetically Modified T Cells. BIOCHEMISTRY (MOSCOW) 2019; 84:695-710. [PMID: 31509722 DOI: 10.1134/s0006297919070022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This review focuses on the structure and molecular action mechanisms of chimeric antigen receptors (CARs) and major aspects of the manufacturing and clinical application of products for the CAR-T (CAR-modified T lymphocyte) therapy of hematological and solid tumors with special emphasis on the strategies for combined use of CAR-T therapy with immuno-oncological monoclonal antibodies (checkpoint inhibitors) and cytokines to boost survival, persistence, and antitumor efficacy of CAR-T therapy. The review also summarizes preclinical and clinical data on the additive effects of the combined use of CAR-T therapy with interleukins and monoclonal antibodies targeting immune checkpoints.
Collapse
Affiliation(s)
- P M Gershovich
- CJSC Biocad, St. Petersburg, 198515, Russia. .,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A V Karabelskii
- CJSC Biocad, St. Petersburg, 198515, Russia.,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A B Ulitin
- CJSC Biocad, St. Petersburg, 198515, Russia
| | - R A Ivanov
- CJSC Biocad, St. Petersburg, 198515, Russia
| |
Collapse
|
15
|
Nitinol thin films functionalized with CAR-T cells for the treatment of solid tumours. Nat Biomed Eng 2019; 4:195-206. [PMID: 31819155 DOI: 10.1038/s41551-019-0486-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 11/07/2019] [Indexed: 12/11/2022]
Abstract
Micropatterned nickel titanium (commonly known as nitinol) thin films with complex designs, high structural resolution and excellent biocompatibility can be cheaply fabricated using magnetron sputtering. Here, we show that these benefits can be leveraged to fabricate micromesh implants that are loaded with tumour-specific human chimeric antigen receptor (CAR)-T cells for the treatment of solid tumours. In a mouse model of non-resectable ovarian cancer, the cell-loaded nitinol thin films spatially conformed to the implantation site, fostered the rapid expansion of T cells, delivered a high density of T cells directly to the tumour and significantly improved animal survival. We also show that self-expandable stents that were coated with T-cell-loaded films and implanted into subcutaneous tumours in mice improved the duration of stent patency by delaying tumour ingrowth. By providing direct access to tumours, CAR-T-cell-loaded micropatterned nitinol thin films can improve the effects of cell-based therapies.
Collapse
|
16
|
Springuel L, Lonez C, Alexandre B, Van Cutsem E, Machiels JPH, Van Den Eynde M, Prenen H, Hendlisz A, Shaza L, Carrasco J, Canon JL, Opyrchal M, Odunsi K, Rottey S, Gilham DE, Flament A, Lehmann FF. Chimeric Antigen Receptor-T Cells for Targeting Solid Tumors: Current Challenges and Existing Strategies. BioDrugs 2019; 33:515-537. [PMID: 31363930 PMCID: PMC6790340 DOI: 10.1007/s40259-019-00368-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chimeric antigen receptor-T cells (CAR-Ts) are an exciting new cancer treatment modality exemplified by the recent regulatory approval of two CD19-targeted CAR-T therapies for certain B cell malignancies. However, this success in the hematological setting has yet to translate to a significant level of objective clinical responses in the solid tumor setting. The reason for this lack of translation undoubtedly lies in the substantial challenges raised by solid tumors to all therapies, including CAR-T, that differ from B cell malignancies. For instance, intravenously infused CAR-Ts are likely to make rapid contact with cancerous B cells since both tend to reside in the same vascular compartments within the body. By contrast, solid cancers tend to form discrete tumor masses with an immune-suppressive tumor microenvironment composed of tumor cells and non-tumor stromal cells served by abnormal vasculature that restricts lymphocyte infiltration and suppresses immune function, expansion, and persistence. Moreover, the paucity of uniquely and homogeneously expressed tumor antigens and inherent plasticity of cancer cells provide major challenges to the specificity, potency, and overall effectiveness of CAR-T therapies. This review focuses on the major preclinical and clinical strategies currently being pursued to tackle these challenges in order to drive the success of CAR-T therapy against solid tumors.
Collapse
Affiliation(s)
| | | | | | | | | | - Marc Van Den Eynde
- Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Hans Prenen
- University Hospital Antwerp (UZ Antwerp), Antwerp, Belgium
| | - Alain Hendlisz
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | - Leila Shaza
- Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | - Kunle Odunsi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | | | | | | | | |
Collapse
|
17
|
Li D, Li X, Zhou WL, Huang Y, Liang X, Jiang L, Yang X, Sun J, Li Z, Han WD, Wang W. Genetically engineered T cells for cancer immunotherapy. Signal Transduct Target Ther 2019; 4:35. [PMID: 31637014 PMCID: PMC6799837 DOI: 10.1038/s41392-019-0070-9] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
T cells in the immune system protect the human body from infection by pathogens and clear mutant cells through specific recognition by T cell receptors (TCRs). Cancer immunotherapy, by relying on this basic recognition method, boosts the antitumor efficacy of T cells by unleashing the inhibition of immune checkpoints and expands adaptive immunity by facilitating the adoptive transfer of genetically engineered T cells. T cells genetically equipped with chimeric antigen receptors (CARs) or TCRs have shown remarkable effectiveness in treating some hematological malignancies, although the efficacy of engineered T cells in treating solid tumors is far from satisfactory. In this review, we summarize the development of genetically engineered T cells, outline the most recent studies investigating genetically engineered T cells for cancer immunotherapy, and discuss strategies for improving the performance of these T cells in fighting cancers.
Collapse
Affiliation(s)
- Dan Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xue Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Wei-Lin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Yong Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xiao Liang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
- Department of Medical Oncology, Cancer Center, West China Hospital, West China Medical School, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Lin Jiang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Xiao Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| | - Jie Sun
- Department of Cell Biology, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, 310058 Zhejiang, China
- Institute of Hematology, Zhejiang University & Laboratory of Stem cell and Immunotherapy Engineering, 310058 Zhejing, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, 200032 Shanghai, China
- CARsgen Therapeutics, 200032 Shanghai, China
| | - Wei-Dong Han
- Molecular & Immunological Department, Biotherapeutic Department, Chinese PLA General Hospital, No. 28 Fuxing Road, 100853 Beijing, China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and the Collaborative Innovation Center for Biotherapy, 610041 Chengdu, China
| |
Collapse
|
18
|
Rotolo R, Leuci V, Donini C, Cykowska A, Gammaitoni L, Medico G, Valabrega G, Aglietta M, Sangiolo D. CAR-Based Strategies beyond T Lymphocytes: Integrative Opportunities for Cancer Adoptive Immunotherapy. Int J Mol Sci 2019; 20:ijms20112839. [PMID: 31212634 PMCID: PMC6600566 DOI: 10.3390/ijms20112839] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/27/2019] [Accepted: 05/28/2019] [Indexed: 12/26/2022] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T lymphocytes (CAR Ts) produced impressive clinical results against selected hematological malignancies, but the extension of CAR T cell therapy to the challenging field of solid tumors has not, so far, replicated similar clinical outcomes. Many efforts are currently dedicated to improve the efficacy and safety of CAR-based adoptive immunotherapies, including application against solid tumors. A promising approach is CAR engineering of immune effectors different from αβT lymphocytes. Herein we reviewed biological features, therapeutic potential, and safety of alternative effectors to conventional CAR T cells: γδT, natural killer (NK), NKT, or cytokine-induced killer (CIK) cells. The intrinsic CAR-independent antitumor activities, safety profile, and ex vivo expansibility of these alternative immune effectors may favorably contribute to the clinical development of CAR strategies. The proper biological features of innate immune response effectors may represent an added value in tumor settings with heterogeneous CAR target expression, limiting the risk of tumor clonal escape. All these properties bring out CAR engineering of alternative immune effectors as a promising integrative option to be explored in future clinical studies.
Collapse
Affiliation(s)
- Ramona Rotolo
- Department of Oncology, University of Torino, 10140 Torino, Italy.
| | - Valeria Leuci
- Department of Oncology, University of Torino, 10140 Torino, Italy.
- Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo TO, Italy.
| | - Chiara Donini
- Department of Oncology, University of Torino, 10140 Torino, Italy.
| | - Anna Cykowska
- Department of Oncology, University of Torino, 10140 Torino, Italy.
| | | | - Giovanni Medico
- Department of Oncology, University of Torino, 10140 Torino, Italy.
| | - Giorgio Valabrega
- Department of Oncology, University of Torino, 10140 Torino, Italy.
- Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo TO, Italy.
| | - Massimo Aglietta
- Department of Oncology, University of Torino, 10140 Torino, Italy.
- Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo TO, Italy.
| | - Dario Sangiolo
- Department of Oncology, University of Torino, 10140 Torino, Italy.
- Candiolo Cancer Institute FPO-IRCCS, 10060 Candiolo TO, Italy.
| |
Collapse
|
19
|
Abstract
BACKGROUND Melanoma-associated antigen-A (MAGE-A) was recognized as high-expressed in many solid tumors including esophageal carcinoma (EC), nevertheless, was reported to be low/not-expressed in normal tissues. Thus, it was considered as an extraordinary appropriate target for treatment especially in immunotherapy. Therefore, it demanded more detail knowledge on the precise function of MAGE-A. METHODS In this study, we used the data from the Cancer Genome Atlas dataset (TCGA-ESCA) to analyze the expression and survival for MAGE A3/4/11 (the subtype of MAGE-A) using the online tool of UALCAN. Furthermore, the high-throughput sequencing data of the patients with esophageal squamous-cell carcinoma (ESCC) from TCGA dataset were performed to analyze the correlation test, gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment of MAGE A3/4/9/11 using LinkeDomics (online tool) and ClueGO (inner software of Cytoscape). Finally, relative gene expressions of MAGE A3/4/9/11 were verified by quantitative real-time PCR (q-PCR) in the patients with EC. RESULTS MAGE A3/4/11 was high-expressed in tissues of patients with ESCC, and there was no difference in survival time for patients between the high-expressed with the low/medium-expressed. The Go enrichment analysis showed that the 4 MAGE-A subtypes (MAGE-A3/4/9/11) were enriched in the regulation of the adaptive immune response, translational initiation, interleukin-4 production, response to type I interferon, and skin development, respectively. The KEGG results showed that they were enriched in T cell receptor signaling pathway (MAGE-A3), Th1 and Th2 differentiation, antigen processing and presentation (MAGE-A4), cytokine-cytokine receptor interaction (MAGE-A9), and chemokine signaling pathway (MAGE-A11). CONCLUSION MAGE A3/4/9/11 was high-expressed in EC, and were enrolled in the regulation of immune response. They may consider as candidate immune target for EC treatment and provided the messages for further research in the function of MAGE-A.
Collapse
Affiliation(s)
- Xiaohua Chen
- Oncology of Panyu Central Hospital, Panyu Cancer Institute
| | - Sina Cai
- Oncology of The Hospital of Third Affiliated Southern Medical University, Guangzhou, Guangdong
| | - Liping Wang
- The First People's Hospital of Chenzhou, Chenzhou, Hunan
| | - Xiaona Zhang
- Graceland Medical Center, The Sixth Affiliated Hospital of Sun Yat–Sen University, Guangzhou, Guangdong, China
| | - Wenhui Li
- Oncology of Panyu Central Hospital, Panyu Cancer Institute
| | - Xiaolong Cao
- Oncology of Panyu Central Hospital, Panyu Cancer Institute
| |
Collapse
|
20
|
Mirzaei HR, Mirzaei H, Namdar A, Rahmati M, Till BG, Hadjati J. Predictive and therapeutic biomarkers in chimeric antigen receptor T‐cell therapy: A clinical perspective. J Cell Physiol 2018; 234:5827-5841. [DOI: 10.1002/jcp.27519] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Hamid Reza Mirzaei
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology School of Medicine, Mashhad University of Medical Sciences Mashahd Iran
| | - Afshin Namdar
- Department of Dentistry Faculty of Medicine and Dentistry, University of Alberta Edmonton Canada
| | - Majid Rahmati
- Cancer Prevention Research Center Shahroud University of Medical Sciences Shahroud Iran
| | - Brian G. Till
- Clinical Research Division Fred Hutchinson Cancer Research Center Seattle WA United States
| | - Jamshid Hadjati
- Department of Medical Immunology School of Medicine, Tehran University of Medical Sciences Tehran Iran
| |
Collapse
|
21
|
Ajina A, Maher J. Strategies to Address Chimeric Antigen Receptor Tonic Signaling. Mol Cancer Ther 2018; 17:1795-1815. [PMID: 30181329 PMCID: PMC6130819 DOI: 10.1158/1535-7163.mct-17-1097] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/19/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
Abstract
Adoptive cell transfer using chimeric antigen receptors (CAR) has emerged as one of the most promising new therapeutic modalities for patients with relapsed or refractory B-cell malignancies. Thus far, results in patients with advanced solid tumors have proven disappointing. Constitutive tonic signaling in the absence of ligand is an increasingly recognized complication when deploying these synthetic fusion receptors and can be a cause of poor antitumor efficacy, impaired survival, and reduced persistence in vivo In parallel, ligand-dependent tonic signaling can mediate toxicity and promote T-cell anergy, exhaustion, and activation-induced cell death. Here, we review the mechanisms underpinning CAR tonic signaling and highlight the wide variety of effects that can emerge after making subtle structural changes or altering the methodology of CAR transduction. We highlight strategies to prevent unconstrained tonic signaling and address its deleterious consequences. We also frame this phenomenon in the context of endogenous TCR tonic signaling, which has been shown to regulate peripheral tolerance, facilitate the targeting of foreign antigens, and suggest opportunities to coopt ligand-dependent CAR tonic signaling to facilitate in vivo persistence and efficacy. Mol Cancer Ther; 17(9); 1795-815. ©2018 AACR.
Collapse
MESH Headings
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Adam Ajina
- CAR Mechanics Group, King's College London, London, United Kingdom.
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
| | - John Maher
- CAR Mechanics Group, King's College London, London, United Kingdom
- School of Cancer and Pharmaceutical Studies, Guy's Hospital, London, United Kingdom
- Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, London, United Kingdom
- Department of Immunology, Eastbourne Hospital, East Sussex, United Kingdom
| |
Collapse
|
22
|
Murad JM, Graber DJ, Sentman CL. Advances in the use of natural receptor- or ligand-based chimeric antigen receptors (CARs) in haematologic malignancies. Best Pract Res Clin Haematol 2018; 31:176-183. [PMID: 29909918 DOI: 10.1016/j.beha.2018.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/12/2018] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptors (CAR)-T cell therapy has recently made promising advances towards treatment of B-cell malignancies. This approach makes use of an antibody-derived single chain variable fragment (scFv)-based CAR to target the CD19 antigen. Currently scFvs are the most common strategy for creation of CARs, but tumor cells can also be targeted using non-antibody based approaches with designs focused on the interaction between natural receptors and their ligands. This emerging strategy has been used in unique ways to target multiple tumor types, including solid and haematological malignancies. In this review, we will highlight the performance of receptor-ligand combinations as designs for CARs to treat cancer, with a particular focus on haematologic malignancies.
Collapse
Affiliation(s)
- Joana M Murad
- Celdara Medical LLC, Lebanon, NH, 16 Cavendish Ct Suite 240, Lebanon, NH 03766, USA.
| | - David J Graber
- Center for Synthetic Immunity and Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Dr., Lebanon, NH 03765, USA.
| | - Charles L Sentman
- Center for Synthetic Immunity and Department of Microbiology & Immunology, Geisel School of Medicine at Dartmouth, One Medical Center Dr., Lebanon, NH 03765, USA.
| |
Collapse
|
23
|
Huang Y, Li D, Qin DY, Gou HF, Wei W, Wang YS, Wei YQ, Wang W. Interleukin-armed chimeric antigen receptor-modified T cells for cancer immunotherapy. Gene Ther 2017; 25:192-197. [DOI: 10.1038/gt.2017.81] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 04/10/2017] [Accepted: 07/28/2017] [Indexed: 01/01/2023]
|
24
|
Thayaparan T, Petrovic RM, Achkova DY, Zabinski T, Davies DM, Klampatsa A, Parente-Pereira AC, Whilding LM, van der Stegen SJ, Woodman N, Sheaff M, Cochran JR, Spicer JF, Maher J. CAR T-cell immunotherapy of MET-expressing malignant mesothelioma. Oncoimmunology 2017; 6:e1363137. [PMID: 29209570 DOI: 10.1080/2162402x.2017.1363137] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/24/2017] [Accepted: 07/31/2017] [Indexed: 12/21/2022] Open
Abstract
Mesothelioma is an incurable cancer for which effective therapies are required. Aberrant MET expression is prevalent in mesothelioma, although targeting using small molecule-based therapeutics has proven disappointing. Chimeric antigen receptors (CARs) couple the HLA-independent binding of a cell surface target to the delivery of a tailored T-cell activating signal. Here, we evaluated the anti-tumor activity of MET re-targeted CAR T-cells against mesothelioma. Using immunohistochemistry, MET was detected in 67% of malignant pleural mesotheliomas, most frequently of epithelioid or biphasic subtype. The presence of MET did not influence patient survival. Candidate MET-specific CARs were engineered in which a CD28+CD3ζ endodomain was fused to one of 3 peptides derived from the N and K1 domains of hepatocyte growth factor (HGF), which represents the minimum MET binding element present in this growth factor. Using an NIH3T3-based artificial antigen-presenting cell system, we found that all 3 candidate CARs demonstrated high specificity for MET. By contrast, these CARs did not mediate T-cell activation upon engagement of other HGF binding partners, namely CD44v6 or heparan sulfate proteoglycans, including Syndecan-1. NK1-targeted CARs demonstrated broadly similar in vitro potency, indicated by destruction of MET-expressing mesothelioma cell lines, accompanied by cytokine release. In vivo anti-tumor activity was demonstrated following intraperitoneal delivery to mice with an established mesothelioma xenograft. Progressive tumor regression occurred without weight loss or other clinical indicators of toxicity. These data confirm the frequent expression of MET in malignant pleural mesothelioma and demonstrate that this can be targeted effectively and safely using a CAR T-cell immunotherapeutic strategy.
Collapse
Affiliation(s)
- Thivyan Thayaparan
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Roseanna M Petrovic
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Daniela Y Achkova
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Tomasz Zabinski
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - David M Davies
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Astero Klampatsa
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.,Pulmonary, Allergy & Critical Care Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ana C Parente-Pereira
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Lynsey M Whilding
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | | | - Natalie Woodman
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Michael Sheaff
- Department of Histopathology, Barts Health NHS Trust, The Royal London Hospital, London E1 2ES, UK
| | - Jennifer R Cochran
- Department of Bioengineering and Chemical Engineering, Stanford Cancer Institute, 443 Via Ortega, Room 356, Stanford, CA, USA
| | - James F Spicer
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.,Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - John Maher
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.,Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK.,Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex, BN21 2UD, UK
| |
Collapse
|
25
|
Gilham DE, Maher J. 'Atypical' CAR T cells: NKG2D and Erb-B as examples of natural receptor/ligands to target recalcitrant solid tumors. Immunotherapy 2017; 9:723-733. [PMID: 28771104 DOI: 10.2217/imt-2017-0045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has recently been recommended for approval for certain B-cell malignancies bringing the approach closer to mainstream cancer treatment. This rapid rise to prominence has been driven by impressive clinical results and the means to successfully commercialize the approach now being actively pursued. The current success of CAR T cells in B-cell malignancies relies upon the absolute lineage specificity of the CD19 antigen. CARs can also be targeted using non-antibody approaches, including the use of receptors and ligands to provide target specificity that have different specificities and binding kinetics. The specific examples of NKG2D and Erb-B are used that provide different characteristics and target profiles for CAR T-cell therapy of cancer.
Collapse
MESH Headings
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Cancer Vaccines/immunology
- Genetic Therapy
- Humans
- Immunotherapy, Adoptive/methods
- Leukemia, B-Cell/genetics
- Leukemia, B-Cell/immunology
- Leukemia, B-Cell/therapy
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Neoplasm Recurrence, Local
- Receptor, ErbB-2/immunology
- Receptor, ErbB-2/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins/genetics
- T-Lymphocytes/physiology
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- David E Gilham
- Research & Development, Celyad S.A., Axis Business Park, Rue Edouard Belin 2, B-1435 Mont Saint Guibert, Belgium
| | - John Maher
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
- Department of Clinical Immunology & Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex, BN21 2UD, UK
| |
Collapse
|
26
|
Mo Z, Du P, Wang G, Wang Y. The Multi-Purpose Tool of Tumor Immunotherapy: Gene-Engineered T Cells. J Cancer 2017; 8:1690-1703. [PMID: 28775789 PMCID: PMC5535725 DOI: 10.7150/jca.18681] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 02/27/2017] [Indexed: 02/05/2023] Open
Abstract
A detailed summary of the published clinical trials of chimeric antigen receptor T cells (CAR-T) and TCR-transduced T cells (TCR-T) was constructed to understand the development trend of adoptive T cell therapy (ACT). In contrast to TCR-T, the number of CAR-T clinical trials has increased dramatically in China in the last three years. The ACT seems to be very prosperous. But, the multidimensional interaction of tumor, tumor associated antigen (TAA) and normal tissue exacerbates the uncontrolled outcome of T cells gene therapy. It reminds us the importance that optimizing treatment security to prevent the fatal serious adverse events. How to balance the safety and effectiveness of the ACT? At least six measures can potentially optimize the safety of ACT. At the same time, with the application of gene editing techniques, more endogenous receptors are disrupted while more exogenous receptors are expressed on T cells. As a multi-purpose tool of tumor immunotherapy, gene-engineered T cells (GE-T) have been given different functional weapons. A network which is likely to link radiation therapy, tumor vaccines, CAR-T and TCR-T is being built. Moreover, more and more evidences indicated that the combination of the ACT and other therapies would further enhance the anti-tumor capacity of the GE-T.
Collapse
Affiliation(s)
- Zeming Mo
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| | - Peixin Du
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| | - Guoping Wang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| | - Yongsheng Wang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, China 610041
| |
Collapse
|
27
|
Klampatsa A, Achkova DY, Davies DM, Parente-Pereira AC, Woodman N, Rosekilly J, Osborne G, Thayaparan T, Bille A, Sheaf M, Spicer JF, King J, Maher J. Intracavitary 'T4 immunotherapy' of malignant mesothelioma using pan-ErbB re-targeted CAR T-cells. Cancer Lett 2017; 393:52-59. [PMID: 28223167 DOI: 10.1016/j.canlet.2017.02.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 12/11/2022]
Abstract
Malignant mesothelioma remains an incurable cancer. We demonstrated that mesotheliomas expressed EGFR (79.2%), ErbB4 (49.0%) and HER2 (6.3%), but lacked ErbB3. At least one ErbB family member was expressed in 88% of tumors. To exploit ErbB dysregulation in this disease, patient T-cells were engineered by retroviral transduction to express a panErbB-targeted chimeric antigen receptor (CAR), co-expressed with a chimeric cytokine receptor that allows interleukin (IL)-4 mediated CAR T-cell proliferation. This combination is referred to as T4 immunotherapy. T-cells from mesothelioma patients were uniformly amenable to T4 genetic modification and expansion/enrichment thereafter using IL-4. Patient-derived T4+ T-cells were activated upon contact with a panel of four mesothelioma cell lines, leading to cytotoxicity and cytokine release in all cases. Adoptive transfer of T4 immunotherapy to SCID Beige mice with an established bioluminescent LO68 mesothelioma xenograft was followed by regression or eradication of disease in all animals. Despite the established ability of T4 immunotherapy to elicit cytokine release syndrome in SCID Beige mice, therapy was very well tolerated. These findings provide a strong rationale for the clinical evaluation of intracavitary T4 immunotherapy to treat mesothelioma.
Collapse
Affiliation(s)
- Astero Klampatsa
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Daniela Y Achkova
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - David M Davies
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Ana C Parente-Pereira
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Natalie Woodman
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - James Rosekilly
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Georgina Osborne
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Thivyan Thayaparan
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Andrea Bille
- Department of Thoracic Surgery, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Michael Sheaf
- Department of Histopathology, Barts Health NHS Trust, The Royal London Hospital, London E1 2ES, UK
| | - James F Spicer
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Juliet King
- Department of Thoracic Surgery, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - John Maher
- King's College London, Division of Cancer Studies, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK; Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex BN21 2UD, UK.
| |
Collapse
|
28
|
Karnieli O, Friedner OM, Allickson JG, Zhang N, Jung S, Fiorentini D, Abraham E, Eaker SS, Yong TK, Chan A, Griffiths S, Wehn AK, Oh S, Karnieli O. A consensus introduction to serum replacements and serum-free media for cellular therapies. Cytotherapy 2016; 19:155-169. [PMID: 28017599 DOI: 10.1016/j.jcyt.2016.11.011] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 10/09/2016] [Accepted: 11/09/2016] [Indexed: 02/06/2023]
Abstract
The cell therapy industry is a fast-growing industry targeted toward a myriad of clinical indications. As the cell therapy industry matures and clinical trials hit their pivotal Phase 3 studies, there will be a significant need for scale-up, process validation, and critical raw material quality assurance. Part of the well discussed challenges of upscaling manufacturing processes there is a less discussed issue relating to the availability of raw materials in the needed quality and quantities. The FDA recently noted that over 80% of the 66 investigational new drug (IND) applications for mesenchymal stem cell (MSC) products analyzed described the use of FBS during manufacturing. Accumulated data from the past years show an acceleration in serum consumption by at least 10%-15% annually, which suggests that the global demand for serum may soon exceed the supply. Ongoing concerns of safety issues due to risks of various pathogen contaminations, as well as issues related to the aforementioned serum variability that can affect final product reproducibility, are strong motivators to search for serum substitutes or serum-free media. it is important to note that there are no accepted definitions for most of these terms which leads to misleading's and misunderstandings, where the same term might be defined differently by different vendors, manufacturer, and users. It is the drug developer's responsibility to clarify what the supplied labels mean and to identify the correct questions and audits to ensure quality. The paper reviews the available serum replacements, main components, basic strategies for replacement of serum and suggests definitions.
Collapse
Affiliation(s)
| | | | - Julie G Allickson
- Regenerative Medicine Clinical Center, Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Nan Zhang
- Hematology Branch, National Heart, Lung, and Blood Institute National Institute of Health, Bethesda, Maryland, USA
| | - Sunghoon Jung
- Cell Therapy Research & Technology Lonza Walkersville, Walkersville, Maryland, USA
| | | | - Eytan Abraham
- Cell Therapy Research & Technology Lonza Walkersville, Walkersville, Maryland, USA
| | - Shannon S Eaker
- GE Healthcare Cell Therapy Division, Marlborough, Massachusetts, USA
| | | | - Allan Chan
- Bioprocessing Technology Institute, Singapore
| | | | - Amy K Wehn
- Irvine Scientific, Santa Ana, California, USA
| | - Steve Oh
- Bioprocessing Technology Institute, Singapore
| | | |
Collapse
|
29
|
Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: Summary and perspective. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jocit.2016.08.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
30
|
Lordick F, Janjigian YY. Clinical impact of tumour biology in the management of gastroesophageal cancer. Nat Rev Clin Oncol 2016; 13:348-60. [PMID: 26925958 PMCID: PMC5521012 DOI: 10.1038/nrclinonc.2016.15] [Citation(s) in RCA: 111] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The characterization of oesophageal and gastric cancer into subtypes based on genotype has evolved in the past decade. Insights into the molecular landscapes of gastroesophageal cancer provide a roadmap to assist the development of new drugs and their use in combinations, for patient stratification, and for trials of targeted therapies. Trastuzumab is the only approved treatment for gastroesophageal cancers that overexpress HER2. Acquired resistance usually limits the duration of response to this treatment, although a number of new agents directed against HER2 have the potential to overcome or prolong the time until resistance occurs. Beyond that, anti-VEGFR2 therapy with ramucirumab was the first biological treatment strategy to produce a survival benefit in an unselected population of patients with chemotherapy-refractory gastroesophageal cancer. Large initiatives are starting to address the role of biomarker-driven targeted therapy in the metastatic and in the perioperative setting for patients with this disease. Immunotherapy also holds promise, and our understanding of subsets of gastroesophageal cancer based on patterns of immune response continues to evolve. Efforts are underway to identify more relevant genomic subsets through genomic screening, functional studies, and molecular characterization. Herein, we provide an overview of the key developments in the treatment of gastroesophageal cancer, and discuss potential strategies to further optimize therapy by targeting disease subtypes.
Collapse
Affiliation(s)
- Florian Lordick
- University Cancer Center Leipzig, University Medicine Leipzig, Liebigstraße 20 D, 04103 Leipzig, Germany
| | - Yelena Y Janjigian
- Gastrointestinal Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, 1275 York Avenue, New York, New York 10065, USA
| |
Collapse
|
31
|
Abstract
It was estimated that 59,340 new cases of head and neck cancer would be diagnosed in the US alone in 2015 and that 12,290 deaths would be attributed to the disease. Local and regional recurrences may be treated with chemotherapy and radiation; however, metastatic head and neck cancer is fatal and is treated with chemotherapy for palliation. Recent successful treatment of a variety of solid and hematological malignancies by immunotherapeutic approaches (i.e. harnessing the body's own immune system to combat disease) has added a fourth therapeutic option for the treatment of cancer. This commentary will review the status of immunotherapies in clinical development for the specific treatment of head and neck cancer.
Collapse
Affiliation(s)
- Carolina Soto Chervin
- Department of Medicine, NorthShore University HealthSystem, Evanston, Ilinois, 60201, USA
| | - Bruce Brockstein
- Department of Medicine, NorthShore University HealthSystem, Evanston, Ilinois, 60201, USA; Department of Medicine, University of Chicago Pritzker School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
32
|
Abstract
A letter in response to: Milena Kalaitsidou, Gray Kueberuwa, Antje Schütt & David Edward Gilham. CAR T-cell therapy: toxicity and the relevance of preclinical models. Immunotherapy 7(5), 487–497 (2015).
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of Hematology & Oncology, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| |
Collapse
|
33
|
Whilding LM, Maher J. CAR T-cell immunotherapy: The path from the by-road to the freeway? Mol Oncol 2015; 9:1994-2018. [PMID: 26563646 PMCID: PMC5528729 DOI: 10.1016/j.molonc.2015.10.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 10/13/2015] [Accepted: 10/14/2015] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptors are genetically encoded artificial fusion molecules that can re-program the specificity of peripheral blood polyclonal T-cells against a selected cell surface target. Unparallelled clinical efficacy has recently been demonstrated using this approach to treat patients with refractory B-cell malignancy. However, the approach is technically challenging and can elicit severe toxicity in patients. Moreover, solid tumours have largely proven refractory to this approach. In this review, we describe the important structural features of CARs and how this may influence function. Emerging clinical experience is summarized in both solid tumours and haematological malignancies. Finally, we consider the particular challenges imposed by solid tumours to the successful development of CAR T-cell immunotherapy, together with a number of innovative strategies that have been developed in an effort to reverse the balance in favour of therapeutic benefit.
Collapse
Affiliation(s)
- Lynsey M Whilding
- King's College London, King's Health Partners Integrated Cancer Centre, Department of Research Oncology, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK.
| | - John Maher
- King's College London, King's Health Partners Integrated Cancer Centre, Department of Research Oncology, Guy's Hospital Campus, Great Maze Pond, London SE1 9RT, UK; Department of Immunology, Barnet Hospital, Royal Free London NHS Foundation Trust, Barnet, Hertfordshire, EN5 3DJ, UK; Department of Clinical Immunology and Allergy, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|