1
|
Schamel WW, Zintchenko M, Nguyen T, Fehse B, Briquez PS, Minguet S. The potential of γδ CAR and TRuC T cells: An unearthed treasure. Eur J Immunol 2024; 54:e2451074. [PMID: 39192467 DOI: 10.1002/eji.202451074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
Recent years have witnessed the success of αβ T cells engineered to express chimeric antigen receptors (CARs) in treating haematological cancers. CARs combine the tumour antigen binding capability of antibodies with the signalling functions of the T-cell receptor (TCR) ζ chain and co-stimulatory receptors. Despite the success, αβ CAR T cells face limitations. Possible solutions would be the use of γδ T cells and new chimeric receptors, such as TCR fusion constructs (TRuCs). Notably, γδ CAR T cells are gaining traction in pre-clinical and clinical studies, demonstrating a promising safety profile in several pilot studies. This review delves into the current understanding of γδ CAR and TCR fusion construct T cells, exploring the opportunities and challenges they present for cancer treatment.
Collapse
Affiliation(s)
- Wolfgang W Schamel
- Signaling Research Centres BIOSS and CIBSS; Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Clinics Freiburg, Freiburg, Germany
| | - Marina Zintchenko
- Signaling Research Centres BIOSS and CIBSS; Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Trang Nguyen
- Signaling Research Centres BIOSS and CIBSS; Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, and Hamburg Centre for Translational Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Centre for Translational Immunology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Priscilla S Briquez
- Department of General and Visceral Surgery, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany
| | - Susana Minguet
- Signaling Research Centres BIOSS and CIBSS; Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University Clinics Freiburg, Freiburg, Germany
| |
Collapse
|
2
|
Minguet S, Maus MV, Schamel WW. From TCR fundamental research to innovative chimeric antigen receptor design. Nat Rev Immunol 2024:10.1038/s41577-024-01093-7. [PMID: 39433885 DOI: 10.1038/s41577-024-01093-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2024] [Indexed: 10/23/2024]
Abstract
Engineered T cells that express chimeric antigen receptors (CARs) have transformed the treatment of haematological cancers. CARs combine the tumour-antigen-binding function of antibodies with the signalling functions of the T cell receptor (TCR) ζ chain and co-stimulatory receptors. The resulting constructs aim to mimic the TCR-based and co-receptor-based activation of T cells. Although these have been successful for some types of cancer, new CAR formats are needed, to limit side effects and broaden their use to solid cancers. Insights into the mechanisms of TCR signalling, including the identification of signalling motifs that are not present in the TCR ζ chain and mechanistic insights in TCR activation, have enabled the development of CAR formats that outcompete the current CARs in preclinical mouse models and clinical trials. In this Perspective, we explore the mechanistic rationale behind new CAR designs.
Collapse
Affiliation(s)
- Susana Minguet
- Signalling Research Centers BIOSS and CIBSS, Freiburg, Germany.
- Department of Synthetic Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Marcela V Maus
- Cellular Immunotherapy Program and Krantz Family Center for Cancer Research, Mass General Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Wolfgang W Schamel
- Signalling Research Centers BIOSS and CIBSS, Freiburg, Germany.
- Centre for Chronic Immunodeficiency (CCI), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Rus Bakarurraini NAA, Kamarudin AA, Jamal R, Abu N. Engineered T cells for Colorectal Cancer. Immunotherapy 2024; 16:987-998. [PMID: 39229803 PMCID: PMC11485792 DOI: 10.1080/1750743x.2024.2391733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
Colorectal cancer (CRC) is a major contributor to global cancer incidence and mortality. Conventional treatments have limitations; hence, innovative approaches are imperative. Recent advancements in cancer research have led to the development of personalized targeted therapies and immunotherapies. Immunotherapy, in particular, T cell-based therapies, exhibited to be promising in enhancing cancer treatment outcomes. This review focuses on the landscape of engineered T cells as a potential option for the treatment of CRC. It highlights the approaches, challenges and current advancements in this field. As the understanding of molecular mechanisms increases, engineered T cells hold great potential in revolutionizing cancer treatment. To fully explore their safety efficacy in improving patient outcomes, further research and clinical trials are necessary.
Collapse
Affiliation(s)
| | - Ammar Akram Kamarudin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| |
Collapse
|
4
|
Yun KM, Bazhenova L. Emerging New Targets in Systemic Therapy for Malignant Pleural Mesothelioma. Cancers (Basel) 2024; 16:1252. [PMID: 38610930 PMCID: PMC11011044 DOI: 10.3390/cancers16071252] [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: 02/01/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Malignant pleural mesothelioma (MPM) is a heterogeneous cancer composed of distinct molecular and pathologic subtypes. Unfortunately, MPM is aggressive, and current therapies for advanced, unresectable disease remain limited to cytotoxic chemotherapy and immunotherapy. Our understanding of the genomic landscape of MPM is steadily growing, while the discovery of effective targeted therapies in MPM has advanced more slowly than in other solid tumors. Given the prevalence of alterations in tumor suppressor genes in MPM, it has been challenging to identify actionable targets. However, efforts to characterize the genetic signatures in MPM over the last decade have led to a range of novel targeted therapeutics entering early-phase clinical trials. In this review, we discuss the advancements made thus far in targeted systemic therapies in MPM and the future direction of targeted strategies in patients with advanced MPM.
Collapse
Affiliation(s)
- Karen M. Yun
- Division of Hematology-Oncology, Moores Cancer Center at UC San Diego Health, La Jolla, CA 92093, USA;
| | | |
Collapse
|
5
|
Juraske C, Krissmer SM, Teuber ES, Parigiani MA, Strietz J, Wesch D, Kabelitz D, Minguet S, Schamel WW. Reprogramming of human γδ T cells by expression of an anti-CD19 TCR fusion construct (εTRuC) to enhance tumor killing. J Leukoc Biol 2024; 115:293-305. [PMID: 38149982 DOI: 10.1093/jleuko/qiad128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/23/2023] [Accepted: 10/05/2023] [Indexed: 12/28/2023] Open
Abstract
We have developed a new format of a chimeric antigen receptor for αβ T cells, in which the single-chain variable fragment recognizing the tumor antigen is directly fused to the T cell receptor, called T cell receptor fusion construct (TRuC). Here, we express an anti-CD19 εTRuC in primary γδ T cells that were expanded using zoledronate (Zol) or concanavalin A. We show that the resulting εTRuC γδ T cells were reprogrammed to better recognize CD19-positive B cell tumors and-in case of the Zol-expanded cells-a CD19-expressing colon adenocarcinoma-derived cell line in vitro. This resulted in enhanced tumor killing, upregulation of the activation marker CD25, and secretion of cytokines. We found that the transduction efficiency of the concanavalin A-expanded cells was better than the one of the Zol-expanded ones. Our in vitro cytotoxicity data suggest that the Vδ2 T cells were better killers than the Vδ1 T cells. Finally, addition of vitamin C promoted the recovery of larger γδ T cell numbers after lentiviral transduction, as used for the expression of the εTRuC. In conclusion, the generation and use of γδ εTRuC T cells might be a new approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Claudia Juraske
- Centre for Biological Signalling Studies BIOSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency CCI, Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 115, 79106 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine SGBM, University of Freiburg, Albertstraße 19A, 79104 Freiburg, Germany
| | - Sonia M Krissmer
- Centre for Biological Signalling Studies BIOSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency CCI, Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 115, 79106 Freiburg, Germany
| | - Evelyn S Teuber
- Centre for Biological Signalling Studies BIOSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency CCI, Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 115, 79106 Freiburg, Germany
| | - Maria A Parigiani
- Centre for Biological Signalling Studies BIOSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency CCI, Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 115, 79106 Freiburg, Germany
| | - Juliane Strietz
- Centre for Biological Signalling Studies BIOSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency CCI, Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 115, 79106 Freiburg, Germany
| | - Daniela Wesch
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts University of Kiel and University Hospital Schleswig-Holstein Campus Kiel, Arnold-Heller-Straße 3, 24105 Kiel, Germany
| | - Susana Minguet
- Centre for Biological Signalling Studies BIOSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency CCI, Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 115, 79106 Freiburg, Germany
| | - Wolfgang W Schamel
- Centre for Biological Signalling Studies BIOSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Integrative Biological Signalling Studies CIBSS, Faculty of Biology, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
- Centre for Chronic Immunodeficiency CCI, Medical Centre Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Str. 115, 79106 Freiburg, Germany
| |
Collapse
|
6
|
McCarthy D, Lofgren M, Watt A, Horton H, Kieffer-Kwon P, Ding J, Kobold S, Baeuerle PA, Hofmeister R, Gutierrez DA, Tighe R. Functional enhancement of mesothelin-targeted TRuC-T cells by a PD1-CD28 chimeric switch receptor. Cancer Immunol Immunother 2023; 72:4195-4207. [PMID: 37848682 PMCID: PMC10700406 DOI: 10.1007/s00262-023-03556-7] [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: 05/04/2023] [Accepted: 09/26/2023] [Indexed: 10/19/2023]
Abstract
T cells expressing a mesothelin (MSLN)-specific T cell receptor fusion construct (TRuC®), called TC-210, have demonstrated robust antitumor activity in preclinical models of mesothelioma, ovarian cancer, and lung cancer. However, they are susceptible to suppression by the programmed cell death protein 1 (PD-1)/programmed cell death protein ligand 1 (PD-L1) axis and lack intrinsic costimulatory signaling elements. To enhance the function of anti-MSLN TRuC-T cells, chimeric switch receptors (CSRs) have been designed to co-opt the immunosuppressive PD-1/PD-L1 axis and to deliver a CD28-mediated costimulatory signal. Here, we report that coexpression of the PD1-CD28 CSR in TRuC-T cells enhanced T cell receptor signaling, increased proinflammatory effector cytokines, decreased anti-inflammatory cytokines, and sustained effector function in the presence of PD-L1 when compared with TC-210. Anti-MSLN TRuC-T cells engineered to coexpress PD1-CD28 CSRs comprising the ectodomain of PD-1 and the intracellular domain of CD28 linked by the transmembrane domain of PD-1 were selected for integration into an anti-MSLN TRuC-T cell therapy product called TC-510. In vitro, TC-510 showed significant improvements in persistence and resistance to exhaustion upon chronic stimulation by tumor cells expressing MSLN and PD-L1 when compared with TC-210. In vivo, TC-510 showed a superior ability to provide durable protection following tumor rechallenge, versus TC-210. These data demonstrate that integration of a PD1-CD28 CSR into TRuC-T cells improves effector function, resistance to exhaustion, and prolongs persistence. Based on these findings, TC-510 is currently being evaluated in patients with MSLN-expressing solid tumors.
Collapse
Affiliation(s)
- Derrick McCarthy
- TCR2 Therapeutics, Inc., 100 Binney Street, Suite 710, Cambridge, MA, 02142, USA
| | | | - Amy Watt
- Adaptimmune, Philadelphia, PA, USA
| | - Holly Horton
- TCR2 Therapeutics, Inc., 100 Binney Street, Suite 710, Cambridge, MA, 02142, USA
| | | | - Jian Ding
- TCR2 Therapeutics, Inc., 100 Binney Street, Suite 710, Cambridge, MA, 02142, USA
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS‑M) and Division of Clinical Pharmacology, Department of Medicine IV, Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Patrick A Baeuerle
- TCR2 Therapeutics, Inc., 100 Binney Street, Suite 710, Cambridge, MA, 02142, USA
- Institute of Immunology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Robert Hofmeister
- TCR2 Therapeutics, Inc., 100 Binney Street, Suite 710, Cambridge, MA, 02142, USA
| | - Dario A Gutierrez
- TCR2 Therapeutics, Inc., 100 Binney Street, Suite 710, Cambridge, MA, 02142, USA
| | - Robert Tighe
- TCR2 Therapeutics, Inc., 100 Binney Street, Suite 710, Cambridge, MA, 02142, USA.
| |
Collapse
|
7
|
Requejo Cier CJ, Valentini N, Lamarche C. Unlocking the potential of Tregs: innovations in CAR technology. Front Mol Biosci 2023; 10:1267762. [PMID: 37900916 PMCID: PMC10602912 DOI: 10.3389/fmolb.2023.1267762] [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: 07/27/2023] [Accepted: 09/20/2023] [Indexed: 10/31/2023] Open
Abstract
Regulatory T cells (Tregs) adoptive immunotherapy is emerging as a viable treatment option for both autoimmune and alloimmune diseases. However, numerous challenges remain, including limitations related to cell number, availability of target-specific cells, stability, purity, homing ability, and safety concerns. To address these challenges, cell engineering strategies have emerged as promising solutions. Indeed, it has become feasible to increase Treg numbers or enhance their stability through Foxp3 overexpression, post-translational modifications, or demethylation of the Treg-specific demethylated region (TSDR). Specificity can be engineered by the addition of chimeric antigen receptors (CARs), with new techniques designed to fine-tune specificity (tandem chimeric antigen receptors, universal chimeric antigen receptors, synNotch chimeric antigen receptors). The introduction of B-cell targeting antibody receptor (BAR) Tregs has paved the way for effective regulation of B cells and plasma cells. In addition, other constructs have emerged to enhance Tregs activation and function, such as optimized chimeric antigen receptors constructs and the use of armour proteins. Chimeric antigen receptor expression can also be better regulated to limit tonic signaling. Furthermore, various opportunities exist for enhancing the homing capabilities of CAR-Tregs to improve therapy outcomes. Many of these genetic modifications have already been explored for conventional CAR-T therapy but need to be further considered for CAR-Tregs therapies. This review highlights innovative CAR-engineering strategies that have the potential to precisely and efficiently manage immune responses in autoimmune diseases and improve transplant outcomes. As these strategies are further explored and optimized, CAR-Treg therapies may emerge as powerful tools for immune intervention.
Collapse
Affiliation(s)
- Christopher J. Requejo Cier
- Department of Microbiology, Infectiology and Immunology, Hôpital Maisonneuve-Rosemont Research Institute, Université de Montréal, Montreal, QC, Canada
| | - Nicolas Valentini
- Department of Microbiology, Infectiology and Immunology, Hôpital Maisonneuve-Rosemont Research Institute, Université de Montréal, Montreal, QC, Canada
| | - Caroline Lamarche
- Department of Medicine, Hôpital Maisonneuve-Rosemont Research Institute, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
8
|
Dagogo-Jack I. Targeted Approaches to Treatment of Pleural Mesothelioma: A Review. JCO Precis Oncol 2023; 7:e2300344. [PMID: 37992257 PMCID: PMC10681489 DOI: 10.1200/po.23.00344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/16/2023] [Accepted: 09/11/2023] [Indexed: 11/24/2023] Open
Abstract
Pleural mesothelioma is an aggressive disease that is enriched for inactivating alterations in tumor suppressor genes. Systemic therapeutic strategies for pleural mesothelioma generally involve chemotherapies and immunotherapies that are chosen without consideration of the tumor's molecular profile. As this generalized approach to treatment rarely yields durable responses, alternative therapeutic regimens are urgently indicated. Preclinical studies have identified synthetic lethal protein and metabolic interactions, recurrently overexpressed proteins, and frequent pathway perturbations that may be therapeutically exploited in mesothelioma. This review discusses the mechanism of action of emerging investigational therapies and summarizes findings from phase I-II clinical trials exploring selective, biomarker-driven therapeutic strategies for mesothelioma, with a focus on five common targets. Finally, using lessons learned from these clinical trials, imperatives for successful implementation of targeted therapy in mesothelioma are discussed.
Collapse
Affiliation(s)
- Ibiayi Dagogo-Jack
- Department of Medicine, Massachusetts General Hospital Cancer Center, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Hassan R, Butler M, O'Cearbhaill RE, Oh DY, Johnson M, Zikaras K, Smalley M, Ross M, Tanyi JL, Ghafoor A, Shah NN, Saboury B, Cao L, Quintás-Cardama A, Hong D. Mesothelin-targeting T cell receptor fusion construct cell therapy in refractory solid tumors: phase 1/2 trial interim results. Nat Med 2023; 29:2099-2109. [PMID: 37501016 PMCID: PMC10427427 DOI: 10.1038/s41591-023-02452-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/08/2023] [Indexed: 07/29/2023]
Abstract
The T cell receptor fusion construct (TRuC) gavocabtagene autoleucel (gavo-cel) consists of single-domain anti-mesothelin antibody that integrates into the endogenous T cell receptor (TCR) and engages the signaling capacity of the entire TCR upon mesothelin binding. Here we describe phase 1 results from an ongoing phase1/2 trial of gavo-cel in patients with treatment-refractory mesothelin-expressing solid tumors. The primary objectives were to evaluate safety and determine the recommended phase 2 dose (RP2D). Secondary objectives included efficacy. Thirty-two patients received gavo-cel at increasing doses either as a single agent (n = 3) or after lymphodepletion (LD, n = 29). Dose-limiting toxicities of grade 3 pneumonitis and grade 5 bronchioalveolar hemorrhage were noted. The RP2D was determined as 1 × 108 cells per m2 after LD. Grade 3 or higher pneumonitis was seen in 16% of all patients and in none at the RP2D; grade 3 or higher cytokine release syndrome occurred in 25% of all patients and in 15% at the RP2D. In 30 evaluable patients, the overall response rate and disease control rate were 20% (13% confirmed) and 77%, respectively, and the 6-month overall survival rate was 70%. Gavo-cel warrants further study in patients with mesothelin-expressing cancers given its encouraging anti-tumor activity, but it may have a narrow therapeutic window. ClinicalTrials.gov identifier: NCT03907852 .
Collapse
Affiliation(s)
- Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Marcus Butler
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Roisin E O'Cearbhaill
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY, USA
| | - David Y Oh
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | | | | | | | | | - Janos L Tanyi
- Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA, USA
| | - Azam Ghafoor
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - David Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
10
|
Maher J. Chimeric Antigen Receptor (CAR) T-Cell Therapy for Patients with Lung Cancer: Current Perspectives. Onco Targets Ther 2023; 16:515-532. [PMID: 37425981 PMCID: PMC10327905 DOI: 10.2147/ott.s341179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR)-engineered T-cells has achieved unprecedented efficacy in selected hematological cancers. However, solid tumors such as lung cancer impose several additional challenges to the attainment of clinical success using this emerging therapeutic modality. Lung cancer is the biggest cause of cancer-related mortality worldwide, accounting for approximately 1.8 million deaths worldwide each year. Obstacles to the development of CAR T-cell immunotherapy for lung cancer include the selection of safe tumor-selective targets, accounting for the large number of candidates that have been evaluated thus far. Tumor heterogeneity is also a key hurdle, meaning that single target-based approaches are susceptible to therapeutic failure through the emergence of antigen null cancers. There is also a need to enable CAR T-cells to traffic efficiently to sites of disease, to infiltrate tumor deposits and to operate within the hostile tumor microenvironment formed by solid tumors, resisting the onset of exhaustion. Multiple immune, metabolic, physical and chemical barriers operate at the core of malignant lesions, with potential for further heterogeneity and evolution in the face of selective therapeutic pressures. Although the extraordinarily adaptable nature of lung cancers has recently been unmasked, immunotherapy using immune checkpoint blockade can achieve long-term disease control in a small number of patients, establishing clinical proof of concept that immunotherapies can control advanced lung carcinomas. This review summarizes pre-clinical CAR T-cell research that is specifically focused on lung cancer in addition to published and ongoing clinical trial activity. A number of advanced engineering strategies are also described which are designed to bridge the gap to the attainment of meaningful efficacy using genetically engineered T-cells.
Collapse
Affiliation(s)
- John Maher
- King’s College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, Guy’s Cancer Centre, Great Maze Pond, London, SE1 9RT, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London, SE1 9RT, UK
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex, BN21 2UD, UK
| |
Collapse
|
11
|
De Pauw T, De Mey L, Debacker JM, Raes G, Van Ginderachter JA, De Groof TWM, Devoogdt N. Current status and future expectations of nanobodies in oncology trials. Expert Opin Investig Drugs 2023; 32:705-721. [PMID: 37638538 DOI: 10.1080/13543784.2023.2249814] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/16/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Monoclonal antibodies have revolutionized personalized medicine for cancer in recent decades. Despite their broad application in oncology, their large size and complexity may interfere with successful tumor targeting for certain applications of cancer diagnosis and therapy. Nanobodies have unique structural and pharmacological features compared to monoclonal antibodies and have successfully been used as complementary anti-cancer diagnostic and/or therapeutic tools. AREAS COVERED Here, an overview is given of the nanobody-based diagnostics and therapeutics that have been or are currently being tested in oncological clinical trials. Furthermore, preclinical developments, which are likely to be translated into the clinic in the near future, are highlighted. EXPERT OPINION Overall, the presented studies show the application potential of nanobodies in the field of oncology, making it likely that more nanobodies will be clinically approved in the upcoming future.
Collapse
Affiliation(s)
- Tessa De Pauw
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Lynn De Mey
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Jens M Debacker
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Geert Raes
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Jo A Van Ginderachter
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium
- Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Timo W M De Groof
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging Laboratory, Department of Medical Imaging, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|