1
|
Lussana F, Magnani CF, Galimberti S, Gritti G, Gaipa G, Belotti D, Cabiati B, Napolitano S, Ferrari S, Moretti A, Buracchi C, Borleri GM, Rambaldi B, Rizzuto G, Grassi A, Paganessi M, Meli C, Tettamanti S, Risca G, Pais G, Spinozzi G, Benedicenti F, Cazzaniga G, Capelli C, Gotti E, Introna M, Golay J, Montini E, Balduzzi A, Valsecchi MG, Dastoli G, Rambaldi A, Biondi A. Donor-derived CARCIK-CD19 cells engineered with Sleeping Beauty transposon in acute lymphoblastic leukemia relapsed after allogeneic transplantation. Blood Cancer J 2025; 15:54. [PMID: 40180925 PMCID: PMC11968829 DOI: 10.1038/s41408-025-01260-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
Non-viral engineering can ease CAR-T cell production and reduce regulatory and cost requirements. We utilized Sleeping Beauty transposon to engineer donor-derived anti-CD19.CD28.OX40.CD3zeta T cells differentiated in cytokine-induced killer (CARCIK-CD19) for B-cell precursor acute lymphoblastic leukemia (BCP-ALL) patients relapsed after allogeneic hematopoietic stem cell transplantation (alloHSCT). We report the results of CARCIK-CD19 observed in 36 patients (4 children and 32 adults) treated according to the final recommended dose. Cytokine release syndrome of grade 2 or lower occurred in 15 patients, ICANS grade 2 in 1 patient, and late-onset peripheral neurotoxicity of grade 3 in 2 patients. GVHD never occurred after treatment with allogeneic CARCIK-CD19. Complete remission was achieved by 30 out of 36 patients (83.3%), with MRD negativity in 89% of responders. With a median follow-up of 2.2 years, the 1-year overall survival was 57.0%, and event-free survival was 32.0%. The median duration of response at 1 year was 38.6%. CAR-T cells expanded rapidly after infusion and remained detectable for over 2 years. Integration site analysis after infusion showed a high clonal diversity. These data demonstrated that SB-engineered CAR-T cells are safe and induce durable remission in heavily pretreated patients with BCP-ALL relapsed after alloHSCT. Trial registration: The phase 1/2 and phase II trials are registered at www.clinicaltrials.gov as NCT#03389035 and NCT#05252403.
Collapse
Affiliation(s)
- Federico Lussana
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Chiara F Magnani
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medical Oncology and Hematology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Stefania Galimberti
- Department of Medicine and Surgery, Bicocca Bioinformatics, Biostatistics and Bioimaging Centre B4, University of Milano-Bicocca, Milan, Italy
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giuseppe Gritti
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Giuseppe Gaipa
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Daniela Belotti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Benedetta Cabiati
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Laboratorio di Terapia Cellulare e Genica Stefano Verri, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Sara Napolitano
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Silvia Ferrari
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Alex Moretti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Chiara Buracchi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Gian Maria Borleri
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Benedetta Rambaldi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Giuliana Rizzuto
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Anna Grassi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Muriel Paganessi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Cristian Meli
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Sarah Tettamanti
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Risca
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Pais
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Spinozzi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giovanni Cazzaniga
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Chiara Capelli
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Elisa Gotti
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Martino Introna
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Josée Golay
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- USS Centro di Terapia Cellulare "G. Lanzani", Bergamo, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Adriana Balduzzi
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy
| | - Maria Grazia Valsecchi
- Department of Medicine and Surgery, Bicocca Bioinformatics, Biostatistics and Bioimaging Centre B4, University of Milano-Bicocca, Milan, Italy
- Biostatistics and Clinical Epidemiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giuseppe Dastoli
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology, University of Milan, Milan, Italy
| | - Andrea Biondi
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- Department of Pediatrics, University of Milano-Bicocca, Fondazione MBBM, Monza, Italy.
| |
Collapse
|
2
|
van den Berg J, Läubli H, Khanna N, Jeker LT, Holbro A. Basic Concepts and Indications of CAR T Cells. Hamostaseologie 2025; 45:14-23. [PMID: 39970899 DOI: 10.1055/a-2491-3652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized cancer immunotherapy, particularly for hematological malignancies. This personalized approach is based on genetically engineering T cells derived from the patient to target antigens expressed-among others-on malignant cells. Nowadays they offer new hope where conventional therapies, such as chemotherapy and radiation, have often failed. Since the first FDA approval in 2017, CAR T cell therapy has rapidly expanded, proving highly effective against previously refractory diseases with otherwise a dismal outcome. Despite its promise, CAR T cell therapy continues to face significant challenges, including complex manufacturing, the management of toxicities, resistance mechanisms that impact long-term efficacy, and limited access as well as high costs, which continue to shape ongoing research and clinical applications. This review aims to provide an overview of CAR T cell therapy, including its fundamental concepts, clinical applications, current challenges, and future directions in hematological malignancies.
Collapse
Affiliation(s)
- Jana van den Berg
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Nina Khanna
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, Basel, Switzerland
| | - Lukas T Jeker
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
- Transplantation Immunology and Nephrology, University Hospital Basel, Basel, Switzerland
| | - Andreas Holbro
- Division of Hematology, University Hospital Basel, Basel, Switzerland
- Innovation Focus Cell Therapies, University Hospital Basel, Basel, Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel, Switzerland
| |
Collapse
|
3
|
Feuchtinger T, Bader P, Subklewe M, Breidenbach M, Willier S, Metzler M, Gökbuget N, Hauer J, Müller F, Schlegel PG, Frühwald M, Schmid C, Troeger A, Baldus C, Meisel R, Künkele A, Topp M, Bourquin JP, Cario G, Von Stackelberg A, Peters C. Approaches for bridging therapy prior to chimeric antigen receptor T cells for relapsed/refractory acute lymphoblastic B-lineage leukemia in children and young adults. Haematologica 2024; 109:3892-3903. [PMID: 38356450 PMCID: PMC11609793 DOI: 10.3324/haematol.2023.283780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 02/09/2024] [Indexed: 02/16/2024] Open
Abstract
The ongoing development of immunotherapies, including chimeric antigen receptor (CAR) T cells, has revolutionized cancer treatment. In pediatric relapsed/refractory B-lineage acute leukemia antiCD19-CAR induce impressive initial response rates, with event-free survival plateauing at 30-50% according to long-term follow-up data. During the interval between diagnosis of relapse or refractoriness and CAR T-cell infusion, patients require a bridging therapy. To date, this therapy has consisted of highly variable approaches based on local experience. Here, in an European collaborative effort of pediatric and adult hematologists, we summarize current knowledge with the aim of establishing guidance for bridging therapy. We discuss treatment strategies for different subgroups of patients, the advantages and disadvantages of low- and high-intensity regimens, and the potential impact of bridging therapy on outcomes after CAR T-cell infusion. This guidance is a step towards cross-institutional harmonization of bridging therapy, including personalized approaches. This will allow better comparability of clinical data and increase the level of evidence for the treatment of children and young adults with relapsed/ refractory B-lineage acute leukemia until they can receive CAR T-cell infusion.
Collapse
Affiliation(s)
- Tobias Feuchtinger
- Department of Paediatric Haematology, Oncology, Hemostaseology and Stem Cell Transplantation, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Germany; Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg.
| | - Peter Bader
- Goethe University, University Hospital, Department for Children and Adolescents, Division for Stern Cell Transplantation, Immunology and Intensive Care, Frankfurt
| | - Marion Subklewe
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Medicine III, University Hospital, LMU Munich, Munich
| | - Maike Breidenbach
- Department of Paediatric Haematology, Oncology, Hemostaseology and Stem Cell Transplantation, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Germany; Bavarian Cancer Research Center (BZKF), R/R ALL Study Group
| | - Semjon Willier
- Department of Paediatric Haematology, Oncology, Hemostaseology and Stem Cell Transplantation, Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Germany; Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology and Oncology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg
| | - Markus Metzler
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Erlangen
| | - Nicola Gökbuget
- Department of Medicine II, Haematology/Oncology, Goethe University, Frankfurt
| | - Julia Hauer
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Pediatrics and Children's Cancer Research Centre, TUM School of Medicine, Children's Hospital Munich Schwabing, Technical University of Munich, Munich
| | - Fabian Müller
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Internal Medicine 5, Haematology and Oncology, University Hospital of Erlangen, Friedrich- Alexander University of Erlangen- Nuremberg (FAU), Erlangen
| | - Paul-Gerhardt Schlegel
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; University Children's Hospital Wuerzburg, Wuerzburg
| | - Michael Frühwald
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Pediatrics and Adolescent Medicine, Swabian Children's Cancer Center, University Medical Center Augsburg, Augsburg
| | - Christoph Schmid
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Internal Medicine, University Medical Centre Augsburg, Augsburg
| | - Anja Troeger
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Paediatric Haematology, Oncology and Stem Cell Transplantation, University of Regensburg, Regensburg
| | - Claudia Baldus
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel
| | - Roland Meisel
- Division of Paediatric Stern Cell Therapy, Department of Paediatric Oncology, Haematology and Clinical Immunology, Medical Faculty, Heinrich-Heine- University, Duesseldorf
| | - Annette Künkele
- Charite-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Paediatric Oncology and Haematology, Berlin
| | - Max Topp
- Bavarian Cancer Research Center (BZKF), R/R ALL Study Group; Department of Medicine II, University Hospital of Wuerzburg, Wuerzburg
| | | | - Gunnar Cario
- Department of Pediatrics, University Medical Center Schleswig-Holstein, Campus Kiel, Germany, Kiel, Schleswig-Holstein
| | - Arend Von Stackelberg
- Charite-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Department of Paediatric Oncology and Haematology, Berlin
| | - Christina Peters
- St. Anna Children's Hospital, St. Anna Children's Research Institute, Medical University Vienna, Vienna
| |
Collapse
|
4
|
Boretti A. The transformative potential of AI-driven CRISPR-Cas9 genome editing to enhance CAR T-cell therapy. Comput Biol Med 2024; 182:109137. [PMID: 39260044 DOI: 10.1016/j.compbiomed.2024.109137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/31/2024] [Accepted: 09/08/2024] [Indexed: 09/13/2024]
Abstract
This narrative review examines the promising potential of integrating artificial intelligence (AI) with CRISPR-Cas9 genome editing to advance CAR T-cell therapy. AI algorithms offer unparalleled precision in identifying genetic targets, essential for enhancing the therapeutic efficacy of CAR T-cell treatments. This precision is critical for eliminating negative regulatory elements that undermine therapy effectiveness. Additionally, AI streamlines the manufacturing process, significantly reducing costs and increasing accessibility, thereby encouraging further research and development investment. A key benefit of AI integration is improved safety; by predicting and minimizing off-target effects, AI enhances the specificity of CRISPR-Cas9 edits, contributing to safer CAR T-cell therapy. This advancement is crucial for patient safety and broader clinical adoption. The convergence of AI and CRISPR-Cas9 has transformative potential, poised to revolutionize personalized immunotherapy. These innovations could expand the application of CAR T-cell therapy beyond hematologic malignancies to various solid tumors and other non-hematologic conditions, heralding a new era in cancer treatment that substantially improves patient outcomes.
Collapse
|
5
|
Chang JF, Wellhausen N, Engel NW, Landmann JH, Hopkins CR, Salas-McKee J, Bear AS, Selli ME, Agarwal S, Jadlowsky JK, Linette GP, Gill S, June CH, Fraietta JA, Singh N. Identification of Core Techniques That Enhance Genome Editing of Human T Cells Expressing Synthetic Antigen Receptors. Cancer Immunol Res 2024; 12:1136-1146. [PMID: 38869428 DOI: 10.1158/2326-6066.cir-24-0251] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/15/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024]
Abstract
Genome editing technologies have seen remarkable progress in recent years, enabling precise regulation of exogenous and endogenous genes. These advances have been extensively applied to the engineering of human T lymphocytes, leading to the development of practice changing therapies for patients with cancer and the promise of synthetic immune cell therapies for a variety of nonmalignant diseases. Many distinct conceptual and technical approaches have been used to edit T-cell genomes, however targeted assessments of which techniques are most effective for manufacturing, gene editing, and transgene expression are rarely reported. Through extensive comparative evaluation, we identified methods that most effectively enhance engineering of research-scale and preclinical T-cell products at critical stages of manufacturing.
Collapse
Affiliation(s)
- Ju-Fang Chang
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Center for Gene and Cellular Immunotherapy, Washington University School of Medicine, St. Louis, Missouri
| | - Nils Wellhausen
- Division of Hematology and Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Nils W Engel
- Division of Hematology and Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jack H Landmann
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Center for Gene and Cellular Immunotherapy, Washington University School of Medicine, St. Louis, Missouri
| | - Caitlin R Hopkins
- Deparment of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - January Salas-McKee
- Deparment of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Adham S Bear
- Division of Hematology and Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Mehmet E Selli
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Center for Gene and Cellular Immunotherapy, Washington University School of Medicine, St. Louis, Missouri
| | - Sangya Agarwal
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Julie K Jadlowsky
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Gerald P Linette
- Division of Hematology and Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Saar Gill
- Division of Hematology and Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Carl H June
- Division of Hematology and Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Joseph A Fraietta
- Deparment of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
- Center for Cellular Immunotherapies, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Nathan Singh
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Center for Gene and Cellular Immunotherapy, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
6
|
Atimango AO, Wesana J, Kalule SW, Verbeke W, De Steur H. Genome editing in food and agriculture: from regulations to consumer perspectives. Curr Opin Biotechnol 2024; 87:103127. [PMID: 38564970 DOI: 10.1016/j.copbio.2024.103127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 04/04/2024]
Abstract
Genome editing (GE) has emerged as a technology that could revolutionize food and agricultural production. While its advent has evoked enthusiasm for a more sustainable food system, there exists heterogeneity in regulations and public opinions regarding the technology. This review discusses evidence on the implications of government regulations on GE, and perceptions of genome-edited (GEd) food and related regulations. The review highlights consumers' positive attitude and preference for GEd foods when compared with genetically modified foods, despite the limited awareness and knowledge of GE technology. While policy changes might trigger debates, providing tailored benefits, information to consumers could further improve their attitude toward GE.
Collapse
Affiliation(s)
- Alice O Atimango
- Department of Agricultural Economics, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; Department of Rural Development and Agribusiness, Faculty of Agriculture and Environment, Gulu University, P.O. Box 166, Gulu, Uganda
| | - Joshua Wesana
- Department of Agricultural Economics, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium; Faculty of Agriculture and Environmental Sciences, Mountains of the Moon University, Kasindikwa Village, Lake Saaka, Fort-Portal, Uganda
| | - Stephen W Kalule
- Department of Rural Development and Agribusiness, Faculty of Agriculture and Environment, Gulu University, P.O. Box 166, Gulu, Uganda
| | - Wim Verbeke
- Department of Agricultural Economics, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium
| | - Hans De Steur
- Department of Agricultural Economics, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, B-9000 Ghent, Belgium.
| |
Collapse
|
7
|
Andreu-Saumell I, Rodriguez-Garcia A, Mühlgrabner V, Gimenez-Alejandre M, Marzal B, Castellsagué J, Brasó-Maristany F, Calderon H, Angelats L, Colell S, Nuding M, Soria-Castellano M, Barbao P, Prat A, Urbano-Ispizua A, Huppa JB, Guedan S. CAR affinity modulates the sensitivity of CAR-T cells to PD-1/PD-L1-mediated inhibition. Nat Commun 2024; 15:3552. [PMID: 38670972 PMCID: PMC11053011 DOI: 10.1038/s41467-024-47799-z] [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: 10/06/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy for solid tumors faces significant hurdles, including T-cell inhibition mediated by the PD-1/PD-L1 axis. The effects of disrupting this pathway on T-cells are being actively explored and controversial outcomes have been reported. Here, we hypothesize that CAR-antigen affinity may be a key factor modulating T-cell susceptibility towards the PD-1/PD-L1 axis. We systematically interrogate CAR-T cells targeting HER2 with either low (LA) or high affinity (HA) in various preclinical models. Our results reveal an increased sensitivity of LA CAR-T cells to PD-L1-mediated inhibition when compared to their HA counterparts by using in vitro models of tumor cell lines and supported lipid bilayers modified to display varying PD-L1 densities. CRISPR/Cas9-mediated knockout (KO) of PD-1 enhances LA CAR-T cell cytokine secretion and polyfunctionality in vitro and antitumor effect in vivo and results in the downregulation of gene signatures related to T-cell exhaustion. By contrast, HA CAR-T cell features remain unaffected following PD-1 KO. This behavior holds true for CD28 and ICOS but not 4-1BB co-stimulated CAR-T cells, which are less sensitive to PD-L1 inhibition albeit targeting the antigen with LA. Our findings may inform CAR-T therapies involving disruption of PD-1/PD-L1 pathway tailored in particular for effective treatment of solid tumors.
Collapse
Affiliation(s)
- Irene Andreu-Saumell
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Alba Rodriguez-Garcia
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain.
| | - Vanessa Mühlgrabner
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Marta Gimenez-Alejandre
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Berta Marzal
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Joan Castellsagué
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Fara Brasó-Maristany
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Hugo Calderon
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Laura Angelats
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Salut Colell
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Mara Nuding
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Marta Soria-Castellano
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Paula Barbao
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Aleix Prat
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Alvaro Urbano-Ispizua
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Johannes B Huppa
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Sonia Guedan
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain.
| |
Collapse
|
8
|
Niu Z, Wu J, Zhao Q, Zhang J, Zhang P, Yang Y. CAR-based immunotherapy for breast cancer: peculiarities, ongoing investigations, and future strategies. Front Immunol 2024; 15:1385571. [PMID: 38680498 PMCID: PMC11045891 DOI: 10.3389/fimmu.2024.1385571] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
Surgery, chemotherapy, and endocrine therapy have improved the overall survival and postoperative recurrence rates of Luminal A, Luminal B, and HER2-positive breast cancers but treatment modalities for triple-negative breast cancer (TNBC) with poor prognosis remain limited. The effective application of the rapidly developing chimeric antigen receptor (CAR)-T cell therapy in hematological tumors provides new ideas for the treatment of breast cancer. Choosing suitable and specific targets is crucial for applying CAR-T therapy for breast cancer treatment. In this paper, we summarize CAR-T therapy's effective targets and potential targets in different subtypes based on the existing research progress, especially for TNBC. CAR-based immunotherapy has resulted in advancements in the treatment of breast cancer. CAR-macrophages, CAR-NK cells, and CAR-mesenchymal stem cells (MSCs) may be more effective and safer for treating solid tumors, such as breast cancer. However, the tumor microenvironment (TME) of breast tumors and the side effects of CAR-T therapy pose challenges to CAR-based immunotherapy. CAR-T cells and CAR-NK cells-derived exosomes are advantageous in tumor therapy. Exosomes carrying CAR for breast cancer immunotherapy are of immense research value and may provide a treatment modality with good treatment effects. In this review, we provide an overview of the development and challenges of CAR-based immunotherapy in treating different subtypes of breast cancer and discuss the progress of CAR-expressing exosomes for breast cancer treatment. We elaborate on the development of CAR-T cells in TNBC therapy and the prospects of using CAR-macrophages, CAR-NK cells, and CAR-MSCs for treating breast cancer.
Collapse
Affiliation(s)
- Zhipu Niu
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jingyuan Wu
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qiancheng Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jinyu Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Pengyu Zhang
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yiming Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|