1
|
Amiri M, Moaveni AK, Majidi Zolbin M, Shademan B, Nourazarian A. Optimizing cancer treatment: the synergistic potential of CAR-T cell therapy and CRISPR/Cas9. Front Immunol 2024; 15:1462697. [PMID: 39582866 PMCID: PMC11581867 DOI: 10.3389/fimmu.2024.1462697] [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: 07/10/2024] [Accepted: 09/23/2024] [Indexed: 11/26/2024] Open
Abstract
Optimizing cancer treatment has become a pivotal goal in modern oncology, with advancements in immunotherapy and genetic engineering offering promising avenues. CAR-T cell therapy, a revolutionary approach that harnesses the body's own immune cells to target and destroy cancer cells, has shown remarkable success, particularly in treating acute lymphoblastic leukemia (ALL), and in treating other hematologic malignancies. While CAR-T cell therapy has shown promise, challenges such as high cost and manufacturing complexity remain. However, its efficacy in solid tumors remains limited. The integration of CRISPR/Cas9 technology, a powerful and precise genome-editing tool, also raises safety concerns regarding unintended edits and off-target effects, offers a synergistic potential to overcome these limitations. CRISPR/Cas9 can enhance CAR-T cell therapy by improving the specificity and persistence of CAR-T cells, reducing off-target effects, and engineering resistance to tumor-induced immunosuppression. This combination can also facilitate the knockout of immune checkpoint inhibitors, boosting the anti-tumor activity of CAR-T cells. Recent studies have demonstrated that CRISPR/Cas9-edited CAR-T cells can target previously untreatable cancer types, offering new hope for patients with refractory cancers. This synergistic approach not only enhances the efficacy of cancer treatment but also paves the way for personalized therapies tailored to individual genetic profiles. This review highlights the ongoing research efforts to refine this approach and explores its potential to revolutionize cancer treatment across a broader range of malignancies. As research progresses, the integration of CAR-T cell therapy and CRISPR/Cas9 holds the promise of transforming cancer treatment, making it more effective and accessible. This review explores the current advancements, challenges, and future prospects of this innovative therapeutic strategy.
Collapse
Affiliation(s)
- Maryam Amiri
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Kian Moaveni
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric Urology and Regenerative Medicine Research Center, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Shademan
- Medical Journalism, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
2
|
Rojas-Quintero J, Díaz MP, Palmar J, Galan-Freyle NJ, Morillo V, Escalona D, González-Torres HJ, Torres W, Navarro-Quiroz E, Rivera-Porras D, Bermúdez V. Car T Cells in Solid Tumors: Overcoming Obstacles. Int J Mol Sci 2024; 25:4170. [PMID: 38673757 PMCID: PMC11050550 DOI: 10.3390/ijms25084170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/12/2024] [Accepted: 03/13/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR T cell) therapy has emerged as a prominent adoptive cell therapy and a therapeutic approach of great interest in the fight against cancer. This approach has shown notorious efficacy in refractory hematological neoplasm, which has bolstered its exploration in the field of solid cancers. However, successfully managing solid tumors presents considerable intrinsic challenges, which include the necessity of guiding the modified cells toward the tumoral region, assuring their penetration and survival in adverse microenvironments, and addressing the complexity of identifying the specific antigens for each type of cancer. This review focuses on outlining the challenges faced by CAR T cell therapy when used in the treatment of solid tumors, as well as presenting optimizations and emergent approaches directed at improving its efficacy in this particular context. From precise localization to the modulation of the tumoral microenvironment and the adaptation of antigen recognition strategies, diverse pathways will be examined to overcome the current limitations and buttress the therapeutic potential of CAR T cells in the fight against solid tumors.
Collapse
Affiliation(s)
- Joselyn Rojas-Quintero
- Medicine, Pulmonary, Critical Care, and Sleep Medicine Department, Baylor College of Medicine, Houston, TX 77030, USA;
| | - María P. Díaz
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Jim Palmar
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Nataly J. Galan-Freyle
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
| | - Valery Morillo
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Daniel Escalona
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | | | - Wheeler Torres
- Facultad de Medicina, Centro de Investigaciones Endocrino—Metabólicas, Universidad del Zulia, Maracaibo 4001, Venezuela (J.P.); (V.M.); (D.E.); (W.T.)
| | - Elkin Navarro-Quiroz
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias Básicas y Biomédicas, Barranquilla 080002, Colombia
| | - Diego Rivera-Porras
- Facultad de Ciencias Jurídicas y Sociales, Universidad Simón Bolívar, Cúcuta 540001, Colombia;
| | - Valmore Bermúdez
- Centro de Investigaciones en Ciencias de la Vida, Universidad Simón Bolívar, Barranquilla 080002, Colombia; (N.J.G.-F.); (E.N.-Q.)
- Facultad de Ciencias de la Salud, Universidad Simón Bolívar, Barranquilla 080002, Colombia;
| |
Collapse
|
3
|
Schaible P, Bethge W, Lengerke C, Haraszti RA. RNA Therapeutics for Improving CAR T-cell Safety and Efficacy. Cancer Res 2023; 83:354-362. [PMID: 36512627 PMCID: PMC7614194 DOI: 10.1158/0008-5472.can-22-2155] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 11/02/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022]
Abstract
Autologous chimeric antigen receptor (CAR) T cells have recently emerged as potent tools in the fight against cancer, with promising therapeutic efficacy against hematological malignancies. However, several limitations hamper their widespread clinical use, including availability of target antigen, severe toxic effects, primary and secondary resistance, heterogeneous quality of autologous T cells, variable persistence, and low activity against solid tumors. Development of allogeneic off-the-shelf CAR T cells could help address some of these limitations but is impeded by alloimmunity with either rejection and limited expansion of allo-CAR T cells or CAR T cells versus host reactions. RNA therapeutics, such as small interfering RNAs, microRNAs, and antisense oligonucleotides, are able to silence transcripts in a sequence-specific and proliferation-sensitive way, which may offer a way to overcome some of the challenges facing CAR T-cell development and clinical utility. Here, we review how different RNA therapeutics or a combination of RNA therapeutics and genetic engineering could be harnessed to improve the safety and efficacy of autologous and allogeneic CAR T-cell therapy.
Collapse
Affiliation(s)
- Philipp Schaible
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Wolfgang Bethge
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Claudia Lengerke
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Reka Agnes Haraszti
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Mohammadian Gol T, Ureña-Bailén G, Hou Y, Sinn R, Antony JS, Handgretinger R, Mezger M. CRISPR medicine for blood disorders: Progress and challenges in delivery. Front Genome Ed 2023; 4:1037290. [PMID: 36687779 PMCID: PMC9853164 DOI: 10.3389/fgeed.2022.1037290] [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: 09/05/2022] [Accepted: 12/22/2022] [Indexed: 01/09/2023] Open
Abstract
Blood disorders are a group of diseases including hematological neoplasms, clotting disorders and orphan immune deficiency diseases that affects human health. Current improvements in genome editing based therapeutics demonstrated preclinical and clinical proof to treat different blood disorders. Genome editing components such as Cas nucleases, guide RNAs and base editors are supplied in the form of either a plasmid, an mRNA, or a ribonucleoprotein complex. The most common delivery vehicles for such components include viral vectors (e.g., AAVs and RV), non-viral vectors (e.g., LNPs and polymers) and physical delivery methods (e.g., electroporation and microinjection). Each of the delivery vehicles specified above has its own advantages and disadvantages and the development of a safe transferring method for ex vivo and in vivo application of genome editing components is still a big challenge. Moreover, the delivery of genome editing payload to the target blood cells possess key challenges to provide a possible cure for patients with inherited monogenic blood diseases and hematological neoplastic tumors. Here, we critically review and summarize the progress and challenges related to the delivery of genome editing elements to relevant blood cells in an ex vivo or in vivo setting. In addition, we have attempted to provide a future clinical perspective of genome editing to treat blood disorders with possible clinical grade improvements in delivery methods.
Collapse
Affiliation(s)
- Tahereh Mohammadian Gol
- Department of Hematology and Oncology, University Children’s Hospital, University of Tübingen, Tübingen, Germany
| | - Guillermo Ureña-Bailén
- Department of Hematology and Oncology, University Children’s Hospital, University of Tübingen, Tübingen, Germany
| | - Yujuan Hou
- Department of Hematology and Oncology, University Children’s Hospital, University of Tübingen, Tübingen, Germany
| | - Ralph Sinn
- Department of Hematology and Oncology, University Children’s Hospital, University of Tübingen, Tübingen, Germany
| | - Justin S. Antony
- Department of Hematology and Oncology, University Children’s Hospital, University of Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Children’s Hospital, University of Tübingen, Tübingen, Germany,Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates
| | - Markus Mezger
- Department of Hematology and Oncology, University Children’s Hospital, University of Tübingen, Tübingen, Germany,*Correspondence: Markus Mezger,
| |
Collapse
|
5
|
Ureña-Bailén G, Dobrowolski JM, Hou Y, Dirlam A, Roig-Merino A, Schleicher S, Atar D, Seitz C, Feucht J, Antony JS, Mohammadian Gol T, Handgretinger R, Mezger M. Preclinical Evaluation of CRISPR-Edited CAR-NK-92 Cells for Off-the-Shelf Treatment of AML and B-ALL. Int J Mol Sci 2022; 23:12828. [PMID: 36361619 PMCID: PMC9655234 DOI: 10.3390/ijms232112828] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 08/10/2023] Open
Abstract
Acute myeloid leukemia (AML) and B-cell acute lymphocytic leukemia (B-ALL) are severe blood malignancies affecting both adults and children. Chimeric antigen receptor (CAR)-based immunotherapies have proven highly efficacious in the treatment of leukemia. However, the challenge of the immune escape of cancer cells remains. The development of more affordable and ready-to-use therapies is essential in view of the costly and time-consuming preparation of primary cell-based treatments. In order to promote the antitumor function against AML and B-ALL, we transduced NK-92 cells with CD276-CAR or CD19-CAR constructs. We also attempted to enhance cytotoxicity by a gene knockout of three different inhibitory checkpoints in NK cell function (CBLB, NKG2A, TIGIT) with CRISPR-Cas9 technology. The antileukemic activity of the generated cell lines was tested with calcein and luciferase-based cytotoxicity assays in various leukemia cell lines. Both CAR-NK-92 exhibited targeted cytotoxicity and a significant boost in antileukemic function in comparison to parental NK-92. CRISPR-Cas9 knock-outs did not improve B-ALL cytotoxicity. However, triple knock-out CD276-CAR-NK-92 cells, as well as CBLB or TIGIT knock-out NK-92 cells, showed significantly enhanced cytotoxicity against U-937 or U-937 CD19/tag AML cell lines. These results indicate that the CD19-CAR and CD276-CAR-NK-92 cell lines' cytotoxic performance is suitable for leukemia killing, making them promising off-the-shelf therapeutic candidates. The knock-out of CBLB and TIGIT in NK-92 and CD276-CAR-NK-92 should be further investigated for the treatment of AML.
Collapse
MESH Headings
- Humans
- Antigens, CD19
- B7 Antigens/metabolism
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Immunotherapy, Adoptive/methods
- Killer Cells, Natural
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/metabolism
- Lymphoma, B-Cell
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Receptors, Chimeric Antigen
Collapse
Affiliation(s)
- Guillermo Ureña-Bailén
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Jérôme-Maurice Dobrowolski
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Yujuan Hou
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Alicia Dirlam
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | | | - Sabine Schleicher
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Daniel Atar
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Christian Seitz
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72074 Tuebingen, Germany
| | - Judith Feucht
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, 72074 Tuebingen, Germany
| | - Justin S. Antony
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Tahereh Mohammadian Gol
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| | - Markus Mezger
- Department of Hematology and Oncology, Children’s Hospital, University Hospital Tuebingen, 72076 Tuebingen, Germany
| |
Collapse
|
6
|
He X, Zeng XX. Immunotherapy and CRISPR Cas Systems: Potential Cure of COVID-19? Drug Des Devel Ther 2022; 16:951-972. [PMID: 35386853 PMCID: PMC8979261 DOI: 10.2147/dddt.s347297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 03/16/2022] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 has plunged the world into a pandemic that affected millions. The continually emerging new variants of concern raise the question as to whether the existing vaccines will continue to provide sufficient protection for individuals from SARS-CoV-2 during natural infection. This narrative review aims to briefly outline various immunotherapeutic options and discuss the potential of clustered regularly interspaced short palindromic repeat (CRISPR Cas system technology against COVID-19 treatment as specific cure. As the development of vaccine, convalescent plasma, neutralizing antibodies are based on the understanding of human immune responses against SARS-CoV-2, boosting human body immune responses in case of SARS-CoV-2 infection, immunotherapeutics seem feasible as specific cure against COVID-19 if the present challenges are overcome. In cell based therapeutics, apart from the high costs, risks and side effects, there are technical problems such as the production of sufficient potent immune cells and antibodies under limited time to treat the COVID-19 patients in mild conditions prior to progression into a more severe case. The CRISPR Cas technology could be utilized to refine the specificity and safety of CAR-T cells, CAR-NK cells and neutralizing antibodies against SARS-CoV-2 during various stages of the COVID-19 disease progression in infected individuals. Moreover, CRISPR Cas technology are proposed in hypotheses to degrade the viral RNA in order to terminate the infection caused by SARS-CoV-2. Thus personalized cocktails of immunotherapeutics and CRISPR Cas systems against COVID-19 as a strategy might prevent further disease progression and circumvent immunity escape.
Collapse
Affiliation(s)
- Xuesong He
- Department of Cardiology, Changzhou Jintan First People’s Hospital, Changzhou City, Jiangsu Province, 213200, People’s Republic of China
| | - Xiao Xue Zeng
- Department of Health Management, Centre of General Practice, The Seventh Affiliated Hospital, Southern Medical University, Foshan City, Guangdong Province, 528000, People’s Republic of China
| |
Collapse
|
7
|
Antony JS, Daniel-Moreno A, Lamsfus-Calle A, Raju J, Kaftancioglu M, Ureña-Bailén G, Rottenberger J, Hou Y, Santhanakumaran V, Lee JH, Heumos L, Böhringer J, Krägeloh-Mann I, Handgretinger R, Mezger M. A Mutation-Agnostic Hematopoietic Stem Cell Gene Therapy for Metachromatic Leukodystrophy. CRISPR J 2021; 5:66-79. [PMID: 34882002 DOI: 10.1089/crispr.2021.0075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a rare genetic disorder caused by mutations in the Arylsulfatase-A (ARSA) gene. The enzyme plays a key role in sulfatide metabolism in brain cells, and its deficiency leads to neurodegeneration. The clinical manifestations of MLD include stagnation and decline of motor and cognitive function, leading to premature death with limited standard treatment options. Here, we describe a mutation-agnostic hematopoietic stem and progenitor cell (HSPC) gene therapy using CRISPR-Cas9 and AAV6 repair template as a prospective treatment option for MLD. Our strategy achieved efficient insertions and deletions (>87%) and a high level of gene integration (>47%) at the ARSA locus in human bone marrow-derived HSPCs, with no detectable off-target editing. As a proof of concept, we tested our mutation-agnostic therapy in HSPCs derived from two MLD patients with distinct mutations and demonstrated restoration of ARSA enzyme activity (>30-fold improvement) equivalent to healthy adults. In summary, our investigation enabled a mutation-agnostic therapy for MLD patients with proven efficacy and strong potential for clinical translation.
Collapse
Affiliation(s)
- Justin S Antony
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Alberto Daniel-Moreno
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Andrés Lamsfus-Calle
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Janani Raju
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Merve Kaftancioglu
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Guillermo Ureña-Bailén
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Jennifer Rottenberger
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Yujuan Hou
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Vidiyaah Santhanakumaran
- Department of Pediatric Neurology, University Children's Hospital Tübingen, Germany; and University of Tübingen, Tübingen, Germany
| | - Jun-Hoe Lee
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Lukas Heumos
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Judith Böhringer
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany.,Department of Pediatric Neurology, University Children's Hospital Tübingen, Germany; and University of Tübingen, Tübingen, Germany
| | - Ingeborg Krägeloh-Mann
- Department of Pediatric Neurology, University Children's Hospital Tübingen, Germany; and University of Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| | - Markus Mezger
- Department of Hematology and Oncology, University Children's Hospital, University of Tübingen, Germany; University of Tübingen, Tübingen, Germany
| |
Collapse
|
8
|
Büning H, Fehse B, Ivics Z, Kochanek S, Koehl U, Kupatt C, Mussolino C, Nettelbeck DM, Schambach A, Uckert W, Wagner E, Cathomen T. Gene Therapy "Made in Germany": A Historical Perspective, Analysis of the Status Quo, and Recommendations for Action by the German Society for Gene Therapy. Hum Gene Ther 2021; 32:987-996. [PMID: 34662229 DOI: 10.1089/hum.2021.29178.hbu] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gene therapies have been successfully applied to treat severe inherited and acquired disorders. Although research and development are sufficiently well funded in Germany and while the output of scientific publications and patents is comparable with the leading nations in gene therapy, the country lags noticeably behind with regard to the number of both clinical studies and commercialized gene therapy products. In this article, we give a historical perspective on the development of gene therapy in Germany, analyze the current situation from the standpoint of the German Society for Gene Therapy (DG-GT), and define recommendations for action that would enable our country to generate biomedical and economic advantages from innovations in this sector, instead of merely importing advanced therapy medicinal products. Inter alia, we propose (1) to harmonize and simplify regulatory licensing processes to enable faster access to advanced therapies, and (2) to establish novel coordination, support and funding structures that facilitate networking of the key players. Such a center would provide the necessary infrastructure and know-how to translate cell and gene therapies to patients on the one hand, and pave the way for commercialization of these promising and innovative technologies on the other. Hence, these courses of action would not only benefit the German biotech and pharma landscape but also the society and the patients in need of new treatment options.
Collapse
Affiliation(s)
- Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | | | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI) and Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany.,Institute for Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| | - Christian Kupatt
- Klinik und Poliklinik für Innere Medizin I, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Claudio Mussolino
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Dirk M Nettelbeck
- Clinical Cooperation Unit Virotherapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Wolfgang Uckert
- Department of Molecular Cell Biology and Gene Therapy, Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Center for System-based Drug Research, Center for NanoScience (CeNS), Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Faculty, University of Freiburg, Freiburg, Germany
| |
Collapse
|
9
|
Zhi L, Su X, Yin M, Zhang Z, Lu H, Niu Z, Guo C, Zhu W, Zhang X. Genetical engineering for NK and T cell immunotherapy with CRISPR/Cas9 technology: Implications and challenges. Cell Immunol 2021; 369:104436. [PMID: 34500148 DOI: 10.1016/j.cellimm.2021.104436] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/07/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022]
Abstract
Immunotherapy has become one of the most promising strategies in cancer therapies. Among the therapeutic alternatives, genetically engineered NK/T cell therapies have emerged as powerful and innovative therapeutic modalities for cancer patients with precise targeting and impressive efficacy. Nonetheless, this approach still faces multiple challenges, such as immunosuppressive tumor microenvironment, exhaustion of immune effector cells in tumors, off-target effects manufacturing complexity, and poor infiltration of effector cells, all of which need to be overcome for further utilization to cancers. Recently, CRISPR/Cas9 genome editing technology, with the goal of enhancing the efficacy and increasing the availability of engineered effector cell therapies, has shown considerable potential in the novel strategies and options to overcome these limitations. Here we review the current progress of the applications of CRISPR in cancer immunotherapy. Furthermore, we discuss issues related to the NK/T cell applications, gene delivery methods, efficiency, challenges, and implications of CRISPR/Cas9.
Collapse
Affiliation(s)
- Lingtong Zhi
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Xin Su
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Meichen Yin
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Zikang Zhang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Hui Lu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Zhiyuan Niu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Changjiang Guo
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China
| | - Wuling Zhu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province, PR China.
| | - Xuan Zhang
- Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, Henan, PR China.
| |
Collapse
|
10
|
Chimeric Antigen Receptor-Engineered Natural Killer (CAR NK) Cells in Cancer Treatment; Recent Advances and Future Prospects. Stem Cell Rev Rep 2021; 17:2081-2106. [PMID: 34472037 PMCID: PMC8410173 DOI: 10.1007/s12015-021-10246-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2021] [Indexed: 12/28/2022]
Abstract
Natural Killer (NK) cells are critical members of the innate immunity lymphocytes and have a critical role in host defense against malignant cells. Adoptive cell therapy (ACT) using chimeric antigen receptor (CAR) redirects the specificity of the immune cell against a target-specific antigen. ACT has recently created an outstanding opportunity for cancer treatment. Unlike CAR-armored T cells which hadnsome shortcomings as the CAR-receiving construct, Major histocompatibility complex (MHC)-independency, shorter lifespan, the potential to produce an off-the-shelf immune product, and potent anti-tumor properties of the NK cells has introduced NK cells as a potent alternative target for expression of CAR. Here, we aim to provide an updated overview on the current improvements in CAR NK design and immunobiology and describe the potential of CAR-modified NK cells as an alternative “off-the-shelf” carrier of CAR. We also provide lists for the sources of NK cells in the process of CAR NK cell production, different methods for transduction of the CAR genetic sequence to NK cells, the differences between CAR T and CAR NK, and CAR NK-targeted tumor antigens in current studies. Additionally, we provide data on recently published preclinical and clinical studies of CAR NK therapy and a list of finished and ongoing clinical trials. For achieving CAR NK products with higher efficacy and safety, we discuss current challenges in transduction and expansion of CAR NK cells, CAR NK therapy side effects, and challenges that limit the optimal efficacy of CAR NK cells and recommend possible solutions to enhance the persistence, function, safety, and efficacy of CAR NK cells with a special focus on solid tumors.
Collapse
|
11
|
Zuo S, Song J, Zhang J, He Z, Sun B, Sun J. Nano-immunotherapy for each stage of cancer cellular immunity: which, why, and what? Theranostics 2021; 11:7471-7487. [PMID: 34158861 PMCID: PMC8210608 DOI: 10.7150/thno.59953] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy provides a new avenue for combating cancer. Current research in anticancer immunotherapy is primary based on T cell-mediated cellular immunity, which can be divided into seven steps and is named the cancer-immunity cycle. Unfortunately, clinical applications of cancer immunotherapies are restricted by inefficient drug delivery, low response rates, and unmanageable adverse reactions. In response to these challenges, the combination of nanotechnology and immunotherapy (nano-immunotherapy) has been extensively studied in recent years. Rational design of advanced nano-immunotherapies requires in-depth consideration of "which" immune step is targeted, "why" it needs to be further enhanced, and "what" nanotechnology can do for immunotherapy. However, the applications and effects of nanotechnology in the cancer-immunity cycle have not been well reviewed. Herein, we summarize the current developments in nano-immunotherapy for each stage of cancer cellular immunity, with special attention on the which, why and what. Furthermore, we summarize the advantages of nanotechnology for combination immunotherapy in two categories: enhanced efficacy and reduced toxicity. Finally, we discuss the challenges of nano-immunotherapy in detail and provide a perspective.
Collapse
Affiliation(s)
| | | | | | | | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
12
|
Chimeric antigen receptor T cell therapy in oncology – Pipeline at a glance: Analysis of the ClinicalTrials.gov database. Crit Rev Oncol Hematol 2021; 159:103239. [DOI: 10.1016/j.critrevonc.2021.103239] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
|
13
|
Yilmaz A, Cui H, Caligiuri MA, Yu J. Chimeric antigen receptor-engineered natural killer cells for cancer immunotherapy. J Hematol Oncol 2020; 13:168. [PMID: 33287875 PMCID: PMC7720606 DOI: 10.1186/s13045-020-00998-9] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are a critical component of the innate immune system. Chimeric antigen receptors (CARs) re-direct NK cells toward tumor cells carrying corresponding antigens, creating major opportunities in the fight against cancer. CAR NK cells have the potential for use as universal CAR cells without the need for human leukocyte antigen matching or prior exposure to tumor-associated antigens. Exciting data from recent clinical trials have renewed interest in the field of cancer immunotherapy due to the potential of CAR NK cells in the production of "off-the-shelf" anti-cancer immunotherapeutic products. Here, we provide an up-to-date comprehensive overview of the recent advancements in key areas of CAR NK cell research and identify under-investigated research areas. We summarize improvements in CAR design and structure, advantages and disadvantages of using CAR NK cells as an alternative to CAR T cell therapy, and list sources to obtain NK cells. In addition, we provide a list of tumor-associated antigens targeted by CAR NK cells and detail challenges in expanding and transducing NK cells for CAR production. We additionally discuss barriers to effective treatment and suggest solutions to improve CAR NK cell function, proliferation, persistence, therapeutic effectiveness, and safety in solid and liquid tumors.
Collapse
Affiliation(s)
- Ahmet Yilmaz
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Hanwei Cui
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 E. Duarte Road, KCRB, Bldg. 158, 3rd Floor, Room 3017, Los Angeles, CA, 91010, USA
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Los Angeles, CA, 91010, USA
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Los Angeles, CA, 91010, USA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, 1500 E. Duarte Road, KCRB, Bldg. 158, 3rd Floor, Room 3017, Los Angeles, CA, 91010, USA.
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, CA, 91010, USA.
- Department of Immuno-Oncology, City of Hope Beckman Research Institute, Los Angeles, CA, 91010, USA.
- City of Hope Comprehensive Cancer Center and Beckman Research Institute, Los Angeles, CA, 91010, USA.
| |
Collapse
|
14
|
Wu W, Huo Y, Ding X, Zhou Y, Gu S, Gao Y. Identification of the risks in CAR T-cell therapy clinical trials in China: a Delphi study. Ther Adv Med Oncol 2020; 12:1758835920966574. [PMID: 33149770 PMCID: PMC7580145 DOI: 10.1177/1758835920966574] [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: 01/31/2020] [Accepted: 07/13/2020] [Indexed: 11/16/2022] Open
Abstract
Aims: Within the past few years, there has been tremendous growth in clinical trials of chimeric antigen receptor (CAR) T-cell therapies. Unlike those of many small-molecule pharmaceuticals, CAR T-cell therapy clinical trials are fraught with risks due to the use of live cell products. The aim of this study is to reach a consensus with experts on the most relevant set of risks that practically occur in CAR T-cell therapy clinical trials. Methods: A Delphi method of consensus development was used to identify the risks in CAR T-cell therapy clinical trials, comprising three survey rounds. The expert panel consisted of principal investigators, clinical research physicians, members of institutional ethics committees, and Good Clinical Practice managers. Results: Of the 24 experts invited to participate in this Delphi study, 20 participants completed Round 1, Round 2, and Round 3. Finally, consensus (defined as >80% agreement) was achieved for 54 risks relating to CAR T-cell clinical trials. Effective interventions related to these risks are needed to ensure the proper protection of subject health and safety. Conclusion: The Delphi method was successful in gaining a consensus on risks relevant to CAR T-cell clinical trials in a geographically diverse expert association. It is hoped that this work can benefit future risk-based quality management in clinical trials and can potentially promote the better development of CAR T-cell therapy products.
Collapse
Affiliation(s)
- Weijia Wu
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Shanghai, China
| | - Yan Huo
- National Institution of Food and Drug Control, National Medical Products Administration, Beijing, China
| | - Xueying Ding
- Engineering Technology Research Center of Cell Therapy and Clinical Translation, Shanghai Science and Technology Committee
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shengying Gu
- Clinical Research Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Gao
- Department of Clinical Pharmacy and Pharmaceutical Management, School of Pharmacy, Fudan University, Pudong District, Shanghai, 200433, China
| |
Collapse
|
15
|
Qi T, Fu J, Zhang W, Cui W, Xu X, Yue J, Wang Q, Tian X. Mutation of PD-1 immune receptor tyrosine-based switch motif (ITSM) enhances the antitumor activity of cytotoxic T cells. Transl Cancer Res 2020; 9:6811-6819. [PMID: 35117290 PMCID: PMC8798335 DOI: 10.21037/tcr-20-2118] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/09/2020] [Indexed: 11/06/2022]
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1), as an immune checkpoint cell membrane receptor, negatively regulates T cell activation via its immune receptor, the tyrosine-based switch motif (ITSM). The purpose of this research was to evaluate the antitumor activity T cells with the ITSM mutation of PD-1 on non-small cell lung cancer (NSCLC) in vitro and in vivo. METHODS In this study, the tyrosine of ITSM in cytotoxic T cells was mutated using the adenine base editor (ABE)-xCas9 system to evaluate its effect on the antitumor activity of T cells against NSCLC. RESULTS Results showed that the PD-1-deficient T cells enhanced the death of the cocultured NSCLC cells compared with the normal T cells and saline solution. PD-1-deficient T cells also changed the interleukin 2(IL-2), interferon γ (IFN-γ), tumor necrosis factor α (TNF-α), and granulocyte-macrophage colony-stimulating factor (GM-CSF) secretion of T cells compared with those of the normal T cells. The effectiveness of ITSM mutation in enhancing the antitumor activity of PD-1-deficient T cells was verified in vivo by using a mouse xenograft model. The xenografted mice treated with PD-1-deficient T cells demonstrated repressed tumor growth of the NSCLC cells compared with those treated with normal T cells and saline solution. CONCLUSIONS The mutation of ITSM in cytotoxic T cell via the ABE-xCas9 system can significantly enhance the antitumor activity of T cells.
Collapse
Affiliation(s)
- Tongbing Qi
- Key Laboratory of Biomedical Engineering & Technology of Shandong High School, Qilu Medical University, Zibo, China
| | - Juan Fu
- Shandong Bio-focus Gene Science and Technology Ltd., Zibo, China
| | - Wen Zhang
- Clinical laboratory of Zibo Central Hospital, Zibo, China
| | - Weitong Cui
- Key Laboratory of Biomedical Engineering & Technology of Shandong High School, Qilu Medical University, Zibo, China
| | - Xiaosong Xu
- Shandong Bio-focus Gene Science and Technology Ltd., Zibo, China
| | - Jianmei Yue
- Department of Endocrinology, the First Hospital of Zibo City, Qilu Medical University, Zibo, China
| | - Qinglu Wang
- Key Laboratory of Biomedical Engineering & Technology of Shandong High School, Qilu Medical University, Zibo, China
| | - Xuewen Tian
- College of Sports and Health, Shandong Sport University, Jinan, China
| |
Collapse
|
16
|
Sun JY, Hu HB, Cheng YX, Lu XJ. CRISPR in medicine: applications and challenges. Brief Funct Genomics 2020; 19:151-153. [PMID: 32432687 DOI: 10.1093/bfgp/elaa011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|