1
|
Diop MP, van der Stegen SJC. The Pluripotent Path to Immunotherapy. Exp Hematol 2024; 139:104648. [PMID: 39251182 DOI: 10.1016/j.exphem.2024.104648] [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: 04/02/2024] [Revised: 08/14/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Adoptive cell therapy (ACT) enhances the patient's own immune cells' ability to identify and eliminate cancer cells. Several immune cell types are currently being applied in autologous ACT, including T cells, natural killer (NK) cells, and macrophages. The cells' inherent antitumor capacity can be used, or they can be targeted toward tumor-associated antigen through expression of a chimeric antigen receptor (CAR). Although CAR-based ACT has achieved great results in hematologic malignancies, the accessibility of ACT is limited by the autologous nature of the therapy. Induced pluripotent stem cells (iPSCs) hold the potential to address this challenge, because they can provide an unlimited source for the in vitro generation of immune cells. Various immune subsets have been generated from iPSC for application in ACT, including several T-cell subsets (αβT cells, mucosal-associated invariant T cells, invariant NKT [iNKT] cells, and γδT cells), as well as NK cells, macrophages, and neutrophils. iPSC-derived αβT, NK, and iNKT cells are currently being tested in phase I clinical trials. The ability to perform (multiplexed) gene editing at the iPSC level and subsequent differentiation into effector populations not only expands the arsenal of ACT but allows for development of ACT utilizing cell types which cannot be efficiently obtained from peripheral blood or engineered and expanded in vitro.
Collapse
Affiliation(s)
- Mame P Diop
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
2
|
Morel VJ, Rössler J, Bernasconi M. Targeted immunotherapy and nanomedicine for rhabdomyosarcoma: The way of the future. Med Res Rev 2024; 44:2730-2773. [PMID: 38885148 DOI: 10.1002/med.22059] [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: 06/29/2023] [Revised: 04/17/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma of childhood. Histology separates two main subtypes: embryonal RMS (eRMS; 60%-70%) and alveolar RMS (aRMS; 20%-30%). The aggressive aRMS carry one of two characteristic chromosomal translocations that result in the expression of a PAX3::FOXO1 or PAX7::FOXO1 fusion transcription factor; therefore, aRMS are now classified as fusion-positive (FP) RMS. Embryonal RMS have a better prognosis and are clinically indistinguishable from fusion-negative (FN) RMS. Next to histology and molecular characteristics, RMS risk groupings are now available defining low risk tumors with excellent outcomes and advanced stage disease with poor prognosis, with an overall survival of about only 20% despite intensified multimodal treatment. Therefore, development of novel effective targeted strategies to increase survival and to decrease long-term side effects is urgently needed. Recently, immunotherapies and nanomedicine have been emerging for potent and effective tumor treatments with minimal side effects, raising hopes for effective and safe cures for RMS patients. This review aims to describe the most relevant preclinical and clinical studies in immunotherapy and targeted nanomedicine performed so far in RMS and to provide an insight in future developments.
Collapse
Affiliation(s)
- Victoria Judith Morel
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Jochen Rössler
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Michele Bernasconi
- Department of Pediatric Hematology and Oncology, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Heim C, Hartig L, Weinelt N, Moser LM, Salzmann-Manrique E, Merker M, Wels WS, Tonn T, Bader P, Klusmann JH, van Wijk SJ, Rettinger E. Bortezomib promotes the TRAIL-mediated killing of resistant rhabdomyosarcoma by ErbB2/Her2-targeted CAR-NK-92 cells via DR5 upregulation. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200802. [PMID: 38706988 PMCID: PMC11067460 DOI: 10.1016/j.omton.2024.200802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 04/08/2024] [Indexed: 05/07/2024]
Abstract
Treatment resistance and immune escape are hallmarks of metastatic rhabdomyosarcoma (RMS), underscoring the urgent medical need for therapeutic agents against this disease entity as a key challenge in pediatric oncology. Chimeric antigen receptor (CAR)-based immunotherapies, such as the ErbB2 (Her2)-CAR-engineered natural killer (NK) cell line NK-92/5.28.z, provide antitumor cytotoxicity primarily through CAR-mediated cytotoxic granule release and thereafter-even in cases with low surface antigen expression or tumor escape-by triggering intrinsic NK cell-mediated apoptosis induction via additional ligand/receptors. In this study, we showed that bortezomib increased susceptibility toward apoptosis in clinically relevant RMS cell lines RH30 and RH41, and patient-derived RMS tumor organoid RMS335, by upregulation of the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) receptor DR5 in these metastatic, relapsed/refractory (r/r) RMS tumors. Subsequent administration of NK-92/5.28.z cells significantly enhanced antitumor activity in vitro. Applying recombinant TRAIL instead of NK-92/5.28.z cells confirmed that the synergistic antitumor effects of the combination treatment were mediated via TRAIL. Western blot analyses indicated that the combination treatment with bortezomib and NK-92/5.28.z cells increased apoptosis by interacting with the nuclear factor κB, JNK, and caspase pathways. Overall, bortezomib pretreatment can sensitize r/r RMS tumors to CAR- and, by upregulating DR5, TRAIL-mediated cytotoxicity of NK-92/5.28.z cells.
Collapse
Affiliation(s)
- Catrin Heim
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Leonie Hartig
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Nadine Weinelt
- Institute for Experimental Paediatric Haematology and Oncology (EPOH), 60528 Frankfurt am Main, Germany
| | - Laura M. Moser
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Emilia Salzmann-Manrique
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Michael Merker
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Winfried S. Wels
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, 60596 Frankfurt am Main, Germany
| | - Torsten Tonn
- DRK-Blutspendedienst Baden-Württemberg/Hessen gemeinnützige GmbH, 60505 Frankfurt am Main, Germany
| | - Peter Bader
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Jan-Henning Klusmann
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
- Goethe University Frankfurt, Department of Pediatrics, 60590 Frankfurt am Main, Germany
| | - Sjoerd J.L. van Wijk
- Institute for Experimental Paediatric Haematology and Oncology (EPOH), 60528 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
| | - Eva Rettinger
- Goethe University Frankfurt, Department of Pediatrics, Division of Stem Cell Transplantation and Immunology, 60590 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt am Main, a partnership between DKFZ and University Hospital and Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT) Frankfurt Marburg, 60590 Frankfurt am Main, Germany
| |
Collapse
|
4
|
Liu F, Miao X, Han L, Song X. Advances in CAR-NK cell therapy for lung cancer: is it a better choice in the future? Front Oncol 2024; 14:1390006. [PMID: 38863635 PMCID: PMC11165017 DOI: 10.3389/fonc.2024.1390006] [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: 02/22/2024] [Accepted: 04/22/2024] [Indexed: 06/13/2024] Open
Abstract
Lung cancer remains one of the leading causes of cancer-related mortality worldwide necessitating the development of innovative therapeutic strategies. Chimeric antigen receptor (CAR) natural killer (NK) cell therapy represents a promising advancement in the field of oncology offering a novel approach to target and eliminate tumor cells with high specificity and reduced risk of immune-related adverse effects. This paper reviews the mechanism, potential targets, and recent advances in CAR-NK cell therapy for lung cancer, including the design and engineering of CAR-NK cells, preclinical studies, and the outcomes of early-phase clinical trials. We highlight the unique advantages of using NK cells, such as their innate ability to recognize and kill cancer cells and their reduced potential for inducing graft-versus-host disease (GvHD) and cytokine release syndrome (CRS) compared to CAR T-cell therapies. Results from recent studies demonstrate significant antitumor activity in lung cancer models with improved targeting and persistence of CAR-NK cells observed in vitro and in vivo. Finally, we discuss the challenges in optimizing CAR-NK cell therapies, including the potential resistance mechanisms. The paper concludes with an outlook on the future directions of CAR-NK cell research and its implications for lung cancer treatment emphasizing the importance of continued innovation and collaboration in the field.
Collapse
Affiliation(s)
- Fengqin Liu
- The Third Department of Geriatrics, Weifang People’s Hospital, Weifang, Shandong, China
| | - Xia Miao
- Central Supply Service Department (CSSD), Weifang People’s Hospital, Weifang, Shandong, China
| | - Lu Han
- The Third Department of Geriatrics, Weifang People’s Hospital, Weifang, Shandong, China
| | - Xiao Song
- Department of Gastroenterology, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
5
|
Li T, Niu M, Zhang W, Qin S, Zhou J, Yi M. CAR-NK cells for cancer immunotherapy: recent advances and future directions. Front Immunol 2024; 15:1361194. [PMID: 38404574 PMCID: PMC10884099 DOI: 10.3389/fimmu.2024.1361194] [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: 12/25/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Natural Killer (NK) cells, intrinsic to the innate immune system, are pivotal in combating cancer due to their independent cytotoxic capabilities in antitumor immune response. Unlike predominant treatments that target T cell immunity, the limited success of T cell immunotherapy emphasizes the urgency for innovative approaches, with a spotlight on harnessing the potential of NK cells. Despite tumors adapting mechanisms to evade NK cell-induced cytotoxicity, there is optimism surrounding Chimeric Antigen Receptor (CAR) NK cells. This comprehensive review delves into the foundational features and recent breakthroughs in comprehending the dynamics of NK cells within the tumor microenvironment. It critically evaluates the potential applications and challenges associated with emerging CAR-NK cell therapeutic strategies, positioning them as promising tools in the evolving landscape of precision medicine. As research progresses, the unique attributes of CAR-NK cells offer a new avenue for therapeutic interventions, paving the way for a more effective and precise approach to cancer treatment.
Collapse
Affiliation(s)
- Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijiang Zhang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Shuang Qin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|