51
|
Ali A, DiPersio JF. ReCARving the future: bridging CAR T-cell therapy gaps with synthetic biology, engineering, and economic insights. Front Immunol 2024; 15:1432799. [PMID: 39301026 PMCID: PMC11410633 DOI: 10.3389/fimmu.2024.1432799] [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: 05/14/2024] [Accepted: 08/12/2024] [Indexed: 09/22/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has revolutionized the treatment of hematologic malignancies, offering remarkable remission rates in otherwise refractory conditions. However, its expansion into broader oncological applications faces significant hurdles, including limited efficacy in solid tumors, safety concerns related to toxicity, and logistical challenges in manufacturing and scalability. This review critically examines the latest advancements aimed at overcoming these obstacles, highlighting innovations in CAR T-cell engineering, novel antigen targeting strategies, and improvements in delivery and persistence within the tumor microenvironment. We also discuss the development of allogeneic CAR T cells as off-the-shelf therapies, strategies to mitigate adverse effects, and the integration of CAR T cells with other therapeutic modalities. This comprehensive analysis underscores the synergistic potential of these strategies to enhance the safety, efficacy, and accessibility of CAR T-cell therapies, providing a forward-looking perspective on their evolutionary trajectory in cancer treatment.
Collapse
Affiliation(s)
- Alaa Ali
- Stem Cell Transplant and Cellular Immunotherapy Program, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, United States
| | - John F DiPersio
- Center for Gene and Cellular Immunotherapy, Washington University in Saint Louis, Saint Louis, MO, United States
| |
Collapse
|
52
|
Vandecandelaere G, Ramapriyan R, Gaffey M, Richardson LG, Steuart SJ, Tazhibi M, Kalaw A, Grewal EP, Sun J, Curry WT, Choi BD. Pre-Clinical Models for CAR T-Cell Therapy for Glioma. Cells 2024; 13:1480. [PMID: 39273050 PMCID: PMC11394304 DOI: 10.3390/cells13171480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/28/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Immunotherapy represents a transformative shift in cancer treatment. Among myriad immune-based approaches, chimeric antigen receptor (CAR) T-cell therapy has shown promising results in treating hematological malignancies. Despite aggressive treatment options, the prognosis for patients with malignant brain tumors remains poor. Research leveraging CAR T-cell therapy for brain tumors has surged in recent years. Pre-clinical models are crucial in evaluating the safety and efficacy of these therapies before they advance to clinical trials. However, current models recapitulate the human tumor environment to varying degrees. Novel in vitro and in vivo techniques offer the opportunity to validate CAR T-cell therapies but also have limitations. By evaluating the strengths and weaknesses of various pre-clinical glioma models, this review aims to provide a roadmap for the development and pre-clinical testing of CAR T-cell therapies for brain tumors.
Collapse
Affiliation(s)
- Gust Vandecandelaere
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
- Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Rishab Ramapriyan
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Matthew Gaffey
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Leland Geoffrey Richardson
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Samuel Jeffrey Steuart
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Masih Tazhibi
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Adrian Kalaw
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Eric P. Grewal
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Jing Sun
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - William T. Curry
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| | - Bryan D. Choi
- Brain Tumor Immunotherapy Lab, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (G.V.)
| |
Collapse
|
53
|
Ius T, Somma T, Pasqualetti F, Berardinelli J, Vitulli F, Caccese M, Cella E, Cenciarelli C, Pozzoli G, Sconocchia G, Zeppieri M, Gerardo C, Caffo M, Lombardi G. Local therapy in glioma: An evolving paradigm from history to horizons (Review). Oncol Lett 2024; 28:440. [PMID: 39081966 PMCID: PMC11287108 DOI: 10.3892/ol.2024.14573] [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: 02/13/2024] [Accepted: 06/14/2024] [Indexed: 08/02/2024] Open
Abstract
Despite the implementation of multimodal treatments after surgery, glioblastoma (GBM) remains an incurable disease, posing a significant challenge in neuro-oncology. In this clinical setting, local therapy (LT), a developing paradigm, has received significant interest over time due to its potential to overcome the drawbacks of conventional therapy options for GBM. The present review aimed to trace the historical development, highlight contemporary advances and provide insights into the future horizons of LT in GBM management. In compliance with the Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols criteria, a systematic review of the literature on the role of LT in GBM management was conducted. A total of 2,467 potentially relevant articles were found and, after removal of duplicates, 2,007 studies were screened by title and abstract (Cohen's κ coefficient=0.92). Overall, it emerged that 15, 10 and 6 clinical studies explored the clinical efficiency of intraoperative local treatment modalities, local radiotherapy and local immunotherapy, respectively. GBM recurrences occur within 2 cm of the radiation field in 80% of cases, emphasizing the significant influence of local factors on recurrence. This highlights the urgent requirement for LT strategies. In total, three primary reasons have thus led to the development of numerous LT solutions in recent decades: i) Intratumoral implants allow the blood-brain barrier to be bypassed, resulting in limited systemic toxicity; ii) LT facilitates bridging therapy between surgery and standard treatments; and iii) given the complexity of GBM, targeting multiple components of the tumor microenvironment through ligands specific to various elements could have a synergistic effect in treatments. Considering the spatial and temporal heterogeneity of GBM, the disease prognosis could be significantly improved by a combination of therapeutic strategies in the era of precision medicine.
Collapse
Affiliation(s)
- Tamara Ius
- Unit of Neurosurgery, Head-Neck and Neurosciences Department, University Hospital of Udine, I-33100 Udine, Italy
| | - Teresa Somma
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | | | - Jacopo Berardinelli
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | - Francesca Vitulli
- Division of Neurosurgery, Department of Neurosciences, Reproductive and Odontostomatological Sciences, Federico II University, I-80128 Naples, Italy
| | - Mario Caccese
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
| | - Eugenia Cella
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
- Medical Oncology 2, San Martino Hospital-IRCCS, I-16131 Genoa Italy
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology, National Research Council, I-00133 Roma, Italy
| | - Giacomo Pozzoli
- Section of Pharmacology, Department of Healthcare Surveillance and Bioethics, Catholic University Medical School, Fondazione Policlinico Universitario A. Gemelli IRCCS, I-00168 Rome, Italy
| | - Giuseppe Sconocchia
- Institute of Translational Pharmacology, National Research Council, I-00133 Roma, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, I-33100 Udine, Italy
| | - Caruso Gerardo
- Unit of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University Hospital of Messina, I-98125 Messina, Italy
| | - Maria Caffo
- Unit of Neurosurgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University Hospital of Messina, I-98125 Messina, Italy
| | - Giuseppe Lombardi
- Medical Oncology 1, Veneto Institute of Oncology-IRCCS, I-35128 Padua, Italy
| |
Collapse
|
54
|
Hawly J, Murcar MG, Schcolnik-Cabrera A, Issa ME. Glioblastoma stem cell metabolism and immunity. Cancer Metastasis Rev 2024; 43:1015-1035. [PMID: 38530545 DOI: 10.1007/s10555-024-10183-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 03/09/2024] [Indexed: 03/28/2024]
Abstract
Despite enormous efforts being invested in the development of novel therapies for brain malignancies, there remains a dire need for effective treatments, particularly for pediatric glioblastomas. Their poor prognosis has been attributed to the fact that conventional therapies target tumoral cells, but not glioblastoma stem cells (GSCs). GSCs are characterized by self-renewal, tumorigenicity, poor differentiation, and resistance to therapy. These characteristics represent the fundamental tools needed to recapitulate the tumor and result in a relapse. The mechanisms by which GSCs alter metabolic cues and escape elimination by immune cells are discussed in this article, along with potential strategies to harness effector immune cells against GSCs. As cellular immunotherapy is making significant advances in a variety of cancers, leveraging this underexplored reservoir may result in significant improvements in the treatment options for brain malignancies.
Collapse
Affiliation(s)
- Joseph Hawly
- Faculty of Medicine and Medical Sciences, University of Balamand, Dekouaneh, Lebanon
| | - Micaela G Murcar
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Mark E Issa
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.
| |
Collapse
|
55
|
Zaninelli S, Panna S, Tettamanti S, Melita G, Doni A, D’Autilia F, Valgardsdottir R, Gotti E, Rambaldi A, Golay J, Introna M. Functional Activity of Cytokine-Induced Killer Cells Enhanced by CAR-CD19 Modification or by Soluble Bispecific Antibody Blinatumomab. Antibodies (Basel) 2024; 13:71. [PMID: 39311376 PMCID: PMC11417890 DOI: 10.3390/antib13030071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/26/2024] Open
Abstract
Strategies to increase the anti-tumor efficacy of cytokine-induced killer cells (CIKs) include genetic modification with chimeric antigen receptors (CARs) or the addition of soluble T-cell engaging bispecific antibodies (BsAbs). Here, CIKs were modified using a transposon system integrating two distinct anti-CD19 CARs (CAR-MNZ and CAR-BG2) or combined with soluble CD3xCD19 BsAb blinatumomab (CIK + Blina). CAR-MNZ bearing the CD28-OX40-CD3ζ signaling modules, and CAR-BG2, designed on the Tisagenlecleucel CAR sequence (Kymriah®), carrying the 4-1BB and CD3ζ signaling elements, were employed. After transfection and CIK expansion, cells expressed CAR-CD19 to a similar extent (35.9% CAR-MNZ and 17.7% CAR-BG2). In vitro evaluations demonstrated robust proliferation and cytotoxicity (~50% cytotoxicity) of CARCIK-MNZ, CARCIK-BG2, and CIK + Blina against CD19+ target cells, suggesting similar efficacy. All effectors formed an increased number of synapses, activated NFAT and NFkB, and secreted IL-2 and IFN-ɣ upon encountering targets. CIK + Blina displayed strongest NFAT and IFN-ɣ induction, whereas CARCIK-BG2 demonstrated superior synapse formation. All the effectors have shown therapeutic activity in vivo against the CD19+ Daudi tumor model, with CARCIK cells showing a more durable response compared to CIK + Blina, likely due to the short half-life of Blina in this model.
Collapse
Affiliation(s)
- Silvia Zaninelli
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Silvia Panna
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Sarah Tettamanti
- M. Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Giusi Melita
- M. Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Andrea Doni
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, 20089 Milano, Italy
| | - Francesca D’Autilia
- Unit of Multiscale and Nanostructural Imaging, IRCCS Humanitas Research Hospital, 20089 Milano, Italy
| | - Rut Valgardsdottir
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Elisa Gotti
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII Hospital, 24127 Bergamo, Italy
- Department of Oncology and Hematology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Josée Golay
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| | - Martino Introna
- Center of Cellular Therapy “G. Lanzani”, Division of Hematology, ASST Papa Giovanni XXIII, 24122 Bergamo, Italy; (S.Z.)
| |
Collapse
|
56
|
Zeng X, Zhang H, Guo J, Yang D, Zhu Y, Liu N, Tang J, Liu T, Zhao X. A novel bispecific T-cell engager using the ligand-target csGRP78 against acute myeloid leukemia. Cell Mol Life Sci 2024; 81:371. [PMID: 39196413 PMCID: PMC11358366 DOI: 10.1007/s00018-024-05410-0] [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: 01/03/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 08/29/2024]
Abstract
Current medical therapies for treating acute myeloid leukemia (AML) remain unmet, and AML patients may benefit from targeted immunotherapy approaches that focus on specific tumor antigens. GRP78, which is upregulated in various malignant tumors such as AML, is partially expressed as cell surface GRP78 (csGRP78) on the cell membrane, making it an ideal target for redirecting T cells, including T-cell engagers. However, considering the conventional approach of using two scFv segments to construct a bispecific T-cell engager (BiTE), we have undertaken the development of a novel BiTE that utilizes a cyclic peptide ligand to specifically target csGRP78, which we refer to as GRP78-CD3/BiTE. We studied the effects of GRP78-CD3/BiTE on treatments for AML in vitro and in vivo and assessed the pharmacokinetics of this engager. Our findings demonstrated that GRP78-CD3/BiTE could not only effectively mediate the cytotoxicity of T cells against csGRP78-expressing AML cells but also specifically eliminate primary AML tumor cells in vitro. Furthermore, GRP78-CD3/BiTE exhibited a longer half-life despite having a lower molecular weight than CD19-CD3/BiTE. In a xenograft mouse model of AML, treatment with GRP78-CD3/BiTE prolonged the survival time of the mice. Our findings demonstrate that GRP78-CD3/BiTE is effective and selective for eliminating csGRP78-expressing AML cells and suggest that this approach to targeted immunotherapy could lead to effective new treatments for AML.
Collapse
MESH Headings
- Endoplasmic Reticulum Chaperone BiP
- Humans
- Animals
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Mice
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/pharmacology
- CD3 Complex/immunology
- Heat-Shock Proteins/immunology
- Heat-Shock Proteins/metabolism
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Ligands
- Female
- Mice, SCID
- Immunotherapy/methods
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Xiaozhu Zeng
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hang Zhang
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jing Guo
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dong Yang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yongjie Zhu
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Nan Liu
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jie Tang
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Ting Liu
- Department of Hematology, Institute of Hematology, West China Hospital of Sichuan University, Chengdu, Sichuan, 610041, China.
| | - Xudong Zhao
- Department of Targeting Therapy & Immunology and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Respiratory Health and Multimorbidity and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
57
|
Bernstock JD, Gerstl JVE, Valdés PA, Friedman GK, Chiocca E. Next-generation CAR T cell therapies for glioblastoma. Sci Transl Med 2024; 16:eadp2660. [PMID: 39196960 DOI: 10.1126/scitranslmed.adp2660] [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: 03/15/2024] [Accepted: 08/05/2024] [Indexed: 08/30/2024]
Abstract
Interim results from two phase 1 trials demonstrate progress in the use of chimeric antigen receptor (CAR) T cell therapy for recurrent glioblastoma (GBM).
Collapse
Affiliation(s)
- Joshua D Bernstock
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Neurosurgery, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jakob V E Gerstl
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pablo A Valdés
- Department of Neurosurgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Gregory K Friedman
- Division of Pediatrics, Neuro-Oncology Section, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - E Chiocca
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
58
|
Taha Z, Crupi MJF, Alluqmani N, MacKenzie D, Vallati S, Whelan JT, Fareez F, Alwithenani A, Petryk J, Chen A, Spinelli MM, Ng K, Sobh J, de Souza CT, Bharadwa PR, Lee TKH, Thomas DA, Huang BZ, Kassas O, Poutou J, Gilchrist VH, Boulton S, Thomson M, Marius R, Hooshyar M, McComb S, Arulanandam R, Ilkow CS, Bell JC, Diallo JS. Complementary dual-virus strategy drives synthetic target and cognate T-cell engager expression for endogenous-antigen agnostic immunotherapy. Nat Commun 2024; 15:7267. [PMID: 39179564 PMCID: PMC11343834 DOI: 10.1038/s41467-024-51498-0] [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: 10/12/2023] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
Targeted antineoplastic immunotherapies have achieved remarkable clinical outcomes. However, resistance to these therapies due to target absence or antigen shedding limits their efficacy and excludes tumours from candidacy. To address this limitation, here we engineer an oncolytic rhabdovirus, vesicular stomatitis virus (VSVΔ51), to express a truncated targeted antigen, which allows for HER2-targeting with trastuzumab. The truncated HER2 (HER2T) lacks signaling capabilities and is efficiently expressed on infected cell surfaces. VSVΔ51-mediated HER2T expression simulates HER2-positive status in tumours, enabling effective treatment with the antibody-drug conjugate trastuzumab emtansine in vitro, ex vivo, and in vivo. Additionally, we combine VSVΔ51-HER2T with an oncolytic vaccinia virus expressing a HER2-targeted T-cell engager. This dual-virus therapeutic strategy demonstrates potent curative efficacy in vivo in female mice using CD3+ infiltrate for anti-tumour immunity. Our findings showcase the ability to tailor the tumour microenvironment using oncolytic viruses, thereby enhancing compatibility with "off-the-shelf" targeted therapies.
Collapse
Affiliation(s)
- Zaid Taha
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| | - Mathieu Joseph François Crupi
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| | - Nouf Alluqmani
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Duncan MacKenzie
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Sydney Vallati
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Jack Timothy Whelan
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Faiha Fareez
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Akram Alwithenani
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Julia Petryk
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Andrew Chen
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Marcus Mathew Spinelli
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Kristy Ng
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Judy Sobh
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | | | - Priya Rose Bharadwa
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Timothy Kit Hin Lee
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Dylan Anthony Thomas
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Ben Zhen Huang
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Omar Kassas
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Joanna Poutou
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Victoria Heather Gilchrist
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - Stephen Boulton
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Max Thomson
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Ricardo Marius
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Mohsen Hooshyar
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Scott McComb
- Cancer Immunology Team, National Research Council of Canada, Human Health Therapeutics, Ottawa, ON, K1A 0R6, Canada
| | - Rozanne Arulanandam
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
| | - Carolina Solange Ilkow
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
| | - John Cameron Bell
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| | - Jean-Simon Diallo
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, K1H 8L6, Canada.
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
59
|
Testa U, Castelli G, Pelosi E. CAR-T Cells in the Treatment of Nervous System Tumors. Cancers (Basel) 2024; 16:2913. [PMID: 39199683 PMCID: PMC11352247 DOI: 10.3390/cancers16162913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Chimeric antigen receptor T cells (CAR-Ts) have shown a remarkable efficacy in hematological malignancies but limited responses in solid tumors. Among solid tumors, CAR-T cell therapy has been particularly explored in brain tumors. CAR-T cells have shown a limited clinical efficacy in various types of brain tumors due to several factors that have hampered their activity, including tumor antigen heterogeneity, the limited access of CAR-T cells to brain tumor cells, limited CAR-T cell trafficking and in vivo persistence and the presence of a highly immunosuppressive tumor microenvironment. Despite these considerations, some recent studies have shown promising antitumor activity of GD2-CAR-T cells on diffuse midline gliomas and neuroblastomas and of CARv3-TEAM-E cells in glioblastomas. However, strategies are required to improve the effect of CAR-T cells in brain tumors, including advanced CAR-T cell design with multiple antigenic targeting and incorporation of combination therapies.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (G.C.); (E.P.)
| | | | | |
Collapse
|
60
|
Hill TF, Narvekar P, Asher GD, Edelstein JN, Camp ND, Grimm A, Thomas KR, Leiken MD, Molloy KM, Cook PJ, Arlauckas SP, Morgan RA, Tasian SK, Rawlings DJ, James RG. Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. Mol Ther 2024; 32:2676-2691. [PMID: 38959896 PMCID: PMC11405176 DOI: 10.1016/j.ymthe.2024.06.004] [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: 01/23/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Bispecific antibodies are an important tool for the management and treatment of acute leukemias. As a next step toward clinical translation of engineered plasma cells, we describe approaches for secretion of bispecific antibodies by human plasma cells. We show that human plasma cells expressing either fragment crystallizable domain-deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of B acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19-bispecific secretion by plasma cells and prevents self-targeting. Plasma cells secreting anti-CD19-bispecific antibodies elicited in vivo control of acute lymphoblastic leukemia patient-derived xenografts in immunodeficient mice co-engrafted with autologous T cells. In these studies, we found that leukemic control elicited by engineered plasma cells was similar to CD19-targeted chimeric antigen receptor-expressing T cells. Finally, the steady-state concentration of anti-CD19 bispecifics in serum 1 month after cell delivery and tumor eradication was comparable with that observed in patients treated with a steady-state infusion of blinatumomab. These findings support further development of ePCs for use as a durable delivery system for the treatment of acute leukemias, and potentially other cancers.
Collapse
Affiliation(s)
- Tyler F Hill
- University of Washington, Medical Scientist Training Program, Seattle, WA, USA; Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Parnal Narvekar
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Gregory D Asher
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | - Nathan D Camp
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Annaiz Grimm
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Kerri R Thomas
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Peter J Cook
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Sarah K Tasian
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research, Philadelphia, PA, USA; Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David J Rawlings
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Immunology, Seattle, WA, USA
| | - Richard G James
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Pharmacology, Seattle, WA, USA.
| |
Collapse
|
61
|
Chen T, Ma W, Wang X, Ye Q, Hou X, Wang Y, Jiang C, Meng X, Sun Y, Cai J. Insights of immune cell heterogeneity, tumor-initiated subtype transformation, drug resistance, treatment and detecting technologies in glioma microenvironment. J Adv Res 2024:S2090-1232(24)00315-1. [PMID: 39097088 DOI: 10.1016/j.jare.2024.07.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 06/30/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024] Open
Abstract
BACKGROUND With the gradual understanding of glioma development and the immune microenvironment, many immune cells have been discovered. Despite the growing comprehension of immune cell functions and the clinical application of immunotherapy, the precise roles and characteristics of immune cell subtypes, how glioma induces subtype transformation of immune cells and its impact on glioma progression have yet to be understood. AIM OF THE REVIEW In this review, we comprehensively center on the four major immune cells within the glioma microenvironment, particularly neutrophils, macrophages, lymphocytes, myeloid-derived suppressor cells (MDSCs), and other significant immune cells. We discuss (1) immune cell subtype markers, (2) glioma-induced immune cell subtype transformation, (3) the mechanisms of each subtype influencing chemotherapy resistance, (4) therapies targeting immune cells, and (5) immune cell-associated single-cell sequencing. Eventually, we identified the characteristics of immune cell subtypes in glioma, comprehensively summarized the exact mechanism of glioma-induced immune cell subtype transformation, and concluded the progress of single-cell sequencing in exploring immune cell subtypes in glioma. KEY SCIENTIFIC CONCEPTS OF REVIEW In conclusion, we have analyzed the mechanism of chemotherapy resistance detailly, and have discovered prospective immunotherapy targets, excavating the potential of novel immunotherapies approach that synergistically combines radiotherapy, chemotherapy, and surgery, thereby paving the way for improved immunotherapeutic strategies against glioma and enhanced patient outcomes.
Collapse
Affiliation(s)
- Tongzheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenbin Ma
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qile Ye
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xintong Hou
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiwei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China; The Six Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangqi Meng
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Ying Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
62
|
Polak R, Zhang ET, Kuo CJ. Cancer organoids 2.0: modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer 2024; 24:523-539. [PMID: 38977835 DOI: 10.1038/s41568-024-00706-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2024] [Indexed: 07/10/2024]
Abstract
The development of neoplasia involves a complex and continuous interplay between malignantly transformed cells and the tumour microenvironment (TME). Cancer immunotherapies targeting the immune TME have been increasingly validated in clinical trials but response rates vary substantially between tumour histologies and are often transient, idiosyncratic and confounded by resistance. Faithful experimental models of the patient-specific tumour immune microenvironment, capable of recapitulating tumour biology and immunotherapy effects, would greatly improve patient selection, target identification and definition of resistance mechanisms for immuno-oncology therapeutics. In this Review, we discuss currently available and rapidly evolving 3D tumour organoid models that capture important immune features of the TME. We highlight diverse opportunities for organoid-based investigations of tumour immunity, drug development and precision medicine.
Collapse
Affiliation(s)
- Roel Polak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa T Zhang
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
63
|
Chen M, Chen F, Gao Z, Li X, Hu L, Yang S, Zhao S, Song Z. CAFs and T cells interplay: The emergence of a new arena in cancer combat. Biomed Pharmacother 2024; 177:117045. [PMID: 38955088 DOI: 10.1016/j.biopha.2024.117045] [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: 04/25/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/04/2024] Open
Abstract
The interaction between the immune system and the tumor matrix has a huge impact on the progression and treatment of cancer. This paper summarizes and discusses the crosstalk between T cells and cancer-associated fibroblasts (CAFs). CAFs can also produce inhibitors that counteract the function of T cells and promote tumor immune escape, while T cells can also engage in complex two-way interactions with CAFs through direct cell contact, the exchange of soluble factors such as cytokines, and the remodeling of the extracellular matrix. Precise targeted intervention can effectively reverse tumor-promoting crosstalk between T cells and CAFs, improve anti-tumor immune response, and provide a new perspective for cancer treatment. Therefore, it is important to deeply understand the mechanism of crosstalk between T cells and CAFs. This review aims to outline the underlying mechanisms of these interactions and discuss potential therapeutic strategies that may become fundamental tools in the treatment of cancer, especially hard-to-cure cancers.
Collapse
Affiliation(s)
- Minjie Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Fei Chen
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Zhaofeng Gao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Xiaoping Li
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Lingyu Hu
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Shuying Yang
- Department of intensive medicine, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Siqi Zhao
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| | - Zhengwei Song
- Department of Surgery, the Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China.
| |
Collapse
|
64
|
Mog BJ, Marcou N, DiNapoli SR, Pearlman AH, Nichakawade TD, Hwang MS, Douglass J, Hsiue EHC, Glavaris S, Wright KM, Konig MF, Paul S, Wyhs N, Ge J, Miller MS, Azurmendi P, Watson E, Pardoll DM, Gabelli SB, Bettegowda C, Papadopoulos N, Kinzler KW, Vogelstein B, Zhou S. Preclinical studies show that Co-STARs combine the advantages of chimeric antigen and T cell receptors for the treatment of tumors with low antigen densities. Sci Transl Med 2024; 16:eadg7123. [PMID: 38985855 DOI: 10.1126/scitranslmed.adg7123] [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: 01/16/2023] [Revised: 04/01/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
Two types of engineered T cells have been successfully used to treat patients with cancer, one with an antigen recognition domain derived from antibodies [chimeric antigen receptors (CARs)] and the other derived from T cell receptors (TCRs). CARs use high-affinity antigen-binding domains and costimulatory domains to induce T cell activation but can only react against target cells with relatively high amounts of antigen. TCRs have a much lower affinity for their antigens but can react against target cells displaying only a few antigen molecules. Here, we describe a new type of receptor, called a Co-STAR (for costimulatory synthetic TCR and antigen receptor), that combines aspects of both CARs and TCRs. In Co-STARs, the antigen-recognizing components of TCRs are replaced by high-affinity antibody fragments, and costimulation is provided by two modules that drive NF-κB signaling (MyD88 and CD40). Using a TCR-mimic antibody fragment that targets a recurrent p53 neoantigen presented in a common human leukocyte antigen (HLA) allele, we demonstrate that T cells equipped with Co-STARs can kill cancer cells bearing low densities of antigen better than T cells engineered with conventional CARs and patient-derived TCRs in vitro. In mouse models, we show that Co-STARs mediate more robust T cell expansion and more durable tumor regressions than TCRs similarly modified with MyD88 and CD40 costimulation. Our data suggest that Co-STARs may have utility for other peptide-HLA antigens in cancer and other targets where antigen density may limit the efficacy of engineered T cells.
Collapse
Affiliation(s)
- Brian J Mog
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Nikita Marcou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sarah R DiNapoli
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Alexander H Pearlman
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Tushar D Nichakawade
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218, USA
| | - Michael S Hwang
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jacqueline Douglass
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Emily Han-Chung Hsiue
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Stephanie Glavaris
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Katharine M Wright
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Maximilian F Konig
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | - Suman Paul
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Nicolas Wyhs
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jiaxin Ge
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michelle S Miller
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - P Azurmendi
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Evangeline Watson
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Sandra B Gabelli
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chetan Bettegowda
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nickolas Papadopoulos
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Kenneth W Kinzler
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| | - Bert Vogelstein
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shibin Zhou
- Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg~Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA
| |
Collapse
|
65
|
Geurts M, Preusser M. Locoregional delivery of chimeric antigen receptor-T cells: Breaking the spell in glioblastoma? Neuro Oncol 2024; 26:1177-1180. [PMID: 38592708 PMCID: PMC11226876 DOI: 10.1093/neuonc/noae063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Indexed: 04/10/2024] Open
Affiliation(s)
- Marjolein Geurts
- The Brain Tumour Center at the Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Matthias Preusser
- Division of Oncology, Department of Medicine 1, Medical University, Vienna, Austria
| |
Collapse
|
66
|
Shi Y, Kotchetkov IS, Dobrin A, Hanina SA, Rajasekhar VK, Healey JH, Sadelain M. GLUT1 overexpression enhances CAR T cell metabolic fitness and anti-tumor efficacy. Mol Ther 2024; 32:2393-2405. [PMID: 38720457 PMCID: PMC11286825 DOI: 10.1016/j.ymthe.2024.05.006] [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: 12/04/2023] [Revised: 04/02/2024] [Accepted: 05/03/2024] [Indexed: 05/20/2024] Open
Abstract
The tumor microenvironment presents many obstacles to effective chimeric antigen receptor (CAR) T cell therapy, including glucose competition from tumor and myeloid cells. Using mouse models of acute lymphoblastic leukemia (ALL), renal cell carcinoma (RCC), and glioblastoma (GBM), we show that enforced expression of the glucose transporter GLUT1 enhances anti-tumor efficacy and promotes favorable CAR-T cell phenotypes for two clinically relevant CAR designs, 19-28z and IL13Rα2-BBz. In the NALM6 ALL model, 19-28z-GLUT1 promotes T stem cell-like memory formation and prolongs survival. RNA sequencing of these CAR-T cells reveals that the overexpression of GLUT1, but not GLUT3, enriches for genes involved in glycolysis, mitochondrial respiration, and memory precursor phenotypes. Extending these data, 19-28z-GLUT1 CAR-T cells improve tumor control and response to rechallenge in an RCC patient-derived xenograft model. Furthermore, IL13Rα2-BBz CAR-T cells overexpressing GLUT1 prolong the survival of mice bearing orthotopic GBMs and exhibit decreased exhaustion markers. This novel engineering approach can offer a competitive advantage to CAR-T cells in harsh tumor environments where glucose is limiting.
Collapse
Affiliation(s)
- Yuzhe Shi
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ivan S Kotchetkov
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anton Dobrin
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sophie A Hanina
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Vinagolu K Rajasekhar
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - John H Healey
- Orthopedic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
67
|
Kuznetsova AV, Glukhova XA, Popova OP, Beletsky IP, Ivanov AA. Contemporary Approaches to Immunotherapy of Solid Tumors. Cancers (Basel) 2024; 16:2270. [PMID: 38927974 PMCID: PMC11201544 DOI: 10.3390/cancers16122270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
In recent years, the arrival of the immunotherapy industry has introduced the possibility of providing transformative, durable, and potentially curative outcomes for various forms of malignancies. However, further research has shown that there are a number of issues that significantly reduce the effectiveness of immunotherapy, especially in solid tumors. First of all, these problems are related to the protective mechanisms of the tumor and its microenvironment. Currently, major efforts are focused on overcoming protective mechanisms by using different adoptive cell therapy variants and modifications of genetically engineered constructs. In addition, a complex workforce is required to develop and implement these treatments. To overcome these significant challenges, innovative strategies and approaches are necessary to engineer more powerful variations of immunotherapy with improved antitumor activity and decreased toxicity. In this review, we discuss recent innovations in immunotherapy aimed at improving clinical efficacy in solid tumors, as well as strategies to overcome the limitations of various immunotherapies.
Collapse
Affiliation(s)
- Alla V. Kuznetsova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia
| | - Xenia A. Glukhova
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Olga P. Popova
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| | - Igor P. Beletsky
- Onni Biotechnologies Ltd., Aalto University Campus, Metallimiehenkuja 10, 02150 Espoo, Finland; (X.A.G.); (I.P.B.)
| | - Alexey A. Ivanov
- Laboratory of Molecular and Cellular Pathology, Russian University of Medicine (Formerly A.I. Evdokimov Moscow State University of Medicine and Dentistry), Ministry of Health of the Russian Federation, Bld 4, Dolgorukovskaya Str, 1127006 Moscow, Russia; (A.V.K.); (O.P.P.)
| |
Collapse
|
68
|
Sun Z, Wang C, Zhao Y, Ling Q. CAR-T cell therapy in advanced thyroid cancer: from basic to clinical. Front Immunol 2024; 15:1411300. [PMID: 38911868 PMCID: PMC11190081 DOI: 10.3389/fimmu.2024.1411300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/27/2024] [Indexed: 06/25/2024] Open
Abstract
The majority of patients with thyroid cancer can attain a favorable prognosis with a comprehensive treatment program based on surgical treatment. However, the current treatment options for advanced thyroid cancer are still limited. In recent years, chimeric antigen receptor-modified T-cell (CAR-T) therapy has received widespread attention in the field of oncology treatment. It has achieved remarkable results in the treatment of hematologic tumors. However, due to the constraints of multiple factors, the therapeutic efficacy of CAR-T therapy for solid tumors, including thyroid cancer, has not yet met expectations. This review outlines the fundamental structure and treatment strategies of CAR-T cells, provides an overview of the advancements in both preclinical investigations and clinical trials focusing on targets associated with CAR-T cell therapy in treating thyroid cancer, and discusses the challenges and solutions to CAR-T cell therapy for thyroid cancer. In conclusion, CAR-T cell therapy is a promising therapeutic approach for thyroid cancer, and we hope that our review will provide a timely and updated study of CAR-T cell therapy for thyroid cancer to advance the field.
Collapse
|
69
|
Wang X, Wang P, Liao Y, Zhao X, Hou R, Li S, Guan Z, Jin Y, Ma W, Liu D, Zheng J, Shi M. Expand available targets for CAR-T therapy to overcome tumor drug resistance based on the "Evolutionary Traps". Pharmacol Res 2024; 204:107221. [PMID: 38768669 DOI: 10.1016/j.phrs.2024.107221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/17/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Based on the concept of "Evolutionary Traps", targeting survival essential genes obtained during tumor drug resistance can effectively eliminate resistant cells. While, it still faces limitations. In this study, lapatinib-resistant cells were used to test the concept of "Evolutionary Traps" and no suitable target stand out because of the identified genes without accessible drug. However, a membrane protein PDPN, which is low or non-expressed in normal tissues, is identified as highly expressed in lapatinib-resistant tumor cells. PDPN CAR-T cells were developed and showed high cytotoxicity against lapatinib-resistant tumor cells in vitro and in vivo, suggesting that CAR-T may be a feasible route for overcoming drug resistance of tumor based on "Evolutionary Trap". To test whether this concept is cell line or drug dependent, we analyzed 21 drug-resistant tumor cell expression profiles reveal that JAG1, GPC3, and L1CAM, which are suitable targets for CAR-T treatment, are significantly upregulated in various drug-resistant tumor cells. Our findings shed light on the feasibility of utilizing CAR-T therapy to treat drug-resistant tumors and broaden the concept of the "Evolutionary Trap".
Collapse
Affiliation(s)
- Xu Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Pu Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Ying Liao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Xuan Zhao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Rui Hou
- College of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sijin Li
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Zhangchun Guan
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Yuhang Jin
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Wen Ma
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China
| | - Dan Liu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China; Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu 221002, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
70
|
Fowler D, Barisa M, Southern A, Nattress C, Hawkins E, Vassalou E, Kanouta A, Counsell J, Rota E, Vlckova P, Draper B, De Mooij T, Farkas A, Brezovjakova H, Baker AT, Scotlandi K, Manara MC, Tape C, Chester K, Anderson J, Fisher J. Payload-delivering engineered γδ T cells display enhanced cytotoxicity, persistence, and efficacy in preclinical models of osteosarcoma. Sci Transl Med 2024; 16:eadg9814. [PMID: 38809963 DOI: 10.1126/scitranslmed.adg9814] [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: 02/02/2023] [Accepted: 04/23/2024] [Indexed: 05/31/2024]
Abstract
T cell-based cancer immunotherapy has typically relied on membrane-bound cytotoxicity enhancers such as chimeric antigen receptors expressed in autologous αβ T cells. These approaches are limited by tonic signaling of synthetic constructs and costs associated with manufacturing. γδ T cells are an emerging alternative for cellular therapy, having innate antitumor activity, potent antibody-dependent cellular cytotoxicity, and minimal alloreactivity. We present an immunotherapeutic platform technology built around the innate properties of the Vγ9Vδ2 T cell, harnessing specific characteristics of this cell type and offering an allocompatible cellular therapy that recruits bystander immunity. We engineered γδ T cells to secrete synthetic tumor-targeting opsonins in the form of an scFv-Fc fusion protein and a mitogenic IL-15Rα-IL-15 fusion protein (stIL15). Using GD2 as a model antigen, we show that GD2-specific opsonin-secreting Vγ9Vδ2 T cells (stIL15-OPS-γδ T cells) have enhanced cytotoxicity and promote bystander activity of other lymphoid and myeloid cells. Secretion of stIL-15 abrogated the need for exogenous cytokine supplementation and further mediated activation of bystander natural killer cells. Compared with unmodified γδ T cells, stIL15-OPS-γδ T cells exhibited superior in vivo control of subcutaneous tumors and persistence in the blood. Moreover, stIL15-OPS-γδ T cells were efficacious against patient-derived osteosarcomas in animal models and in vitro, where efficacy could be boosted with the addition of zoledronic acid. Together, the data identify stIL15-OPS-γδ T cells as a candidate allogeneic cell therapy platform combining direct cytolysis with bystander activation to promote tumor control.
Collapse
Affiliation(s)
- Daniel Fowler
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Marta Barisa
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Alba Southern
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Callum Nattress
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Elizabeth Hawkins
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Eleni Vassalou
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Angeliki Kanouta
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | | | - Enrique Rota
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Petra Vlckova
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Benjamin Draper
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Tessa De Mooij
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Andrea Farkas
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Helena Brezovjakova
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Alfie T Baker
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Katia Scotlandi
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, Via di Barbiano 1/10, 40136 Bologna Italy
| | - Maria C Manara
- IRCCS Istituto Ortopedico Rizzoli, Experimental Oncology Laboratory, Via di Barbiano 1/10, 40136 Bologna Italy
| | - Chris Tape
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - Kerry Chester
- UCL Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, WC1E 6DD London, UK
| | - John Anderson
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| | - Jonathan Fisher
- UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research, 20 Guilford Street, WC1N 1DZ London, UK
| |
Collapse
|
71
|
Mohan N, Ayinde S, Peng H, Dutta S, Shen Y, Falkowski VM, Biel TG, Ju T, Wu WJ. Structural and functional characterization of IgG- and non-IgG-based T-cell-engaging bispecific antibodies. Front Immunol 2024; 15:1376096. [PMID: 38863707 PMCID: PMC11165055 DOI: 10.3389/fimmu.2024.1376096] [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: 01/24/2024] [Accepted: 05/06/2024] [Indexed: 06/13/2024] Open
Abstract
Bispecific T-cell-engaging antibodies are a growing class of therapeutics with numerous molecules being tested in clinical trials and, currently, seven of them have received market approval. They are structurally complex and function as adaptors to redirect the cytotoxicity of T cells to kill tumor cells. T-cell-engaging bispecific antibodies can be generally divided into two categories: IgG/IgG-like and non-IgG-like formats. Different formats may have different intrinsic potencies and physiochemical properties, and comprehensive studies are needed to gain a better understanding of how the differences in formats impact on structural and functional characteristics. In this study, we designed and generated bispecific T-cell-engaging antibodies with IgG-like (DVD-Ig) and non-IgG (BiTE) formats. Both target the same pair of antigens (EGFR and CD3) to minimize the possible influence of targets on functional characterization. We performed a side-by-side comparison to assess differences in the physiochemical and biological properties of these two bispecific T-cell-engaging antibodies using a variety of breast and ovarian cancer cell-based functional assays to delineate the structural-functional relationships and anti-tumor activities/potency. We found that the Fc portion of T-cell-engaging bispecific antibodies can significantly impact antigen binding activity, potency, and stability in addition to eliciting different mechanisms of action that contribute the killing of cancer cells.
Collapse
Affiliation(s)
- Nishant Mohan
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Safiat Ayinde
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Hanjing Peng
- Office of Pharmaceutical Manufacturing Assessment, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Shraboni Dutta
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Yi Shen
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Vincent M. Falkowski
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Thomas G. Biel
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Tongzhong Ju
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| | - Wen Jin Wu
- Office of Pharmaceutical Quality Research, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, United States
| |
Collapse
|
72
|
Sadowski K, Jażdżewska A, Kozłowski J, Zacny A, Lorenc T, Olejarz W. Revolutionizing Glioblastoma Treatment: A Comprehensive Overview of Modern Therapeutic Approaches. Int J Mol Sci 2024; 25:5774. [PMID: 38891962 PMCID: PMC11172387 DOI: 10.3390/ijms25115774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/21/2024] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in the adult population, with an average survival of 12.1 to 14.6 months. The standard treatment, combining surgery, radiotherapy, and chemotherapy, is not as efficient as we would like. However, the current possibilities are no longer limited to the standard therapies due to rapid advancements in biotechnology. New methods enable a more precise approach by targeting individual cells and antigens to overcome cancer. For the treatment of glioblastoma, these are gamma knife therapy, proton beam therapy, tumor-treating fields, EGFR and VEGF inhibitors, multiple RTKs inhibitors, and PI3K pathway inhibitors. In addition, the increasing understanding of the role of the immune system in tumorigenesis and the ability to identify tumor-specific antigens helped to develop immunotherapies targeting GBM and immune cells, including CAR-T, CAR-NK cells, dendritic cells, and immune checkpoint inhibitors. Each of the described methods has its advantages and disadvantages and faces problems, such as the inefficient crossing of the blood-brain barrier, various neurological and systemic side effects, and the escape mechanism of the tumor. This work aims to present the current modern treatments of glioblastoma.
Collapse
Affiliation(s)
- Karol Sadowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Adrianna Jażdżewska
- The Department of Anatomy and Neurobiology, Medical University of Gdansk, Dębinki 1, 80-211 Gdansk, Poland;
| | - Jan Kozłowski
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Aleksandra Zacny
- The Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland; (K.S.)
| | - Tomasz Lorenc
- Department of Radiology I, The Maria Sklodowska-Curie National Research Institute of Oncology, Roentgena 5, 02-781 Warsaw, Poland
| | - Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
73
|
Gordon KS, Perez CR, Garmilla A, Lam MSY, Aw JJ, Datta A, Lauffenburger DA, Pavesi A, Birnbaum ME. Pooled screening for CAR function identifies novel IL13Rα2-targeted CARs for treatment of glioblastoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.586240. [PMID: 38766252 PMCID: PMC11100612 DOI: 10.1101/2024.04.04.586240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Chimeric antigen receptor therapies have demonstrated potent efficacy in treating B cell malignancies, but have yet to meaningfully translate to solid tumors. Here, we utilize our pooled screening platform, CARPOOL, to expedite the discovery of CARs with anti-tumor functions necessary for solid tumor efficacy. We performed selections in primary human T cells expressing a library of 1.3×10 6 3 rd generation CARs targeting IL13Rα2, a cancer testis antigen commonly expressed in glioblastoma. Selections were performed for cytotoxicity, proliferation, memory formation, and persistence upon repeated antigen challenge. Each enriched CAR robustly produced the phenotype for which it was selected, and one enriched CAR triggered potent cytotoxicity and long-term proliferation upon in vitro tumor rechallenge. It also showed significantly improved persistence and comparable antigen-specific tumor control in a microphysiological human in vitro model and a xenograft model of human glioblastoma. Taken together, this work demonstrates the utility of extending CARPOOL to diseases beyond hematological malignancies and represents the largest exploration of signaling combinations in human primary cells to date.
Collapse
|
74
|
Martín-Antonio B, Blanco B, González-Murillo Á, Hidalgo L, Minguillón J, Pérez-Chacón G. Newer generations of multi-target CAR and STAb-T immunotherapeutics: NEXT CART Consortium as a cooperative effort to overcome current limitations. Front Immunol 2024; 15:1386856. [PMID: 38779672 PMCID: PMC11109416 DOI: 10.3389/fimmu.2024.1386856] [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: 02/16/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Adoptive T cellular immunotherapies have emerged as relevant approaches for treating cancer patients who have relapsed or become refractory (R/R) to traditional cancer treatments. Chimeric antigen receptor (CAR) T-cell therapy has improved survival in various hematological malignancies. However, significant limitations still impede the widespread adoption of these therapies in most cancers. To advance in this field, six research groups have created the "NEXT Generation CART MAD Consortium" (NEXT CART) in Madrid's Community, which aims to develop novel cell-based immunotherapies for R/R and poor prognosis cancers. At NEXT CART, various basic and translational research groups and hospitals in Madrid concur to share and synergize their basic expertise in immunotherapy, gene therapy, and immunological synapse, and clinical expertise in pediatric and adult oncology. NEXT CART goal is to develop new cell engineering approaches and treatments for R/R adult and pediatric neoplasms to evaluate in multicenter clinical trials. Here, we discuss the current limitations of T cell-based therapies and introduce our perspective on future developments. Advancement opportunities include developing allogeneic products, optimizing CAR signaling domains, combining cellular immunotherapies, multi-targeting strategies, and improving tumor-infiltrating lymphocytes (TILs)/T cell receptor (TCR) therapy. Furthermore, basic studies aim to identify novel tumor targets, tumor molecules in the tumor microenvironment that impact CAR efficacy, and strategies to enhance the efficiency of the immunological synapse between immune and tumor cells. Our perspective of current cellular immunotherapy underscores the potential of these treatments while acknowledging the existing hurdles that demand innovative solutions to develop their potential for cancer treatment fully.
Collapse
Affiliation(s)
- Beatriz Martín-Antonio
- Department of Experimental Hematology, Instituto de Investigación Sanitaria-Fundación Jiménez Diaz (IIS-FJD), Madrid, Spain
| | - Belén Blanco
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - África González-Murillo
- Department of Pediatric Hematology and Oncology, Advanced Therapies Unit, Fundación Investigación Biomédica Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Laura Hidalgo
- Cellular Biotechnology Unit, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jordi Minguillón
- La Paz Hospital Institute for Health Research (IdiPAZ), Hospital Universitario La Paz. Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Gema Pérez-Chacón
- Immunity, Immunopathology and Emergent Therapies Group. Instituto de Investigaciones Biomedicas Sols-Morreale. CSIC-UAM, Madrid, Spain
| |
Collapse
|
75
|
Vazaios K, van Berkum RE, Calkoen FG, van der Lugt J, Hulleman E. OV Modulators of the Paediatric Brain TIME: Current Status, Combination Strategies, Limitations and Future Directions. Int J Mol Sci 2024; 25:5007. [PMID: 38732225 PMCID: PMC11084613 DOI: 10.3390/ijms25095007] [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: 03/12/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Oncolytic viruses (OVs) are characterised by their preference for infecting and replicating in tumour cells either naturally or after genetic modification, resulting in oncolysis. Furthermore, OVs can elicit both local and systemic anticancer immune responses while specifically infecting and lysing tumour cells. These characteristics render them a promising therapeutic approach for paediatric brain tumours (PBTs). PBTs are frequently marked by a cold tumour immune microenvironment (TIME), which suppresses immunotherapies. Recent preclinical and clinical studies have demonstrated the capability of OVs to induce a proinflammatory immune response, thereby modifying the TIME. In-depth insights into the effect of OVs on different cell types in the TIME may therefore provide a compelling basis for using OVs in combination with other immunotherapy modalities. However, certain limitations persist in our understanding of oncolytic viruses' ability to regulate the TIME to enhance anti-tumour activity. These limitations primarily stem from the translational limitations of model systems, the difficulties associated with tracking reliable markers of efficacy throughout the course of treatment and the role of pre-existing viral immunity. In this review, we describe the different alterations observed in the TIME in PBTs due to OV treatment, combination therapies of OVs with different immunotherapies and the hurdles limiting the development of effective OV therapies while suggesting future directions based on existing evidence.
Collapse
Affiliation(s)
| | | | | | | | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (K.V.); (F.G.C.); (J.v.d.L.)
| |
Collapse
|
76
|
Wang AF, Hsueh B, Choi BD, Gerstner ER, Dunn GP. Immunotherapy for Brain Tumors: Where We Have Been, and Where Do We Go From Here? Curr Treat Options Oncol 2024; 25:628-643. [PMID: 38649630 DOI: 10.1007/s11864-024-01200-9] [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] [Accepted: 03/18/2024] [Indexed: 04/25/2024]
Abstract
OPINION STATEMENT Immunotherapy for glioblastoma (GBM) remains an intensive area of investigation. Given the seismic impact of cancer immunotherapy across a range of malignancies, there is optimism that harnessing the power of immunity will influence GBM as well. However, despite several phase 3 studies, there are still no FDA-approved immunotherapies for GBM. Importantly, the field has learned a great deal from the randomized studies to date. Today, we are continuing to better understand the disease-specific features of the microenvironment in GBM-as well as the exploitable antigenic characteristic of the tumor cells themselves-that are informing the next generation of immune-based therapeutic strategies. The coming phase of next-generation immunotherapies is thus poised to bring us closer to treatments that will improve the lives of patients with GBM.
Collapse
Affiliation(s)
- Alexander F Wang
- Department of Neurosurgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Brian Hsueh
- Department of Neurosurgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
| | - Bryan D Choi
- Department of Neurosurgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA
- Brain Tumor Immunology and Immunotherapy Program, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Elizabeth R Gerstner
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Stephen E. and Catherine Pappas Center for Neuro-Oncology, Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Gavin P Dunn
- Department of Neurosurgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, USA.
- Brain Tumor Immunology and Immunotherapy Program, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
77
|
Wehrli M, Guinn S, Birocchi F, Kuo A, Sun Y, Larson RC, Almazan AJ, Scarfò I, Bouffard AA, Bailey SR, Anekal PV, Llopis PM, Nieman LT, Song Y, Xu KH, Berger TR, Kann MC, Leick MB, Silva H, Salas-Benito D, Kienka T, Grauwet K, Armstrong TD, Zhang R, Zhu Q, Fu J, Schmidts A, Korell F, Jan M, Choi BD, Liss AS, Boland GM, Ting DT, Burkhart RA, Jenkins RW, Zheng L, Jaffee EM, Zimmerman JW, Maus MV. Mesothelin CAR T Cells Secreting Anti-FAP/Anti-CD3 Molecules Efficiently Target Pancreatic Adenocarcinoma and its Stroma. Clin Cancer Res 2024; 30:1859-1877. [PMID: 38393682 PMCID: PMC11062832 DOI: 10.1158/1078-0432.ccr-23-3841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 02/25/2024]
Abstract
PURPOSE Targeting solid tumors with chimeric antigen receptor (CAR) T cells remains challenging due to heterogenous target antigen expression, antigen escape, and the immunosuppressive tumor microenvironment (TME). Pancreatic cancer is characterized by a thick stroma generated by cancer-associated fibroblasts (CAF), which may contribute to the limited efficacy of mesothelin-directed CAR T cells in early-phase clinical trials. To provide a more favorable TME for CAR T cells to target pancreatic ductal adenocarcinoma (PDAC), we generated T cells with an antimesothelin CAR and a secreted T-cell-engaging molecule (TEAM) that targets CAF through fibroblast activation protein (FAP) and engages T cells through CD3 (termed mesoFAP CAR-TEAM cells). EXPERIMENTAL DESIGN Using a suite of in vitro, in vivo, and ex vivo patient-derived models containing cancer cells and CAF, we examined the ability of mesoFAP CAR-TEAM cells to target PDAC cells and CAF within the TME. We developed and used patient-derived ex vivo models, including patient-derived organoids with patient-matched CAF and patient-derived organotypic tumor spheroids. RESULTS We demonstrated specific and significant binding of the TEAM to its respective antigens (CD3 and FAP) when released from mesothelin-targeting CAR T cells, leading to T-cell activation and cytotoxicity of the target cell. MesoFAP CAR-TEAM cells were superior in eliminating PDAC and CAF compared with T cells engineered to target either antigen alone in our ex vivo patient-derived models and in mouse models of PDAC with primary or metastatic liver tumors. CONCLUSIONS CAR-TEAM cells enable modification of tumor stroma, leading to increased elimination of PDAC tumors. This approach represents a promising treatment option for pancreatic cancer.
Collapse
Affiliation(s)
- Marc Wehrli
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Samantha Guinn
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Filippo Birocchi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Adam Kuo
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yi Sun
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Rebecca C. Larson
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Antonio J. Almazan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Irene Scarfò
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Amanda A. Bouffard
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Stefanie R. Bailey
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | | | | | - Linda T. Nieman
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Yuhui Song
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Katherine H. Xu
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Trisha R. Berger
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Michael C. Kann
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Mark B. Leick
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Harrison Silva
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Diego Salas-Benito
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Tamina Kienka
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Korneel Grauwet
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Todd D. Armstrong
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Rui Zhang
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Qingfeng Zhu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Juan Fu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Andrea Schmidts
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Max Jan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Bryan D. Choi
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - Andrew S. Liss
- Division of Gastrointestinal and Oncologic Surgery, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Genevieve M. Boland
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School; Boston, MA, USA
| | - David T. Ting
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Richard A. Burkhart
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Russell W. Jenkins
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Elizabeth M. Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Jacquelyn W. Zimmerman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University; Baltimore, MD, USA
- Cancer Convergence Institute and Bloomberg Kimmel Institute at Johns Hopkins; University, Baltimore, MD, USA
| | - Marcela V. Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Cancer Center, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
- Blood and Marrow Transplant Program, Massachusetts General Hospital; Harvard Medical School; Boston, MA, USA
| |
Collapse
|
78
|
Yang Z, Hou Y, Grande G, Cho JH, Wang C, Shi Y, Zak J, Wan Y, Qin K, Liu D, Teijaro JR, Lerner RA, Wu P. Targeted desialylation and cytolysis of tumour cells by fusing a sialidase to a bispecific T-cell engager. Nat Biomed Eng 2024; 8:499-512. [PMID: 38693431 PMCID: PMC11577304 DOI: 10.1038/s41551-024-01202-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 03/26/2024] [Indexed: 05/03/2024]
Abstract
Bispecific T-cell engagers (BiTEs) bring together tumour cells and cytotoxic T cells by binding to specific cell-surface tumour antigens and T-cell receptors, and have been clinically successful for the treatment of B-cell malignancies. Here we show that a BiTE-sialidase fusion protein enhances the susceptibility of solid tumours to BiTE-mediated cytolysis of tumour cells via targeted desialylation-that is, the removal of terminal sialic acid residues on glycans-at the BiTE-induced T-cell-tumour-cell interface. In xenograft and syngeneic mouse models of leukaemia and of melanoma and breast cancer, and compared with the parental BiTE molecules, targeted desialylation via the BiTE-sialidase fusion proteins enhanced the formation of immunological synapses, T-cell activation and T-cell-mediated tumour-cell cytolysis in the presence of the target antigen. The targeted desialylation of tumour cells may enhance the potency of therapies relying on T-cell engagers.
Collapse
Affiliation(s)
- Zhuo Yang
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Yingqin Hou
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Geramie Grande
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Jong Hyun Cho
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - Chao Wang
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Yujie Shi
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jaroslav Zak
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Yue Wan
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Ke Qin
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Rutgers University-New Jersey Medical School, Newark, NJ, USA
| | - John R Teijaro
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Richard A Lerner
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Peng Wu
- Department of Molecular and Cellular Biology, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
79
|
Long X, Zhang Z, Li Y, Deng K, Gao W, Huang M, Wang X, Lin X, She X, Zhao Y, Zhang M, Huang C, Wang S, Du Y, Du P, Chen S, Liu Q, Wu M. ScRNA-seq reveals novel immune-suppressive T cells and investigates CMV-TCR-T cells cytotoxicity against GBM. J Immunother Cancer 2024; 12:e008967. [PMID: 38688579 PMCID: PMC11086384 DOI: 10.1136/jitc-2024-008967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a fatal primary brain malignancy in adults. Previous studies have shown that cytomegalovirus (CMV) is a risk factor for tumorigenesis and aggressiveness for glioblastoma. However, little is known about how CMV infection affects immune cells in the tumor microenvironment of GBM. Furthermore, there has been almost no engineered T-cell receptor (TCR)-T targeting CMV for GBM research to date. METHODS We evaluated the CMV infection status of patients with GBM's tumor tissue by immune electron microscopy, immunofluorescence, and droplet digital PCR. We performed single-cell RNA sequencing for CMV-infected GBM to investigate the effects of CMV on the GBM immune microenvironment. CellChat was applied to analyze the interaction between cells in the GBM tumor microenvironment. Additionally, we conducted single-cell TCR/B cell receptor (BCR) sequencing and Grouping of Lymphocyte Interactions with Paratope Hotspots 2 algorithms to acquire specific CMV-TCR sequences. Genetic engineering was used to introduce CMV-TCR into primary T cells derived from patients with CMV-infected GBM. Flow cytometry was used to measure the proportion and cytotoxicity status of T cells in vitro. RESULTS We identified two novel immune cell subpopulations in CMV-infected GBM, which were bipositive CD68+SOX2+ tumor-associated macrophages and FXYD6+ T cells. We highlighted that the interaction between bipositive TAMs or cancer cells and T cells was predominantly focused on FXYD6+ T cells rather than regulatory T cells (Tregs), whereas, FXYD6+ T cells were further identified as a group of novel immunosuppressive T cells. CMV-TCR-T cells showed significant therapeutic effects on the human-derived orthotopic GBM mice model. CONCLUSIONS These findings provided an insight into the underlying mechanism of CMV infection promoting the GBM immunosuppression, and provided a novel potential immunotherapy strategy for patients with GBM.
Collapse
Affiliation(s)
- Xinmiao Long
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Zuping Zhang
- Department of Pathogeny Biology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Yuzhe Li
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, Hunan, China
- Chinese Academy of Medical Sciences and Peking Union Medical College, Peking University, Beijing, China
| | - Kun Deng
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Wei Gao
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Meng Huang
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, Hunan, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China Medical Research Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiangyu Wang
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Lin
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiaoling She
- Department of Pathology in Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yiming Zhao
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathogeny Biology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Minfu Zhang
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cheng Huang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Shiyi Wang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Yinfei Du
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| | - Peng Du
- Department of Neurosurgery, The Second Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Shuai Chen
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qing Liu
- Department of Neurosurgery in Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Minghua Wu
- Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
- FuRong Laboratory, Central South University, Changsha, Hunan, China
| |
Collapse
|
80
|
Mitra A, Kumar A, Amdare NP, Pathak R. Current Landscape of Cancer Immunotherapy: Harnessing the Immune Arsenal to Overcome Immune Evasion. BIOLOGY 2024; 13:307. [PMID: 38785789 PMCID: PMC11118874 DOI: 10.3390/biology13050307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Cancer immune evasion represents a leading hallmark of cancer, posing a significant obstacle to the development of successful anticancer therapies. However, the landscape of cancer treatment has significantly evolved, transitioning into the era of immunotherapy from conventional methods such as surgical resection, radiotherapy, chemotherapy, and targeted drug therapy. Immunotherapy has emerged as a pivotal component in cancer treatment, harnessing the body's immune system to combat cancer and offering improved prognostic outcomes for numerous patients. The remarkable success of immunotherapy has spurred significant efforts to enhance the clinical efficacy of existing agents and strategies. Several immunotherapeutic approaches have received approval for targeted cancer treatments, while others are currently in preclinical and clinical trials. This review explores recent progress in unraveling the mechanisms of cancer immune evasion and evaluates the clinical effectiveness of diverse immunotherapy strategies, including cancer vaccines, adoptive cell therapy, and antibody-based treatments. It encompasses both established treatments and those currently under investigation, providing a comprehensive overview of efforts to combat cancer through immunological approaches. Additionally, the article emphasizes the current developments, limitations, and challenges in cancer immunotherapy. Furthermore, by integrating analyses of cancer immunotherapy resistance mechanisms and exploring combination strategies and personalized approaches, it offers valuable insights crucial for the development of novel anticancer immunotherapeutic strategies.
Collapse
Affiliation(s)
- Ankita Mitra
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, Uttar Pradesh, India
| | - Nitin P. Amdare
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
81
|
Montoya M, Gallus M, Phyu S, Haegelin J, de Groot J, Okada H. A Roadmap of CAR-T-Cell Therapy in Glioblastoma: Challenges and Future Perspectives. Cells 2024; 13:726. [PMID: 38727262 PMCID: PMC11083543 DOI: 10.3390/cells13090726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/20/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
Glioblastoma (GBM) is the most common primary malignant brain tumor, with a median overall survival of less than 2 years and a nearly 100% mortality rate under standard therapy that consists of surgery followed by combined radiochemotherapy. Therefore, new therapeutic strategies are urgently needed. The success of chimeric antigen receptor (CAR) T cells in hematological cancers has prompted preclinical and clinical investigations into CAR-T-cell treatment for GBM. However, recent trials have not demonstrated any major success. Here, we delineate existing challenges impeding the effectiveness of CAR-T-cell therapy for GBM, encompassing the cold (immunosuppressive) microenvironment, tumor heterogeneity, T-cell exhaustion, local and systemic immunosuppression, and the immune privilege inherent to the central nervous system (CNS) parenchyma. Additionally, we deliberate on the progress made in developing next-generation CAR-T cells and novel innovative approaches, such as low-intensity pulsed focused ultrasound, aimed at surmounting current roadblocks in GBM CAR-T-cell therapy.
Collapse
Affiliation(s)
- Megan Montoya
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Marco Gallus
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Su Phyu
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Jeffrey Haegelin
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - John de Groot
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
| | - Hideho Okada
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94158, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94129, USA
| |
Collapse
|
82
|
Dobersberger M, Sumesgutner D, Zajc CU, Salzer B, Laurent E, Emminger D, Sylvander E, Lehner E, Teufl M, Seigner J, Bobbili MR, Kunert R, Lehner M, Traxlmayr MW. An engineering strategy to target activated EGFR with CAR T cells. CELL REPORTS METHODS 2024; 4:100728. [PMID: 38492569 PMCID: PMC11045874 DOI: 10.1016/j.crmeth.2024.100728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/18/2024] [Accepted: 02/16/2024] [Indexed: 03/18/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have shown remarkable response rates in hematological malignancies. In contrast, CAR T cell treatment of solid tumors is associated with several challenges, in particular the expression of most tumor-associated antigens at lower levels in vital organs, resulting in on-target/off-tumor toxicities. Thus, innovative approaches to improve the tumor specificity of CAR T cells are urgently needed. Based on the observation that many human solid tumors activate epidermal growth factor receptor (EGFR) on their surface through secretion of EGFR ligands, we developed an engineering strategy for CAR-binding domains specifically directed against the ligand-activated conformation of EGFR. We show, in several experimental systems, that the generated binding domains indeed enable CAR T cells to distinguish between active and inactive EGFR. We anticipate that this engineering concept will be an important step forward to improve the tumor specificity of CAR T cells directed against EGFR-positive solid cancers.
Collapse
Affiliation(s)
- Markus Dobersberger
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria
| | - Delia Sumesgutner
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Charlotte U Zajc
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Benjamin Salzer
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria
| | - Elisabeth Laurent
- BOKU Core Facility Biomolecular & Cellular Analysis, BOKU University, 1190 Vienna, Austria
| | - Dominik Emminger
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria
| | - Elise Sylvander
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria
| | - Elisabeth Lehner
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Magdalena Teufl
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria
| | - Jacqueline Seigner
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; Department of Biotechnology, Institute of Animal Cell Technology and Systems Biology, BOKU University, 1190 Vienna, Austria
| | - Madhusudhan Reddy Bobbili
- Department of Biotechnology, Institute of Molecular Biotechnology, BOKU University, 1190 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, Research Center in Cooperation with AUVA, 1200 Vienna, Austria
| | - Renate Kunert
- Department of Biotechnology, Institute of Animal Cell Technology and Systems Biology, BOKU University, 1190 Vienna, Austria
| | - Manfred Lehner
- CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria; St. Anna Children's Cancer Research Institute, CCRI, 1090 Vienna, Austria; St. Anna Children's Hospital, Department of Pediatrics, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael W Traxlmayr
- Department of Chemistry, Institute of Biochemistry, BOKU University, 1190 Vienna, Austria; CD Laboratory for Next Generation CAR T Cells, 1090 Vienna, Austria.
| |
Collapse
|
83
|
Cech P, Skórka K, Dziki L, Giannopoulos K. T-Cell Engagers-The Structure and Functional Principle and Application in Hematological Malignancies. Cancers (Basel) 2024; 16:1580. [PMID: 38672662 PMCID: PMC11048836 DOI: 10.3390/cancers16081580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Recent advancements in cancer immunotherapy have made directing the cellular immune response onto cancer cells a promising strategy for the treatment of hematological malignancies. The introduction of monoclonal antibody-based (mAbs) targeted therapy has significantly improved the prognosis for hematological patients. Facing the issues of mAb-based therapies, a novel bispecific antibody (BsAb) format was developed. T-cell engagers (TCEs) are BsAbs, which simultaneously target tumor-associated antigens on tumor cells and CD3 molecules present on T-cells. This mechanism allows for the direct activation of T-cells and their anti-tumor features, ultimately resulting in the lysis of tumor cells. In 2014, the FDA approved blinatumomab, a TCE directed to CD3 and CD19 for treatment of acute lymphoblastic leukemia. Since then, numerous TCEs have been developed, allowing for treating different hematological malignancies such as acute myeloid leukemia, multiple myeloma, and non-Hodgkin lymphoma and Hodgkin lymphoma. As of November 2023, seven clinically approved TCE therapies are on the market. TCE-based therapies still have their limitations; however, improving the properties of TCEs, as well as combining TCE-based therapies with other forms of treatment, give hope to find the cures for currently terminal diseases. In this paper, we summarized the technical basis of the TCE technology, its application in hematology, and its current issues and prospects.
Collapse
Affiliation(s)
| | - Katarzyna Skórka
- Department of Experimental Hematooncology, Medical University of Lublin, 20-093 Lublin, Poland; (P.C.); (L.D.); (K.G.)
| | | | | |
Collapse
|
84
|
Tapia-Galisteo A, Sánchez-Rodríguez I, Narbona J, Iglesias-Hernández P, Aragón-García S, Jiménez-Reinoso A, Compte M, Khan S, Tsuda T, Chames P, Lacadena J, Álvarez-Vallina L, Sanz L. Combination of T cell-redirecting strategies with a bispecific antibody blocking TGF-β and PD-L1 enhances antitumor responses. Oncoimmunology 2024; 13:2338558. [PMID: 38623463 PMCID: PMC11018002 DOI: 10.1080/2162402x.2024.2338558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/30/2024] [Indexed: 04/17/2024] Open
Abstract
T cell-based immunotherapies for solid tumors have not achieved the clinical success observed in hematological malignancies, partially due to the immunosuppressive effect promoted by the tumor microenvironment, where PD-L1 and TGF-β play a pivotal role. However, durable responses to immune checkpoint inhibitors remain limited to a minority of patients, while TGF-β inhibitors have not reached the market yet. Here, we describe a bispecific antibody for dual blockade of PD-L1 and TFG-β, termed AxF (scFv)2, under the premise that combination with T cell redirecting strategies would improve clinical benefit. The AxF (scFv)2 antibody was well expressed in mammalian and yeast cells, bound both targets and inhibited dose-dependently the corresponding signaling pathways in luminescence-based cellular reporter systems. Moreover, combined treatment with trispecific T-cell engagers (TriTE) or CAR-T cells significantly boosted T cell activation status and cytotoxic response in breast, lung and colorectal (CRC) cancer models. Importantly, the combination of an EpCAMxCD3×EGFR TriTE with the AxF (scFv)2 delayed CRC tumor growth in vivo and significantly enhanced survival compared to monotherapy with the trispecific antibody. In summary, we demonstrated the feasibility of concomitant blockade of PD-L1 and TGF-β by a single molecule, as well as its therapeutic potential in combination with different T cell redirecting agents to overcome tumor microenvironment-mediated immunosuppression.
Collapse
Affiliation(s)
- Antonio Tapia-Galisteo
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Iñigo Sánchez-Rodríguez
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Javier Narbona
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Patricia Iglesias-Hernández
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Saray Aragón-García
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Anaïs Jiménez-Reinoso
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Marta Compte
- Department of Antibody Engineering, Leadartis SL, Madrid, Spain
| | - Shaukat Khan
- Nemours Children’s Health Delaware, Wilmington, Delaware, USA
| | - Takeshi Tsuda
- Nemours Children’s Health Delaware, Wilmington, Delaware, USA
| | - Patrick Chames
- Aix Marseille Univ, CNRS, INSERM, Institute Paoli-Calmettes, CRCM, Marseille, France
| | - Javier Lacadena
- Department of Biochemistry and Molecular Biology, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Álvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Hospital Universitario 12 de Octubre, Madrid, Spain
- Immuno-oncology and Immunotherapy Group, Biomedical Research Institute Hospital 12 de Octubre, Madrid, Spain
- H12O-CNIO Cancer Immunotherapy Clinical Research Unit, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Laura Sanz
- Molecular Immunology Unit, Biomedical Research Institute Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
85
|
Jenkins MR, Drummond KJ. CAR T-Cell Therapy for Glioblastoma. N Engl J Med 2024; 390:1329-1332. [PMID: 38598802 DOI: 10.1056/nejme2401307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Affiliation(s)
- Misty R Jenkins
- From the Immunology Division, Walter and Eliza Hall Institute, and the Department of Medical Biology, University of Melbourne, Parkville, VIC (M.R.J.), and the Department of Surgery, University of Melbourne, Melbourne, VIC (K.J.D.) - all in Australia
| | - Katharine J Drummond
- From the Immunology Division, Walter and Eliza Hall Institute, and the Department of Medical Biology, University of Melbourne, Parkville, VIC (M.R.J.), and the Department of Surgery, University of Melbourne, Melbourne, VIC (K.J.D.) - all in Australia
| |
Collapse
|
86
|
Choi BD, Gerstner ER, Frigault MJ, Leick MB, Mount CW, Balaj L, Nikiforow S, Carter BS, Curry WT, Gallagher K, Maus MV. Intraventricular CARv3-TEAM-E T Cells in Recurrent Glioblastoma. N Engl J Med 2024; 390:1290-1298. [PMID: 38477966 PMCID: PMC11162836 DOI: 10.1056/nejmoa2314390] [Citation(s) in RCA: 121] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
In this first-in-human, investigator-initiated, open-label study, three participants with recurrent glioblastoma were treated with CARv3-TEAM-E T cells, which are chimeric antigen receptor (CAR) T cells engineered to target the epidermal growth factor receptor (EGFR) variant III tumor-specific antigen, as well as the wild-type EGFR protein, through secretion of a T-cell-engaging antibody molecule (TEAM). Treatment with CARv3-TEAM-E T cells did not result in adverse events greater than grade 3 or dose-limiting toxic effects. Radiographic tumor regression was dramatic and rapid, occurring within days after receipt of a single intraventricular infusion, but the responses were transient in two of the three participants. (Funded by Gateway for Cancer Research and others; INCIPIENT ClinicalTrials.gov number, NCT05660369.).
Collapse
Affiliation(s)
- Bryan D Choi
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Elizabeth R Gerstner
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Matthew J Frigault
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Mark B Leick
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Christopher W Mount
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Leonora Balaj
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Sarah Nikiforow
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Bob S Carter
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - William T Curry
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Kathleen Gallagher
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| | - Marcela V Maus
- From the Cellular Immunotherapy Program (B.D.C., M.J.F., M.B.L., C.W.M., K.G., M.V.M.) and Krantz Family Center for Cancer Research (M.B.L., K.G., M.V.M.), Mass General Cancer Center, and the Departments of Neurology (E.R.G.), Pathology (C.W.M., K.G.), Neurosurgery (B.D.C., L.B., B.S.C., W.T.C.), and Medicine (M.J.F., M.B.L., M.V.M.), Massachusetts General Hospital and Harvard Medical School, and Connell and O'Reilly Families Cell Manipulation Core Facility, Dana-Farber/Harvard Cancer Center (S.N.) - both in Boston
| |
Collapse
|
87
|
Chen M, Zhang J, Li H, Deng Y, Huang Y, Shen W, Zeng Y, Ci T. Engineered platelet-based immune engager for tumor post-surgery treatment. BIOMATERIALS ADVANCES 2024; 158:213796. [PMID: 38342024 DOI: 10.1016/j.bioadv.2024.213796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/13/2024]
Abstract
Tumor metastasis and recurrence are principal reasons for the high mortality and poor prognosis of cancers. Inefficient engagement between T cell and tumor cell, as well as the universal existence of immune checkpoints, are important factors to the limited immunological surveillance of the immune systems to tumor cells. Herein, an immune engager based on engineered platelets with CD3 antibody modification (P-aCD3) was constructed to facilitate the contact between T cell and tumor cell via providing the anchoring sites of above two cells. Combined with the immune checkpoint blockade strategy, P-aCD3 effectively enhanced T cell mediated cytotoxicity and inhibited tumor recurrence and metastasis in mice melanoma postoperative model and breast cancer model, resulting in significantly prolonged survival of mice.
Collapse
Affiliation(s)
- Mo Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Jinniu Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huangjuan Li
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Yueyang Deng
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, Jiangsu Province 210009, China
| | - Yun Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenhao Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yixing Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tianyuan Ci
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
88
|
Kembuan GJ, Kim JY, Maus MV, Jan M. Targeting solid tumor antigens with chimeric receptors: cancer biology meets synthetic immunology. Trends Cancer 2024; 10:312-331. [PMID: 38355356 PMCID: PMC11006585 DOI: 10.1016/j.trecan.2024.01.003] [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: 12/05/2022] [Revised: 01/02/2024] [Accepted: 01/05/2024] [Indexed: 02/16/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a medical breakthrough in the treatment of B cell malignancies. There is intensive focus on developing solid tumor-targeted CAR-T cell therapies. Although clinically approved CAR-T cell therapies target B cell lineage antigens, solid tumor targets include neoantigens and tumor-associated antigens (TAAs) with diverse roles in tumor biology. Multiple early-stage clinical trials now report encouraging signs of efficacy for CAR-T cell therapies that target solid tumors. We review the landscape of solid tumor target antigens from the perspective of cancer biology and gene regulation, together with emerging clinical data for CAR-T cells targeting these antigens. We then discuss emerging synthetic biology strategies and their application in the clinical development of novel cellular immunotherapies.
Collapse
Affiliation(s)
- Gabriele J Kembuan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Joanna Y Kim
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Max Jan
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, USA; Harvard Medical School, Boston, MA, USA; Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
89
|
Klein C, Brinkmann U, Reichert JM, Kontermann RE. The present and future of bispecific antibodies for cancer therapy. Nat Rev Drug Discov 2024; 23:301-319. [PMID: 38448606 DOI: 10.1038/s41573-024-00896-6] [Citation(s) in RCA: 114] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/22/2024] [Indexed: 03/08/2024]
Abstract
Bispecific antibodies (bsAbs) enable novel mechanisms of action and/or therapeutic applications that cannot be achieved using conventional IgG-based antibodies. Consequently, development of these molecules has garnered substantial interest in the past decade and, as of the end of 2023, 14 bsAbs have been approved: 11 for the treatment of cancer and 3 for non-oncology indications. bsAbs are available in different formats, address different targets and mediate anticancer function via different molecular mechanisms. Here, we provide an overview of recent developments in the field of bsAbs for cancer therapy. We focus on bsAbs that are approved or in clinical development, including bsAb-mediated dual modulators of signalling pathways, tumour-targeted receptor agonists, bsAb-drug conjugates, bispecific T cell, natural killer cell and innate immune cell engagers, and bispecific checkpoint inhibitors and co-stimulators. Finally, we provide an outlook into next-generation bsAbs in earlier stages of development, including trispecifics, bsAb prodrugs, bsAbs that induce degradation of tumour targets and bsAbs acting as cytokine mimetics.
Collapse
Affiliation(s)
- Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich, Schlieren, Switzerland.
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University Stuttgart, Stuttgart, Germany.
| |
Collapse
|
90
|
Yang JL, Yamada-Hunter SA, Labanieh L, Sotillo E, Cheah JS, Roberts DS, Mackall CL, Bertozzi CR, Ting AY. Directed evolution of genetically encoded LYTACs for cell-mediated delivery. Proc Natl Acad Sci U S A 2024; 121:e2320053121. [PMID: 38513100 PMCID: PMC10990137 DOI: 10.1073/pnas.2320053121] [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: 11/15/2023] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Lysosome-targeting chimeras (LYTACs) are a promising therapeutic modality to drive the degradation of extracellular proteins. However, early versions of LYTAC contain synthetic glycopeptides that cannot be genetically encoded. Here, we present our designs for a fully genetically encodable LYTAC (GELYTAC), making our tool compatible with integration into therapeutic cells for targeted delivery at diseased sites. To achieve this, we replaced the glycopeptide portion of LYTACs with the protein insulin-like growth factor 2 (IGF2). After showing initial efficacy with wild-type IGF2, we increased the potency of GELYTAC using directed evolution. Subsequently, we demonstrated that our engineered GELYTAC construct not only secretes from HEK293T cells but also from human primary T-cells to drive the uptake of various targets into receiver cells. Immune cells engineered to secrete GELYTAC thus represent a promising avenue for spatially selective targeted protein degradation.
Collapse
Affiliation(s)
- Jonathan Lee Yang
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Sean A. Yamada-Hunter
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
| | - Louai Labanieh
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Bioengineering, Stanford University, Stanford, CA94305
- Parker Institute for Cancer Immunotherapy, San Francisco, CA94305
| | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
- Department of Bioengineering, Stanford University, Stanford, CA94305
| | - Joleen S. Cheah
- Department of Biology, Stanford University, Stanford, CA94305
| | - David S. Roberts
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
| | - Crystal L. Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA94305
- Parker Institute for Cancer Immunotherapy, San Francisco, CA94305
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA94305
- Department of Medicine, Stanford University School of Medicine, Stanford, CA94305
| | - Carolyn R. Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- HHMI, Stanford University, Stanford, CA94305
| | - Alice Y. Ting
- Department of Chemistry, Stanford University, Stanford, CA94305
- Sarafan ChEM-H, Stanford University, Stanford, CA94305
- Department of Biology, Stanford University, Stanford, CA94305
- Department of Genetics, Stanford University, Stanford, CA94305
- Chan Zuckerberg Biohub-San Francisco, San Francisco, CA94158
| |
Collapse
|
91
|
Salvato I, Marchini A. Immunotherapeutic Strategies for the Treatment of Glioblastoma: Current Challenges and Future Perspectives. Cancers (Basel) 2024; 16:1276. [PMID: 38610954 PMCID: PMC11010873 DOI: 10.3390/cancers16071276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/14/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Despite decades of research and the best up-to-date treatments, grade 4 Glioblastoma (GBM) remains uniformly fatal with a patient median overall survival of less than 2 years. Recent advances in immunotherapy have reignited interest in utilizing immunological approaches to fight cancer. However, current immunotherapies have so far not met the anticipated expectations, achieving modest results in their journey from bench to bedside for the treatment of GBM. Understanding the intrinsic features of GBM is of crucial importance for the development of effective antitumoral strategies to improve patient life expectancy and conditions. In this review, we provide a comprehensive overview of the distinctive characteristics of GBM that significantly influence current conventional therapies and immune-based approaches. Moreover, we present an overview of the immunotherapeutic strategies currently undergoing clinical evaluation for GBM treatment, with a specific emphasis on those advancing to phase 3 clinical studies. These encompass immune checkpoint inhibitors, adoptive T cell therapies, vaccination strategies (i.e., RNA-, DNA-, and peptide-based vaccines), and virus-based approaches. Finally, we explore novel innovative strategies and future prospects in the field of immunotherapy for GBM.
Collapse
Affiliation(s)
- Ilaria Salvato
- NORLUX Neuro-Oncology Laboratory, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg;
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine (FSTM), University of Luxembourg, L-4367 Belvaux, Luxembourg
| | - Antonio Marchini
- Laboratory of Oncolytic Virus Immuno-Therapeutics (LOVIT), Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Laboratory of Oncolytic Virus Immuno-Therapeutics, German Cancer Research Center, 69120 Heidelberg, Germany
| |
Collapse
|
92
|
Dougé A, El Ghazzi N, Lemal R, Rouzaire P. Adoptive T Cell Therapy in Solid Tumors: State-of-the Art, Current Challenges, and Upcoming Improvements. Mol Cancer Ther 2024; 23:272-284. [PMID: 37903371 DOI: 10.1158/1535-7163.mct-23-0310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/18/2023] [Accepted: 10/25/2023] [Indexed: 11/01/2023]
Abstract
In solid tumors, three main complementary approaches of adoptive T-cell therapies were successively developed: tumor-infiltrating lymphocytes, chimeric antigen receptor engineered T cells, and high-affinity T-cell receptor engineered T cells. In this review, we summarized rational and main results of these three adoptive T-cell therapies in solid tumors field and gave an overview of encouraging data and their limits. Then, we listed the major remaining challenges (including tumor antigen loss, on-target/off-tumor effect, tumor access difficulties and general/local immunosubversion) and their lines of research. Finally, we gave insight into the ongoing trials in solid tumor.
Collapse
Affiliation(s)
- Aurore Dougé
- Medical Oncology Department, University Hospital, Clermont-Ferrand, France
- EA(UR)7453 CHELTER - Clermont Auvergne University, Clermont-Ferrand, France
| | - Nathan El Ghazzi
- Medical Oncology Department, University Hospital, Clermont-Ferrand, France
| | - Richard Lemal
- EA(UR)7453 CHELTER - Clermont Auvergne University, Clermont-Ferrand, France
- Histocompatibility and Immunogenetic Department, University Hospital, Clermont-Ferrand, France
| | - Paul Rouzaire
- EA(UR)7453 CHELTER - Clermont Auvergne University, Clermont-Ferrand, France
- Histocompatibility and Immunogenetic Department, University Hospital, Clermont-Ferrand, France
| |
Collapse
|
93
|
Teng F, Cui T, Zhou L, Gao Q, Zhou Q, Li W. Programmable synthetic receptors: the next-generation of cell and gene therapies. Signal Transduct Target Ther 2024; 9:7. [PMID: 38167329 PMCID: PMC10761793 DOI: 10.1038/s41392-023-01680-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/22/2023] [Accepted: 10/11/2023] [Indexed: 01/05/2024] Open
Abstract
Cell and gene therapies hold tremendous promise for treating a range of difficult-to-treat diseases. However, concerns over the safety and efficacy require to be further addressed in order to realize their full potential. Synthetic receptors, a synthetic biology tool that can precisely control the function of therapeutic cells and genetic modules, have been rapidly developed and applied as a powerful solution. Delicately designed and engineered, they can be applied to finetune the therapeutic activities, i.e., to regulate production of dosed, bioactive payloads by sensing and processing user-defined signals or biomarkers. This review provides an overview of diverse synthetic receptor systems being used to reprogram therapeutic cells and their wide applications in biomedical research. With a special focus on four synthetic receptor systems at the forefront, including chimeric antigen receptors (CARs) and synthetic Notch (synNotch) receptors, we address the generalized strategies to design, construct and improve synthetic receptors. Meanwhile, we also highlight the expanding landscape of therapeutic applications of the synthetic receptor systems as well as current challenges in their clinical translation.
Collapse
Affiliation(s)
- Fei Teng
- University of Chinese Academy of Sciences, Beijing, 101408, China.
| | - Tongtong Cui
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Li Zhou
- University of Chinese Academy of Sciences, Beijing, 101408, China
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qingqin Gao
- University of Chinese Academy of Sciences, Beijing, 101408, China
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Zhou
- University of Chinese Academy of Sciences, Beijing, 101408, China.
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Wei Li
- University of Chinese Academy of Sciences, Beijing, 101408, China.
- State Key Laboratory of Stem Cell and Regenerative Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| |
Collapse
|
94
|
Albelda SM. CAR T cell therapy for patients with solid tumours: key lessons to learn and unlearn. Nat Rev Clin Oncol 2024; 21:47-66. [PMID: 37904019 DOI: 10.1038/s41571-023-00832-4] [Citation(s) in RCA: 118] [Impact Index Per Article: 118.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/09/2023] [Indexed: 11/01/2023]
Abstract
Chimeric antigen receptor (CAR) T cells have been approved for use in patients with B cell malignancies or relapsed and/or refractory multiple myeloma, yet efficacy against most solid tumours remains elusive. The limited imaging and biopsy data from clinical trials in this setting continues to hinder understanding, necessitating a reliance on imperfect preclinical models. In this Perspective, I re-evaluate current data and suggest potential pathways towards greater success, drawing lessons from the few successful trials testing CAR T cells in patients with solid tumours and the clinical experience with tumour-infiltrating lymphocytes. The most promising approaches include the use of pluripotent stem cells, co-targeting multiple mechanisms of immune evasion, employing multiple co-stimulatory domains, and CAR ligand-targeting vaccines. An alternative strategy focused on administering multiple doses of short-lived CAR T cells in an attempt to pre-empt exhaustion and maintain a functional effector pool should also be considered.
Collapse
Affiliation(s)
- Steven M Albelda
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Pulmonary and Critical Care Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
95
|
Majumder A. Evolving CAR-T-Cell Therapy for Cancer Treatment: From Scientific Discovery to Cures. Cancers (Basel) 2023; 16:39. [PMID: 38201467 PMCID: PMC10777914 DOI: 10.3390/cancers16010039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
In recent years, chimeric antigen receptor (CAR)-T-cell therapy has emerged as the most promising immunotherapy for cancer that typically uses patients' T cells and genetically engineered them to target cancer cells. Although recent improvements in CAR-T-cell therapy have shown remarkable success for treating hematological malignancies, the heterogeneity in tumor antigens and the immunosuppressive nature of the tumor microenvironment (TME) limits its efficacy in solid tumors. Despite the enormous efforts that have been made to make CAR-T-cell therapy more effective and have minimal side effects for treating hematological malignancies, more research needs to be conducted regarding its use in the clinic for treating various other types of cancer. The main concern for CAR-T-cell therapy is severe toxicities due to the cytokine release syndrome, whereas the other challenges are associated with complexity and immune-suppressing TME, tumor antigen heterogeneity, the difficulty of cell trafficking, CAR-T-cell exhaustion, and reduced cytotoxicity in the tumor site. This review discussed the latest discoveries in CAR-T-cell therapy strategies and combination therapies, as well as their effectiveness in different cancers. It also encompasses ongoing clinical trials; current challenges regarding the therapeutic use of CAR-T-cell therapy, especially for solid tumors; and evolving treatment strategies to improve the therapeutic application of CAR-T-cell therapy.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Medicine, University of California San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
96
|
Park JJ, Lee KAV, Lam SZ, Tang K, Chen S. Genome Engineering for Next-Generation Cellular Immunotherapies. Biochemistry 2023; 62:3455-3464. [PMID: 35930700 PMCID: PMC11320893 DOI: 10.1021/acs.biochem.2c00340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Over the past decade, cellular immunotherapies such as CAR-T, TCR-T, and NK cell therapies have achieved tremendous success in cancer treatment. However, various challenges and obstacles remain, including antigen escape, immunosuppression in the tumor microenvironment, toxicities, and on-target off-tumor effects. Recent strategies for overcoming these roadblocks have included the use of genome engineering. Multiplexed CRISPR-Cas and synthetic biology approaches facilitate the development of cell therapies with higher potency and sophisticated modular control; they also offer a toolkit for allogeneic therapy development. Engineering approaches have targeted genetic modifications to enhance long-term persistence through cytokine modulation, knockout of genes mediating immunosuppressive signals, and genes such as the endogenous TCR and MHC-I that elicit adverse host-graft interactions in an allogeneic context. Genome engineering approaches for other immune cell types are also being explored, such as CAR macrophages and CAR-NK cells. Future therapeutic development of cellular immunotherapies may also be guided by novel target discovery through unbiased CRISPR genetic screening approaches.
Collapse
Affiliation(s)
- Jonathan J Park
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- System Biology Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Center for Cancer Systems Biology, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- M.D.-Ph.D. Program, Yale University, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Molecular Cell Biology, Genetics, and Development Program, Yale University, 333 Cedar Street, New Haven, Connecticut 06520, United States
| | - Kyoung A V Lee
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- System Biology Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Center for Cancer Systems Biology, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Department of Biostatistics, Yale School of Public Health, 60 College Street, New Haven, Connecticut 06510, United States
| | - Stanley Z Lam
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- System Biology Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Center for Cancer Systems Biology, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
| | - Kaiyuan Tang
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- System Biology Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Center for Cancer Systems Biology, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- System Biology Institute, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- Center for Cancer Systems Biology, Yale University, 850 West Campus Drive, West Haven, Connecticut 06516, United States
- M.D.-Ph.D. Program, Yale University, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Molecular Cell Biology, Genetics, and Development Program, Yale University, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Immunobiology Program, Yale University, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Yale Comprehensive Cancer Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Yale Stem Cell Center, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
- Yale Center for Biomedical Data Science, Yale University School of Medicine, 333 Cedar Street, New Haven, Connecticut 06520, United States
| |
Collapse
|
97
|
Nasiri F, Farrokhi K, Safarzadeh Kozani P, Mahboubi Kancha M, Dashti Shokoohi S, Safarzadeh Kozani P. CAR-T cell immunotherapy for ovarian cancer: hushing the silent killer. Front Immunol 2023; 14:1302307. [PMID: 38146364 PMCID: PMC10749368 DOI: 10.3389/fimmu.2023.1302307] [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: 09/26/2023] [Accepted: 11/13/2023] [Indexed: 12/27/2023] Open
Abstract
As the most lethal gynecologic oncological indication, carcinoma of the ovary has been ranked as the 5th cause of cancer-related mortality in women, with a high percentage of the patients being diagnosed at late stages of the disease and a five-year survival of ~ 30%. Ovarian cancer patients conventionally undergo surgery for tumor removal followed by platinum- or taxane-based chemotherapy; however, a high percentage of patients experience tumor relapse. Cancer immunotherapy has been regarded as a silver lining in the treatment of patients with various immunological or oncological indications; however, mirvetuximab soravtansine (a folate receptor α-specific mAb) and bevacizumab (a VEGF-A-specific mAb) are the only immunotherapeutics approved for the treatment of ovarian cancer patients. Chimeric antigen receptor T-cell (CAR-T) therapy has achieved tremendous clinical success in the treatment of patients with certain B-cell lymphomas and leukemias, as well as multiple myeloma. In the context of solid tumors, CAR-T therapies face serious obstacles that limit their therapeutic benefit. Such hindrances include the immunosuppressive nature of solid tumors, impaired tumor infiltration, lack of qualified tumor-associated antigens, and compromised stimulation and persistence of CAR-Ts following administration. Over the past years, researchers have made arduous attempts to apply CAR-T therapy to ovarian cancer. In this review, we outline the principles of CAR-T therapy and then highlight its limitations in the context of solid tumors. Ultimately, we focus on preclinical and clinical findings achieved in CAR-T-mediated targeting of different ovarian cancer-associated target antigens.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Khadijeh Farrokhi
- Department of Microbial Biotechnology, Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maral Mahboubi Kancha
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Setareh Dashti Shokoohi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
98
|
Ruella M, Korell F, Porazzi P, Maus MV. Mechanisms of resistance to chimeric antigen receptor-T cells in haematological malignancies. Nat Rev Drug Discov 2023; 22:976-995. [PMID: 37907724 PMCID: PMC10965011 DOI: 10.1038/s41573-023-00807-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 11/02/2023]
Abstract
Chimeric antigen receptor (CAR)-T cells have recently emerged as a powerful therapeutic approach for the treatment of patients with chemotherapy-refractory or relapsed blood cancers, including acute lymphoblastic leukaemia, diffuse large B cell lymphoma, follicular lymphoma, mantle cell lymphoma and multiple myeloma. Nevertheless, resistance to CAR-T cell therapies occurs in most patients. In this Review, we summarize the resistance mechanisms to CAR-T cell immunotherapy by analysing CAR-T cell dysfunction, intrinsic tumour resistance and the immunosuppressive tumour microenvironment. We discuss current research strategies to overcome multiple resistance mechanisms, including optimization of the CAR design, improvement of in vivo T cell function and persistence, modulation of the immunosuppressive tumour microenvironment and synergistic combination strategies.
Collapse
Affiliation(s)
- Marco Ruella
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Felix Korell
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Patrizia Porazzi
- Division of Hematology and Oncology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
99
|
Keshavarz S, Wall JR, Keshavarz S, Vojoudi E, Jafari-Shakib R. Breast cancer immunotherapy: a comprehensive review. Clin Exp Med 2023; 23:4431-4447. [PMID: 37658246 DOI: 10.1007/s10238-023-01177-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023]
Abstract
Cancer remains a major health problem despite numerous new medical interventions that have been introduced in recent years. One of the major choices for cancer therapy is so-called adoptive cell therapy (ACT). ACT can be performed using both innate immune cells, including dendritic cells (DCs), natural killer (NK) cells, and γδ T cells and acquired immune T cells. It has become possible to utilize these cells in both their native and modified states in clinical studies. Because of considerable success in cancer treatment, ACT now plays a role in advanced therapy protocols. Genetic engineering of autologous and allogeneic immune cells (T lymphocytes, NK cells, macrophages, etc.) with chimeric antigen receptors (CAR) is a powerful new tool to target specific antigens on cancer cells. The Food and Drug Administration (FDA) in the US has approved certain CAR-T cells for hematologic malignancies and it is hoped that their use can be extended to incorporate a variety of cells, in particular NK cells. However, the ACT method has some limitations, such as the risk of rejection in allogeneic engrafts. Accordingly, numerous efforts are being made to eliminate or minimize this and other complications. In the present review, we have developed a guide to breast cancer (BC) therapy from conventional therapy, through to cell-based approaches, in particular novel technologies including CAR with emphasis on NK cells as a new and safer candidate in this field as well as the more recent aptamer technology, which can play a major role in BC immunotherapy.
Collapse
Affiliation(s)
- Samaneh Keshavarz
- School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Jack R Wall
- University of Notre Dame Australia, Sydney, Australia
| | - Somayeh Keshavarz
- School of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Elham Vojoudi
- Regenerative Medicine, Organ Procurement and Transplantation Multidisciplinary Center, Razi Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| | - Reza Jafari-Shakib
- Department of Immunology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
100
|
Ibanez J, Hebbar N, Thanekar U, Yi Z, Houke H, Ward M, Nevitt C, Tian L, Mack SC, Sheppard H, Chiang J, Velasquez MP, Krenciute G. GRP78-CAR T cell effector function against solid and brain tumors is controlled by GRP78 expression on T cells. Cell Rep Med 2023; 4:101297. [PMID: 37992682 PMCID: PMC10694756 DOI: 10.1016/j.xcrm.2023.101297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 08/25/2023] [Accepted: 10/26/2023] [Indexed: 11/24/2023]
Abstract
Lack of targetable antigens is a key limitation for developing successful T cell-based immunotherapies. Members of the unfolded protein response (UPR) represent ideal immunotherapy targets because the UPR regulates the ability of cancer cells to resist cell death, sustain proliferation, and metastasize. Glucose-regulated protein 78 (GRP78) is a key UPR regulator that is overexpressed and translocated to the cell surface of a wide variety of cancers in response to elevated endoplasmic reticulum (ER) stress. We show that GRP78 is highly expressed on the cell surface of multiple solid and brain tumors, making cell surface GRP78 a promising chimeric antigen receptor (CAR) T cell target. We demonstrate that GRP78-CAR T cells can recognize and kill GRP78+ brain and solid tumors in vitro and in vivo. Additionally, our findings demonstrate that GRP78 is upregulated on CAR T cells upon T cell activation; however, this expression is tumor-cell-line specific and results in heterogeneous GRP78-CAR T cell therapeutic response.
Collapse
Affiliation(s)
- Jorge Ibanez
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Nikhil Hebbar
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Unmesha Thanekar
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Zhongzhen Yi
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Haley Houke
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Meghan Ward
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Chris Nevitt
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Liqing Tian
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Stephen C Mack
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Heather Sheppard
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Jason Chiang
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - M Paulina Velasquez
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | - Giedre Krenciute
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|