101
|
Schaible P. Modifying enzyme replacement therapy - A perspective. J Cell Mol Med 2023; 27:165-173. [PMID: 36566487 PMCID: PMC9843529 DOI: 10.1111/jcmm.17653] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/26/2022] Open
Abstract
Several diseases are caused by the lack of functional proteins, including lysosomal storage diseases or haemophilia A and B. Patients suffering from one of these diseases are treated via enzyme replacement therapies to restore the missing protein. Although this treatment strategy prevents some disease symptoms, enzyme replacement therapies are very expensive and require very frequent infusions, which can cause infusion adverse reactions and massively impair the quality of life of the patients. This review proposes a technology to sustainably produce proteins within the patient to potentially make frequent protein-infusions redundant. This technology is based on blood circulating immune cells as producers of the needed therapeutic protein. To ensure a stable protein concentration over time the cells are equipped with a system, which induces cell proliferation when low therapeutic protein levels are detected and a system inhibiting cell proliferation when high therapeutic protein levels are detected.
Collapse
|
102
|
Wang Y, Hu Q. Bio‐Orthogonal Chemistry in Cell Engineering. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Yixin Wang
- Pharmaceutical Sciences Division School of Pharmacy University of Wisconsin-Madison Madison WI 53705 USA
- Carbone Cancer Center School of Medicine and Public Health University of Wisconsin-Madison Madison WI 53705 USA
- Wisconsin Center for NanoBioSystems School of Pharmacy University of Wisconsin-Madison Madison WI 53705 USA
| | - Quanyin Hu
- Pharmaceutical Sciences Division School of Pharmacy University of Wisconsin-Madison Madison WI 53705 USA
- Carbone Cancer Center School of Medicine and Public Health University of Wisconsin-Madison Madison WI 53705 USA
- Wisconsin Center for NanoBioSystems School of Pharmacy University of Wisconsin-Madison Madison WI 53705 USA
| |
Collapse
|
103
|
Dalal RJ, Ohnsorg ML, Panda S, Reineke TM. Hydrophilic Surface Modification of Cationic Unimolecular Bottlebrush Vectors Moderate pDNA and RNP Bottleplex Stability and Delivery Efficacy. Biomacromolecules 2022; 23:5179-5192. [PMID: 36445696 DOI: 10.1021/acs.biomac.2c00999] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A cationic unimolecular bottlebrush polymer with chemically modified end-groups was synthesized to understand the impact of hydrophilicity on colloidal stability, nucleic acid delivery performance, and toxicity. The bottlebrush polymer template was synthesized using grafting-through techniques and was therefore composed of a polynorbornene backbone with poly(2-(dimethylamino)ethyl methacrylate) side chains with dodecyl trithiocarbonate end-groups. Postpolymerization modification was performed to fully remove the end-groups or install hydroxy and methoxy poly(ethylene glycol) functional groups on the bottlebrush exterior. The bottlebrush family was preformulated with biological payloads of pDNA and CRISPR-Cas9 RNP in both water and PBS to understand binding, aggregation kinetics, cytotoxicity, and delivery efficacy. Increasing end-group hydrophilicity and preformulation of bottleplexes in PBS increased colloidal stability and cellular viability; however, this did not always result in increased transfection efficiency. The bottlebrush family exemplifies how formulation conditions, polymer loading, and end-group functionality of bottlebrushes can be tuned to balance expression with cytotoxicity ratios and result in enhanced overall performance.
Collapse
Affiliation(s)
- Rishad J Dalal
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Monica L Ohnsorg
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Sidharth Panda
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
104
|
Chen T, Xue Y, Wang S, Lu J, Zhou H, Zhang W, Zhou Z, Li B, Li Y, Wang Z, Li C, Eloy Y, Sun H, Shen Y, Diarra MD, Ge C, Chai X, Mou H, Lin P, Yu X, Ye Z. Enhancement of T cell infiltration via tumor-targeted Th9 cell delivery improves the efficacy of antitumor immunotherapy of solid tumors. Bioact Mater 2022; 23:508-523. [PMID: 36514387 PMCID: PMC9727594 DOI: 10.1016/j.bioactmat.2022.11.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 11/13/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022] Open
Abstract
Insufficient infiltration of T cells severely compromises the antitumor efficacy of adoptive cell therapy (ACT) against solid tumors. Here, we present a facile immune cell surface engineering strategy aiming to substantially enhance the anti-tumor efficacy of Th9-mediated ACT by rapidly identifying tumor-specific binding ligands and improving the infiltration of infused cells into solid tumors. Non-genetic decoration of Th9 cells with tumor-targeting peptide screened from phage display not only allowed precise targeted ACT against highly heterogeneous solid tumors but also substantially enhanced infiltration of CD8+ T cells, which led to improved antitumor outcomes. Mechanistically, infusion of Th9 cells modified with tumor-specific binding ligands facilitated the enhanced distribution of tumor-killing cells and remodeled the immunosuppressive microenvironment of solid tumors via IL-9 mediated immunomodulation. Overall, we presented a simple, cost-effective, and cell-friendly strategy to enhance the efficacy of ACT against solid tumors with the potential to complement the current ACT.
Collapse
Affiliation(s)
- Tao Chen
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Yucheng Xue
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Shengdong Wang
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Jinwei Lu
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Hao Zhou
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Wenkan Zhang
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Zhiyi Zhou
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310009, China
| | - Binghao Li
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Yong Li
- Qingtian People's Hospital, Department of Orthopedics, Lishui, 323900, China
| | - Zenan Wang
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Changwei Li
- Department of Orthopedics, Shanghai Key Laboratory for the Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 20025, China
| | - Yinwang Eloy
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Hangxiang Sun
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Yihang Shen
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Mohamed Diaty Diarra
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Chang Ge
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Xupeng Chai
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Haochen Mou
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China
| | - Peng Lin
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China,Corresponding author. Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Xiaohua Yu
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China,Corresponding author. Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Zhaoming Ye
- Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China,Orthopaedic Research Institute, Zhejiang University, Hangzhou, 310009, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310009, China,Corresponding author. Orthopaedic Oncology Services, Department of Orthopaedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
105
|
Yi W, Xiao P, Liu X, Zhao Z, Sun X, Wang J, Zhou L, Wang G, Cao H, Wang D, Li Y. Recent advances in developing active targeting and multi-functional drug delivery systems via bioorthogonal chemistry. Signal Transduct Target Ther 2022; 7:386. [PMID: 36460660 PMCID: PMC9716178 DOI: 10.1038/s41392-022-01250-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Bioorthogonal chemistry reactions occur in physiological conditions without interfering with normal physiological processes. Through metabolic engineering, bioorthogonal groups can be tagged onto cell membranes, which selectively attach to cargos with paired groups via bioorthogonal reactions. Due to its simplicity, high efficiency, and specificity, bioorthogonal chemistry has demonstrated great application potential in drug delivery. On the one hand, bioorthogonal reactions improve therapeutic agent delivery to target sites, overcoming off-target distribution. On the other hand, nanoparticles and biomolecules can be linked to cell membranes by bioorthogonal reactions, providing approaches to developing multi-functional drug delivery systems (DDSs). In this review, we first describe the principle of labeling cells or pathogenic microorganisms with bioorthogonal groups. We then highlight recent breakthroughs in developing active targeting DDSs to tumors, immune systems, or bacteria by bioorthogonal chemistry, as well as applications of bioorthogonal chemistry in developing functional bio-inspired DDSs (biomimetic DDSs, cell-based DDSs, bacteria-based and phage-based DDSs) and hydrogels. Finally, we discuss the difficulties and prospective direction of bioorthogonal chemistry in drug delivery. We expect this review will help us understand the latest advances in the development of active targeting and multi-functional DDSs using bioorthogonal chemistry and inspire innovative applications of bioorthogonal chemistry in developing smart DDSs for disease treatment.
Collapse
Affiliation(s)
- Wenzhe Yi
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Ping Xiao
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Xiaochen Liu
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Zitong Zhao
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Xiangshi Sun
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Jue Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Lei Zhou
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Guanru Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Haiqiang Cao
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China
| | - Dangge Wang
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai, 264000 China
| | - Yaping Li
- grid.9227.e0000000119573309State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203 China ,Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264000 China
| |
Collapse
|
106
|
MacDonald KN, Salim K, Levings MK. Manufacturing next-generation regulatory T-cell therapies. Curr Opin Biotechnol 2022; 78:102822. [PMID: 36332342 DOI: 10.1016/j.copbio.2022.102822] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/11/2022] [Accepted: 09/19/2022] [Indexed: 12/14/2022]
Abstract
Regulatory T-cell (Treg) therapy has shown promise in treating autoimmune diseases, transplant rejection, or graft-versus-host disease in early clinical trials. These trials have demonstrated that cell therapy using polyclonal Tregs is feasible and safe, however, the field has been limited by the lack of polyclonal cell specificity and consequent large cell numbers required, and the difficulty in generating autologous products for some patients. Thus, the field is moving toward 'next generation' Treg cell therapies that include genetic modification strategies to engineer specificity and/or modify function, as well as methods to generate Tregs in vitro. In this review, we describe how genetic modification of Tregs using viral transduction or gene editing may be incorporated into Treg manufacturing protocols. We also describe how Tregs may be generated via FOXP3 gene editing or overexpression, or by differentiation from pluripotent stem cells. The application of these various types of engineered Tregs is discussed.
Collapse
Affiliation(s)
- Katherine N MacDonald
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Kevin Salim
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Surgery, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
107
|
PAT strategies and applications for cell therapy processing. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
108
|
Mikhael J, Fowler J, Shah N. Chimeric Antigen Receptor T-Cell Therapies: Barriers and Solutions to Access. JCO Oncol Pract 2022; 18:800-807. [PMID: 36130152 DOI: 10.1200/op.22.00315] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapies are relatively new treatments for patients with heavily pretreated hematologic malignancies. Although these innovative therapies can offer substantial benefit to patients with limited alternative treatment options, patient-access barriers exist. Conventional clinical trials are time-consuming and may be limited by strict patient eligibility criteria, resources, and availability of enrollment slots. Because of the complexity of the CAR-T administration process, treatment delivery can be associated with additional burden for the patient, including requiring patients to reside close to treatment centers and remain with a caregiver after infusion. Manufacturing of CAR-T cells is completed in specialized facilities and depends on the availability of reagents, manufacturing workforce, and timely transportation. CAR-T therapy is costly, and many US health plans restrict coverage of cell and gene therapies. Several of the existing challenges because of these barriers have been exacerbated during the COVID-19 pandemic. This review discusses these barriers and proposes some potential solutions to improving patient access, including innovation in clinical trial design and manufacturing, location of treatment delivery, and key stakeholder opinions regarding treatment and reimbursement. We propose a call to action for key stakeholder groups to address these barriers to CAR-T therapy to expand treatment access for patients. Future collaboration between key stakeholders, including payers, regulatory agencies, and industry/academia, will be critical to continue to address these barriers and enhance patient access to these therapies.
Collapse
Affiliation(s)
- Joseph Mikhael
- Translational Genomics Research Institute (TGen), Applied Cancer Research and Drug Discovery Division, Phoenix, AZ
| | | | - Nina Shah
- Bone Marrow Transplantation and Hematologic Malignancy Unit, Division of Hematology-Oncology, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
109
|
Abstract
Immune cells are being engineered to recognize and respond to disease states, acting as a "living drug" when transferred into patients. Therapies based on engineered immune cells are now a clinical reality, with multiple engineered T cell therapies approved for treatment of hematologic malignancies. Ongoing preclinical and clinical studies are testing diverse strategies to modify the fate and function of immune cells for applications in cancer, infectious disease, and beyond. Here, we discuss current progress in treating human disease with immune cell therapeutics, emerging strategies for immune cell engineering, and challenges facing the field, with a particular emphasis on the treatment of cancer, where the most effort has been applied to date.
Collapse
Affiliation(s)
- Darrell J. Irvine
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Marcela V. Maus
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.,Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston MA, USA
| | - David J. Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, U.S.A.,Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, MA, U.S.A
| | - Wilson W. Wong
- Department of Biomedical Engineering and Biological Design Center, Boston University, Boston, MA, USA
| |
Collapse
|
110
|
Rahimmanesh I, Tavangar M, Zahedi SN, Azizi Y, Khanahmad Shahreza H. Optimization of Culture Media for Ex vivo T-Cell Expansion for Adoptive T-Cell Therapy. Adv Biomed Res 2022; 11:94. [PMID: 36518860 PMCID: PMC9744083 DOI: 10.4103/abr.abr_349_21] [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: 11/01/2021] [Revised: 01/12/2022] [Accepted: 01/31/2022] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Adoptive T-cell therapy is a promising treatment strategy for cancer immunotherapy. The ability of immunotherapy based on the adoptive cell transfer of genetically modified T cells to generate powerful clinical responses has been highlighted by recent clinical success. Techniques which are used to expand large numbers of T cells from different sources are critical in adoptive cell therapy. In this study, we evaluated the expansion, proliferation, activation of T lymphocytes, in the presence of various concentrations of interleukin-2, phytohemagglutinin (PHA), and insulin. MATERIALS AND METHODS The effect of different supplemented culture media on T cell expansion was evaluated using MTT assay. The expression level of the Ki-67 proliferation marker was evaluated by real-time polymerase chain reaction. In addition, flow cytometry analysis was performed to access T cell subpopulations. RESULTS Our results showed that supplemented culture media with an optimized concentration of PHA and interleukin-2 increased total fold expansion of T cells up to 500-fold with approximately 90% cell viability over 7 days. The quantitative assessment of Ki-67 in expanded T cells showed a significant elevation of this proliferation marker. Flow cytometry was also used to assess the proportion of CD4+ and CD8+ cells, and the main expanded population was CD3+ CD8+ cells. CONCLUSIONS Based on these findings, we introduced a low-cost and rapid method to support the efficient expansion of T cells for adoptive cell therapy and other in vivo experiments.
Collapse
Affiliation(s)
- Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrsa Tavangar
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyedeh Noushin Zahedi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yadollah Azizi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Khanahmad Shahreza
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
111
|
Triantafyllou N, Bernardi A, Lakelin M, Shah N, Papathanasiou MM. A digital platform for the design of patient-centric supply chains. Sci Rep 2022; 12:17365. [PMID: 36253394 PMCID: PMC9576774 DOI: 10.1038/s41598-022-21290-5] [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: 12/20/2021] [Accepted: 09/26/2022] [Indexed: 01/10/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T cell therapies have received increasing attention, showing promising results in the treatment of acute lymphoblastic leukaemia and aggressive B cell lymphoma. Unlike typical cancer treatments, autologous CAR T cell therapies are patient-specific; this makes them a unique therapeutic to manufacture and distribute. In this work, we focus on the development of a computer modelling tool to assist the design and assessment of supply chain structures that can reliably and cost-efficiently deliver autologous CAR T cell therapies. We focus on four demand scales (200, 500, 1000 and 2000 patients annually) and we assess the tool's capabilities with respect to the design of responsive supply chain candidate solutions while minimising cost.
Collapse
Affiliation(s)
- Niki Triantafyllou
- grid.7445.20000 0001 2113 8111Sargent Centre for Process System Engineering, Imperial College London, London, SW7 2AZ UK ,grid.7445.20000 0001 2113 8111 Department of Chemical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Andrea Bernardi
- grid.7445.20000 0001 2113 8111Sargent Centre for Process System Engineering, Imperial College London, London, SW7 2AZ UK ,grid.7445.20000 0001 2113 8111 Department of Chemical Engineering, Imperial College London, London, SW7 2AZ UK
| | | | - Nilay Shah
- grid.7445.20000 0001 2113 8111Sargent Centre for Process System Engineering, Imperial College London, London, SW7 2AZ UK ,grid.7445.20000 0001 2113 8111 Department of Chemical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Maria M. Papathanasiou
- grid.7445.20000 0001 2113 8111Sargent Centre for Process System Engineering, Imperial College London, London, SW7 2AZ UK ,grid.7445.20000 0001 2113 8111 Department of Chemical Engineering, Imperial College London, London, SW7 2AZ UK
| |
Collapse
|
112
|
Wolff JH, Mikkelsen JG. Delivering genes with human immunodeficiency virus-derived vehicles: still state-of-the-art after 25 years. J Biomed Sci 2022; 29:79. [PMID: 36209077 PMCID: PMC9548131 DOI: 10.1186/s12929-022-00865-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 09/29/2022] [Indexed: 11/10/2022] Open
Abstract
Viruses are naturally endowed with the capacity to transfer genetic material between cells. Following early skepticism, engineered viruses have been used to transfer genetic information into thousands of patients, and genetic therapies are currently attracting large investments. Despite challenges and severe adverse effects along the way, optimized technologies and improved manufacturing processes are driving gene therapy toward clinical translation. Fueled by the outbreak of AIDS in the 1980s and the accompanying focus on human immunodeficiency virus (HIV), lentiviral vectors derived from HIV have grown to become one of the most successful and widely used vector technologies. In 2022, this vector technology has been around for more than 25 years. Here, we celebrate the anniversary by portraying the vector system and its intriguing properties. We dive into the technology itself and recapitulate the use of lentiviral vectors for ex vivo gene transfer to hematopoietic stem cells and for production of CAR T-cells. Furthermore, we describe the adaptation of lentiviral vectors for in vivo gene delivery and cover the important contribution of lentiviral vectors to basic molecular research including their role as carriers of CRISPR genome editing technologies. Last, we dwell on the emerging capacity of lentiviral particles to package and transfer foreign proteins.
Collapse
Affiliation(s)
- Jonas Holst Wolff
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark
| | - Jacob Giehm Mikkelsen
- Department of Biomedicine, Aarhus University, Høegh-Guldbergs Gade 10, 8000, Aarhus C, Denmark.
| |
Collapse
|
113
|
Abstract
Chimeric antigen receptor T (CAR-T) cells therapy has revolutionized the treatment paradigms for hematological malignancies, with multi-line therapy-refractory patients achieving durable complete remissions (CR) and relatively high objective response rate (ORR). So far, many CAR-T products, such as Kymriah, Yescarta and Tecartus, have been developed and got the unprecedented results. However, some patients may relapse afterwards, driving intense investigations into promoting the development of novel strategies to overcome resistance and mechanisms of relapse. Notable technical progress, such as nanobodies and CRISPR-Case9, has also taken place to ensure CAR-T cell therapy fully satisfies its medical potential. In this review, we outline the basic principles for the development and manufacturing processes of CAR-T cell therapy, summarize the similarities and differences in efficacy of different products as well as their corresponding clinical results, and discuss CAR-T immunotherapy combined with other clinical effects of drug therapy.
Collapse
Affiliation(s)
- Junru Lu
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
114
|
Bailly S, Cartron G, Chaganti S, Córdoba R, Corradini P, Düll J, Ferrarini I, Osborne W, Rosenwald A, Sancho J, Tilly H, Van Den Neste E, Viardot A, Visco C. Targeting CD19 in diffuse large B-cell lymphoma: An expert opinion paper. Hematol Oncol 2022; 40:505-517. [PMID: 35488888 PMCID: PMC9796473 DOI: 10.1002/hon.3013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 04/07/2022] [Indexed: 01/01/2023]
Abstract
The ubiquitous, early-stage expression, efficient internalization, limited off-target effects, and high disease specificity of CD19 make it an attractive therapeutic target. Currently available anti-CD19 therapies have demonstrated particular promise in patients with relapsed or refractory B-cell non-Hodgkin lymphoma. Selection of the most appropriate treatment strategy should be based on individual patient characteristics and the goal of therapy. However, evidence and knowledge about the sequencing of anti-CD19 therapies are limited. Here, we review the current evidence for CD19 as a target in diffuse large B-cell lymphoma and consider approaches to the use of anti-CD19 therapy.
Collapse
Affiliation(s)
- Sarah Bailly
- Département d’HématologieCliniques Universitaires Saint‐LucBrusselsBelgium
| | - Guillaume Cartron
- Department of HaematologyCentre Hospitalier Universitaire de MontpellierUMR‐CNRS 5535MontpellierFrance
| | | | - Raul Córdoba
- Department of HematologyFundación Jiménez Díaz University HospitalHealth Research Institute IIS‐FJDMadridSpain
| | - Paolo Corradini
- Fondazione IRCCS Istituto Nazionale dei TumoriUniversity of MilanMilanItaly
| | - Johannes Düll
- Medizinische Klinik und Poliklinik IIUniversitätsklinikum WürzburgWürzburgGermany
| | - Isacco Ferrarini
- Department of MedicineSection of HematologyUniversity of VeronaVeronaItaly
| | - Wendy Osborne
- Newcastle Upon Tyne Hospitals NHS Foundation TrustNewcastleUK
| | - Andreas Rosenwald
- Institute of PathologyUniversity of Würzburg, and Comprehensive Cancer Center MainfrankenWürzburgGermany
| | | | - Hervé Tilly
- Department of Hematology and U1245Centre Henri Becquerel and University of RouenRouenFrance
| | - Eric Van Den Neste
- Département d’HématologieCliniques Universitaires Saint‐LucBrusselsBelgium
| | - Andreas Viardot
- Department of Internal Medicine IIIUniversity Hospital UlmUlmGermany
| | - Carlo Visco
- Department of MedicineSection of HematologyUniversity of VeronaVeronaItaly
| |
Collapse
|
115
|
Ding M, Lin J, Qin C, Wei P, Tian J, Lin T, Xu T. Application of synthetic biology in bladder cancer. Chin Med J (Engl) 2022; 135:2178-2187. [PMID: 36209735 PMCID: PMC9771244 DOI: 10.1097/cm9.0000000000002344] [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: 04/19/2022] [Indexed: 12/24/2022] Open
Abstract
ABSTRACT Bladder cancer (BC) is the most common malignant tumor of the genitourinary system. The age of individuals diagnosed with BC tends to decrease in recent years. A variety of standard therapeutic options are available for the clinical management of BC, but limitations exist. It is difficult to surgically eliminate small lesions, while radiation and chemotherapy damage normal tissues, leading to severe side effects. Therefore, new approaches are required to improve the efficacy and specificity of BC treatment. Synthetic biology is a field emerging in the last decade that refers to biological elements, devices, and materials that are artificially synthesized according to users' needs. In this review, we discuss how to utilize genetic elements to regulate BC-related gene expression periodically and quantitatively to inhibit the initiation and progression of BC. In addition, the design and construction of gene circuits to distinguish cancer cells from normal cells to kill the former but spare the latter are elaborated. Then, we introduce the development of genetically modified T cells for targeted attacks on BC. Finally, synthetic nanomaterials specializing in detecting and killing BC cells are detailed. This review aims to describe the innovative details of the clinical diagnosis and treatment of BC from the perspective of synthetic biology.
Collapse
Affiliation(s)
- Mengting Ding
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
| | - Jiaxing Lin
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
| | - Caipeng Qin
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
| | - Ping Wei
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Jiahe Tian
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 528403, China
| | - Tao Xu
- Department of Urology, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
116
|
Kim S, Kim K. Lipid-mediated ex vivo cell surface engineering for augmented cellular functionalities. BIOMATERIALS ADVANCES 2022; 140:213059. [PMID: 35961186 DOI: 10.1016/j.bioadv.2022.213059] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/23/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
Once administrated, intercellular adhesion to recognize and/or arrest target cells is essential for specific treatments, especially for cancer or tumor. However, immune cells administrated into the tumor-microenvironment could lose their intrinsic functionalities such as target recognition ability, resulting in an ineffective cancer immunotherapy. Various manipulation techniques for decorating functional moieties onto cell surface and enhancing target recognition have been developed. A hydrophobic interaction-mediated ex-vivo cell surface engineering using lipid-based biomaterials could be a state-of-the-art engineering technique that could achieve high-efficiency cell surface modification by a single method without disturbance of intrinsic characteristics of cells. In this regard, this review provides design principles for the development of lipid-based biomaterials with a linear structure of lipid, polyethylene glycol, and functional group, strategies for the synthesis process, and their practical applications in biomedical engineering. Especially, we provide new insights into the development of a novel surface coating techniques for natural killer (NK) cells with engineering decoration of cancer targeting moieties on their cell surfaces. Among immune cells, NK cells are interesting cell population for substituting T cells because of their excellent safety and independent anticancer efficacy. Thus, optimal strategies to select cancer-type-specific targeting moieties and present them onto the surface of immune cells (especially, NK cells) using lipid-based biomaterials could provide additional tools to capture cancer cells for developing novel immune cell therapy products. Enhanced anticancer efficacies by surface-engineered NK cells have been demonstrated both in vitro and in vivo. Therefore, it could be speculated that recent progresses in cell surface modification technology via lipid-based biomaterials could strengthen immune surveillance and immune synapses for utilization in a next-generation cancer immunotherapy, beyond currently available genetic engineering tool such as chimeric antigen receptor-mediated immune cell modulation.
Collapse
Affiliation(s)
- Sungjun Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea
| | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, Seoul, Republic of Korea.
| |
Collapse
|
117
|
Cunningham AW, Jones M, Frank N, Sethi D, Miller MM. Stem-like memory T cells are generated during hollow fiber perfusion-based expansion and enriched after cryopreservation in an automated modular cell therapy manufacturing process. Cytotherapy 2022; 24:1148-1157. [PMID: 36031522 DOI: 10.1016/j.jcyt.2022.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/02/2022] [Accepted: 07/26/2022] [Indexed: 01/20/2023]
Abstract
BACKGROUND AIMS Modular automation is a flexible and reliable option to build the foundation of a new or evolving process or to introduce automation to a process that is already established. Herein the authors demonstrate that modular automation provides both high-quality and high-yield T-cell products. METHODS Cells from three individual donors collected on an automated continuous flow centrifugation system were successfully expanded in a functionally closed, automated, perfusion-based hollow fiber bioreactor. These cells were then prepared for cryopreservation in an automated closed-system device that maintains temperature and aliquots a mixed cell product and cryoprotectant into product bags. Cell product bags were thawed and expanded in flasks. Samples taken throughout this manufacturing process were analyzed for cell phenotype, exhaustion markers and functionality. The proportion of CD4+ and CD8+ T cells was maintained through each step, from pre-expansion and post-expansion to immediately after thaw and 24 h after thaw. RESULTS Interestingly, phenotypic markers such as CD45RO, CD45RA and CCR7 evolved throughout the process and stem-like memory T cells emerged as the predominant phenotype in the clinically relevant 24-h post-thaw sample. CONCLUSIONS Modular automation supported the generation of stem-like memory T cells that were not terminally exhausted and were able to produce effector cytokines upon restimulation.
Collapse
Affiliation(s)
| | - Mark Jones
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA
| | - Nathan Frank
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA
| | - Dalip Sethi
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA.
| | - Mindy M Miller
- Terumo Blood and Cell Technologies, Lakewood, Colorado, USA.
| |
Collapse
|
118
|
Hu JF, Wang ZW, Liao CY, Chen ZW, Kang FP, Lin CF, Lin TS, Huang L, Tian YF, Chen S. Induced expression of CCL19 promotes the anti-tumor ability of CAR-T cells by increasing their infiltration ability. Front Immunol 2022; 13:958960. [PMID: 35990619 PMCID: PMC9391072 DOI: 10.3389/fimmu.2022.958960] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 07/08/2022] [Indexed: 12/05/2022] Open
Abstract
Background Chimeric antigen receptor-engineered T cell (CAR-T) therapy has shown promising potential for anti-cancer treatment. However, for pancreatic ductal adenocarcinoma (PDAC), the lack of infiltrative ability of these CAR-T cells leads to sub-optimal treatment outcome. Methods Chemokine (C-C motif) ligand 19 (CCL19), the expression of which is regulated by the nuclear factor of activated T cell pathway, was transfected into targeting mesothelin CAR-T cells (mesoCAR-N19) using NFAT regulating element. It was expressed in activated CAR-T cells by OKT3 or mesothelin+ tumor cells but not in inactive cells. The migratory ability of these CAR-T cells was then measured. Subsequently, functional identification of these CAR-T cells was performed in vivo. In addition, the tumor lytic activity and proliferation of the CAR-T cells were measured in vitro. The degree of CAR-T cell infiltration and distribution into the PDAC tumors was examined using the immunohistochemical staining of hCD3 and the detection of CAR gene copy number by quantitative PCR. Finally, the functional assessment of chemokine (C-C motif) receptor 7 knock-out was performed in the CAR-T cells. Results Through in vitro Transwell assays, it was demonstrated that mesoCAR-N19 can be specifically expressed in CAR-T cells activated by tumor cells compared with conventional mesothelin CAR-T (mesoCAR) cells. We also observed that upregulating the expression of CCL19 can increase the recruitment of additional T cells. In vivo studies subsequently revealed that this highly specific recruitment of T cell infiltration is associated with enhanced tumor-suppressive activities downstream. Conclusion Induced expression of CCL19 can promote the anti-tumor ability of CAR-T cells by increasing their infiltrative ability. This study potentially uncovered novel method of activating CAR-T cells to enhance their infiltrative capacities, which offers a novel direction for PDAC treatment.
Collapse
Affiliation(s)
- Jian-fei Hu
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
| | - Zu-wei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
| | - Cheng-yu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
| | - Zhi-wen Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
| | - Feng-ping Kang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
| | - Cai-feng Lin
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Tian-sheng Lin
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Long Huang
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, China
| | - Yi-feng Tian
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, China
- *Correspondence: Yi-feng Tian, ; Shi Chen,
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, China
- *Correspondence: Yi-feng Tian, ; Shi Chen,
| |
Collapse
|
119
|
Cheng EL, Cardle II, Kacherovsky N, Bansia H, Wang T, Zhou Y, Raman J, Yen A, Gutierrez D, Salipante SJ, des Georges A, Jensen MC, Pun SH. Discovery of a Transferrin Receptor 1-Binding Aptamer and Its Application in Cancer Cell Depletion for Adoptive T-Cell Therapy Manufacturing. J Am Chem Soc 2022; 144:13851-13864. [PMID: 35875870 PMCID: PMC10024945 DOI: 10.1021/jacs.2c05349] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The clinical manufacturing of chimeric antigen receptor (CAR) T cells includes cell selection, activation, gene transduction, and expansion. While the method of T-cell selection varies across companies, current methods do not actively eliminate the cancer cells in the patient's apheresis product from the healthy immune cells. Alarmingly, it has been found that transduction of a single leukemic B cell with the CAR gene can confer resistance to CAR T-cell therapy and lead to treatment failure. In this study, we report the identification of a novel high-affinity DNA aptamer, termed tJBA8.1, that binds transferrin receptor 1 (TfR1), a receptor broadly upregulated by cancer cells. Using competition assays, high resolution cryo-EM, and de novo model building of the aptamer into the resulting electron density, we reveal that tJBA8.1 shares a binding site on TfR1 with holo-transferrin, the natural ligand of TfR1. We use tJBA8.1 to effectively deplete B lymphoma cells spiked into peripheral blood mononuclear cells with minimal impact on the healthy immune cell composition. Lastly, we present opportunities for affinity improvement of tJBA8.1. As TfR1 expression is broadly upregulated in many cancers, including difficult-to-treat T-cell leukemias and lymphomas, our work provides a facile, universal, and inexpensive approach for comprehensively removing cancerous cells from patient apheresis products for safe manufacturing of adoptive T-cell therapies.
Collapse
Affiliation(s)
- Emmeline L Cheng
- Department of Bioengineering, University of Washington, Seattle, Washington 98195-5061, United States
| | - Ian I Cardle
- Department of Bioengineering, University of Washington, Seattle, Washington 98195-5061, United States.,Seattle Children's Therapeutics, Seattle, Washington 98101, United States
| | - Nataly Kacherovsky
- Department of Bioengineering, University of Washington, Seattle, Washington 98195-5061, United States
| | - Harsh Bansia
- Structural Biology Initiative, CUNY Advanced Science Research Center, City University of New York, New York, New York 10031, United States
| | - Tong Wang
- Nanoscience Initiative, CUNY Advanced Science Research Center, City University of New York, New York, New York 10031, United States
| | - Yunshi Zhou
- Department of Bioengineering, University of Washington, Seattle, Washington 98195-5061, United States
| | - Jai Raman
- Department of Bioengineering, University of Washington, Seattle, Washington 98195-5061, United States
| | - Albert Yen
- Department of Bioengineering, University of Washington, Seattle, Washington 98195-5061, United States
| | - Dominique Gutierrez
- Structural Biology Initiative, CUNY Advanced Science Research Center, City University of New York, New York, New York 10031, United States.,Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York (CUNY), New York, New York 10016, United States
| | - Stephen J Salipante
- Department of Laboratory Medicine, University of Washington, Seattle, Washington 98195-7110, United States
| | - Amédée des Georges
- Structural Biology Initiative, CUNY Advanced Science Research Center, City University of New York, New York, New York 10031, United States.,Ph.D. Programs in Biochemistry and Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States.,Department of Chemistry and Biochemistry, City College of New York, New York, New York 10031, United States
| | - Michael C Jensen
- Seattle Children's Therapeutics, Seattle, Washington 98101, United States.,Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, United States
| | - Suzie H Pun
- Department of Bioengineering, University of Washington, Seattle, Washington 98195-5061, United States
| |
Collapse
|
120
|
Zeng Q, Liu Z, Niu T, He C, Qu Y, Qian Z. Application of nanotechnology in CAR-T-cell immunotherapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
121
|
Miwa H, Dimatteo R, de Rutte J, Ghosh R, Di Carlo D. Single-cell sorting based on secreted products for functionally defined cell therapies. MICROSYSTEMS & NANOENGINEERING 2022; 8:84. [PMID: 35874174 PMCID: PMC9303846 DOI: 10.1038/s41378-022-00422-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/18/2022] [Accepted: 06/13/2022] [Indexed: 05/13/2023]
Abstract
Cell therapies have emerged as a promising new class of "living" therapeutics over the last decade and have been particularly successful for treating hematological malignancies. Increasingly, cellular therapeutics are being developed with the aim of treating almost any disease, from solid tumors and autoimmune disorders to fibrosis, neurodegenerative disorders and even aging itself. However, their therapeutic potential has remained limited due to the fundamental differences in how molecular and cellular therapies function. While the structure of a molecular therapeutic is directly linked to biological function, cells with the same genetic blueprint can have vastly different functional properties (e.g., secretion, proliferation, cell killing, migration). Although there exists a vast array of analytical and preparative separation approaches for molecules, the functional differences among cells are exacerbated by a lack of functional potency-based sorting approaches. In this context, we describe the need for next-generation single-cell profiling microtechnologies that allow the direct evaluation and sorting of single cells based on functional properties, with a focus on secreted molecules, which are critical for the in vivo efficacy of current cell therapies. We first define three critical processes for single-cell secretion-based profiling technology: (1) partitioning individual cells into uniform compartments; (2) accumulating secretions and labeling via reporter molecules; and (3) measuring the signal associated with the reporter and, if sorting, triggering a sorting event based on these reporter signals. We summarize recent academic and commercial technologies for functional single-cell analysis in addition to sorting and industrial applications of these technologies. These approaches fall into three categories: microchamber, microfluidic droplet, and lab-on-a-particle technologies. Finally, we outline a number of unmet needs in terms of the discovery, design and manufacturing of cellular therapeutics and how the next generation of single-cell functional screening technologies could allow the realization of robust cellular therapeutics for all patients.
Collapse
Affiliation(s)
- Hiromi Miwa
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Robert Dimatteo
- Department of Chemical and Biomolecular Engineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Joseph de Rutte
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- Partillion Bioscience, Los Angeles, CA 90095 USA
| | - Rajesh Ghosh
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
| | - Dino Di Carlo
- Department of Bioengineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California - Los Angeles, Los Angeles, CA 90095 USA
- California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
122
|
Grimme CJ, Hanson MG, Corcoran LG, Reineke TM. Polycation Architecture Affects Complexation and Delivery of Short Antisense Oligonucleotides: Micelleplexes Outperform Polyplexes. Biomacromolecules 2022; 23:3257-3271. [PMID: 35862267 DOI: 10.1021/acs.biomac.2c00338] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Herein, we examine the complexation and biological delivery of a short single-stranded antisense oligonucleotide (ASO) payload with four polymer derivatives that form two architectural variants (polyplexes and micelleplexes): a homopolymer poly(2-dimethylaminoethyl methacrylate) (D), a diblock polymer poly(ethylene glycol)methylether methacrylate-block-poly(2-dimethylaminoethyl methacrylate) (ObD), and two micelle-forming variants, poly(2-dimethylaminoethyl methacrylate)-block-poly(n-butyl methacrylate) (DB) and poly(ethylene glycol)methylether methacrylate-block-poly(2-dimethylaminoethyl methacrylate)-block-poly(n-butyl methacrylate) (ObDB). Both polyplexes and micelleplexes complexed ASOs, and the incorporation of an Ob brush enhances colloidal stability. Micellplexes are templated by the size and shape of the unloaded micelle and that micelle-ASO complexation is not sensitive to formulation/mixing order, allowing ease, versatility, and reproducibility in packaging short oligonucleotides. The DB micelleplexes promoted the largest gene silencing, internalization, and tolerable toxicity while the ObDB micelleplexes displayed enhanced colloidal stability and highly efficient payload trafficking despite having lower cellular uptake. Overall, this work demonstrates that cationic micelles are superior delivery vehicles for ASOs denoting the importance of vehicle architecture in biological performance.
Collapse
Affiliation(s)
- Christian J Grimme
- Department of Chemical Engineering & Materials Science, University of Minnesota, 421 Washington Avenue SE, Minneapolis, Minnesota 55455, United States
| | - Mckenna G Hanson
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Louis G Corcoran
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota, 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
123
|
Yang J, He J, Zhang X, Li J, Wang Z, Zhang Y, Qiu L, Wu Q, Sun Z, Ye X, Yin W, Cao W, Shen L, Sersch M, Lu P. Next-day manufacture of a novel anti-CD19 CAR-T therapy for B-cell acute lymphoblastic leukemia: first-in-human clinical study. Blood Cancer J 2022; 12:104. [PMID: 35798714 PMCID: PMC9262977 DOI: 10.1038/s41408-022-00694-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/30/2022] [Accepted: 06/10/2022] [Indexed: 12/02/2022] Open
Abstract
To improve clinical outcomes and shorten the vein-to-vein time of chimeric antigen receptor T (CAR-T) cells, we developed the FasT CAR-T (F-CAR-T) next-day manufacturing platform. We report the preclinical and first-in-human clinical studies evaluating the safety, feasibility, and preliminary efficacy of CD19 F-CAR-T in B-cell acute lymphoblastic leukemia (B-ALL). CD19 F-CAR-T cells demonstrated excellent proliferation with a younger cellular phenotype, less exhaustion, and more effective tumor elimination compared to conventional CAR-T cells in the preclinical study. In our phase I study (NCT03825718), F-CAR-T cells were successfully manufactured and infused in all of the 25 enrolled pediatric and adult patients with B-ALL. CD19 F-CAR-T safety profile was manageable with 24% grade 3 cytokine release syndrome (CRS) and 28% grade 3/4 neurotoxicity occurring predominantly in pediatric patients. On day 14, 23/25 patients achieved minimal residual disease (MRD)-negative complete remission (CR), and 20 subsequently underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT) within 3 months post F-CAR-T therapy. Fifteen of 20 patients were disease-free with a median remission duration of 734 days. One patient relapsed and 4/20 died from transplant-related mortality. Of the three patients who did not undergo allo-HSCT, two remained in CR until 10 months post-F-CAR-T. Our data indicate that anti-CD19 FasT CAR-T shows promising early efficacy for B-ALL. Further evaluations in larger clinical studies are needed.
Collapse
Affiliation(s)
- Junfang Yang
- Hebei Yanda Lu Daopei Hospital, Langfang, Hebei, China.,Beijing Lu Daopei Institute of Hematology, Beijing, China
| | - Jiaping He
- Gracell Biotechnologies Co., Ltd, Shanghai, China
| | - Xian Zhang
- Hebei Yanda Lu Daopei Hospital, Langfang, Hebei, China.,Beijing Lu Daopei Institute of Hematology, Beijing, China
| | - Jingjing Li
- Hebei Yanda Lu Daopei Hospital, Langfang, Hebei, China.,Beijing Lu Daopei Institute of Hematology, Beijing, China
| | | | | | - Liyuan Qiu
- Hebei Yanda Lu Daopei Hospital, Langfang, Hebei, China
| | - Qionglu Wu
- Gracell Biotechnologies Co., Ltd, Shanghai, China
| | - Zhe Sun
- Gracell Biotechnologies Co., Ltd, Shanghai, China
| | - Xun Ye
- Gracell Biotechnologies Co., Ltd, Shanghai, China
| | - Wenjie Yin
- Gracell Biotechnologies Co., Ltd, Shanghai, China
| | - Wei Cao
- Gracell Biotechnologies Co., Ltd, Shanghai, China
| | - Lianjun Shen
- Gracell Biotechnologies Co., Ltd, Shanghai, China.
| | | | - Peihua Lu
- Hebei Yanda Lu Daopei Hospital, Langfang, Hebei, China. .,Beijing Lu Daopei Institute of Hematology, Beijing, China.
| |
Collapse
|
124
|
Sudarsanam H, Buhmann R, Henschler R. Influence of Culture Conditions on Ex Vivo Expansion of T Lymphocytes and Their Function for Therapy: Current Insights and Open Questions. Front Bioeng Biotechnol 2022; 10:886637. [PMID: 35845425 PMCID: PMC9277485 DOI: 10.3389/fbioe.2022.886637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/16/2022] [Indexed: 01/03/2023] Open
Abstract
Ex vivo expansion of T lymphocytes is a central process in the generation of cellular therapies targeted at tumors and other disease-relevant structures, which currently cannot be reached by established pharmaceuticals. The influence of culture conditions on T cell functions is, however, incompletely understood. In clinical applications of ex vivo expanded T cells, so far, a relatively classical standard cell culture methodology has been established. The expanded cells have been characterized in both preclinical models and clinical studies mainly using a therapeutic endpoint, for example antitumor response and cytotoxic function against cellular targets, whereas the influence of manipulations of T cells ex vivo including transduction and culture expansion has been studied to a much lesser detail, or in many contexts remains unknown. This includes the circulation behavior of expanded T cells after intravenous application, their intracellular metabolism and signal transduction, and their cytoskeletal (re)organization or their adhesion, migration, and subsequent intra-tissue differentiation. This review aims to provide an overview of established T cell expansion methodologies and address unanswered questions relating in vivo interaction of ex vivo expanded T cells for cellular therapy.
Collapse
Affiliation(s)
| | | | - Reinhard Henschler
- Institute of Transfusion Medicine, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|
125
|
Novel CD19 chimeric antigen receptor T cells manufactured next-day for acute lymphoblastic leukemia. Blood Cancer J 2022; 12:96. [PMID: 35750687 PMCID: PMC9232607 DOI: 10.1038/s41408-022-00688-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/23/2022] [Accepted: 05/31/2022] [Indexed: 11/23/2022] Open
Abstract
Chimeric antigen receptor-engineered T (CAR-T) cells have shown promising efficacy in patients with relapsed/refractory B cell acute lymphoblastic leukemia (R/R B-ALL). However, challenges remain including long manufacturing processes that need to be overcome. We presented the CD19-targeting CAR-T cell product GC007F manufactured next-day (FasTCAR-T cells) and administered to patients with R/R B-ALL. A total of 21 patients over 14 years of age with CD19+ R/R B-ALL were screened, enrolled and infused with a single infusion of GC007F CAR-T at three different dose levels. The primary objective of the study was to assess safety, secondary objectives included pharmacokinetics of GC007F cells in patients with R/R B-ALL and preliminary efficacy. We were able to demonstrate in preclinical studies that GC007F cells exhibited better proliferation and tumor killing than conventional CAR-T (C-CAR-T) cells. In this investigator-initiated study all 18 efficacy-evaluable patients achieved a complete remission (CR) (18/18, 100.00%) by day 28, with 17 of the patients (94.4%) achieving CR with minimal residual disease (MRD) negative. Fifteen (83.3%) remained disease free at the 3-month assessment, 14 patients (77.8%) maintaining MRD negative at month 3. Among all 21 enrolled patients, the median peak of CAR-T cell was on day 10, with a median peak copy number of 104899.5/µg DNA and a median persistence period of 56 days (range: 7–327 days). The incidence of cytokine release syndrome (CRS) was 95.2% (n = 20), with severe CRS occurring in 52.4% (n = 11) of the patients. Six patients (28.6%) developed neurotoxicity of any grade. GC007F demonstrated superior expansion capacity and a less exhausted phenotype as compared to (C-CAR-T) cells. Moreover, this first-in-human clinical study showed that the novel, next-day manufacturing FasTCAR-T cells was feasible with a manageable toxicity profile in patients with R/R B-ALL.
Collapse
|
126
|
Wang Z, Wang H, Lin S, Ahmed S, Angers S, Sargent EH, Kelley SO. Nanoparticle Amplification Labeling for High-Performance Magnetic Cell Sorting. NANO LETTERS 2022; 22:4774-4783. [PMID: 35639489 DOI: 10.1021/acs.nanolett.2c01018] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Magnetic cell sorting is an enabling tool for the isolation of specific cellular subpopulations for downstream applications and requires the cells to be labeled by a sufficient number of magnetic nanoparticles to leverage magnetophoresis for efficient separation. This requirement makes it challenging to target weakly expressed biomarkers. Here, we developed a new approach that selectively and efficiently amplifies the magnetic labeling on cells through sequentially connected antibodies and nanoparticles delivered to the surface or interior of the cell. Using this approach, we achieved amplification up to 100-fold for surface and intracellular markers. We also demonstrated the utility of this assay for enabling high-performance magnetic cell sorting when it is applied to the analysis of rare tumor cells for cancer diagnosis and the purification of transfected CAR T cells for immunotherapy. The data presented demonstrate a useful tool for the stratification of rare cell subpopulations.
Collapse
Affiliation(s)
- Zongjie Wang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada
| | - Hansen Wang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
| | - Sichun Lin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, Canada
| | - Sharif Ahmed
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
| | - Stephane Angers
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Terrence Donnelly Centre for Cellular & Biomolecular Research, University of Toronto, Toronto M5S 3E1, Canada
| | - Edward H Sargent
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
| | - Shana O Kelley
- Institute of Biomedical Engineering, University of Toronto, Toronto M5S 3G9, Canada
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto M5S 1A8, Canada
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| |
Collapse
|
127
|
Hydroxycholesterol substitution in ionizable lipid nanoparticles for mRNA delivery to T cells. J Control Release 2022; 347:521-532. [PMID: 35569584 DOI: 10.1016/j.jconrel.2022.05.020] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022]
Abstract
Delivery of nucleic acids, such as mRNA, to immune cells has become a major focus in the past decade with ionizable lipid nanoparticles (LNPs) emerging as a clinically-validated delivery platform. LNPs-typically composed of ionizable lipids, cholesterol, phospholipids, and polyethylene glycol lipids -have been designed and optimized for a variety of applications including cancer therapies, vaccines, and gene editing. However, LNPs have only recently been investigated for delivery to T cells, which has various therapeutic applications including the engineering of T cell immunotherapies. While several LNP formulations have been evaluated for mRNA delivery, recent work has demonstrated that the utilization of cholesterol analogs may enhance mRNA delivery. Other studies have shown that cholesterols modified with hydroxyl groups can alter endocytic recycling mechanisms. Here, we engineered a library of LNPs incorporating hydroxycholesterols to evaluate their impact on mRNA delivery to T cells by leveraging endosomal trafficking mechanisms. Substitution of 25% and 50% 7α-hydroxycholesterol for cholesterol in LNPs enhanced mRNA delivery to primary human T cells ex vivo by 1.8-fold and 2.0-fold, respectively. Investigation of endosomal trafficking revealed that these modifications also increase late endosome production and reduce the presence of recycling endosomes. These results suggest that hydroxyl modification of cholesterol molecules incorporated into LNP formulations provides a mechanism for improving delivery of nucleic acid cargo to T cells for a range of immunotherapy applications.
Collapse
|
128
|
Michels A, Ho N, Buchholz CJ. Precision Medicine: In Vivo CAR Therapy as a Showcase for Receptor-Targeted Vector Platforms. Mol Ther 2022; 30:2401-2415. [PMID: 35598048 DOI: 10.1016/j.ymthe.2022.05.018] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/13/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells are a cancer immunotherapy of extremes: Unprecedentedly effective, but complex and costly to manufacture, they are not yet a therapeutic option for all who would benefit. This disparity has motivated worldwide efforts to simplify treatment. Among the proposed solutions, the generation of CAR T cells directly in the patient, i.e. in vivo, is arguably simultaneously the most technically challenging and clinically useful approach to convert CAR therapy from a cell-based autologous medicinal product into a universally applicable off-the-shelf treatment. Here we review the current state-of-the-art of in vivo CAR therapy, focusing especially on the vector technologies used. These cover lentiviral vectors, adenovirus-associated vectors as well as synthetic polymer nanocarriers and lipid nanoparticles. Proof-of-concept, i.e. the generation of CAR cells directly in mouse models, has been demonstrated for all vector platforms. Receptor-targeting of vector particles is crucial, as it can prevent CAR gene delivery into off-target cells, thus reducing toxicities. We discuss the properties of the vector platforms, such as their immunogenicity, potency, and modes of CAR delivery (permanent versus transient). Finally, we outline the work required to advance in vivo CAR therapy from proof-of-concept to a robust, scalable technology for clinical testing.
Collapse
Affiliation(s)
- Alexander Michels
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - Naphang Ho
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany
| | - Christian J Buchholz
- Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, Paul-Ehrlich-Straße 51-59, 63225 Langen, Germany;; Frankfurt Cancer Institute (FCI), Goethe-University, Paul-Ehrlich-Straße 42-44, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
129
|
Zhang H, Zhang L, An C, Zhang Y, Shao F, Gao Y, Zhang Y, Li H, Zhang Y, Ren C, Sun K, He W, Cheng F, Wang H, Weitz DA. Large-scale single-cell encapsulation in microgels through metastable droplet-templating combined with microfluidic-integration. Biofabrication 2022; 14. [PMID: 35593920 DOI: 10.1088/1758-5090/ac7168] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 05/05/2022] [Indexed: 11/11/2022]
Abstract
Current techniques for the generation of cell-laden microgels are limited by numerous challenges, including poorly uncontrolled batch-to-batch variations, processes that are both labor- and time-consuming, the high expense of devices and reagents, and low production rates; this hampers the translation of laboratory findings to clinical applications. To address these challenges, we develop a droplet-based microfluidic strategy based on metastable droplet-templating and microchannel integration for the substantial large-scale production of single cell-laden alginate microgels. Specifically, we present a continuous processing method for microgel generation by introducing amphiphilic perfluoronated alcohols to obtain metastable emulsion droplets as sacrificial templates. In addition, to adapt to the metastable emulsion system, integrated microfluidic chips containing 80 drop-maker units are designed and optimized based on the computational fluid dynamics simulation. This strategy allows single cell encapsulation in microgels at a maximum production rate of 10 ml hr-1 of cell suspension while retaining cell viability and functionality. These results represent a significant advance toward using cell-laden microgels for clinical-relevant applications, including cell therapy, tissue regeneration and 3D bioprinting.
Collapse
Affiliation(s)
- Haoyue Zhang
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Liyuan Zhang
- Harvard School of Engineering and Applied Sciences, Harvard University, Pierce Hall, 29 Oxford Street, Cambridge, MA 02138, Cambridge, Massachusetts, MA 02138, UNITED STATES
| | - Chuanfeng An
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Yang Zhang
- Shenzhen University, Laboratory of Regenerative Biomaterials, Department of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, 518037, P.R. China, Shenzhen, Guangdong, 518037, CHINA
| | - Fei Shao
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Yijie Gao
- Dalian Municipal Central Hospital Affiliated of Dalian Medical University, 42 Xuegong Street, Shahekou district, Dalian, Liaoning province, Dalian, Liaoning, 116033, CHINA
| | - Yonghao Zhang
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Hanting Li
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Yujie Zhang
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Changle Ren
- Dalian Municipal Central Hospital Affiliated of Dalian Medical University, 42 Xuegong Street, Shahekou district, Dalian, Liaoning province, Dalian, Liaoning, 116033, CHINA
| | - Kai Sun
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Wei He
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Fang Cheng
- Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, Liaoning, 116024, CHINA
| | - Huanan Wang
- dalian university of technology, Dalian University of Technology, Dalian University of Technology, No.2 Linggong Road, High-tech District, Dalian, 116024, P.R.China, Dalian, 116024, CHINA
| | - David A Weitz
- Harvard School of Engineering and Applied Sciences, Harvard University, Pierce Hall, 29 Oxford Street, Cambridge, MA 02138, Cambridge, Massachusetts, MA 02138, UNITED STATES
| |
Collapse
|
130
|
Lombard-Banek C, Pohl KI, Kwee EJ, Elliott JT, Schiel JE. A Sensitive and Controlled Data-Independent Acquisition Method for Proteomic Analysis of Cell Therapies. J Proteome Res 2022; 21:1229-1239. [PMID: 35404046 PMCID: PMC9087334 DOI: 10.1021/acs.jproteome.1c00887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Indexed: 11/29/2022]
Abstract
Mass spectrometry (MS)-based proteomic measurements are uniquely poised to impact the development of cell and gene therapies. With the adoption of rigorous instrumental performance qualifications (PQs), large-scale proteomics can move from a research to a manufacturing control tool. Especially suited, data-independent acquisition (DIA) approaches have distinctive qualities to extend multiattribute method (MAM) principles to characterize the proteome of cell therapies. Here, we describe the development of a DIA method for the sensitive identification and quantification of proteins on a Q-TOF instrument. Using the improved acquisition parameters, we defined a control strategy and highlighted some metrics to improve the reproducibility of SWATH acquisition-based proteomic measurements. Finally, we applied the method to analyze the proteome of Jurkat cells that here serves as a model for human T-cells. Raw and processed data were deposited in PRIDE (PXD029780).
Collapse
Affiliation(s)
- Camille Lombard-Banek
- National
Institute of Standards and Technology, Material and Measurements Laboratory, Gaithersburg, Maryland 20899, United States
- Institute
for Bioscience and Bioengineering Research, Rockville, Maryland 20850, United States
| | | | - Edward J. Kwee
- National
Institute of Standards and Technology, Material and Measurements Laboratory, Gaithersburg, Maryland 20899, United States
| | - John T. Elliott
- National
Institute of Standards and Technology, Material and Measurements Laboratory, Gaithersburg, Maryland 20899, United States
| | - John E. Schiel
- National
Institute of Standards and Technology, Material and Measurements Laboratory, Gaithersburg, Maryland 20899, United States
- Institute
for Bioscience and Bioengineering Research, Rockville, Maryland 20850, United States
| |
Collapse
|
131
|
Teoh J, Brown LF. Developing lisocabtagene maraleucel chimeric antigen receptor T-cell manufacturing for improved process, product quality and consistency across CD19+ hematologic indications. Cytotherapy 2022; 24:962-973. [DOI: 10.1016/j.jcyt.2022.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 11/26/2022]
|
132
|
Chen N, He Y, Zang M, Zhang Y, Lu H, Zhao Q, Wang S, Gao Y. Approaches and materials for endocytosis-independent intracellular delivery of proteins. Biomaterials 2022; 286:121567. [DOI: 10.1016/j.biomaterials.2022.121567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/26/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
|
133
|
Yao M, Yang JL, Wang DF, Wang L, Chen Y, Yao DF. Encouraging specific biomarkers-based therapeutic strategies for hepatocellular carcinoma. World J Clin Cases 2022; 10:3321-3333. [PMID: 35611205 PMCID: PMC9048543 DOI: 10.12998/wjcc.v10.i11.3321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/10/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
The prevention, early discovery and effective treatment of patients with hepatocellular carcinoma (HCC) remain a global medical challenge. At present, HCC is still mainly treated by surgery, supplemented by vascular embolization, radio frequency, radiotherapy, chemotherapy and biotherapy. The application of multikinase inhibitor sorafenib, chimeric antigen receptor T cells, or PD-1/PD-L1 inhibitors can prolong the median survival of HCC patients. However, the treatment efficacy is still unsatisfactory due to HCC metastasis and postoperative recurrence. During the process of hepatocyte malignant transformation, HCC tissues can express and secrete many types of specific biomarkers, or oncogenic antigen molecules into blood, for example, alpha-fetoprotein, glypican-3, Wnt3a (one of the key signaling molecules in the Wnt/β-catenin pathway), insulin-like growth factor (IGF)-II or IGF-I receptor, vascular endothelial growth factor, secretory clusterin and so on. In addition, combining immunotherapy with non-coding RNAs might improve anti-cancer efficacy. These biomarkers not only contribute to HCC diagnosis or prognosis, but may also become molecular targets for HCC therapy under developing or clinical trials. This article reviews the progress in emerging biomarkers in basic research or clinical trials for HCC immunotherapy.
Collapse
Affiliation(s)
- Min Yao
- Research Center of Clinical Medicine & Department of Immunology, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Jun-Ling Yang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - De-Feng Wang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Li Wang
- Department of Medical Informatics, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Ying Chen
- Department of Oncology, Affiliated Second Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Deng-Fu Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
134
|
Mi J, Ye Q, Min Y. Advances in Nanotechnology Development to Overcome Current Roadblocks in CAR-T Therapy for Solid Tumors. Front Immunol 2022; 13:849759. [PMID: 35401561 PMCID: PMC8983935 DOI: 10.3389/fimmu.2022.849759] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/01/2022] [Indexed: 11/17/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy for the treatment of hematologic tumors has achieved remarkable success, with five CAR-T therapies approved by the United States Food and Drug Administration. However, the efficacy of CAR-T therapy against solid tumors is not satisfactory. There are three existing hurdles in CAR-T cells for solid tumors. First, the lack of a universal CAR to recognize antigens at the site of solid tumors and the compact tumor structure make it difficult for CAR-T cells to locate in solid tumors. Second, soluble inhibitors and suppressive immune cells in the tumor microenvironment can inhibit or even inactivate T cells. Third, low survival and proliferation rates of CAR-T cells in vivo significantly influence the therapeutic effect. As an emerging method, nanotechnology has a great potential to enhance cell proliferation, activate T cells, and restarting the immune response. In this review, we discuss how nanotechnology can modify CAR-T cells through variable methods to improve the therapeutic effect of solid tumors.
Collapse
Affiliation(s)
- Juan Mi
- Department of Pathology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qing Ye
- Department of Pathology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanzeng Min
- CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China.,Department of Endocrinology, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Science at the Microscale, University of Science and Technology of China, Hefei, China
| |
Collapse
|
135
|
Qian S, Villarejo-Campos P, Guijo I, Hernández-Villafranca S, García-Olmo D, González-Soares S, Guadalajara H, Jiménez-Galanes S, Qian C. Update for Advance CAR-T Therapy in Solid Tumors, Clinical Application in Peritoneal Carcinomatosis From Colorectal Cancer and Future Prospects. Front Immunol 2022; 13:841425. [PMID: 35401510 PMCID: PMC8990899 DOI: 10.3389/fimmu.2022.841425] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Latest advances in the field of cancer immunotherapy have developed the (Chimeric Antigen Receptor) CAR-T cell therapy. This therapy was first used in hematological malignancies which obtained promising results; therefore, the use of CAR-T cells has become a popular approach for treating non-solid tumors. CAR-T cells consist of T-lymphocytes that are engineered to express an artificial receptor against any surface antigen of our choice giving us the capacity of offering precise and personalized treatment. This leaded to the development of CAR-T cells for treating solid tumors with the hope of obtaining the same result; however, their use in solid tumor and their efficacy have not achieved the expected results. The reason of these results is because solid tumors have some peculiarities that are not present in hematological malignancies. In this review we explain how CAR-T cells are made, their mechanism of action, adverse effect and how solid tumors can evade their action, and also we summarize their use in colorectal cancer and peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Siyuan Qian
- Department of Surgery, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | | | - Ismael Guijo
- Department of Surgery, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | | | - Damián García-Olmo
- Department of Surgery, Fundación Jimenez Diaz University Hospital, Madrid, Spain
- Department of Surgery, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sara González-Soares
- Department of Surgery, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | - Héctor Guadalajara
- Department of Surgery, Fundación Jimenez Diaz University Hospital, Madrid, Spain
| | | | - Cheng Qian
- Chongqing Precision Biotechnology Co. Ltd, Chongqing, China
| |
Collapse
|
136
|
Michelozzi IM, Sufi J, Adejumo TA, Amrolia PJ, Tape CJ, Giustacchini A. High-dimensional functional phenotyping of preclinical human CAR T cells using mass cytometry. STAR Protoc 2022; 3:101174. [PMID: 35199038 PMCID: PMC8844283 DOI: 10.1016/j.xpro.2022.101174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Here, we present a comprehensive protocol for the generation and functional characterization of chimeric antigen receptor (CAR) T cells and their products by mass cytometry in a reproducible and scalable manner. We describe the production of CAR T cells from human peripheral blood mononuclear cells. We then detail a three-step staining protocol with metal-labeled antibodies and the subsequent mass cytometry analysis. This protocol allows simultaneous characterization of CAR T cell intracellular signaling, activation, proliferation, cytokine production, and phenotype in a single assay.
Collapse
Affiliation(s)
- Ilaria M. Michelozzi
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, Zayed Centre For Research into Rare Disease in Children, WC1N 1DZ London, UK
| | - Jahangir Sufi
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, WC1E 6DD London, UK
| | | | - Persis J. Amrolia
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, Zayed Centre For Research into Rare Disease in Children, WC1N 1DZ London, UK
- Department of Bone Marrow Transplant, Great Ormond Street Hospital for Children, WC1N 3JH London, UK
| | - Christopher J. Tape
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, WC1E 6DD London, UK
| | - Alice Giustacchini
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, Zayed Centre For Research into Rare Disease in Children, WC1N 1DZ London, UK
| |
Collapse
|
137
|
Xin T, Cheng L, Zhou C, Zhao Y, Hu Z, Wu X. In-Vivo Induced CAR-T Cell for the Potential Breakthrough to Overcome the Barriers of Current CAR-T Cell Therapy. Front Oncol 2022; 12:809754. [PMID: 35223491 PMCID: PMC8866962 DOI: 10.3389/fonc.2022.809754] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T cell) therapy has shown impressive success in the treatment of hematological malignancies, but the systemic toxicity and complex manufacturing process of current autologous CAR-T cell therapy hinder its broader applications. Universal CAR-T cells have been developed to simplify the production process through isolation and editing of allogeneic T cells from healthy persons, but the allogeneic CAR-T cells have recently encountered safety concerns, and clinical trials have been halted by the FDA. Thus, there is an urgent need to seek new ways to overcome the barriers of current CAR-T cell therapy. In-vivo CAR-T cells induced by nanocarriers loaded with CAR-genes and gene-editing tools have shown efficiency for regressing leukemia and reducing systemic toxicity in a mouse model. The in-situ programming of autologous T-cells avoids the safety concerns of allogeneic T cells, and the manufacture of nanocarriers can be easily standardized. Therefore, the in-vivo induced CAR-T cells can potentially overcome the abovementioned limitations of current CAR-T cell therapy. Here, we provide a review on CAR structures, gene-editing tools, and gene delivery techniques applied in immunotherapy to help design and develop new in-vivo induced CAR-T cells.
Collapse
Affiliation(s)
- Tianqing Xin
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li Cheng
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chuchao Zhou
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yimeng Zhao
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhenhua Hu
- Department of Health and Nursing, Nanfang College of Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Wu
- Department of Pediatrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
138
|
Koedam J, Wermke M, Ehninger A, Cartellieri M, Ehninger G. Chimeric antigen receptor T-cell therapy in acute myeloid leukemia. Curr Opin Hematol 2022; 29:74-83. [PMID: 35013048 PMCID: PMC8815830 DOI: 10.1097/moh.0000000000000703] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Treatment outcome of relapsed or refractory AML patients remains dismal and new treatment options are needed. Adoptive cell therapy using CAR-T cells is a potentially interesting approach in this. RECENT FINDINGS Several potentially interesting AML targets are being investigated with CAR-T therapy with over 60 clinical trials listed on clinicaltrials.gov. The first clinical data are only just emerging with mixed results, once more proving that further research is needed. SUMMARY Adoptive cell therapy using chimeric antigen receptor T cells is being investigated in AML through many clinical trials. So far, no AML-specific antigen has been identified, requiring additional strategies to mitigate on-target off-tumor toxicity and to increase efficacy. Focus point is to acquire control over the CAR T cells once administered. Strategies to do so include biodegradable CARs, inducible CARs, suicide-switch containing CARs and two-component modular CARs. Limited and mixed results are available, confirming the risk of lasting toxicity for nonswitchable CARs. Initial results of modular CARs suggest toxicity can be mitigated whilst maintaining CAR activity by the use of modular CAR concepts that allows for 'ON' and 'OFF' switching.
Collapse
Affiliation(s)
| | - Martin Wermke
- Division of Hematology, Oncology and Stem Cell Transplantation, Medical Clinic I, Department of Medicine I, University Hospital Carl Gustav Carus
- National Center for Tumor Diseases
| | | | | | | |
Collapse
|
139
|
Poorebrahim M, Quiros-Fernandez I, Fakhr E, Cid-Arregui A. Generation of CAR-T cells using lentiviral vectors. Methods Cell Biol 2022; 167:39-69. [PMID: 35152998 DOI: 10.1016/bs.mcb.2021.07.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer immunotherapy is nowadays largely focused on the development of therapeutic antibodies and chimeric antigen receptors (CARs). Two CARs targeting CD19 have been approved recently for the treatment of some hematological malignancies. This demonstrates the capability of engineered CAR T cells in generating effective tumor responses. Furthermore, several hundred ongoing clinical trials are exploring the feasibility of CAR-based approaches to target tumor-associated antigens in solid tumors. However, there still remain significant challenges and limitations in the design and production of CAR-modified T cells that need to be addressed, such as more effective transduction methods, expression and exhaustion issues, reliable in vitro and in vivo characterization methods, etc. Here we describe current techniques for generating CAR T cells using lentiviral vectors as well as detailed protocols for their functional characterization.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Isaac Quiros-Fernandez
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elham Fakhr
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angel Cid-Arregui
- Targeted Tumor Vaccines Group, Clinical Cooperation Unit Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
140
|
Gurney M, Kundu S, Pandey S, O’Dwyer M. Feeder Cells at the Interface of Natural Killer Cell Activation, Expansion and Gene Editing. Front Immunol 2022; 13:802906. [PMID: 35222382 PMCID: PMC8873083 DOI: 10.3389/fimmu.2022.802906] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
Genome engineered natural killer (NK) cell therapies are emerging as a promising cancer immunotherapy platform with potential advantages and remaining uncertainties. Feeder cells induce activation and proliferation of NK cells via cell surface receptor-ligand interactions, supported by cytokines. Feeder cell expanded NK cell products have supported several NK cell adoptive transfer clinical trials over the past decade. Genome engineered NK cell therapies, including CAR-NK cells, seek to combine innate and alloreactive NK cell anti-tumor activity with antigen specific targeting or additional modifications aimed at improving NK cell persistence, homing or effector function. The profound activating and expansion stimulus provided by feeder cells is integral to current applications of clinical-scale genome engineering approaches in donor-derived, primary NK cells. Herein we explore the complex interactions that exist between feeder cells and both viral and emerging non-viral genome editing technologies in NK cell engineering. We focus on two established clinical-grade feeder systems; Epstein-Barr virus transformed lymphoblastoid cell lines and genetically engineered K562.mbIL21.4-1BBL feeder cells.
Collapse
Affiliation(s)
- Mark Gurney
- Department: Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland
| | - Soumyadipta Kundu
- Department: Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland
- ONK Therapeutics, Galway, Ireland
| | | | - Michael O’Dwyer
- Department: Apoptosis Research Centre, National University of Ireland Galway, Galway, Ireland
- ONK Therapeutics, Galway, Ireland
- *Correspondence: Michael O’Dwyer,
| |
Collapse
|
141
|
Smirnov S, Petukhov A, Levchuk K, Kulemzin S, Staliarova A, Lepik K, Shuvalov O, Zaritskey A, Daks A, Fedorova O. Strategies to Circumvent the Side-Effects of Immunotherapy Using Allogeneic CAR-T Cells and Boost Its Efficacy: Results of Recent Clinical Trials. Front Immunol 2022; 12:780145. [PMID: 34975869 PMCID: PMC8714645 DOI: 10.3389/fimmu.2021.780145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 11/22/2021] [Indexed: 12/27/2022] Open
Abstract
Despite the outstanding results of treatment using autologous chimeric antigen receptor T cells (CAR-T cells) in hematological malignancies, this approach is endowed with several constraints. In particular, profound lymphopenia in some patients and the inability to manufacture products with predefined properties or set of cryopreserved batches of cells directed to different antigens in advance. Allogeneic CAR-T cells have the potential to address these issues but they can cause life-threatening graft-versus-host disease or have shorter persistence due to elimination by the host immune system. Novel strategies to create an “off the shelf” allogeneic product that would circumvent these limitations are an extensive area of research. Here we review CAR-T cell products pioneering an allogeneic approach in clinical trials.
Collapse
Affiliation(s)
- Sergei Smirnov
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia
| | - Alexey Petukhov
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia.,Institute of Cytology, Laboratory of Gene Expression Regulation, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Ksenia Levchuk
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia
| | - Sergey Kulemzin
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia.,Institute of Molecular and Cellular Biology SB Russian Academy of Science (RAS), Department of Molecular Immunology, Laboratory of Immunogenetics, Novosibirsk, Russia
| | - Alena Staliarova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Oncological Department 3, Borovliani, Minsk Region, Belarus
| | - Kirill Lepik
- RM Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Chemotherapy and Bone Marrow Transplantation Department, Saint Petersburg, Russia.,Pavlov University, Department of Hematology, Transfusiology and Transplantology, Saint Petersburg, Russia
| | - Oleg Shuvalov
- Institute of Cytology, Laboratory of Gene Expression Regulation, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Andrey Zaritskey
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia
| | - Alexandra Daks
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia.,Institute of Cytology, Laboratory of Gene Expression Regulation, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Olga Fedorova
- Almazov National Medical Research Centre, Personalized Medicine Centre, Saint Petersburg, Russia.,Institute of Cytology, Laboratory of Gene Expression Regulation, Russian Academy of Sciences, Saint Petersburg, Russia
| |
Collapse
|
142
|
Stock S, Kluever AK, Endres S, Kobold S. Enhanced Chimeric Antigen Receptor T Cell Therapy through Co-Application of Synergistic Combination Partners. Biomedicines 2022; 10:biomedicines10020307. [PMID: 35203517 PMCID: PMC8869718 DOI: 10.3390/biomedicines10020307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/18/2022] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable response rates and revolutionized the treatment of patients suffering from defined hematological malignancies. However, many patients still do not respond to this therapy or relapse after an initial remission, underscoring the need for improved efficacy. Insufficient in vivo activity, persistence, trafficking, and tumor infiltration of CAR T cells, as well as antigen escape and treatment-associated adverse events, limit the therapeutic success. Multiple strategies and approaches have been investigated to further improve CAR T cell therapy. Besides genetic modification of the CAR itself, the combination with other treatment modalities has the potential to improve this approach. In particular, combining CAR T cells with clinically approved compounds such as monoclonal antibodies and small molecule inhibitors might be a promising strategy. Combination partners could already be applied during the production process to influence the cellular composition and immunophenotype of the final CAR T cell product. Alternatively, simultaneous administration of clinically approved compounds with CAR T cells would be another feasible avenue. In this review, we will discuss current strategies to combine CAR T cells with compounds to overcome recent limitations and further enhance this promising cancer therapy, potentially broadening its application beyond hematology.
Collapse
Affiliation(s)
- Sophia Stock
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, 81337 Munich, Germany
- Correspondence: (S.S.); (S.K.)
| | - Anna-Kristina Kluever
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
| | - Stefan Endres
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 80336 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, 80337 Munich, Germany; (A.-K.K.); (S.E.)
- German Center for Translational Cancer Research (DKTK), Partner Site Munich, 80336 Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
- Correspondence: (S.S.); (S.K.)
| |
Collapse
|
143
|
Cao W, Geng ZZ, Wang N, Pan Q, Guo S, Xu S, Zhou J, Liu WR. A Reversible Chemogenetic Switch for Chimeric Antigen Receptor T Cells**. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202109550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Wenyue Cao
- Department of Hematology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei China
- The Texas A&M Drug Discovery Laboratory Department of Chemistry Texas A&M University College Station TX 77843-3255 USA
| | - Zhi Zachary Geng
- The Texas A&M Drug Discovery Laboratory Department of Chemistry Texas A&M University College Station TX 77843-3255 USA
| | - Na Wang
- Department of Hematology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei China
| | - Quan Pan
- The Texas A&M Drug Discovery Laboratory Department of Chemistry Texas A&M University College Station TX 77843-3255 USA
| | - Shaodong Guo
- The Texas A&M Drug Discovery Laboratory Department of Chemistry Texas A&M University College Station TX 77843-3255 USA
| | - Shiqing Xu
- The Texas A&M Drug Discovery Laboratory Department of Chemistry Texas A&M University College Station TX 77843-3255 USA
| | - Jianfeng Zhou
- Department of Hematology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei China
| | - Wenshe Ray Liu
- The Texas A&M Drug Discovery Laboratory Department of Chemistry Texas A&M University College Station TX 77843-3255 USA
- Institute of Biosciences and Technology and Department of Translational Medical Sciences College of Medicine Texas A&M University Houston TX 77030 USA
- Department of Biochemistry and Biophysics Texas A&M University Houston TX 77843 USA
- Department of Molecular and Cellular Medicine College of Medicine Texas A&M University Houston TX 77843 USA
| |
Collapse
|
144
|
Guha P, Katz SC. Strategies for manufacturing cell therapy products aligned with patient needs. Methods Cell Biol 2022; 167:203-226. [DOI: 10.1016/bs.mcb.2021.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
145
|
Cai Y, Prochazkova M, Jiang C, Song HW, Jin J, Moses L, Gkitsas N, Somerville RP, Highfill SL, Panch S, Stroncek DF, Jin P. Establishment and validation of in-house cryopreserved CAR/TCR-T cell flow cytometry quality control. J Transl Med 2021; 19:523. [PMID: 34952597 PMCID: PMC8705121 DOI: 10.1186/s12967-021-03193-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 12/11/2021] [Indexed: 11/10/2022] Open
Abstract
Background Chimeric antigen receptor (CAR) or T-cell receptor (TCR) engineered T-cell therapy has recently emerged as a promising adoptive immunotherapy approach for the treatment of hematologic malignancies and solid tumors. Multiparametric flow cytometry-based assays play a critical role in monitoring cellular manufacturing steps. Since manufacturing CAR/TCR T-cell products must be in compliance with current good manufacturing practices (cGMP), a standard or quality control for flow cytometry assays should be used to ensure the accuracy of flow cytometry results, but none is currently commercially available. Therefore, we established a procedure to generate an in-house cryopreserved CAR/TCR T-cell products for use as a flow cytometry quality control and validated their use. Methods Two CAR T-cell products: CD19/CD22 bispecific CAR T-cells and FGFR4 CAR T-cells and one TCR-engineered T-cell product: KK-LC-1 TCR T-cells were manufactured in Center for Cellular Engineering (CCE), NIH Clinical Center. The products were divided in aliquots, cryopreserved and stored in the liquid nitrogen. The cryopreserved flow cytometry quality controls were tested in flow cytometry assays which measured post-thaw viability, CD3, CD4 and CD8 frequencies as well as the transduction efficiency and vector identity. The long-term stability and shelf-life of cryopreserved quality control cells were evaluated. In addition, the sensitivity as well as the precision assay were also assessed on the cryopreserved quality control cells. Results After thawing, the viability of the cryopreserved CAR/TCR T-cell controls was found to be greater than 50%. The expression of transduction efficiency and vector identity markers by the cryopreserved control cells were stable for at least 1 year; with post-thaw values falling within ± 20% range of the values measured at time of cryopreservation. After thawing and storage at room temperature, the stability of these cryopreserved cells lasted at least 6 h. In addition, our cryopreserved CAR/TCR-T cell quality controls showed a strong correlation between transduction efficiency expression and dilution factors. Furthermore, the results of flow cytometric analysis of the cryopreserved cells among different laboratory technicians and different flow cytometry instruments were comparable, highlighting the reproducibility and reliability of these quality control cells. Conclusion We developed and validated a feasible and reliable procedure to establish a bank of cryopreserved CAR/TCR T-cells for use as flow cytometry quality controls, which can serve as a quality control standard for in-process and lot-release testing of CAR/TCR T-cell products. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03193-7.
Collapse
Affiliation(s)
- Yihua Cai
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Michaela Prochazkova
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Chunjie Jiang
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Hannah W Song
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Jianjian Jin
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Larry Moses
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Nikolaos Gkitsas
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Robert P Somerville
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Steven L Highfill
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Sandhya Panch
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - David F Stroncek
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA
| | - Ping Jin
- Department of Transfusion Medicine and Cellular Engineering, Center for Cellular Engineering, NIH Clinical Center, NIH, 10 Center Drive-MSC-1184, Building 10, Room 3C720, Bethesda, MD, 20892-1184, USA.
| |
Collapse
|
146
|
Maru B, Nadeau L, McKeague M. Enhancing CAR-T Cell Therapy with Functional Nucleic Acids. ACS Pharmacol Transl Sci 2021; 4:1716-1727. [PMID: 34927006 DOI: 10.1021/acsptsci.1c00188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Indexed: 02/07/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a relatively new form of immunotherapy that has had success in treating patients with hematologic malignancies, leading to three recent United States Food and Drug Administration approvals. However, several challenges hinder the widespread use of CAR-T therapy. Here, we review the application of functional nucleic acids such as aptamers and ribozymes as novel tools to improve a variety of steps in CAR-T cell therapy development. We critically examine key studies that highlight the benefits of functional nucleic acids at different stages of cell-based therapy and discuss the feasibility of their practical clinical application. Finally, we offer insights into potential opportunities where chemists can significantly contribute to the innovative incorporation of functional nucleic acids to overcome challenges associated with this cutting-edge immunotherapy.
Collapse
Affiliation(s)
- Bruktawit Maru
- Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada
| | - Lea Nadeau
- Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada
| | - Maureen McKeague
- Pharmacology and Therapeutics, Faculty of Medicine, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada.,Department of Chemistry, Faculty of Science, McGill University, 801 Sherbrooke Street West, Montreal, QC H3A 0B8, Canada
| |
Collapse
|
147
|
Li YR, Dunn ZS, Zhou Y, Lee D, Yang L. Development of Stem Cell-Derived Immune Cells for Off-the-Shelf Cancer Immunotherapies. Cells 2021; 10:cells10123497. [PMID: 34944002 PMCID: PMC8700013 DOI: 10.3390/cells10123497] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/04/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
Cell-based cancer immunotherapy has revolutionized the treatment of hematological malignancies. Specifically, autologous chimeric antigen receptor-engineered T (CAR-T) cell therapies have received approvals for treating leukemias, lymphomas, and multiple myeloma following unprecedented clinical response rates. A critical barrier to the widespread usage of current CAR-T cell products is their autologous nature, which renders these cellular products patient-selective, costly, and challenging to manufacture. Allogeneic cell products can be scalable and readily administrable but face critical concerns of graft-versus-host disease (GvHD), a life-threatening adverse event in which therapeutic cells attack host tissues, and allorejection, in which host immune cells eliminate therapeutic cells, thereby limiting their antitumor efficacy. In this review, we discuss recent advances in developing stem cell-engineered allogeneic cell therapies that aim to overcome the limitations of current autologous and allogeneic cell therapies, with a special focus on stem cell-engineered conventional αβ T cells, unconventional T (iNKT, MAIT, and γδ T) cells, and natural killer (NK) cells.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Z.); (D.L.)
| | - Zachary Spencer Dunn
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA;
| | - Yang Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Z.); (D.L.)
| | - Derek Lee
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Z.); (D.L.)
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (Y.-R.L.); (Y.Z.); (D.L.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
- Correspondence:
| |
Collapse
|
148
|
Li Z, Sun G, Sun G, Cheng Y, Wu L, Wang Q, Lv C, Zhou Y, Xia Y, Tang W. Various Uses of PD1/PD-L1 Inhibitor in Oncology: Opportunities and Challenges. Front Oncol 2021; 11:771335. [PMID: 34869005 PMCID: PMC8635629 DOI: 10.3389/fonc.2021.771335] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022] Open
Abstract
The occurrence and development of cancer are closely related to the immune escape of tumor cells and immune tolerance. Unlike previous surgical, chemotherapy, radiotherapy and targeted therapy, tumor immunotherapy is a therapeutic strategy that uses various means to stimulate and enhance the immune function of the body, and ultimately achieves the goal of controlling tumor cells.With the in-depth understanding of tumor immune escape mechanism and tumor microenvironment, and the in-depth study of tumor immunotherapy, immune checkpoint inhibitors represented by Programmed Death 1/Programmed cell Death-Ligand 1(PD-1/PD-L1) inhibitors are becoming increasingly significant in cancer medication treatment. employ a variety of ways to avoid detection by the immune system, a single strategy is not more effective in overcoming tumor immune evasion and metastasis. Combining different immune agents or other drugs can effectively address situations where immunotherapy is not efficacious, thereby increasing the chances of success and alternative access to alternative immunotherapy. Immune combination therapies for cancer have become a hot topic in cancer treatment today. In this paper, several combination therapeutic modalities of PD1/PD-L1 inhibitors are systematically reviewed. Finally, an analysis and outlook are provided in the context of the recent advances in combination therapy with PD1/PD-L1 inhibitors and the pressing issues in this field.
Collapse
Affiliation(s)
- Zhitao Li
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guoqiang Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Guangshun Sun
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Ye Cheng
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liangliang Wu
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qian Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Chengyu Lv
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yichan Zhou
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongxiang Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, China
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation, Nanjing Medical University, Nanjing, China
| |
Collapse
|
149
|
Deng B, Pan J, Liu Z, Liu S, Chen Y, Qu X, Zhang Y, Lin Y, Zhang Y, Yu X, Zhang Z, Niu X, Luan R, Ma M, Li X, Liu T, Wu X, Niu H, Chang AH, Tong C. Peripheral leukemia burden at time of apheresis negatively affects the clinical efficacy of CART19 in refractory or relapsed B-ALL. Mol Ther Methods Clin Dev 2021; 23:633-643. [PMID: 34901308 PMCID: PMC8640733 DOI: 10.1016/j.omtm.2021.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/07/2021] [Accepted: 10/26/2021] [Indexed: 12/01/2022]
Abstract
Our previous clinical study achieved complete remission (CR) rates of >90% following chimeric antigen receptor T cells targeting CD19 (CART19) treatment of refractory/relapsed B cell acute lymphoblastic leukemia (r/r B-ALL); however, the influence of the leukemia burden in peripheral blood (PB) blasts remains unclear. Here, we retrospectively analyzed 143 patients treated with CART19 (including 36 patients with PB blasts) to evaluate the effect of peripheral leukemia burden at the time of apheresis. One hundred seventeen patients with high disease burdens achieved 91.5% CR or incomplete count recovery CR and 86.3% minimal residual disease-negative CR, and 26 patients with low disease burdens obtained 96.2% MRD− CR. Collectively, 9 of 36 (25%) patients with PB blasts and 2 of 107 (1.87%) patients without PB blasts did not respond to CART19 therapy. The leukemia burden in PB negatively influenced ex vivo cell characteristics, including the transduction efficiency of CD3+ T cells and their fold expansion, and in vivo cell dynamics, including peak CART19 proportion and absolute count, fold expansion, and persistence duration. Further studies showed that these patients had higher programmed death-1 expression in CART19 products. Our data imply that PB blasts negatively affected CART19 production and the clinical efficacy of CART19 therapy in patients with r/r B-ALL.
Collapse
Affiliation(s)
- Biping Deng
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Jing Pan
- Department of Hematology, Beijing Boren Hospital, Beijing 100070, China
| | - Zhaoli Liu
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Shuangyou Liu
- Department of Hematology, Beijing Boren Hospital, Beijing 100070, China
| | - Yunlong Chen
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Xiaomin Qu
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Yu'e Zhang
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Yuehui Lin
- Department of Hematology, Beijing Boren Hospital, Beijing 100070, China
| | - Yanlei Zhang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Xinjian Yu
- Medical Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Zhongxin Zhang
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Xuansha Niu
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Rong Luan
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Ming Ma
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Xiaomei Li
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Tingting Liu
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Xi'ai Wu
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Huan Niu
- Cytology Laboratory, Beijing Boren Hospital, Beijing 100070, China
| | - Alex H. Chang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Corresponding author: Alex H. Chang, Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Chunrong Tong
- Department of Hematology, Beijing Boren Hospital, Beijing 100070, China
- Corresponding author: Chunrong Tong, Department of Hematology, Beijing Boren Hospital, No. 6, South Zhengwangfen, Fengtai District, Beijing 100070, China.
| |
Collapse
|
150
|
Ramos RN, Picanço-Castro V, Oliveira TGM, Mendrone A, De Santis GC, Bonamino MH, Rocha V. Associação Brasileira de Hematologia, Hemoterapia e Terapia Celular Consensus on genetically modified cells. VII. Present and future of technologies for production of CAR cell therapies. Hematol Transfus Cell Ther 2021; 43 Suppl 2:S46-S53. [PMID: 34794797 PMCID: PMC8606694 DOI: 10.1016/j.htct.2021.09.007] [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: 08/24/2021] [Accepted: 09/14/2021] [Indexed: 11/28/2022] Open
Abstract
Chimeric Antigen Receptor T (CAR-T) cells are certainly an important therapy for patients with relapsed and/or refractory hematologic malignancies. Currently, there are five CAR-T cell products approved by the FDA but several research groups and/or biopharmaceutical companies are encouraged to develop new products based on CAR cells using T or other cell types. Production of CAR cells requires intensive work from the basic, pre-clinical to translational levels, aiming to overcome technical difficulties and failure in the production. At least five key common steps are needed for the manipulation of T-lymphocytes (or other cells), such as: cell type selection, activation, gene delivery, cell expansion and final product formulation. However, reproducible manufacturing of high-quality clinical-grade CAR cell products is still required to apply this technology to a greater number of patients. This chapter will discuss the present and future development of new CAR designs that are safer and more effective to improve this therapy, achieving more selective killing of malignant cells and less toxicity to be applied in the clinical setting.
Collapse
Affiliation(s)
- Rodrigo Nalio Ramos
- Laboratório de Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Instituto D'Or de Ensino e Pesquisa, São Paulo, Brazil
| | - Virginia Picanço-Castro
- Fundação Hemocentro de Ribeirão Preto, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, (HC FMRPUSP) Ribeirão Preto, SP, Brazil
| | - Theo Gremen M Oliveira
- Laboratório de Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Fundação Pró-Sangue-Hemocentro de São Paulo, São Paulo, Brazil
| | | | - Gil Cunha De Santis
- Fundação Hemocentro de Ribeirão Preto, Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto da Universidade de São Paulo, (HC FMRPUSP) Ribeirão Preto, SP, Brazil
| | - Martin Hernan Bonamino
- Divisão de Pesquisa Experimental e Translacional, Instituto Nacional do Câncer (INCA), Rio de Janeiro, RJ, Brazil; Vice-Presidência de Pesquisa e Coleções Biológicas da Fundação Oswaldo Cruz ((VPPCB FIOCRUZ), Rio de Janeiro, RJ, Brazil
| | - Vanderson Rocha
- Laboratório de Investigação Médica em Patogênese e Terapia dirigida em Onco-Imuno-Hematologia, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Instituto D'Or de Ensino e Pesquisa, São Paulo, Brazil; Fundação Pró-Sangue-Hemocentro de São Paulo, São Paulo, Brazil.
| |
Collapse
|