1
|
Pandit S, Smith BE, Birnbaum ME, Brudno Y. A biomaterial platform for T cell-specific gene delivery. Acta Biomater 2024; 177:157-164. [PMID: 38364929 PMCID: PMC10948289 DOI: 10.1016/j.actbio.2024.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/18/2024] [Accepted: 02/09/2024] [Indexed: 02/18/2024]
Abstract
Efficient T cell engineering is central to the success of CAR T cell therapy but involves multiple time-consuming manipulations, including T cell isolation, activation, and transduction. These steps add complexity and delay CAR T cell manufacturing, which takes a mean time of 4 weeks. To streamline T cell engineering, we strategically combine two critical engineering solutions - T cell-specific lentiviral vectors and macroporous scaffolds - that enable T cell activation and transduction in a simple, single step. The T cell-specific lentiviral vectors (referred to as STAT virus) target T cells through the display of an anti-CD3 antibody and the CD80 extracellular domain on their surface and provide robust T cell activation. Biocompatible macroporous scaffolds (referred to as Drydux) mediate robust transduction by providing effective interaction between naïve T cells and viral vectors. We show that when unstimulated peripheral blood mononuclear cells (PBMCs) are seeded together with STAT lentivirus on Drydux scaffolds, T cells are activated, selectively transduced, and reprogrammed in a single step. Further, we show that the Drydux platform seeded with PBMCs and STAT lentivirus generates tumor-specific functional CAR T cells. This potent combination of engineered lentivirus and biomaterial scaffold holds promise for an effective, simple, and safe avenue for in vitro and in vivo T cell engineering. STATEMENT OF SIGNIFICANCE: Manufacturing T cell therapies involves lengthy and labor-intensive steps, including T cell selection, activation, and transduction. These steps add complexity to current CAR T cell manufacturing protocols and limit widespread patient access to this revolutionary therapy. In this work, we demonstrate the combination of engineered virus and biomaterial platform that, together, enables selective T cell activation and transduction in a single step, eliminating multistep T cell engineering protocols and significantly simplifying the manufacturing process.
Collapse
Affiliation(s)
- Sharda Pandit
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA
| | - Blake E Smith
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA; Singapore-MIT Alliance for Research and Technology Centre, Singapore, Singapore
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, USA; Comparative Medicine Institute, North Carolina State University, Raleigh, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Kaygisiz K, Rauch‐Wirth L, Iscen A, Hartenfels J, Kremer K, Münch J, Synatschke CV, Weil T. Peptide Amphiphiles as Biodegradable Adjuvants for Efficient Retroviral Gene Delivery. Adv Healthc Mater 2024; 13:e2301364. [PMID: 37947246 PMCID: PMC11468294 DOI: 10.1002/adhm.202301364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 10/20/2023] [Indexed: 11/12/2023]
Abstract
Retroviral gene delivery is the key technique for in vitro and ex vivo gene therapy. However, inefficient virion-cell attachment resulting in low gene transduction efficacy remains a major challenge in clinical applications. Adjuvants for ex vivo therapy settings need to increase transduction efficiency while being easily removed or degraded post-transduction to prevent the risk of venous embolism after infusing the transduced cells back to the bloodstream of patients, yet no such peptide system have been reported thus far. In this study, peptide amphiphiles (PAs) with a hydrophobic fatty acid and a hydrophilic peptide moiety that reveal enhanced viral transduction efficiency are introduced. The PAs form β-sheet-rich fibrils that assemble into positively charged aggregates, promoting virus adhesion to the cell membrane. The block-type amphiphilic sequence arrangement in the PAs ensures efficient cell-virus interaction and biodegradability. Good biodegradability is observed for fibrils forming small aggregates and it is shown that via molecular dynamics simulations, the fibril-fibril interactions of PAs are governed by fibril surface hydrophobicity. These findings establish PAs as additives in retroviral gene transfer, rivalling commercially available transduction enhancers in efficiency and degradability with promising translational options in clinical gene therapy applications.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Lena Rauch‐Wirth
- Institute of Molecular VirologyUlm University Medical CenterMeyerhofstraße 189081UlmGermany
| | - Aysenur Iscen
- Polymer Theory DepartmentMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jan Hartenfels
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Kurt Kremer
- Polymer Theory DepartmentMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Jan Münch
- Institute of Molecular VirologyUlm University Medical CenterMeyerhofstraße 189081UlmGermany
| | - Christopher V. Synatschke
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| | - Tanja Weil
- Department Synthesis of MacromoleculesMax Planck Institute for Polymer ResearchAckermannweg 1055128MainzGermany
| |
Collapse
|
3
|
Loos P, Short L, Savage G, Evgin L. Expansion and Retroviral Transduction of Primary Murine T Cells for CAR T-Cell Therapy. Methods Mol Biol 2024; 2748:41-53. [PMID: 38070106 DOI: 10.1007/978-1-0716-3593-3_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
The development of chimeric antigen receptor (CAR) T cells has been a revolutionary technology for the treatment of relapsed and refractory leukemias and lymphomas. The synthetic CAR molecule redirects T cell function toward tumor surface-expressed antigens through a single-chain variable fragment (scFv) fused to CD3z and intracellular costimulatory domains. Here, we describe a protocol for the generation of CAR T cells using primary mouse T cells and a gammaretroviral vector encoding a CAR transgene. This protocol outlines several transduction and expansion methods based on the use of two transduction enhancers, RetroNectin® and Vectofusin®-1, and cell culture systems such as conventional plates or G-Rex® devices.
Collapse
Affiliation(s)
- Pauline Loos
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Lauralie Short
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Gillian Savage
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Laura Evgin
- Michael Smith Genome Sciences Department, BC Cancer Research Institute, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
4
|
Malach P, Kay C, Tinworth C, Patel F, Joosse B, Wade J, Rosa do Carmo M, Donovan B, Brugman M, Montiel-Equihua C, Francis N. Identification of a small molecule for enhancing lentiviral transduction of T cells. Mol Ther Methods Clin Dev 2023; 31:101113. [PMID: 37790244 PMCID: PMC10544093 DOI: 10.1016/j.omtm.2023.101113] [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: 04/21/2023] [Accepted: 09/13/2023] [Indexed: 10/05/2023]
Abstract
Genetic modification of cells using viral vectors has shown huge therapeutic benefit in multiple diseases. However, inefficient transduction contributes to the high cost of these therapies. Several transduction-enhancing small molecules have previously been identified; however, some may be toxic to the cells or patient, otherwise alter cellular characteristics, or further increase manufacturing complexity. In this study, we aimed to identify molecules capable of enhancing lentiviral transduction of T cells from available small-molecule libraries. We conducted a high-throughput flow-cytometry-based screen of 27,892 compounds, which subsequently was narrowed down to six transduction-enhancing small molecules for further testing with two therapeutic lentiviral vectors used to manufacture GSK's clinical T cell therapy products. We demonstrate enhanced transduction without a negative impact on other product attributes. Furthermore, we present results of transcriptomic analysis, suggesting alteration of ribosome biogenesis, resulting in reduced interferon response, as a potential mechanism of action for the transduction-enhancing activity of the lead compound. Finally, we demonstrate the ability of the lead transduction enhancer to produce a comparable T cell product using a 3-fold reduction in vector volume in our clinical manufacturing process, resulting in a predicted 15% reduction in the overall cost of goods.
Collapse
Affiliation(s)
- Paulina Malach
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| | - Charlotte Kay
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| | - Chris Tinworth
- Medicinal Chemistry, Medicine Design, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| | - Florence Patel
- Screening, Profiling and Molecular Biology, Medicine Design, GSK Upper Providence, Collegeville, PA 19426, USA
| | - Bryan Joosse
- Screening, Profiling and Molecular Biology, Medicine Design, GSK Upper Providence, Collegeville, PA 19426, USA
| | - Jennifer Wade
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| | - Marlene Rosa do Carmo
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| | - Brian Donovan
- Screening, Profiling and Molecular Biology, Medicine Design, GSK Upper Providence, Collegeville, PA 19426, USA
| | - Martijn Brugman
- Analytical Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| | - Claudia Montiel-Equihua
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| | - Natalie Francis
- Product Development, Cell and Gene Therapy, GSK Medicine Research Centre, Stevenage, Hertfordshire SG1 2NY, UK
| |
Collapse
|
5
|
Zhang Y, Zhou W, Yang J, Yang J, Wang W. Chimeric antigen receptor engineered natural killer cells for cancer therapy. Exp Hematol Oncol 2023; 12:70. [PMID: 37563648 PMCID: PMC10413722 DOI: 10.1186/s40164-023-00431-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 07/27/2023] [Indexed: 08/12/2023] Open
Abstract
Natural killer (NK) cells, a unique component of the innate immune system, are inherent killers of stressed and transformed cells. Based on their potent capacity to kill cancer cells and good tolerance of healthy cells, NK cells have been successfully employed in adoptive cell therapy to treat cancer patients. In recent years, the clinical success of chimeric antigen receptor (CAR)-T cells has proven the vast potential of gene-manipulated immune cells as the main force to fight cancer. Following the lessons learned from mature gene-transfer technologies and advanced strategies in CAR-T therapy, NK cells have been rapidly explored as a promising candidate for CAR-based therapy. An exponentially growing number of studies have employed multiple sources of CAR-NK cells to target a wide range of cancer-related antigens, showing remarkable outcomes and encouraging safety profiles. Clinical trials of CAR-NK cells have also shown their impressive therapeutic efficacy in the treatment of hematological tumors, but CAR-NK cell therapy for solid tumors is still in the initial stages. In this review, we present the favorable profile of NK cells as a potential platform for CAR-based engineering and then summarize the outcomes and strategies of CAR-NK therapies in up-to-date preclinical and clinical investigations. Finally, we evaluate the challenges remaining in CAR-NK therapy and describe existing strategies that can assist us in devising future prospective solutions.
Collapse
Affiliation(s)
- Yalan Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Weilin Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
| | - Jiangping Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
- Department of Head and Neck Oncology and Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Jinrong Yang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China
- Hematology Research Laboratory, Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, 610041, People's Republic of China.
| |
Collapse
|
6
|
Maalej KM, Merhi M, Inchakalody VP, Mestiri S, Alam M, Maccalli C, Cherif H, Uddin S, Steinhoff M, Marincola FM, Dermime S. CAR-cell therapy in the era of solid tumor treatment: current challenges and emerging therapeutic advances. Mol Cancer 2023; 22:20. [PMID: 36717905 PMCID: PMC9885707 DOI: 10.1186/s12943-023-01723-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 144.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
In the last decade, Chimeric Antigen Receptor (CAR)-T cell therapy has emerged as a promising immunotherapeutic approach to fight cancers. This approach consists of genetically engineered immune cells expressing a surface receptor, called CAR, that specifically targets antigens expressed on the surface of tumor cells. In hematological malignancies like leukemias, myeloma, and non-Hodgkin B-cell lymphomas, adoptive CAR-T cell therapy has shown efficacy in treating chemotherapy refractory patients. However, the value of this therapy remains inconclusive in the context of solid tumors and is restrained by several obstacles including limited tumor trafficking and infiltration, the presence of an immunosuppressive tumor microenvironment, as well as adverse events associated with such therapy. Recently, CAR-Natural Killer (CAR-NK) and CAR-macrophages (CAR-M) were introduced as a complement/alternative to CAR-T cell therapy for solid tumors. CAR-NK cells could be a favorable substitute for CAR-T cells since they do not require HLA compatibility and have limited toxicity. Additionally, CAR-NK cells might be generated in large scale from several sources which would suggest them as promising off-the-shelf product. CAR-M immunotherapy with its capabilities of phagocytosis, tumor-antigen presentation, and broad tumor infiltration, is currently being investigated. Here, we discuss the emerging role of CAR-T, CAR-NK, and CAR-M cells in solid tumors. We also highlight the advantages and drawbacks of CAR-NK and CAR-M cells compared to CAR-T cells. Finally, we suggest prospective solutions such as potential combination therapies to enhance the efficacy of CAR-cells immunotherapy.
Collapse
Affiliation(s)
- Karama Makni Maalej
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Maysaloun Merhi
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Varghese P. Inchakalody
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Sarra Mestiri
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar
| | - Majid Alam
- grid.413548.f0000 0004 0571 546XTranslational Research Institute, Academic Health System, Dermatology Institute, Hamad Medical Corporation, Doha, Qatar ,grid.413548.f0000 0004 0571 546XDepartment of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar
| | - Cristina Maccalli
- grid.467063.00000 0004 0397 4222Laboratory of Immune and Biological Therapy, Research Department, Sidra Medicine, Doha, Qatar
| | - Honar Cherif
- grid.413548.f0000 0004 0571 546XDepartment of Hematology, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- grid.413548.f0000 0004 0571 546XTranslational Research Institute, Academic Health System, Dermatology Institute, Hamad Medical Corporation, Doha, Qatar
| | - Martin Steinhoff
- grid.413548.f0000 0004 0571 546XTranslational Research Institute, Academic Health System, Dermatology Institute, Hamad Medical Corporation, Doha, Qatar ,grid.413548.f0000 0004 0571 546XDepartment of Dermatology and Venereology, Hamad Medical Corporation, Doha, Qatar ,grid.416973.e0000 0004 0582 4340Department of Dermatology, Weill Cornell Medicine-Qatar, Doha, Qatar ,grid.412603.20000 0004 0634 1084College of Medicine, Qatar University, Doha, Qatar ,grid.5386.8000000041936877XDepartment of Dermatology, Weill Cornell Medicine, New York, USA
| | - Francesco M. Marincola
- grid.418227.a0000 0004 0402 1634Global Head of Research, Kite Pharma, Santa Monica, California USA
| | - Said Dermime
- grid.413548.f0000 0004 0571 546XTranslational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, P.O. Box: 3050, Doha, Qatar ,grid.452146.00000 0004 1789 3191College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
7
|
Fischer A. Gene therapy for inborn errors of immunity: past, present and future. Nat Rev Immunol 2022:10.1038/s41577-022-00800-6. [DOI: 10.1038/s41577-022-00800-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2022] [Indexed: 11/27/2022]
|
8
|
Li Q, Hu W, Liao B, Song C, Li L. Natural high-avidity T-cell receptor efficiently mediates regression of cancer/testis antigen 83 positive common solid cancers. J Immunother Cancer 2022; 10:jitc-2022-004713. [PMID: 35798537 PMCID: PMC9263944 DOI: 10.1136/jitc-2022-004713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND T-cell receptor-engineered T cells (TCR-Ts) have achieved encouraging success in anticancer clinical trials. The antigenic targets, however, were primarily focused on human leukocyte antigen (HLA) A*02:01 restricted epitopes from a few cancer/testis antigens (CTAs) which are not widely expressed in common solid cancers; the tested T-cell receptors (TCRs) were frequently from tumor-infiltrating lymphocytes of old patients and were not assured to have higher avidity. Here, we propose the isolation of high-avidity TCRs against CTAs that are frequently expressed in common solid cancers. METHODS We selected the CT83 protein, which is frequently expressed in common solid cancers, as a model antigen for screening of its specific TCR. The predicted CT83 epitopes with strong or weak binding to HLA-I molecules, popular in the Chinese population, were integrated into three synthetic long peptides. CT83 reactive CD8+ T cells were stimulated with peptide-loaded dendritic cells (DCs) and sorted using the CD137 biomarker for single-cell sequencing to obtain the paired TCRαβ sequence. The higher frequency TCRs were reconstructed for characterization of the CT83 epitope and for assessment of in vitro and in vivo antitumor activities. RESULTS CT83 reactive T cells from young healthy donors (YHDs) were generated by repeated stimulation with DCs and peptides. The single-cell TCR sequencing results of reactive T cells indicated that a single TCR clonotype dominated the paired TCRs. T cells engineered with this dominant TCR led to HLA-A*11:01-restricted recognition of the CT8314-22 epitope, with higher avidity. Functional assays showed powerful cytotoxicity in vitro against the targets of several CT83-positive solid cancer cell lines. Furthermore, TCR-Ts showed therapeutic efficacy in three xenograft solid tumor models. The meta-analysis of gene expression of 92 CTAs indicated that most CTAs did not or at low levels in the thymus, which suggested that those CTAs may experience incomplete thymic central tolerance. CONCLUSIONS High-avidity TCR against CT83 could be isolated from YHDs and efficiently mediate regression of well-established xenograft common solid tumors. The high-avidity TCR repertoire in the peripheral blood of some donors for CT83 and other CTAs provides the basis for the efficient isolation of high-avidity TCRs to target numerous solid cancers.
Collapse
Affiliation(s)
- Qingyang Li
- Department of Clinical Oncology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Wei Hu
- T Cell Immune Technology Co., Ltd, Guangzhou, China
| | - Baoyi Liao
- Department of Clinical Oncology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chanchan Song
- Department of Clinical Oncology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Liangping Li
- Department of Clinical Oncology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
9
|
Haist C, Poschinski Z, Bister A, Hoffmann MJ, Grunewald CM, Hamacher A, Kassack M, Wiek C, Scheckenbach K, Hanenberg H. Engineering a single-chain variable fragment of cetuximab for CAR T-cell therapy against head and neck squamous cell carcinomas. Oral Oncol 2022; 129:105867. [DOI: 10.1016/j.oraloncology.2022.105867] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 01/14/2023]
|
10
|
Gene Engineering T Cells with T-Cell Receptor for Adoptive Therapy. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2453:209-229. [PMID: 35622329 DOI: 10.1007/978-1-0716-2115-8_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Prior to clinical testing of adoptive T-cell therapy with T-cell receptor (TCR)-engineered T cells, TCRs need to be retrieved, annotated, gene-transferred, and extensively tested in vitro to accurately assess specificity and sensitivity of target recognition. Here, we present a fundamental series of protocols that cover critical preclinical parameters, thereby enabling the selection of candidate TCRs for clinical testing.
Collapse
|
11
|
Soldierer M, Bister A, Haist C, Thivakaran A, Cengiz SC, Sendker S, Bartels N, Thomitzek A, Smorra D, Hejazi M, Uhrberg M, Scheckenbach K, Monzel C, Wiek C, Reinhardt D, Niktoreh N, Hanenberg H. Genetic Engineering and Enrichment of Human NK Cells for CAR-Enhanced Immunotherapy of Hematological Malignancies. Front Immunol 2022; 13:847008. [PMID: 35464442 PMCID: PMC9022481 DOI: 10.3389/fimmu.2022.847008] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/28/2022] [Indexed: 01/11/2023] Open
Abstract
The great clinical success of chimeric antigen receptor (CAR) T cells has unlocked new levels of immunotherapy for hematological malignancies. Genetically modifying natural killer (NK) cells as alternative CAR immune effector cells is also highly promising, as NK cells can be transplanted across HLA barriers without causing graft-versus-host disease. Therefore, off-the-shelf usage of CAR NK cell products might allow to widely expand the clinical indications and to limit the costs of treatment per patient. However, in contrast to T cells, manufacturing suitable CAR NK cell products is challenging, as standard techniques for genetically engineering NK cells are still being defined. In this study, we have established optimal lentiviral transduction of primary human NK cells by systematically testing different internal promoters for lentiviral CAR vectors and comparing lentiviral pseudotypes and viral entry enhancers. We have additionally modified CAR constructs recognizing standard target antigens for acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML) therapy—CD19, CD33, and CD123—to harbor a CD34-derived hinge region that allows efficient detection of transduced NK cells in vitro and in vivo and also facilitates CD34 microbead-assisted selection of CAR NK cell products to >95% purity for potential clinical usage. Importantly, as most leukemic blasts are a priori immunogenic for activated primary human NK cells, we developed an in vitro system that blocks the activating receptors NKG2D, DNAM-1, NKp30, NKp44, NKp46, and NKp80 on these cells and therefore allows systematic testing of the specific killing of CAR NK cells against ALL and AML cell lines and primary AML blasts. Finally, we evaluated in an ALL xenotransplantation model in NOD/SCID-gamma (NSG) mice whether human CD19 CAR NK cells directed against the CD19+ blasts are relying on soluble or membrane-bound IL15 production for NK cell persistence and also in vivo leukemia control. Hence, our study provides important insights into the generation of pure and highly active allogeneic CAR NK cells, thereby advancing adoptive cellular immunotherapy with CAR NK cells for human malignancies further.
Collapse
Affiliation(s)
- Maren Soldierer
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Arthur Bister
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Corinna Haist
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Aniththa Thivakaran
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Sevgi Can Cengiz
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephanie Sendker
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nina Bartels
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Antonia Thomitzek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Denise Smorra
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Maryam Hejazi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Kathrin Scheckenbach
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Cornelia Monzel
- Department of Experimental Medical Physics, Heinrich Heine University, Düsseldorf, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Dirk Reinhardt
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Naghmeh Niktoreh
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Helmut Hanenberg
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany.,Department of Otorhinolaryngology, Head & Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| |
Collapse
|
12
|
Karvouni M, Vidal-Manrique M, Lundqvist A, Alici E. Engineered NK Cells Against Cancer and Their Potential Applications Beyond. Front Immunol 2022; 13:825979. [PMID: 35242135 PMCID: PMC8887605 DOI: 10.3389/fimmu.2022.825979] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 01/13/2022] [Indexed: 12/21/2022] Open
Abstract
Cell therapy is an innovative therapeutic concept where viable cells are implanted, infused, or grafted into a patient to treat impaired or malignant tissues. The term was first introduced circa the 19th century and has since resulted in multiple breakthroughs in different fields of medicine, such as neurology, cardiology, and oncology. Lately, cell and gene therapy are merging to provide cell products with additional or enhanced properties. In this context, adoptive transfer of genetically modified cytotoxic lymphocytes has emerged as a novel treatment option for cancer patients. To this day, five cell therapy products have been FDA approved, four of which for CD19-positive malignancies and one for B-cell maturation antigen (BCMA)-positive malignancies. These are personalized immunotherapies where patient T cells are engineered to express chimeric antigen receptors (CARs) with the aim to redirect the cells against tumor-specific antigens. CAR-T cell therapies show impressive objective response rates in clinical trials that, in certain instances, may reach up to 80%. However, the life-threatening side effects associated with T cell toxicity and the manufacturing difficulties of developing personalized therapies hamper their widespread use. Recent literature suggests that Natural Killer (NK) cells, may provide a safer alternative and an 'off-the-shelf' treatment option thanks to their potent antitumor properties and relatively short lifespan. Here, we will discuss the potential of NK cells in CAR-based therapies focusing on the applications of CAR-NK cells in cancer therapy and beyond.
Collapse
Affiliation(s)
- Maria Karvouni
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Marcos Vidal-Manrique
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institute, Stockholm, Sweden
| | - Andreas Lundqvist
- Department of Oncology‐Pathology, Karolinska Institute, Stockholm, Sweden
| | - Evren Alici
- Center for Hematology and Regenerative Medicine, Department of Medicine-Huddinge, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
13
|
Grunewald CM, Haist C, König C, Petzsch P, Bister A, Nößner E, Wiek C, Scheckenbach K, Köhrer K, Niegisch G, Hanenberg H, Hoffmann MJ. Epigenetic Priming of Bladder Cancer Cells With Decitabine Increases Cytotoxicity of Human EGFR and CD44v6 CAR Engineered T-Cells. Front Immunol 2021; 12:782448. [PMID: 34868059 PMCID: PMC8637820 DOI: 10.3389/fimmu.2021.782448] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/26/2021] [Indexed: 11/18/2022] Open
Abstract
Background Treatment of B-cell malignancies with CD19-directed chimeric antigen receptor (CAR) T-cells marked a new era in immunotherapy, which yet has to be successfully adopted to solid cancers. Epigenetic inhibitors of DNA methyltransferases (DNMTi) and histone deacetylases (HDACi) can induce broad changes in gene expression of malignant cells, thus making these inhibitors interesting combination partners for immunotherapeutic approaches. Methods Urothelial carcinoma cell lines (UCC) and benign uroepithelial HBLAK cells pretreated with the DNMTi decitabine or the HDACi romidepsin were co-incubated with CAR T-cells directed against EGFR or CD44v6, and subsequent cytotoxicity assays were performed. Effects on T-cell cytotoxicity and surface antigen expression on UCC were determined by flow cytometry. We also performed next-generation mRNA sequencing of inhibitor-treated UCC and siRNA-mediated knockdown of potential regulators of CAR T-cell killing. Results Exposure to decitabine but not romidepsin enhanced CAR T-cell cytotoxicity towards all UCC lines, but not towards the benign HBLAK cells. Increased killing could neither be attributed to enhanced target antigen expression (EGFR and CD44v6) nor fully explained by changes in the T-cell ligands PD-L1, PD-L2, ICAM-1, or CD95. Instead, gene expression analysis suggested that regulators of cell survival and apoptosis were differentially induced by the treatment. Decitabine altered the balance between survival and apoptosis factors towards an apoptosis-sensitive state associated with increased CAR T-cell killing, while romidepsin, at least partially, tilted this balance in the opposite direction. Knockdown experiments with siRNA in UCC confirmed BID and BCL2L1/BCLX as two key factors for the altered susceptibility of the UCC. Conclusion Our data suggest that the combination of decitabine with CAR T-cell therapy is an attractive novel therapeutic approach to enhance tumor-specific killing of bladder cancer. Since BID and BCL2L1 are essential determinants for the susceptibility of a wide variety of malignant cells, their targeting might be additionally suitable for combination with immunotherapies, e.g., CAR T-cells or checkpoint inhibitors in other malignancies.
Collapse
Affiliation(s)
- Camilla M Grunewald
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Corinna Haist
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carolin König
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Arthur Bister
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Elfriede Nößner
- Immunoanalytics: Tissue Control of Immunocytes, German Research Center for Environmental Health, Helmholtz Zentrum München, Munich, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Kathrin Scheckenbach
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Günter Niegisch
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology and Head and Neck Surgery, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michèle J Hoffmann
- Department of Urology, Medical Faculty, Heinrich Heine University Duesseldorf, Duesseldorf, Germany
| |
Collapse
|
14
|
Qatato M, Venugopalan V, Al-Hashimi A, Rehders M, Valentine AD, Hein Z, Dallto U, Springer S, Brix K. Trace Amine-Associated Receptor 1 Trafficking to Cilia of Thyroid Epithelial Cells. Cells 2021; 10:cells10061518. [PMID: 34208608 PMCID: PMC8234161 DOI: 10.3390/cells10061518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 12/14/2022] Open
Abstract
Trace amine-associated receptor 1 (rodent Taar1/human TAAR1) is a G protein-coupled receptor that is mainly recognized for its functions in neuromodulation. Previous in vitro studies suggested that Taar1 may signal from intracellular compartments. However, we have shown Taar1 to localize apically and on ciliary extensions in rodent thyrocytes, suggesting that at least in the thyroid, Taar1 may signal from the cilia at the apical plasma membrane domain of thyrocytes in situ, where it is exposed to the content of the follicle lumen containing putative Taar1 ligands. This study was designed to explore mouse Taar1 (mTaar1) trafficking, heterologously expressed in human and rat thyroid cell lines in order to establish an in vitro system in which Taar1 signaling from the cell surface can be studied in future. The results showed that chimeric mTaar1-EGFP traffics to the apical cell surface and localizes particularly to spherical structures of polarized thyroid cells, procilia, and primary cilia upon serum-starvation. Moreover, mTaar1-EGFP appears to form high molecular mass forms, possibly homodimers and tetramers, in stably expressing human thyroid cell lines. However, only monomeric mTaar1-EGFP was cell surface biotinylated in polarized human thyrocytes. In polarized rat thyrocytes, mTaar1-EGFP is retained in the endoplasmic reticulum, while cilia were reached by mTaar1-EGFP transiently co-expressed in combination with an HA-tagged construct of the related mTaar5. We conclude that Taar1 trafficking to cilia depends on their integrity. The results further suggest that an in vitro cell model was established that recapitulates Taar1 trafficking in thyrocytes in situ, in principle, and will enable studying Taar1 signaling in future, thus extending our general understanding of its potential significance for thyroid autoregulation.
Collapse
|
15
|
Lis N, Hein Z, Ghanwat SS, Ramnarayan VR, Chambers BJ, Springer S. The murine cytomegalovirus immunoevasin gp40/m152 inhibits NKG2D receptor RAE-1γ by intracellular retention and cell surface masking. J Cell Sci 2021; 134:269012. [PMID: 34085696 DOI: 10.1242/jcs.257428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/14/2021] [Indexed: 12/21/2022] Open
Abstract
NKG2D (also known as KLRK1) is a crucial natural killer (NK) cell-activating receptor, and the murine cytomegalovirus (MCMV) employs multiple immunoevasins to avoid NKG2D-mediated activation. One of the MCMV immunoevasins, gp40 (m152), downregulates the cell surface NKG2D ligand RAE-1γ (also known as Raet1c) thus limiting NK cell activation. This study establishes the molecular mechanism by which gp40 retains RAE-1γ in the secretory pathway. Using flow cytometry and pulse-chase analysis, we demonstrate that gp40 retains RAE-1γ in the early secretory pathway, and that this effect depends on the binding of gp40 to a host protein, TMED10, a member of the p24 protein family. We also show that the TMED10-based retention mechanism can be saturated, and that gp40 has a backup mechanism as it masks RAE-1γ on the cell surface, blocking the interaction with the NKG2D receptor and thus NK cell activation.
Collapse
Affiliation(s)
- Natalia Lis
- Department of Life Sciences and Chemistry, Jacobs University, Bremen 28759, Germany
| | - Zeynep Hein
- Department of Life Sciences and Chemistry, Jacobs University, Bremen 28759, Germany
| | - Swapnil S Ghanwat
- Department of Life Sciences and Chemistry, Jacobs University, Bremen 28759, Germany
| | - Venkat R Ramnarayan
- Department of Life Sciences and Chemistry, Jacobs University, Bremen 28759, Germany
| | - Benedict J Chambers
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm 14152, Sweden
| | - Sebastian Springer
- Department of Life Sciences and Chemistry, Jacobs University, Bremen 28759, Germany
| |
Collapse
|
16
|
CD44v6-targeted CAR T-cells specifically eliminate CD44 isoform 6 expressing head/neck squamous cell carcinoma cells. Oral Oncol 2021; 116:105259. [PMID: 33895463 DOI: 10.1016/j.oraloncology.2021.105259] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 02/04/2021] [Accepted: 02/28/2021] [Indexed: 01/09/2023]
Abstract
Immune checkpoint blockade can cause regression of recurrent and/or refractory head and neck squamous cell carcinoma (HNSCC). As a second type of immunotherapy, adoptive cellular therapy with genetically modified patient's T-cells redirected against the autologous malignant cells by expressing chimeric antigen receptors (CARs) recognizing tumor-associated antigens has been established as highly efficient personalized treatment for hematological malignancies. In solid cancers however, the application of these genetically modified immune effector cells still lacks equal response rates. CD44v6 is an isoform of the hyaluronic receptor CD44 that is almost exclusively expressed at high levels on solid cancers and has been associated with tumorigenesis, tumor cell invasion and metastasis. Here, we established a highly specific CAR against CD44v6 on HNSCC cells that can be expressed on normal T-cells with lentiviral vectors. Using primary human HNSCC cells in combination with CRISPR/Cas9 and overexpression approaches allowed us to confirm the high specificity of our CAR construct for the tumor-associated CD44v6 as target antigen and to demonstrate a direct correlation between CD44v6 expression levels and cytotoxicity of the CAR T-cells. Importantly, the design of our clinically applicable lentiviral vector facilitates to co-express a second transgene for in vivo control of CAR T-cells, if undesired side-effects or toxicities occur.
Collapse
|
17
|
Hoerster K, Uhrberg M, Wiek C, Horn PA, Hanenberg H, Heinrichs S. HLA Class I Knockout Converts Allogeneic Primary NK Cells Into Suitable Effectors for "Off-the-Shelf" Immunotherapy. Front Immunol 2021; 11:586168. [PMID: 33584651 PMCID: PMC7878547 DOI: 10.3389/fimmu.2020.586168] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/04/2020] [Indexed: 11/13/2022] Open
Abstract
Cellular immunotherapy using chimeric antigen receptors (CARs) so far has almost exclusively used autologous peripheral blood-derived T cells as immune effector cells. However, harvesting sufficient numbers of T cells is often challenging in heavily pre-treated patients with malignancies and perturbed hematopoiesis and perturbed hematopoiesis. Also, such a CAR product will always be specific for the individual patient. In contrast, NK cell infusions can be performed in non-HLA-matched settings due to the absence of alloreactivity of these innate immune cells. Still, the infused NK cells are subject to recognition and rejection by the patient's immune system, thereby limiting their life-span in vivo and undermining the possibility for multiple infusions. Here, we designed genome editing and advanced lentiviral transduction protocols to render primary human NK cells unsusceptible/resistant to an allogeneic response by the recipient's CD8+ T cells. After knocking-out surface expression of HLA class I molecules by targeting the B2M gene via CRISPR/Cas9, we also co-expressed a single-chain HLA-E molecule, thereby preventing NK cell fratricide of B2M-knockout (KO) cells via "missing self"-induced lysis. Importantly, these genetically engineered NK cells were functionally indistinguishable from their unmodified counterparts with regard to their phenotype and their natural cytotoxicity towards different AML cell lines. In co-culture assays, B2M-KO NK cells neither induced immune responses of allogeneic T cells nor re-activated allogeneic T cells which had been expanded/primed using irradiated PBMNCs of the respective NK cell donor. Our study demonstrates the feasibility of genome editing in primary allogeneic NK cells to diminish their recognition and killing by mismatched T cells and is an important prerequisite for using non-HLA-matched primary human NK cells as readily available, "off-the-shelf" immune effectors for a variety of immunotherapy indications in human cancer.
Collapse
Affiliation(s)
- Keven Hoerster
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich-Heine University, Düsseldorf, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology & Head/Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
| | - Peter A. Horn
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology & Head/Neck Surgery, University Hospital Düsseldorf, Heinrich Heine University, Düsseldorf, Germany
- Department of Pediatrics III, University Children’s Hospital of Essen, University Duisburg-Essen, Essen, Germany
| | - Stefan Heinrichs
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
18
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
19
|
Lin CY, Gobius I, Souza-Fonseca-Guimaraes F. Natural killer cell engineering – a new hope for cancer immunotherapy. Semin Hematol 2020; 57:194-200. [DOI: 10.1053/j.seminhematol.2020.10.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/20/2020] [Accepted: 10/14/2020] [Indexed: 01/04/2023]
|
20
|
Al-Hashimi A, Venugopalan V, Sereesongsaeng N, Tedelind S, Pinzaru AM, Hein Z, Springer S, Weber E, Führer D, Scott CJ, Burden RE, Brix K. Significance of nuclear cathepsin V in normal thyroid epithelial and carcinoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118846. [PMID: 32910988 DOI: 10.1016/j.bbamcr.2020.118846] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/29/2020] [Indexed: 12/11/2022]
Abstract
Altered expression and/or localization of cysteine cathepsins is believed to involve in thyroid diseases including cancer. Here, we examined the localization of cathepsins B and V in human thyroid tissue sections of different pathological conditions by immunolabeling and morphometry. Cathepsin B was mostly found within endo-lysosomes as expected. In contrast, cathepsin V was detected within nuclei, predominantly in cells of cold nodules, follicular and papillary thyroid carcinoma tissue, while it was less often detected in this unusual localization in hot nodules and goiter tissue. To understand the significance of nuclear cathepsin V in thyroid cells, this study aimed to establish a cellular model of stable nuclear cathepsin V expression. As representative of a specific form lacking the signal peptide and part of the propeptide, N-terminally truncated cathepsin V fused to eGFP recapitulated the nuclear localization of endogenous cathepsin V throughout the cell cycle in Nthy-ori 3-1 cells. Interestingly, the N-terminally truncated cathepsin V-eGFP was more abundant in the nuclei during S phase. These findings suggested a possible contribution of nuclear cathepsin V forms to cell cycle progression. Indeed, we found that N-terminally truncated cathepsin V-eGFP expressing cells were more proliferative than those expressing full-length cathepsin V-eGFP or wild type controls. We conclude that a specific molecular form of cathepsin V localizes to the nucleus of thyroid epithelial and carcinoma cells, where it might involve in deregulated pathways leading to hyperproliferation. These findings highlight the necessity to better understand cathepsin trafficking in health and disease. In particular, cell type specificity of mislocalization of cysteine cathepsins, which otherwise act in a functionally redundant manner, seems to be important to understand their non-canonical roles in cell cycle progression.
Collapse
Affiliation(s)
- Alaa Al-Hashimi
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Vaishnavi Venugopalan
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | | | - Sofia Tedelind
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Alexandra M Pinzaru
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Zeynep Hein
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Sebastian Springer
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany
| | - Ekkehard Weber
- Institute of Physiological Chemistry, Martin Luther University Halle-Wittenberg, Hollystrasse 1, D-06114 Halle-Saale, Germany
| | - Dagmar Führer
- Universität Duisburg-Essen, Universitätsklinikum Essen (AöR), Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Hufeland Strasse 55, D-45177 Essen, Germany
| | - Christopher J Scott
- Patrick G. Johnston Centre for Cancer Research, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Roberta E Burden
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Klaudia Brix
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, D-28759 Bremen, Germany.
| |
Collapse
|
21
|
Fischer A, Hacein-Bey-Abina S. Gene therapy for severe combined immunodeficiencies and beyond. J Exp Med 2020; 217:132743. [PMID: 31826240 PMCID: PMC7041706 DOI: 10.1084/jem.20190607] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/10/2019] [Accepted: 11/06/2019] [Indexed: 12/26/2022] Open
Abstract
This review describes how gene therapy of severe combined immunodeficiency became a reality, primarily based on the expected selective advantage conferred by transduction of hematopoietic progenitor cells. Thus, it resulted in a progressive extension to the treatment of other primary immunodeficiencies. Ex vivo retrovirally mediated gene therapy has been shown within the last 20 yr to correct the T cell immunodeficiency caused by γc-deficiency (SCID X1) and adenosine deaminase (ADA) deficiency. The rationale was brought up by the observation of the revertant of SCIDX1 and ADA deficiency as a kind of natural gene therapy. Nevertheless, the first attempts of gene therapy for SCID X1 were associated with insertional mutagenesis causing leukemia, because the viral enhancer induced transactivation of oncogenes. Removal of this element and use of a promoter instead led to safer but still efficacious gene therapy. It was observed that a fully diversified T cell repertoire could be generated by a limited set (<1,000) of progenitor cells. Further advances in gene transfer technology, including the use of lentiviral vectors, has led to success in the treatment of Wiskott–Aldrich syndrome, while further applications are pending. Genome editing of the mutated gene may be envisaged as an alternative strategy to treat SCID diseases.
Collapse
Affiliation(s)
- Alain Fischer
- Imagine Institute, Paris, France.,Immunology and Pediatric Hematology Department, Assistance Publique-Hôpitaux de Paris, Paris, France.,Institut National de la Santé et de la Recherche Médicale UMR 1163, Paris, France.,Collège de France, Paris, France
| | - Salima Hacein-Bey-Abina
- Unité de Technologies Chimiques et Biologiques pour la Santé, UMR8258 Centre National de la Recherche Scientifique - U1267 Institut National de la Santé et de la Recherche Médicale, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France.,Clinical Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud, Hôpital Kremlin-Bicêtre, Assistance Publique-Hôpitaux de Paris, Le Kremlin Bicêtre, France
| |
Collapse
|
22
|
Agarwalla P, Ogunnaike EA, Ahn S, Ligler FS, Dotti G, Brudno Y. Scaffold-Mediated Static Transduction of T Cells for CAR-T Cell Therapy. Adv Healthc Mater 2020; 9:e2000275. [PMID: 32592454 DOI: 10.1002/adhm.202000275] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/08/2020] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has produced impressive clinical responses in patients with B-cell malignancies. Critical to the success of CAR-T cell therapies is the achievement of robust gene transfer into T cells mediated by viral vectors such as gamma-retroviral vectors. However, current methodologies of retroviral gene transfer rely on spinoculation and the use of retronectin, which may limit the implementation of cost-effective CAR-T cell therapies. Herein, a low-cost, tunable, macroporous, alginate scaffold that transduces T cells with retroviral vectors under static condition is described. CAR-T cells produced by macroporous scaffold-mediated viral transduction exhibit >60% CAR expression, retain effector phenotype, expand to clinically relevant cell numbers, and eradicate CD19+ lymphoma in vivo. Efficient transduction is dependent on scaffold macroporosity. Taken together, the data show that macroporous alginate scaffolds serve as an attractive alternative to current transduction protocols and have high potential for clinical translation to genetically modify T cells for adoptive cellular therapy.
Collapse
Affiliation(s)
- Pritha Agarwalla
- Joint Department of Biomedical Engineering, University of North Carolina ‐ Chapel Hill and North Carolina State University ‐ Raleigh 1840 Enterpreneur Way Raleigh NC 27695 USA
| | - Edikan A. Ogunnaike
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
| | - Sarah Ahn
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina ‐ Chapel Hill and North Carolina State University ‐ Raleigh 1840 Enterpreneur Way Raleigh NC 27695 USA
| | - Gianpietro Dotti
- Department of Microbiology and ImmunologyUniversity of North Carolina at Chapel Hill Chapel Hill NC 27599 USA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina Chapel Hill. 450 West Dr. Chapel Hill NC 27599 USA
| | - Yevgeny Brudno
- Joint Department of Biomedical Engineering, University of North Carolina ‐ Chapel Hill and North Carolina State University ‐ Raleigh 1840 Enterpreneur Way Raleigh NC 27695 USA
- Lineberger Comprehensive Cancer CenterUniversity of North Carolina Chapel Hill. 450 West Dr. Chapel Hill NC 27599 USA
| |
Collapse
|
23
|
Abstract
Adoptive cell therapy with ex vivo expanded tumor infiltrating lymphocytes or gene engineering T cells expressing chimeric antigen receptors (CAR) is a promising treatment for cancer patients. This production utilizes T-cell activation and transduction with activation beads and RetroNectin, respectively. However, the high cost of production is an obstacle for the broad clinical application of novel immunotherapeutic cell products. To facilitate production we refined our approach by using artificial antigen presenting cells (aAPCs) with receptors that ligate CD3, CD28, and the CD137 ligand (CD137L or 41BBL), as well as express the heparin binding domain (HBD), which binds virus for gene-transfer. We have used these aAPC for ex vivo gene engineering and expansion of tumor infiltrating lymphocytes and CAR T cells. We found that aAPCs can support efficacious T-cell expansion and transduction. Moreover, aAPCs expanded T cells exhibit higher production of IFN-γ and lower traits of T-cell exhaustion compared with bead expanded T cells. Our results suggest that aAPC provide a more physiological stimulus for T-cell activation than beads that persistently ligate T cells. The use of a renewable cell line to replace 2 critical reagents (beads and retronectin) for CAR T-cell production can significantly reduce the cost of production and make these therapies more accessible to patients.
Collapse
|
24
|
You Have Got a Fast CAR: Chimeric Antigen Receptor NK Cells in Cancer Therapy. Cancers (Basel) 2020; 12:cancers12030706. [PMID: 32192067 PMCID: PMC7140022 DOI: 10.3390/cancers12030706] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/18/2022] Open
Abstract
The clinical success stories of chimeric antigen receptor (CAR)-T cell therapy against B-cell malignancies have contributed to immunotherapy being at the forefront of cancer therapy today. Their success has fueled interest in improving CAR constructs, identifying additional antigens to target, and clinically evaluating them across a wide range of malignancies. However, along with the exciting potential of CAR-T therapy comes the real possibility of serious side effects. While the FDA has approved commercialized CAR-T cell therapy, challenges associated with manufacturing, costs, and related toxicities have resulted in increased attention being paid to implementing CAR technology in innate cytotoxic natural killer (NK) cells. Here, we review the current landscape of the CAR-NK field, from successful clinical implementation to outstanding challenges which remain to be addressed to deliver the full potential of this therapy to more patients.
Collapse
|
25
|
Müller S, Bexte T, Gebel V, Kalensee F, Stolzenberg E, Hartmann J, Koehl U, Schambach A, Wels WS, Modlich U, Ullrich E. High Cytotoxic Efficiency of Lentivirally and Alpharetrovirally Engineered CD19-Specific Chimeric Antigen Receptor Natural Killer Cells Against Acute Lymphoblastic Leukemia. Front Immunol 2020; 10:3123. [PMID: 32117200 PMCID: PMC7025537 DOI: 10.3389/fimmu.2019.03123] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 12/23/2019] [Indexed: 01/03/2023] Open
Abstract
Autologous chimeric antigen receptor-modified (CAR) T cells with specificity for CD19 showed potent antitumor efficacy in clinical trials against relapsed and refractory B-cell acute lymphoblastic leukemia (B-ALL). Contrary to T cells, natural killer (NK) cells kill their targets in a non-antigen-specific manner and do not carry the risk of inducing graft vs. host disease (GvHD), allowing application of donor-derived cells in an allogenic setting. Hence, unlike autologous CAR-T cells, therapeutic CD19-CAR-NK cells can be generated as an off-the-shelf product from healthy donors. Nevertheless, genetic engineering of peripheral blood (PB) derived NK cells remains challenging and optimized protocols are needed. In our study, we aimed to optimize the generation of CD19-CAR-NK cells by retroviral transduction to improve the high antileukemic capacity of NK cells. We compared two different retroviral vector platforms, the lentiviral and alpharetroviral, both in combination with two different transduction enhancers (Retronectin and Vectofusin-1). We further explored different NK cell isolation techniques (NK cell enrichment and CD3/CD19 depletion) to identify the most efficacious methods for genetic engineering of NK cells. Our results demonstrated that transduction of NK cells with RD114-TR pseudotyped retroviral vectors, in combination with Vectofusin-1 was the most efficient method to generate CD19-CAR-NK cells. Retronectin was potent in enhancing lentiviral/VSV-G gene delivery to NK cells but not alpharetroviral/RD114-TR. Furthermore, the Vectofusin-based transduction of NK cells with CD19-CARs delivered by alpharetroviral/RD114-TR and lentiviral/RD114-TR vectors outperformed lentiviral/VSV-G vectors. The final generated CD19-CAR-NK cells displayed superior cytotoxic activity against CD19-expressing target cells when compared to non-transduced NK cells achieving up to 90% specific killing activity. In summary, our findings present the use of RD114-TR pseudotyped retroviral particles in combination with Vectofusin-1 as a successful strategy to genetically modify PB-derived NK cells to achieve highly cytotoxic CD19-CAR-NK cells at high yield.
Collapse
Affiliation(s)
- Stephan Müller
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division of Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Tobias Bexte
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division of Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
| | - Veronika Gebel
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division of Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Franziska Kalensee
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division of Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Eva Stolzenberg
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division of Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany
| | - Jessica Hartmann
- Division of Medical Biotechnology, Paul-Ehrlich-Institut, Langen, Germany
| | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute of Cellular Therapeutics, Hannover Medical School, Hanover, Germany.,Institute of Clinical Immunology, Faculty of Medicine, University Leipzig, Leipzig, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, Hanover, Germany.,Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Winfried S Wels
- German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| | - Ute Modlich
- Research Group for Gene Modification in Stem Cells, Division of Veterinary Medicine, Paul-Ehrlich Institute, Langen, Germany
| | - Evelyn Ullrich
- Experimental Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,Division of Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine, University Hospital Frankfurt, Goethe University, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK) Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
26
|
Radek C, Bernadin O, Drechsel K, Cordes N, Pfeifer R, Sträßer P, Mormin M, Gutierrez-Guerrero A, Cosset FL, Kaiser AD, Schaser T, Galy A, Verhoeyen E, Johnston IC. Vectofusin-1 Improves Transduction of Primary Human Cells with Diverse Retroviral and Lentiviral Pseudotypes, Enabling Robust, Automated Closed-System Manufacturing. Hum Gene Ther 2019; 30:1477-1493. [PMID: 31578886 PMCID: PMC6919281 DOI: 10.1089/hum.2019.157] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/21/2019] [Indexed: 01/07/2023] Open
Abstract
Cell and gene therapies are finally becoming viable patient treatment options, with both T cell- and hematopoietic stem cell (HSC)-based therapies being approved to market in Europe. However, these therapies, which involve the use of viral vector to modify the target cells, are expensive and there is an urgent need to reduce manufacturing costs. One major cost factor is the viral vector production itself, therefore improving the gene modification efficiency could significantly reduce the amount of vector required per patient. This study describes the use of a transduction enhancing peptide, Vectofusin-1®, to improve the transduction efficiency of primary target cells using lentiviral and gammaretroviral vectors (LV and RV) pseudotyped with a variety of envelope proteins. Using Vectofusin-1 in combination with LV pseudotyped with viral glycoproteins derived from baboon endogenous retrovirus, feline endogenous virus (RD114), and measles virus (MV), a strongly improved transduction of HSCs, B cells and T cells, even when cultivated under low stimulation conditions, could be observed. The formation of Vectofusin-1 complexes with MV-LV retargeted to CD20 did not alter the selectivity in mixed cell culture populations, emphasizing the precision of this targeting technology. Functional, ErbB2-specific chimeric antigen receptor-expressing T cells could be generated using a gibbon ape leukemia virus (GALV)-pseudotyped RV. Using a variety of viral vectors and target cells, Vectofusin-1 performed in a comparable manner to the traditionally used surface-bound recombinant fibronectin. As Vectofusin-1 is a soluble peptide, it was possible to easily transfer the T cell transduction method to an automated closed manufacturing platform, where proof of concept studies demonstrated efficient genetic modification of T cells with GALV-RV and RD114-RV and the subsequent expansion of mainly central memory T cells to a clinically relevant dose.
Collapse
Affiliation(s)
| | - Ornellie Bernadin
- CIRI—International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France
- Inserm, U1111, Ecole Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, CNRS, UMR5308, Lyon, France
| | | | - Nicole Cordes
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Rita Pfeifer
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Pia Sträßer
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Mirella Mormin
- Integrare Research Unit UMR_S951, Genethon, INSERM, University Evry, EPHE, Evry, France
| | - Alejandra Gutierrez-Guerrero
- CIRI—International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France
- Inserm, U1111, Ecole Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - François-loïc Cosset
- CIRI—International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France
- Inserm, U1111, Ecole Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, CNRS, UMR5308, Lyon, France
| | | | - Thomas Schaser
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Anne Galy
- Integrare Research Unit UMR_S951, Genethon, INSERM, University Evry, EPHE, Evry, France
| | - Els Verhoeyen
- CIRI—International Center for Infectiology Research, Team EVIR, Université de Lyon, Lyon, France
- Inserm, U1111, Ecole Normale Supérieure de Lyon, Lyon, France
- Université Lyon 1, CNRS, UMR5308, Lyon, France
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
| | | |
Collapse
|
27
|
Moore N, Chevillet JR, Healey LJ, McBrine C, Doty D, Santos J, Teece B, Truslow J, Mott V, Hsi P, Tandon V, Borenstein JT, Balestrini J, Kotz K. A Microfluidic Device to Enhance Viral Transduction Efficiency During Manufacture of Engineered Cellular Therapies. Sci Rep 2019; 9:15101. [PMID: 31641163 PMCID: PMC6806008 DOI: 10.1038/s41598-019-50981-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 09/23/2019] [Indexed: 02/08/2023] Open
Abstract
The development and approval of engineered cellular therapies are revolutionizing approaches to treatment of diseases. However, these life-saving therapies require extensive use of inefficient bioprocessing equipment and specialized reagents that can drive up the price of treatment. Integration of new genetic material into the target cells, such as viral transduction, is one of the most costly and labor-intensive steps in the production of cellular therapies. Approaches to reducing the costs associated with gene delivery have been developed using microfluidic devices to increase overall efficiency. However, these microfluidic approaches either require large quantities of virus or pre-concentration of cells with high-titer viral particles. Here, we describe the development of a microfluidic transduction device (MTD) that combines microfluidic spatial confinement with advective flow through a membrane to efficiently colocalize target cells and virus particles. We demonstrate that the MTD can improve the efficiency of lentiviral transduction for both T-cell and hematopoietic stem-cell (HSC) targets by greater than two fold relative to static controls. Furthermore, transduction saturation in the MTD is reached with only half the virus required to reach saturation under static conditions. Moreover, we show that MTD transduction does not adversely affect cell viability or expansion potential.
Collapse
Affiliation(s)
- Nathan Moore
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA.
| | - John R Chevillet
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Laura J Healey
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Connor McBrine
- Synthetic Biology, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Daniel Doty
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Jose Santos
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Bryan Teece
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - James Truslow
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Vienna Mott
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Peter Hsi
- Cell and Tissue Engineering, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Vishal Tandon
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | | | - Jenna Balestrini
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| | - Kenneth Kotz
- Biological Microsystems, 555 Technology Square, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
28
|
Schott JW, León-Rico D, Ferreira CB, Buckland KF, Santilli G, Armant MA, Schambach A, Cavazza A, Thrasher AJ. Enhancing Lentiviral and Alpharetroviral Transduction of Human Hematopoietic Stem Cells for Clinical Application. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:134-147. [PMID: 31338385 PMCID: PMC6629974 DOI: 10.1016/j.omtm.2019.05.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/31/2019] [Indexed: 01/27/2023]
Abstract
Ex vivo retroviral gene transfer into CD34+ hematopoietic stem and progenitor cells (HSPCs) has demonstrated remarkable clinical success in gene therapy for monogenic hematopoietic disorders. However, little attention has been paid to enhancement of culture and transduction conditions to achieve reliable effects across patient and disease contexts and to maximize potential vector usage and reduce treatment cost. We systematically tested three HSPC culture media manufactured to cGMP and eight previously described transduction enhancers (TEs) to develop a state-of-the-art clinically applicable protocol. Six TEs enhanced lentiviral (LV) and five TEs facilitated alpharetroviral (ARV) CD34+ HSPC transduction when used alone. Combinatorial TE application tested with LV vectors yielded more potent effects, with up to a 5.6-fold increase in total expression of a reporter gene and up to a 3.8-fold increase in VCN. Application of one of the most promising combinations, the poloxamer LentiBOOST and protamine sulfate, for GMP-compliant manufacturing of a clinical-grade advanced therapy medicinal product (ATMP) increased total VCN by over 6-fold, with no major changes in global gene expression profiles or inadvertent loss of CD34+CD90+ HSPC populations. Application of these defined culture and transduction conditions is likely to significantly improve ex vivo gene therapy manufacturing protocols for HSPCs and downstream clinical efficacy.
Collapse
Affiliation(s)
- Juliane W Schott
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Diego León-Rico
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Carolina B Ferreira
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Karen F Buckland
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Giorgia Santilli
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Myriam A Armant
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Axel Schambach
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany
| | - Alessia Cavazza
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Adrian J Thrasher
- Infection, Immunity and Inflammation Program, Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK.,Great Ormond Street Hospital NHS Foundation Trust, London WC1N 1EH, UK
| |
Collapse
|
29
|
Wilcox DA. Gene Therapy for Platelet Disorders. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
30
|
Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies. J Immunol Res 2018; 2018:4054815. [PMID: 30306093 PMCID: PMC6166361 DOI: 10.1155/2018/4054815] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/26/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are powerful immune effectors whose antitumor activity is regulated through a sophisticated network of activating and inhibitory receptors. As effectors of cancer immunotherapy, NK cells are attractive as they do not attack healthy self-tissues nor do they induce T cell-driven inflammatory cytokine storm, enabling their use as allogeneic adoptive cellular therapies. Clinical responses to adoptive NK-based immunotherapy have been thwarted, however, by the profound immunosuppression induced by the tumor microenvironment, particularly severe in the context of solid tumors. In addition, the short postinfusion persistence of NK cells in vivo has limited their clinical efficacy. Enhancing the antitumor immunity of NK cells through genetic engineering has been fueled by the promise that impaired cytotoxic functionality can be restored or augmented with the use of synthetic genetic approaches. Alongside expressing chimeric antigen receptors to overcome immune escape by cancer cells, enhance their recognition, and mediate their killing, NK cells have been genetically modified to enhance their persistence in vivo by the expression of cytokines such as IL-15, avoid functional and metabolic tumor microenvironment suppression, or improve their homing ability, enabling enhanced targeting of solid tumors. However, NK cells are notoriously adverse to endogenous gene uptake, resulting in low gene uptake and transgene expression with many vector systems. Though viral vectors have achieved the highest gene transfer efficiencies with NK cells, nonviral vectors and gene transfer approaches—electroporation, lipofection, nanoparticles, and trogocytosis—are emerging. And while the use of NK cell lines has achieved improved gene transfer efficiencies particularly with viral vectors, challenges with primary NK cells remain. Here, we discuss the genetic engineering of NK cells as they relate to NK immunobiology within the context of cancer immunotherapy, highlighting the most recent breakthroughs in viral vectors and nonviral approaches aimed at genetic reprogramming of NK cells for improved adoptive immunotherapy of cancer, and, finally, address their clinical status.
Collapse
|
31
|
Hein Z, Borchert B, Tolba Abualrous E, Springer S. Distinct mechanisms survey the structural integrity of HLA-B*27:05 intracellularly and at the surface. PLoS One 2018; 13:e0200811. [PMID: 30071035 PMCID: PMC6071996 DOI: 10.1371/journal.pone.0200811] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 07/03/2018] [Indexed: 02/06/2023] Open
Abstract
HLA-B*27:05 is associated with the development of autoimmune spondyloarthropathies, but the precise causal relationship between the MHC haplotype and disease pathogenesis is yet to be elucidated. Studies focusing on the structure and cellular trafficking of HLA-B*27:05 implicate several links between the onset of inflammation and the unusual conformations of the molecule inside and at the surface of antigen presenting cells. Several lines of evidence emphasize the emergence of those unnatural protein conformations under conditions where peptide loading onto B*27:05 is impaired. To understand how cellular factors distinguish between poorly loaded molecules from the optimally loaded ones, we have investigated the intracellular transport, folding, and cell surface expression of this particular B27 subtype. Our findings show that B*27:05 is structurally unstable in the absence of peptide, and that an artificially introduced disulfide bond between residues 84 and 139 conferred enhanced conformational stability to the suboptimally loaded molecules. Empty or suboptimally loaded B*27:05 can escape intracellular retention and arrive at the cell surface leading to the appearance of increased number of β2m-free heavy chains. Our study reveals a general mechanism found in the early secretory pathways of murine and human cells that apply to the quality control of MHC class I molecules, and it highlights the allotype-specific structural features of HLA-B*27:05 that can be associated with aberrant antigen presentation and that might contribute to the etiology of disease.
Collapse
Affiliation(s)
- Zeynep Hein
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | - Britta Borchert
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
| | - Esam Tolba Abualrous
- Department of Mathematics and Computer Science, Freie Universität, Berlin, Germany
| | - Sebastian Springer
- Department of Life Sciences and Chemistry, Jacobs University, Bremen, Germany
- * E-mail:
| |
Collapse
|
32
|
Cytomegalovirus gp40/m152 Uses TMED10 as ER Anchor to Retain MHC Class I. Cell Rep 2018; 23:3068-3077. [DOI: 10.1016/j.celrep.2018.05.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 03/12/2018] [Accepted: 05/03/2018] [Indexed: 02/03/2023] Open
|
33
|
Park CS, Lacorazza HD. Retroviral Transduction of Quiescent Murine Hematopoietic Stem Cells. Methods Mol Biol 2018; 1686:173-182. [PMID: 29030821 DOI: 10.1007/978-1-4939-7371-2_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hematopoietic stem cells (HSCs) represent an important target cell population in bone marrow transplantation, cell and gene therapy applications, and the development of leukemia models for research. Because the hematopoietic progeny carries the genetic information of HSCs and replenishes the blood and immune system, corrective gene transfer into HSCs provides an ideal therapeutic approach for many monogenic hematological diseases and a useful tool for studies of HSC function and blood formation in normal and malignant hematopoiesis. However, the efficiency of gene transfer into HSCs has been limited by several features of viral vectors, viral titer, methods of viral transduction, and the property of stem cell quiescence. In this chapter, we describe the production of retrovirus using murine stem cell virus (MSCV)-based retroviral vectors and purification and transduction of murine HSCs.
Collapse
Affiliation(s)
- Chun Shik Park
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, 1102 Bates Street, FC830.20, Houston, TX, 77030, USA
| | - H Daniel Lacorazza
- Department of Pathology and Immunology, Baylor College of Medicine, Texas Children's Hospital, 1102 Bates Street, FC830.20, Houston, TX, 77030, USA.
| |
Collapse
|
34
|
Pay SL, Qi X, Willard JF, Godoy J, Sankhavaram K, Horton R, Mitter SK, Quigley JL, Chang LJ, Grant MB, Boulton ME. Improving the Transduction of Bone Marrow-Derived Cells with an Integrase-Defective Lentiviral Vector. Hum Gene Ther Methods 2017; 29:44-59. [PMID: 29160102 PMCID: PMC5806075 DOI: 10.1089/hgtb.2017.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In lentiviral vector (LV) applications where transient transgene expression is sufficient, integrase-defective lentiviral vectors (IDLVs) are beneficial for reducing the potential for off-target effects associated with insertional mutagenesis. It was previously demonstrated that human RPE65 mRNA expression from an integrating lentiviral vector (ILV) induces endogenous Rpe65 and Cralbp mRNA expression in murine bone marrow–derived cells (BMDCs), initiating programming of the cells to retinal pigment epithelium (RPE)-like cells. These cells regenerate RPE in retinal degeneration models when injected systemically. As transient expression of RPE65 is sufficient to activate endogenous RPE-associated genes for programming BMDCs, use of an ILV is an unnecessary risk. In this study, an IDLV expressing RPE65 (IDLV3-RPE65) was generated. Transduction with IDLV3-RPE65 is less efficient than the integrating vector (ILV3-RPE65). Therefore, IDLV3-RPE65 transduction was enhanced with a combination of preloading 20 × -concentrated viral supernatant on RetroNectin at a multiplicity of infection of 50 and transduction of BMDCs by low-speed centrifugation. RPE65 mRNA levels increased from ∼12-fold to ∼25-fold (p < 0.05) after modification of the IDLV3-RPE65 transduction protocol, achieving expression similar to the ∼27-fold (p < 0.05) increase observed with ILV3-RPE65. Additionally, the study shows that the same preparation of RetroNectin can be used to coat up to three wells with no reduction in transduction. Critically, IDLV3-RPE65 transduction initiates endogenous Rpe65 mRNA expression in murine BMDCs and Cralbp/CRALBP mRNA in both murine and human BMDCs, similar to expression observed in ILV3-RPE65-transduced cells. Systemic administration of ILV3-RPE65 or IDLV3-RPE65 programmed BMDCs in a mouse model of retinal degeneration is sufficient to retain visual function and reduce retinal degeneration compared to mice receiving no treatment or naïve BMDC. It is concluded that IDLV3-RPE65 is appropriate for programming BMDCs to RPE-like cells.
Collapse
Affiliation(s)
- S Louise Pay
- 1 Department of Medical and Molecular Genetics, Indiana University School of Medicine , Indianapolis, Indiana.,2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Xiaoping Qi
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Jeffrey F Willard
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Juliana Godoy
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Kavya Sankhavaram
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Ranier Horton
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sayak K Mitter
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Judith L Quigley
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Lung-Ji Chang
- 4 Department of Molecular Genetics and Microbiology, University of Florida , Gainesville, Florida
| | - Maria B Grant
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Michael E Boulton
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
35
|
Vermeer LS, Hamon L, Schirer A, Schoup M, Cosette J, Majdoul S, Pastré D, Stockholm D, Holic N, Hellwig P, Galy A, Fenard D, Bechinger B. Vectofusin-1, a potent peptidic enhancer of viral gene transfer forms pH-dependent α-helical nanofibrils, concentrating viral particles. Acta Biomater 2017; 64:259-268. [PMID: 29017974 DOI: 10.1016/j.actbio.2017.10.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 10/02/2017] [Accepted: 10/06/2017] [Indexed: 01/13/2023]
Abstract
Gene transfer using lentiviral vectors has therapeutic applications spanning from monogenic and infectious diseases to cancer. Such gene therapy has to be improved by enhancing the levels of viral infection of target cells and/or reducing the amount of lentivirus for greater safety and reduced costs. Vectofusin-1, a recently developed cationic amphipathic peptide with a pronounced capacity to enhance such viral transduction, strongly promotes the entry of several retroviral pseudotypes into target cells when added to the culture medium. To clarify the molecular basis of its action the peptide was investigated on a molecular and a supramolecular level by a variety of biophysical approaches. We show that in culture medium vectofusin-1 rapidly forms complexes in the 10 nm range that further assemble into annular and extended nanofibrils. These associate with viral particles allowing them to be easily pelleted for optimal virus-cell interaction. Thioflavin T fluorescence, circular dichroism and infrared spectroscopies indicate that these fibrils have a unique α-helical structure whereas most other viral transduction enhancers form β-amyloid fibrils. A vectofusin-1 derivative (LAH2-A4) is inefficient in biological assays and does not form nanofibrils, suggesting that supramolecular assembly is essential for transduction enhancement. Our observations define vectofusin-1 as a member of a new class of α-helical enhancers of lentiviral infection. Its fibril formation is reversible which bears considerable advantages in handling the peptide in conditions well-adapted to Good Manufacturing Practices and scalable gene therapy protocols.
Collapse
Affiliation(s)
- Louic S Vermeer
- CNRS, Univ. of Strasbourg, Institut de Chimie UMR_7177, Strasbourg, France
| | - Loic Hamon
- INSERM, Univ. of Evry, UMR_S1204, Evry, France
| | | | - Michel Schoup
- CNRS, Univ. of Strasbourg, Institut de Chimie UMR_7177, Strasbourg, France
| | | | - Saliha Majdoul
- Genethon, INSERM, Univ. of Evry, EPHE-PSL Research University, Research Unit Integrare UMR_S951, Evry, France
| | | | - Daniel Stockholm
- Genethon, INSERM, Univ. of Evry, EPHE-PSL Research University, Research Unit Integrare UMR_S951, Evry, France
| | - Nathalie Holic
- Genethon, INSERM, Univ. of Evry, EPHE-PSL Research University, Research Unit Integrare UMR_S951, Evry, France
| | - Petra Hellwig
- CNRS, Univ. of Strasbourg, UMR 7140, Strasbourg, France
| | - Anne Galy
- Genethon, INSERM, Univ. of Evry, EPHE-PSL Research University, Research Unit Integrare UMR_S951, Evry, France
| | | | - Burkhard Bechinger
- CNRS, Univ. of Strasbourg, Institut de Chimie UMR_7177, Strasbourg, France.
| |
Collapse
|
36
|
Morgan RA, Gray D, Lomova A, Kohn DB. Hematopoietic Stem Cell Gene Therapy: Progress and Lessons Learned. Cell Stem Cell 2017; 21:574-590. [PMID: 29100011 PMCID: PMC6039108 DOI: 10.1016/j.stem.2017.10.010] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of allogeneic hematopoietic stem cells (HSCs) to treat genetic blood cell diseases has become a clinical standard but is limited by the availability of suitable matched donors and potential immunologic complications. Gene therapy using autologous HSCs should avoid these limitations and thus may be safer. Progressive improvements in techniques for genetic correction of HSCs, by either vector gene addition or gene editing, are facilitating successful treatments for an increasing number of diseases. We highlight the progress, successes, and remaining challenges toward the development of HSC gene therapies and discuss lessons they provide for the development of future clinical stem cell therapies.
Collapse
Affiliation(s)
- Richard A Morgan
- Charles R. Drew University of Medicine and Science, Los Angeles, CA, 90059; Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, CA, 90095
| | - David Gray
- Molecular Biology Institute Interdepartmental Doctoral Program, University of California, Los Angeles, CA, 90095
| | - Anastasia Lomova
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, CA, 90095
| | - Donald B Kohn
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at University of California, Los Angeles, CA, 90095; Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine at University of California, Los Angeles, CA, 90095; Department of Pediatrics, David Geffen School of Medicine at University of California, Los Angeles, CA, 90095; The Eli & Edythe Broad Center of Regenerative Medicine & Stem Cell Research, University of California, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Majdoul S, Cosette J, Seye AK, Bernard E, Frin S, Holic N, Chazal N, Briant L, Espert L, Galy A, Fenard D. Peptides derived from evolutionarily conserved domains in Beclin-1 and Beclin-2 enhance the entry of lentiviral vectors into human cells. J Biol Chem 2017; 292:18672-18681. [PMID: 28928217 DOI: 10.1074/jbc.m117.800813] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/07/2017] [Indexed: 01/19/2023] Open
Abstract
Autophagy-related proteins such as Beclin-1 are involved in an array of complex processes, including antiviral responses, and may also modulate the efficiency of gene therapy viral vectors. The Tat-Beclin-1 (TB1) peptide has been reported as an autophagy-inducing factor inhibiting the replication of pathogens such as HIV, type 1 (HIV-1). However, autophagy-related proteins are also essential for the early steps of HIV-1 infection. Therefore, we examined the effects of the Beclin-1 evolutionarily conserved domain in TB1 on viral transduction and autophagy in single-round HIV infection or with nonreplicative HIV-1-derived lentiviral vectors. TB1 enhanced transduction with various pseudotypes but without inducing the autophagy process. TB1 augmented the transduction of human CD34+ hematopoietic stem/progenitor cells while maintaining their capacity to engraft in vivo into humanized mice. TB1 was as effective as other transduction additives and functioned by enhancing the adhesion and fusion of viral particles with target cells but not their aggregation. We also found that the N-terminal L1 loop was critical for TB1 transduction-enhancing activity. Interestingly, the Tat-Beclin-2 (TB2) peptide, derived from the human Beclin-2 protein, was even more potent than TB1 in promoting viral transduction and infection. Taken together, our findings suggest that the TB1 and TB2 peptides enhance the viral entry step. Tat-Beclin peptides therefore represent a new family of viral transduction enhancers for potential use in gene therapy.
Collapse
Affiliation(s)
- Saliha Majdoul
- From Integrare Research Unit, UMR_S951, Généthon, INSERM, University of Evry, EPHE, Evry F 91000, France
| | | | - Ababacar K Seye
- From Integrare Research Unit, UMR_S951, Généthon, INSERM, University of Evry, EPHE, Evry F 91000, France
| | - Eric Bernard
- IRIM (ex-CPBS) UMR 9004, Infectious Disease Research Institute of Montpellier, University of Montpellier, CNRS, Montpellier F34293, France
| | - Sophie Frin
- From Integrare Research Unit, UMR_S951, Généthon, INSERM, University of Evry, EPHE, Evry F 91000, France
| | - Nathalie Holic
- From Integrare Research Unit, UMR_S951, Généthon, INSERM, University of Evry, EPHE, Evry F 91000, France
| | - Nathalie Chazal
- IRIM (ex-CPBS) UMR 9004, Infectious Disease Research Institute of Montpellier, University of Montpellier, CNRS, Montpellier F34293, France
| | - Laurence Briant
- IRIM (ex-CPBS) UMR 9004, Infectious Disease Research Institute of Montpellier, University of Montpellier, CNRS, Montpellier F34293, France
| | - Lucile Espert
- IRIM (ex-CPBS) UMR 9004, Infectious Disease Research Institute of Montpellier, University of Montpellier, CNRS, Montpellier F34293, France
| | - Anne Galy
- From Integrare Research Unit, UMR_S951, Généthon, INSERM, University of Evry, EPHE, Evry F 91000, France,
| | - David Fenard
- From Integrare Research Unit, UMR_S951, Généthon, INSERM, University of Evry, EPHE, Evry F 91000, France,
| |
Collapse
|
38
|
He H, Xue J, Wang W, Liu L, Ye C, Cong Z, Kimata JT, Qin C, Zhou P. Efficient Transduction of Human and Rhesus Macaque Primary T Cells by a Modified Human Immunodeficiency Virus Type 1-Based Lentiviral Vector. Hum Gene Ther 2016; 28:271-285. [PMID: 28042947 DOI: 10.1089/hum.2016.135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1)-based lentiviral vectors efficiently transduce genes to human, but not rhesus, primary T cells and hematopoietic stem cells (HSCs). The poor transduction of HIV-1 vectors to rhesus cells is mainly due to species-specific restriction factors such as rhesus TRIM5α. Previously, several strategies to modify HIV-1 vectors were developed to overcome rhesus TRIM5α restriction. While the modified HIV-1 vectors efficiently transduce rhesus HSCs, they remain suboptimal for rhesus primary T cells. Recently, HIV-1 variants that encode combinations of LNEIE mutations in capsid (CA) protein and SIVmac239 Vif were found to replicate efficiently in rhesus primary T cells. Thus, the present study tested whether HIV-1 vectors packaged by a packaging construct containing these CA substitutions could efficiently transduce both human and rhesus primary CD4 T cells. To accomplish this, LNEIE mutations were made in the packaging construct CEMΔ8.9, and recombinant HIV-1 vectors packaged by Δ8.9 WT or Δ8.9 LNEIE were generated. Transduction rates, CA stability, and vector integration in CEMss-CCR5 and CEMss-CCR5-rhTRIM5α/green fluorescent protein cells, as well as transduction rates in human and rhesus primary CD4 T cells by Δ8.9 WT or Δ8.9 LNEIE-packaged HIV-1 vectors, were compared. Finally, the influence of rhesus TRIM5α variations in transduction rates to primary CD4 T cells from a cohort of 37 Chinese rhesus macaques was studied. While it maintains efficient transduction for human T-cell line and primary CD4 T cells, Δ8.9 LNEIE-packaged HIV-1 vector overcomes rhesus TRIM5α-mediated CA degradation, resulting in significantly higher transduction efficiency of rhesus primary CD4 T cells than Δ8.9 WT-packaged HIV-1 vector. Rhesus TRIM5α variations strongly influence transduction efficiency of rhesus primary CD4 T cells by both Δ8.9 WT or Δ8.9 LNEIE-packaged HIV-1 vectors. Thus, it is concluded that Δ8.9 LNEIE-packaged HIV-1 vector overcomes rhesus TRIM5α restriction and efficiently transduces both human and rhesus primary T cells.
Collapse
Affiliation(s)
- Huan He
- 1 The Unit of Anti-Viral Immunity and Genetic Therapy, the Key Laboratory of Molecular Virology and Immunology, the Institut Pasteur of Shanghai , Chinese Academy of Sciences, Shanghai, China
| | - Jing Xue
- 2 Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science , Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Weiming Wang
- 1 The Unit of Anti-Viral Immunity and Genetic Therapy, the Key Laboratory of Molecular Virology and Immunology, the Institut Pasteur of Shanghai , Chinese Academy of Sciences, Shanghai, China
| | - Lihong Liu
- 1 The Unit of Anti-Viral Immunity and Genetic Therapy, the Key Laboratory of Molecular Virology and Immunology, the Institut Pasteur of Shanghai , Chinese Academy of Sciences, Shanghai, China
| | - Chaobaihui Ye
- 1 The Unit of Anti-Viral Immunity and Genetic Therapy, the Key Laboratory of Molecular Virology and Immunology, the Institut Pasteur of Shanghai , Chinese Academy of Sciences, Shanghai, China
| | - Zhe Cong
- 2 Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science , Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jason T Kimata
- 3 Department of Molecular Virology and Microbiology, Baylor College of Medicine , Houston, Texas
| | - Chuan Qin
- 2 Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Remerging Infectious Diseases, Institute of Laboratory Animal Science , Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Paul Zhou
- 1 The Unit of Anti-Viral Immunity and Genetic Therapy, the Key Laboratory of Molecular Virology and Immunology, the Institut Pasteur of Shanghai , Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
39
|
Cavazzana M, Six E, Lagresle-Peyrou C, André-Schmutz I, Hacein-Bey-Abina S. Gene Therapy for X-Linked Severe Combined Immunodeficiency: Where Do We Stand? Hum Gene Ther 2016; 27:108-16. [PMID: 26790362 DOI: 10.1089/hum.2015.137] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
More than 20 years ago, X-linked severe combined immunodeficiency (SCID-X1) appeared to be the best condition to test the feasibility of hematopoietic stem cell gene therapy. The seminal SCID-X1 clinical studies, based on first-generation gammaretroviral vectors, demonstrated good long-term immune reconstitution in most treated patients despite the occurrence of vector-related leukemia in a few of them. This gene therapy has successfully enabled correction of the T cell defect. Natural killer and B cell defects were only partially restored, most likely due to the absence of a conditioning regimen. The success of these pioneering trials paved the way for the extension of gene-based treatment to many other diseases of the hematopoietic system, but the unfortunate serious adverse events led to extensive investigations to define the retrovirus integration profiles. This review puts into perspective the clinical experience of gene therapy for SCID-X1, with the development and implementation of new generations of safer vectors such as self-inactivating gammaretroviral or lentiviral vectors as well as major advances in integrome knowledge.
Collapse
Affiliation(s)
- Marina Cavazzana
- 1 Biotherapy Department, Necker Children's Hospital , Assistance Publique-Hôpitaux de Paris, Paris.,2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Emmanuelle Six
- 2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Chantal Lagresle-Peyrou
- 2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Isabelle André-Schmutz
- 2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,3 Paris Descartes-Sorbonne Paris Cité University, Imagine Institute , Paris.,4 INSERM UMR 1163, Laboratory of Human Lymphohematopoiesis , Paris
| | - Salima Hacein-Bey-Abina
- 1 Biotherapy Department, Necker Children's Hospital , Assistance Publique-Hôpitaux de Paris, Paris.,2 Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest , Assistance Publique-Hôpitaux de Paris, INSERM, Paris.,5 UTCBS CNRS 8258-INSERM U1022, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes , Paris.,6 Immunology Laboratory, Groupe Hospitalier Universitaire Paris-Sud , AP-HP, Le-Kremlin-Bicêtre, France
| |
Collapse
|
40
|
Kailayangiri S, Altvater B, Spurny C, Jamitzky S, Schelhaas S, Jacobs AH, Wiek C, Roellecke K, Hanenberg H, Hartmann W, Wiendl H, Pankratz S, Meltzer J, Farwick N, Greune L, Fluegge M, Rossig C. Targeting Ewing sarcoma with activated and GD2-specific chimeric antigen receptor-engineered human NK cells induces upregulation of immune-inhibitory HLA-G. Oncoimmunology 2016; 6:e1250050. [PMID: 28197367 DOI: 10.1080/2162402x.2016.1250050] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/13/2016] [Accepted: 10/13/2016] [Indexed: 12/22/2022] Open
Abstract
Activated and in vitro expanded natural killer (NK) cells have substantial cytotoxicity against many tumor cells, but their in vivo efficacy to eliminate solid cancers is limited. Here, we used chimeric antigen receptors (CARs) to enhance the activity of NK cells against Ewing sarcomas (EwS) in a tumor antigen-specific manner. Expression of CARs directed against the ganglioside antigen GD2 in activated NK cells increased their responses to GD2+ allogeneic EwS cells in vitro and overcame resistance of individual cell lines to NK cell lysis. Second-generation CARs with 4-1BB and 2B4 co-stimulatory signaling and third-generation CARs combining both co-stimulatory domains were all equally effective. By contrast, adoptive transfer of GD2-specific CAR gene-modified NK cells both by intratumoral and intraperitoneal delivery failed to eliminate GD2-expressing EwS xenografts. Histopathology review revealed upregulation of the immunosuppressive ligand HLA-G in tumor autopsies from mice treated with NK cells compared to untreated control mice. Supporting the relevance of this finding, in vitro co-incubation of NK cells with allogeneic EwS cells induced upregulation of the HLA-G receptor CD85j, and HLA-G1 expressed by EwS cells suppressed the activity of NK cells from three of five allogeneic donors against the tumor cells in vitro. We conclude that HLA-G is a candidate immune checkpoint in EwS where it can contribute to resistance to NK cell therapy. HLA-G deserves evaluation as a potential target for more effective immunotherapeutic combination regimens in this and other cancers.
Collapse
Affiliation(s)
- Sareetha Kailayangiri
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Bianca Altvater
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Christian Spurny
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Silke Jamitzky
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Sonja Schelhaas
- European Institute for Molecular Imaging (EIMI), University of Muenster , Muenster, Germany
| | - Andreas H Jacobs
- European Institute for Molecular Imaging (EIMI), University of Muenster, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital, Heinrich Heine University , Duesseldorf, Germany
| | - Katharina Roellecke
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital, Heinrich Heine University , Duesseldorf, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology, Head and Neck Surgery, Children's Hospital, Heinrich Heine University, Duesseldorf, Germany; Department of Pediatrics III, University Children's Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wolfgang Hartmann
- Gerhard-Domagk Institute for Pathology, University of Muenster , Muenster, Germany
| | - Heinz Wiendl
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster, Germany; Department of Neurology, University Hospital Muenster, Muenster, Germany
| | - Susann Pankratz
- Department of Neurology, University Hospital Muenster , Muenster, Germany
| | - Jutta Meltzer
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Nicole Farwick
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Lea Greune
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Maike Fluegge
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster , Muenster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University of Muenster, Muenster, Germany
| |
Collapse
|
41
|
Hymes JP, Klaenhammer TR. Stuck in the Middle: Fibronectin-Binding Proteins in Gram-Positive Bacteria. Front Microbiol 2016; 7:1504. [PMID: 27713740 PMCID: PMC5031765 DOI: 10.3389/fmicb.2016.01504] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 09/08/2016] [Indexed: 11/13/2022] Open
Abstract
Fibronectin is a multidomain glycoprotein found ubiquitously in human body fluids and extracellular matrices of a variety of cell types from all human tissues and organs, including intestinal epithelial cells. Fibronectin plays a major role in the regulation of cell migration, tissue repair, and cell adhesion. Importantly, fibronectin also serves as a common target for bacterial adhesins in the gastrointestinal tract. Fibronectin-binding proteins (FnBPs) have been identified and characterized in a wide variety of host-associated bacteria. Single bacterial species can contain multiple, diverse FnBPs. In pathogens, some FnBPs contribute to virulence via host cell attachment, invasion, and interference with signaling pathways. Although FnBPs in commensal and probiotic strains are not sufficient to confer virulence, they are essential for attachment to their ecological niches. Here we describe the interaction between human fibronectin and bacterial adhesins by highlighting the FnBPs of Gram-positive pathogens and commensals. We provide an overview of the occurrence and diversity of FnBPs with a focus on the model pathogenic organisms in which FnBPs are most characterized. Continued investigation of FnBPs is needed to fully understand their divergence and specificity in both pathogens and commensals.
Collapse
Affiliation(s)
- Jeffrey P Hymes
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
| | - Todd R Klaenhammer
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University Raleigh, NC, USA
| |
Collapse
|
42
|
Zhang J, Ramadan AM, Griesenauer B, Li W, Turner MJ, Liu C, Kapur R, Hanenberg H, Blazar BR, Tawara I, Paczesny S. ST2 blockade reduces sST2-producing T cells while maintaining protective mST2-expressing T cells during graft-versus-host disease. Sci Transl Med 2016; 7:308ra160. [PMID: 26446957 DOI: 10.1126/scitranslmed.aab0166] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Graft-versus-host disease (GVHD) remains a devastating complication after allogeneic hematopoietic cell transplantation (HCT). We previously identified high plasma soluble suppression of tumorigenicity 2 (sST2) as a biomarker of the development of GVHD and death. sST2 sequesters interleukin-33 (IL-33), limiting its availability to T cells expressing membrane-bound ST2 (mST2) [T helper 2 (TH2) cells and ST2(+)FoxP3(+) regulatory T cells]. We report that blockade of sST2 in the peritransplant period with a neutralizing monoclonal antibody (anti-ST2 mAb) reduced GVHD severity and mortality. We identified intestinal stromal cells and T cells as major sources of sST2 during GVHD. ST2 blockade decreased systemic interferon-γ, IL-17, and IL-23 but increased IL-10 and IL-33 plasma levels. ST2 blockade also reduced sST2 production by IL-17-producing T cells while maintaining protective mST2-expressing T cells, increasing the frequency of intestinal myeloid-derived suppressor cells, and decreasing the frequency of intestinal CD103 dendritic cells. Finally, ST2 blockade preserved graft-versus-leukemia activity in a model of green fluorescent protein (GFP)-positive MLL-AF9 acute myeloid leukemia. Our findings suggest that ST2 is a therapeutic target for severe GVHD and that the ST2/IL-33 pathway could be investigated in other T cell-mediated immune disorders with loss of tolerance.
Collapse
Affiliation(s)
- Jilu Zhang
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Abdulraouf M Ramadan
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brad Griesenauer
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Wei Li
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Matthew J Turner
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN 46202, USA
| | - Chen Liu
- Department of Pathology and Immunology, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Reuben Kapur
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Helmut Hanenberg
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Bruce R Blazar
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| | - Isao Tawara
- Department of Hematology/Oncology, Mie University Hospital, Mie 514-8507, Japan
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA. Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
43
|
Retroviral vector interactions with hematopoietic cells. Curr Opin Virol 2016; 21:41-46. [PMID: 27521874 DOI: 10.1016/j.coviro.2016.07.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 07/27/2016] [Accepted: 07/29/2016] [Indexed: 01/01/2023]
Abstract
Hematopoietic stem cell (HSC) gene therapy using retroviral vectors is a powerful and promising approach to permanently correct many hematopoietic disorders. Increasing the transduction of quiescent HSCs and reducing genotoxicity are major challenges in the field. Retroviral vectors, including lentiviral and foamy vectors, have been extensively modified resulting in improved safety and efficacy. This review will focus on recent advances to improve vector entry, transduction efficiency, control of transgene expression and approaches to improve safety by modifying the retroviral integration profile.
Collapse
|
44
|
Nam K, Jones JP, Lei P, Andreadis ST, Baker OJ. Laminin-111 Peptides Conjugated to Fibrin Hydrogels Promote Formation of Lumen Containing Parotid Gland Cell Clusters. Biomacromolecules 2016; 17:2293-301. [PMID: 27151393 PMCID: PMC5029268 DOI: 10.1021/acs.biomac.6b00588] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Previous studies showed that mouse submandibular gland cells form three-dimensional structures when grown on Laminin-111 gels. The use of Laminin-111 for tissue bioengineering is complicated due to its lack of purity. By contrast, the use of synthetic peptides derived from Laminin-111 is beneficial due to their high purity and easy manipulation. Two Laminin-111 peptides have been identified for salivary cells: the A99 peptide corresponding to the α1 chain from Laminin-111 and the YIGSR peptide corresponding to the β1 chain from Laminin-111, which are important for cell adhesion and migration. We created three-dimensional salivary cell clusters using a modified fibrin hydrogel matrix containing immobilized Laminin-111 peptides. Results indicate that the YIGSR peptide improved morphology and lumen formation in rat parotid Par-C10 cells as compared to cells grown on unmodified fibrin hydrogel. Moreover, a combination of both peptides not only allowed the formation of functional three-dimensional salivary cell clusters but also increased attachment and number of cell clusters. In summary, we demonstrated that fibrin hydrogel decorated with Laminin-111 peptides supports attachment and differentiation of salivary gland cell clusters with mature lumens.
Collapse
Affiliation(s)
- Kihoon Nam
- School of Dentistry, The University of Utah, Salt Lake City, Utah 84108, United States
| | - Joshua P. Jones
- Department of Bioengineering, The University of Utah, Salt Lake City, Utah 84108, United States
| | - Pedro Lei
- Department of Chemical and Biological Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Stelios T. Andreadis
- Department of Chemical and Biological Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
- Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
- Center of Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, New York 14260, United States
| | - Olga J. Baker
- School of Dentistry, The University of Utah, Salt Lake City, Utah 84108, United States
| |
Collapse
|
45
|
Roellecke K, Virts EL, Einholz R, Edson KZ, Altvater B, Rossig C, von Laer D, Scheckenbach K, Wagenmann M, Reinhardt D, Kramm CM, Rettie AE, Wiek C, Hanenberg H. Optimized human CYP4B1 in combination with the alkylator prodrug 4-ipomeanol serves as a novel suicide gene system for adoptive T-cell therapies. Gene Ther 2016; 23:615-26. [PMID: 27092941 DOI: 10.1038/gt.2016.38] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 03/23/2016] [Accepted: 04/05/2016] [Indexed: 12/18/2022]
Abstract
Engineering autologous or allogeneic T cells to express a suicide gene can control potential toxicity in adoptive T-cell therapies. We recently reported the development of a novel human suicide gene system that is based on an orphan human cytochrome P450 enzyme, CYP4B1, and the naturally occurring alkylator prodrug 4-ipomeanol. The goal of this study was to systematically develop a clinically applicable self-inactivating lentiviral vector for efficient co-expression of CYP4B1 as an ER-located protein with two distinct types of cell surface proteins, either MACS selection genes for donor lymphocyte infusions after allogeneic stem cell transplantation or chimeric antigen receptors for retargeting primary T cells. The U3 region of the myeloproliferative sarcoma virus in combination with the T2A site was found to drive high-level expression of our CYP4B1 mutant with truncated CD34 or CD271 as MACS suitable selection markers. This lentiviral vector backbone was also well suited for co-expression of CYP4B1 with a codon-optimized CD19 chimeric antigen receptor (CAR) construct. Finally, 4-ipomeanol efficiently induced apoptosis in primary T cells that co-express mutant CYP4B1 and the divergently located MACS selection and CAR genes. In conclusion, we here developed a clinically suited lentiviral vector that supports high-level co-expression of cell surface proteins with a potent novel human suicide gene.
Collapse
Affiliation(s)
- K Roellecke
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - E L Virts
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - R Einholz
- Institute for Organic Chemistry, University of Tübingen, Tübingen, Germany
| | - K Z Edson
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - B Altvater
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - C Rossig
- Department of Pediatric Hematology and Oncology, University Children's Hospital Muenster, Muenster, Germany
| | - D von Laer
- Institute for Virology, Innsbruck Medical University, Innsbruck, Austria
| | - K Scheckenbach
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - M Wagenmann
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - D Reinhardt
- Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - C M Kramm
- Division of Pediatric Hematology and Oncology, Department of Child and Adolescent Health, University Medical Center Göttingen, Göttingen, Germany
| | - A E Rettie
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA, USA
| | - C Wiek
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany
| | - H Hanenberg
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, Düsseldorf, Germany.,Department of Pediatrics III, University Children's Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
46
|
Li W, Liu L, Gomez A, Zhang J, Ramadan A, Zhang Q, Choi SW, Zhang P, Greenson JK, Liu C, Jiang D, Virts E, Kelich SL, Chu HW, Flynn R, Blazar BR, Hanenberg H, Hanash S, Paczesny S. Proteomics analysis reveals a Th17-prone cell population in presymptomatic graft-versus-host disease. JCI Insight 2016; 1:86660. [PMID: 27195312 DOI: 10.1172/jci.insight.86660] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Gastrointestinal graft-versus-host-disease (GI-GVHD) is a life-threatening complication occurring after allogeneic hematopoietic cell transplantation (HCT), and a blood biomarker that permits stratification of HCT patients according to their risk of developing GI-GVHD would greatly aid treatment planning. Through in-depth, large-scale proteomic profiling of presymptomatic samples, we identified a T cell population expressing both CD146, a cell adhesion molecule, and CCR5, a chemokine receptor that is upregulated as early as 14 days after transplantation in patients who develop GI-GVHD. The CD4+CD146+CCR5+ T cell population is Th17 prone and increased by ICOS stimulation. shRNA knockdown of CD146 in T cells reduced their transmigration through endothelial cells, and maraviroc, a CCR5 inhibitor, reduced chemotaxis of the CD4+CD146+CCR5+ T cell population toward CCL14. Mice that received CD146 shRNA-transduced human T cells did not lose weight, showed better survival, and had fewer CD4+CD146+CCR5+ T cells and less pathogenic Th17 infiltration in the intestine, even compared with mice receiving maraviroc with control shRNA- transduced human T cells. Furthermore, the frequency of CD4+CD146+CCR5+ Tregs was increased in GI-GVHD patients, and these cells showed increased plasticity toward Th17 upon ICOS stimulation. Our findings can be applied to early risk stratification, as well as specific preventative therapeutic strategies following HCT.
Collapse
Affiliation(s)
- Wei Li
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Liangyi Liu
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Jilu Zhang
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Qing Zhang
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Sung W Choi
- University of Michigan, Ann Arbor, Michigan, USA
| | - Peng Zhang
- University of Michigan, Ann Arbor, Michigan, USA
| | | | - Chen Liu
- Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Di Jiang
- National Jewish Health, Denver, Colorado, USA
| | - Elizabeth Virts
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | - Ryan Flynn
- University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Helmut Hanenberg
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | - Sophie Paczesny
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
47
|
Majdoul S, Seye AK, Kichler A, Holic N, Galy A, Bechinger B, Fenard D. Molecular Determinants of Vectofusin-1 and Its Derivatives for the Enhancement of Lentivirally Mediated Gene Transfer into Hematopoietic Stem/Progenitor Cells. J Biol Chem 2015; 291:2161-9. [PMID: 26668323 DOI: 10.1074/jbc.m115.675033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Indexed: 12/20/2022] Open
Abstract
Gene delivery into hCD34+ hematopoietic stem/progenitor cells (HSPCs) using human immunodeficiency virus, type 1-derived lentiviral vectors (LVs) has several promising therapeutic applications. Numerous clinical trials are currently underway. However, the efficiency, safety, and cost of LV gene therapy could be ameliorated by enhancing target cell transduction levels and reducing the amount of LV used on the cells. Several transduction enhancers already exist, such as fibronectin fragments or cationic compounds. Recently, we discovered Vectofusin-1, a new transduction enhancer, also called LAH4-A4, a short histidine-rich amphipathic peptide derived from the LAH4 family of DNA transfection agents. Vectofusin-1 enhances the infectivity of lentiviral and γ-retroviral vectors pseudotyped with various envelope glycoproteins. In this study, we compared a family of Vectofusin-1 isomers and showed that Vectofusin-1 remains the lead peptide for HSPC transduction enhancement with LVs pseudotyped with vesicular stomatitis virus glycoproteins and also with modified gibbon ape leukemia virus glycoproteins. By comparing the capacity of numerous Vectofusin-1 variants to promote the modified gibbon ape leukemia virus glycoprotein-pseudotyped lentiviral vector infectivity of HSPCs, the lysine residues on the N-terminal extremity of Vectofusin-1, a hydrophilic angle of 140° formed by the histidine residues in the Schiffer-Edmundson helical wheel representation, hydrophobic residues consisting of leucine were all found to be essential and helped to define a minimal active sequence. The data also show that the critical determinants necessary for lentiviral transduction enhancement are partially different from those necessary for efficient antibiotic or DNA transfection activity of LAH4 derivatives. In conclusion, these results help to decipher the action mechanism of Vectofusin-1 in the context of hCD34+ cell-based gene therapy.
Collapse
Affiliation(s)
- Saliha Majdoul
- From Généthon, 91000 Evry, France, INSERM UMR_S951, 91000 Evry, France, University of Evry, 91000 Evry, France
| | - Ababacar K Seye
- From Généthon, 91000 Evry, France, INSERM UMR_S951, 91000 Evry, France
| | - Antoine Kichler
- CNRS, UMR_7199, 67401 Illkirch, France, the University of Strasbourg, 67000 Strasbourg, France, and
| | - Nathalie Holic
- From Généthon, 91000 Evry, France, INSERM UMR_S951, 91000 Evry, France, University of Evry, 91000 Evry, France
| | - Anne Galy
- From Généthon, 91000 Evry, France, INSERM UMR_S951, 91000 Evry, France, University of Evry, 91000 Evry, France,
| | - Burkhard Bechinger
- the University of Strasbourg, 67000 Strasbourg, France, and the Institut de Chimie, CNRS, UMR_7177, 67401 Strasbourg, France
| | - David Fenard
- From Généthon, 91000 Evry, France, INSERM UMR_S951, 91000 Evry, France, University of Evry, 91000 Evry, France,
| |
Collapse
|
48
|
Schmidt EM, Wiek C, Parkinson OT, Roellecke K, Freund M, Gombert M, Lottmann N, Steward CA, Kramm CM, Yarov-Yarovoy V, Rettie AE, Hanenberg H. Characterization of an Additional Splice Acceptor Site Introduced into CYP4B1 in Hominoidae during Evolution. PLoS One 2015; 10:e0137110. [PMID: 26355749 PMCID: PMC4565547 DOI: 10.1371/journal.pone.0137110] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 08/12/2015] [Indexed: 01/22/2023] Open
Abstract
CYP4B1 belongs to the cytochrome P450 family 4, one of the oldest P450 families whose members have been highly conserved throughout evolution. The CYP4 monooxygenases typically oxidize fatty acids to both inactive and active lipid mediators, although the endogenous ligand(s) is largely unknown. During evolution, at the transition of great apes to humanoids, the CYP4B1 protein acquired a serine instead of a proline at the canonical position 427 in the meander region. Although this alteration impairs P450 function related to the processing of naturally occurring lung toxins, a study in transgenic mice suggested that an additional serine insertion at position 207 in human CYP4B1 can rescue the enzyme stability and activity. Here, we report that the genomic insertion of a CAG triplet at the intron 5–exon 6 boundary in human CYP4B1 introduced an additional splice acceptor site in frame. During evolution, this change occurred presumably at the stage of Hominoidae and leads to two major isoforms of the CYP4B1 enzymes of humans and great apes, either with or without a serine 207 insertion (insSer207). We further demonstrated that the CYP4B1 enzyme with insSer207 is the dominant isoform (76%) in humans. Importantly, this amino acid insertion did not affect the 4-ipomeanol metabolizing activities or stabilities of the native rabbit or human CYP4B1 enzymes, when introduced as transgenes in human primary cells and cell lines. In our 3D modeling, this functional neutrality of insSer207 is compatible with its predicted location on the exterior surface of CYP4B1 in a flexible side chain. Therefore, the Ser207 insertion does not rescue the P450 functional activity of human CYP4B1 that has been lost during evolution.
Collapse
Affiliation(s)
- Eva M. Schmidt
- Department of Pediatric Hematology, Oncology and Clinical Immunology, Children’s Hospital, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Oliver T. Parkinson
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA 98195, United States of America
| | - Katharina Roellecke
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Marcel Freund
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Michael Gombert
- Department of Pediatric Hematology, Oncology and Clinical Immunology, Children’s Hospital, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Nadine Lottmann
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, 40225 Düsseldorf, Germany
| | | | - Christof M. Kramm
- Division of Pediatric Hematology and Oncology, Department of Child and Adolescent Health, University of Göttingen, 37099 Göttingen, Germany
| | - Vladimir Yarov-Yarovoy
- Departments of Physiology and Membrane Biology, University of California Davis, Davis, CA 95616, United States of America
| | - Allan E. Rettie
- Department of Medicinal Chemistry, School of Pharmacy, University of Washington, Seattle, WA 98195, United States of America
| | - Helmut Hanenberg
- Department of Otorhinolaryngology and Head/Neck Surgery, Heinrich Heine University, 40225 Düsseldorf, Germany
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America
- Department of Pediatrics III, University Children’s Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
- * E-mail:
| |
Collapse
|
49
|
Virts EL, Jankowska A, Mackay C, Glaas MF, Wiek C, Kelich SL, Lottmann N, Kennedy FM, Marchal C, Lehnert E, Scharf RE, Dufour C, Lanciotti M, Farruggia P, Santoro A, Savasan S, Scheckenbach K, Schipper J, Wagenmann M, Lewis T, Leffak M, Farlow JL, Foroud TM, Honisch E, Niederacher D, Chakraborty SC, Vance GH, Pruss D, Timms KM, Lanchbury JS, Alpi AF, Hanenberg H. AluY-mediated germline deletion, duplication and somatic stem cell reversion in UBE2T defines a new subtype of Fanconi anemia. Hum Mol Genet 2015; 24:5093-108. [PMID: 26085575 PMCID: PMC4550815 DOI: 10.1093/hmg/ddv227] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 06/12/2015] [Indexed: 01/09/2023] Open
Abstract
Fanconi anemia (FA) is a rare inherited disorder clinically characterized by congenital malformations, progressive bone marrow failure and cancer susceptibility. At the cellular level, FA is associated with hypersensitivity to DNA-crosslinking genotoxins. Eight of 17 known FA genes assemble the FA E3 ligase complex, which catalyzes monoubiquitination of FANCD2 and is essential for replicative DNA crosslink repair. Here, we identify the first FA patient with biallelic germline mutations in the ubiquitin E2 conjugase UBE2T. Both mutations were aluY-mediated: a paternal deletion and maternal duplication of exons 2-6. These loss-of-function mutations in UBE2T induced a cellular phenotype similar to biallelic defects in early FA genes with the absence of FANCD2 monoubiquitination. The maternal duplication produced a mutant mRNA that could encode a functional protein but was degraded by nonsense-mediated mRNA decay. In the patient's hematopoietic stem cells, the maternal allele with the duplication of exons 2-6 spontaneously reverted to a wild-type allele by monoallelic recombination at the duplicated aluY repeat, thereby preventing bone marrow failure. Analysis of germline DNA of 814 normal individuals and 850 breast cancer patients for deletion or duplication of UBE2T exons 2-6 identified the deletion in only two controls, suggesting aluY-mediated recombinations within the UBE2T locus are rare and not associated with an increased breast cancer risk. Finally, a loss-of-function germline mutation in UBE2T was detected in a high-risk breast cancer patient with wild-type BRCA1/2. Cumulatively, we identified UBE2T as a bona fide FA gene (FANCT) that also may be a rare cancer susceptibility gene.
Collapse
Affiliation(s)
| | | | - Craig Mackay
- Department of MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK
| | - Marcel F Glaas
- Department of Otorhinolaryngology and Head/Neck Surgery (ENT) and
| | - Constanze Wiek
- Department of Otorhinolaryngology and Head/Neck Surgery (ENT) and
| | | | - Nadine Lottmann
- Department of Otorhinolaryngology and Head/Neck Surgery (ENT) and
| | | | | | - Erik Lehnert
- Department of Experimental and Clinical Hemostasis, Hemotherapy and Transfusion Medicine, Heinrich Heine University, Düsseldorf, Germany
| | - Rüdiger E Scharf
- Department of Experimental and Clinical Hemostasis, Hemotherapy and Transfusion Medicine, Heinrich Heine University, Düsseldorf, Germany
| | - Carlo Dufour
- Hematology Unit, G. Gaslini Children's Hospital, Genoa, Italy
| | | | - Piero Farruggia
- Pediatric Hematology and Oncology Unit, A.R.N.A.S. Ospedale Civico, Palermo, Italy
| | | | - Süreyya Savasan
- Department of Pediatrics, Children's Hospital of Michigan, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Jörg Schipper
- Department of Otorhinolaryngology and Head/Neck Surgery (ENT) and
| | - Martin Wagenmann
- Department of Otorhinolaryngology and Head/Neck Surgery (ENT) and
| | - Todd Lewis
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Michael Leffak
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine, Wright State University, Dayton, OH 45435, USA
| | - Janice L Farlow
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Tatiana M Foroud
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ellen Honisch
- Department of Gynecology, Heinrich Heine University, Düsseldorf, Germany and
| | - Dieter Niederacher
- Department of Gynecology, Heinrich Heine University, Düsseldorf, Germany and
| | - Sujata C Chakraborty
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Gail H Vance
- Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | | - Arno F Alpi
- Department of MRC Protein Phosphorylation and Ubiquitylation Unit, College of Life Sciences, University of Dundee, Dundee, UK,
| | - Helmut Hanenberg
- Department of Pediatrics and Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA, Department of Otorhinolaryngology and Head/Neck Surgery (ENT) and
| |
Collapse
|
50
|
Identification of amino acid determinants in CYP4B1 for optimal catalytic processing of 4-ipomeanol. Biochem J 2015; 465:103-14. [PMID: 25247810 DOI: 10.1042/bj20140813] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mammalian CYP4B1 enzymes are cytochrome P450 mono-oxygenases that are responsible for the bioactivation of several exogenous pro-toxins including 4-ipomeanol (4-IPO). In contrast with the orthologous rabbit enzyme, we show here that native human CYP4B1 with a serine residue at position 427 is unable to bioactivate 4-IPO and does not cause cytotoxicity in HepG2 cells and primary human T-cells that overexpress these enzymes. We also demonstrate that a proline residue in the meander region at position 427 in human CYP4B1 and 422 in rabbit CYP4B1 is important for protein stability and rescues the 4-IPO bioactivation of the human enzyme, but is not essential for the catalytic activity of the rabbit CYP4B1 protein. Systematic substitution of native and p.S427P human CYP4B1 with peptide regions from the highly active rabbit enzyme reveals that 18 amino acids in the wild-type rabbit CYP4B1 protein are key for conferring high 4-IPO metabolizing activity. Introduction of 12 of the 18 amino acids that are also present at corresponding positions in other human CYP4 family members into the p.S427P human CYP4B1 protein results in a mutant human enzyme (P+12) that is as stable and as active as the rabbit wild-type CYP4B1 protein. These 12 mutations cluster in the predicted B-C loop through F-helix regions and reveal new amino acid regions important to P450 enzyme stability. Finally, by minimally re-engineering the human CYP4B1 enzyme for efficient activation of 4-IPO, we have developed a novel human suicide gene system that is a candidate for adoptive cellular therapies in humans.
Collapse
|