1
|
Melocchi A, Schmittlein B, Sadhu S, Nayak S, Lares A, Uboldi M, Zema L, di Robilant BN, Feldman SA, Esensten JH. Automated manufacturing of cell therapies. J Control Release 2025; 381:113561. [PMID: 39993639 DOI: 10.1016/j.jconrel.2025.02.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Advanced therapy medicinal products (ATMPs), particularly genetically engineered cell-based therapies, are a major class of drugs with several high-profile Food and Drug Administration (FDA) approvals in the past decade. However, the high cost and limited production capacity of these drugs remain a barrier to access. These costs are primarily due to the complex manufacturing processes (often a single batch for a single patient), which increases personnel and facility expenses, and the challenges associated with tech-transfer from research and development stages to clinical-stage production. In order to scale up and scale out in a cost-effective way, automated solutions capable of multi-step manufacturing have been developed in academia and industry. The aim of the present article is to summarize the design approaches and key features of current multi-step automated systems for cell therapy manufacturing. For each system described in the literature, we will discuss different aspects in detail such as cell specificity, modularity, processing models, manufacturing locations, and integrated quality control. Our analysis highlights that developers need to balance competing needs in an environment where the biological, business, and technological factors are constantly evolving. Thus, designing engineering solutions that align with the pharmaceutical end-user is essential. Adopting a risk-based approach grounded in published data is the most effective strategy to evaluate existing and emerging automated systems.
Collapse
Affiliation(s)
- Alice Melocchi
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy; Multiply Labs, San Francisco, CA, USA.
| | | | | | | | | | - Marco Uboldi
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | - Lucia Zema
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | | | - Steven A Feldman
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan H Esensten
- Advanced Biotherapy Center (ABC), Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
2
|
Beswick L. The health economics of cell and gene therapies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:155-167. [PMID: 39341653 DOI: 10.1016/b978-0-323-90120-8.00004-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The health economics of cell and gene therapies is complex; due to resource-intensive manufacturing, high prices are required for commercial viability that are challenging for healthcare systems to accommodate. Despite high prices, cell and gene therapies can provide value when they deliver substantial clinical benefits and displace long-term healthcare costs compared with existing treatment options. In this chapter, the cost utility approach of economic evaluation is discussed, focusing on the considerations that occur more commonly in cell and gene therapies compared to conventional medicines, how these considerations create challenges in interpreting the evidence and coming to conclusions on value, and what tools exist for understanding the level of decision uncertainty. A summary of the economic evaluation of onasemnogene abeparvovec for spinal muscular atrophy is provided as a real-world example that features the considerations discussed.
Collapse
Affiliation(s)
- Laura Beswick
- Health Economics and Market Access, Cell and Gene Therapy Catapult(†), London, United Kingdom.
| |
Collapse
|
3
|
McCallion O, Bilici M, Hester J, Issa F. Regulatory T-cell therapy approaches. Clin Exp Immunol 2023; 211:96-107. [PMID: 35960852 PMCID: PMC10019137 DOI: 10.1093/cei/uxac078] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 07/26/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Regulatory T cells (Tregs) have enormous therapeutic potential to treat a variety of immunopathologies characterized by aberrant immune activation. Adoptive transfer of ex vivo expanded autologous Tregs continues to progress through mid- to late-phase clinical trials in several disease spaces and has generated promising preliminary safety and efficacy signals to date. However, the practicalities of this strategy outside of the clinical trial setting remain challenging. Here, we review the current landscape of regulatory T-cell therapy, considering emergent approaches and technologies presenting novel ways to engage Tregs, and reflect on the progress necessary to deliver their therapeutic potential to patients.
Collapse
Affiliation(s)
- Oliver McCallion
- Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Merve Bilici
- Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Joanna Hester
- Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fadi Issa
- Correspondence. Fadi Issa, Translational Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Headley Way, Oxford OX3 9DU, UK.
| |
Collapse
|
4
|
Bhatta R, Han J, Liu Y, Bo Y, Wang H. T cell-responsive macroporous hydrogels for in situ T cell expansion and enhanced antitumor efficacy. Biomaterials 2023; 293:121972. [PMID: 36566554 PMCID: PMC9868092 DOI: 10.1016/j.biomaterials.2022.121972] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 11/20/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022]
Abstract
Adoptive T cell therapy has demonstrated great promise for treating cancer and other diseases. While extensive effort has been made to improve ex vivo expansion of T cells, strategies for maintaining the proliferation and function of T cells post adoptive transfer are still lacking. Here we report an injectable T cell-responsive macroporous hydrogel that enables in situ activation and expansion of T cells. The macroporous gel is composed of a polymeric network with dispersed macropores (∼150 μm) that are large enough to home T cells. In the presence of T cells that can gradually disrupt the gel network surrounding the macropores, activation cues can be gradually released for in situ activation and expansion of T cells. This T cell-responsive macroporous gel enables expansion of effector T cells in vivo, is stable over weeks upon subcutaneous injection, and results in enhanced CD8+ T cell response and antitumor efficacy. We further show that the T cell-responsive macroporous gel could achieve comparable antitumor efficacy to conventional T cell therapy with a much lower cell dose. This injectable, T cell-responsive macroporous gel provides a platform for in vivo expansion of engineered T cells in a controlled manner, for timely and effective treatment of diseases.
Collapse
Affiliation(s)
- Rimsha Bhatta
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joonsu Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA; Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
5
|
Koleva-Kolarova R, Buchanan J, Vellekoop H, Huygens S, Versteegh M, Mölken MRV, Szilberhorn L, Zelei T, Nagy B, Wordsworth S, Tsiachristas A. Financing and Reimbursement Models for Personalised Medicine: A Systematic Review to Identify Current Models and Future Options. APPLIED HEALTH ECONOMICS AND HEALTH POLICY 2022; 20:501-524. [PMID: 35368231 PMCID: PMC9206925 DOI: 10.1007/s40258-021-00714-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/28/2021] [Indexed: 05/31/2023]
Abstract
BACKGROUND The number of healthcare interventions described as 'personalised medicine' (PM) is increasing rapidly. As healthcare systems struggle to decide whether to fund PM innovations, it is unclear what models for financing and reimbursement are appropriate to apply in this context. OBJECTIVE To review financing and reimbursement models for PM, summarise their key characteristics, and describe whether they can influence the development and uptake of PM. METHODS A literature review was conducted in Medline, Embase, Web of Science, and Econlit to identify studies published in English between 2009 and 2021, and reviews published before 2009. Grey literature was identified through Google Scholar, Google and subject-specific webpages. Articles that described financing and reimbursement of PM, and financing of non-PM were included. Data were extracted and synthesised narratively to report on the models, as well as facilitators, incentives, barriers and disincentives that could influence PM development and uptake. RESULTS One hundred and fifty-three papers were included. Research and development of PM was financed through both public and private sources and reimbursed largely through traditional models such as single fees, Diagnosis-Related Groups, and bundled payments. Financial-based reimbursement, including rebates and price-volume agreements, was mainly applied to targeted therapies. Performance-based reimbursement was identified mainly for gene and targeted therapies, and some companion diagnostics. Gene therapy manufacturers offered outcome-based rebates for treatment failure for interventions including Luxturna®, Kymriah®, Yescarta®, Zynteglo®, Zolgensma® and Strimvelis®, and coverage with evidence development for Kymriah® and Yescarta®. Targeted testing with OncotypeDX® was granted value-based reimbursement through initial coverage with evidence development. The main barriers and disincentives to PM financing and reimbursement were the lack of strong links between stakeholders and the lack of demonstrable benefit and value of PM. CONCLUSIONS Public-private financing agreements and performance-based reimbursement models could help facilitate the development and uptake of PM interventions with proven clinical benefit.
Collapse
Affiliation(s)
| | - James Buchanan
- Health Economics Research Centre, University of Oxford, Oxford, UK
| | - Heleen Vellekoop
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
| | - Simone Huygens
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
| | - Matthijs Versteegh
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
| | - Maureen Rutten-van Mölken
- Institute for Medical Technology Assessment, Erasmus University Rotterdam, P.O. Box 1738, 3000 DR, Rotterdam, The Netherlands
- Erasmus School of Health Policy and Management, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - László Szilberhorn
- Syreon Research Institute, Budapest, Hungary
- Faculty of Social Sciences, Eötvös Loránd University, Budapest, Hungary
| | - Tamás Zelei
- Syreon Research Institute, Budapest, Hungary
| | - Balázs Nagy
- Syreon Research Institute, Budapest, Hungary
| | - Sarah Wordsworth
- Health Economics Research Centre, University of Oxford, Oxford, UK
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| | - Apostolos Tsiachristas
- Health Economics Research Centre, University of Oxford, Oxford, UK
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
6
|
Pizevska M, Kaeda J, Fritsche E, Elazaly H, Reinke P, Amini L. Advanced Therapy Medicinal Products' Translation in Europe: A Developers' Perspective. Front Med (Lausanne) 2022; 9:757647. [PMID: 35186986 PMCID: PMC8851388 DOI: 10.3389/fmed.2022.757647] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Advanced Therapy Medicinal Products (ATMPs) comprising cell, gene, and tissue-engineered therapies have demonstrated enormous therapeutic benefits. However, their development is complex to be managed efficiently within currently existing regulatory frameworks. Legislation and regulation requirements for ATMPs must strike a balance between the patient safety while promoting innovations to optimize exploitation of these novel therapeutics. This paradox highlights the importance of on-going dynamic dialogue between all stakeholders and regulatory science to facilitate the development of pragmatic ATMP regulatory guidelines.
Collapse
Affiliation(s)
- Maja Pizevska
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Jaspal Kaeda
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Enrico Fritsche
- Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Hisham Elazaly
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| | - Petra Reinke
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Leila Amini
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany.,Berlin Center for Advanced Therapies (BeCAT), Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
7
|
Abou-El-Enein M, Elsallab M, Feldman SA, Fesnak AD, Heslop HE, Marks P, Till BG, Bauer G, Savoldo B. Scalable Manufacturing of CAR T cells for Cancer Immunotherapy. Blood Cancer Discov 2021; 2:408-422. [PMID: 34568831 PMCID: PMC8462122 DOI: 10.1158/2643-3230.bcd-21-0084] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
As of April 2021, there are five commercially available chimeric antigen receptor (CAR) T cell therapies for hematological malignancies. With the current transition of CAR T cell manufacturing from academia to industry, there is a shift toward Good Manufacturing Practice (GMP)-compliant closed and automated systems to ensure reproducibility and to meet the increased demand for cancer patients. In this review we describe current CAR T cells clinical manufacturing models and discuss emerging technological advances that embrace scaling and production optimization. We summarize measures being used to shorten CAR T-cell manufacturing times and highlight regulatory challenges to scaling production for clinical use. Statement of Significance ∣ As the demand for CAR T cell cancer therapy increases, several closed and automated production platforms are being deployed, and others are in development.This review provides a critical appraisal of these technologies that can be leveraged to scale and optimize the production of next generation CAR T cells.
Collapse
Affiliation(s)
- Mohamed Abou-El-Enein
- Division of Medical Oncology, Department of Medicine, and Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Joint USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Magdi Elsallab
- Joint USC/CHLA Cell Therapy Program, University of Southern California, and Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Steven A Feldman
- Stanford Center for Cancer Cell Therapy, Stanford Cancer Institute, Palo Alto, CA
| | - Andrew D Fesnak
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Helen E Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital and Houston Methodist Hospital, Houston, TX, USA
| | - Peter Marks
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, MD, USA
| | - Brian G Till
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Gerhard Bauer
- Institute for Regenerative Cures (IRC), University of California Davis, Sacramento, California, USA
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
8
|
Qiu T, Wang Y, Liang S, Han R, Toumi M. Partnership agreements for regenerative medicines: a database analysis and implications for future innovation. Regen Med 2021; 16:733-755. [PMID: 34431716 DOI: 10.2217/rme-2021-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Partnerships have been leveraged to advance the regenerative medicines (RMs) development. This study analyzed the evolution of partnership landscape for regenerative medicines (RMs). Methods: Partnership agreements publicly announced from January 2014 - June 2020 were described. Results: 1169 partnership agreements with total amount of US$63,496 million were identified. Most agreements concerned RMs that were for oncology (25.3%), in the discovery or preclinical phase (66.9%) and gene-based products (45.3%). The most common partnership type is collaborative agreements without licensing. The partnerships between 'biotechnology companies and not-for-profit organizations' represented the largest number (n = 416; 35.6%). 'Big Pharma' preferred collaboration and licensing agreements with a higher amount. Conclusion: Collaborations between highly specialized players with complementary expertise promote the successful translation of scientific discovery to RMs.
Collapse
Affiliation(s)
- Tingting Qiu
- Department of Public Health, Aix-Marseille University, 27 Boulevard Jean Moulin, Marseille, 13385, France
| | - Yitong Wang
- Department of Public Health, Aix-Marseille University, 27 Boulevard Jean Moulin, Marseille, 13385, France
| | - Shuyao Liang
- Department of Public Health, Aix-Marseille University, 27 Boulevard Jean Moulin, Marseille, 13385, France
| | - Ru Han
- Department of Public Health, Aix-Marseille University, 27 Boulevard Jean Moulin, Marseille, 13385, France
| | - Mondher Toumi
- Department of Public Health, Aix-Marseille University, 27 Boulevard Jean Moulin, Marseille, 13385, France
| |
Collapse
|
9
|
García-González J, Marhuenda-Castillo S, Romero-Carretero S, Beltrán-García J. New era of personalized medicine: Advanced therapy medicinal products in Europe. World J Immunol 2021; 11:1-10. [DOI: 10.5411/wji.v11.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/24/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023] Open
Abstract
Advanced therapy medicinal products are human medical therapies based on genes, cells, or tissues, and due to their characteristics, they offer new innovative opportunities for the treatment of diseases and injuries, especially for diseases beyond the reach of traditional approaches. These therapies are at the forefront of innovation and have historically been very controversial, although in the last decade they have gained prominence while the number of new advanced therapies has increased every year. In this regard, despite the controversy they may generate, they are expected to dominate the market in the coming decades. Technologies based on advanced therapies are the present and future of medicine and bring us closer to the long-awaited precision medicine. Here we review the field as it stands today, with a focus on the molecular mechanisms that guided the different advanced therapies approved by the European Medicines Agency, their current status, and their legal approval.
Collapse
Affiliation(s)
| | | | | | - Jesús Beltrán-García
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia, Valencia 46010, Spain
- Center for Biomedical Research in Rare Diseases Network (CIBERER), Carlos III Health Institute, Valencia 46010, Spain
- INCLIVA Institute of Sanitary Research, Valencia 46010, Spain
| |
Collapse
|
10
|
Abstract
Gene therapy makes it possible to engineer chimeric antigen receptors (CARs) to create T cells that target specific diseases. However, current approaches require elaborate and expensive protocols to manufacture engineered T cells ex vivo, putting this therapy beyond the reach of many patients who might benefit. A solution could be to program T cells in vivo. Here, we evaluate the clinical need for in situ CAR T cell programming, compare competing technologies, review current progress, and provide a perspective on the long-term impact of this emerging and rapidly flourishing biotechnology field.
Collapse
Affiliation(s)
- Neha N Parayath
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Matthias T Stephan
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA.,Department of Bioengineering and Molecular Engineering & Sciences Institute, University of Washington, Seattle, Washington 98195, USA;
| |
Collapse
|
11
|
Hoogduijn MJ, Issa F, Casiraghi F, Reinders MEJ. Cellular therapies in organ transplantation. Transpl Int 2021; 34:233-244. [PMID: 33207013 PMCID: PMC7898347 DOI: 10.1111/tri.13789] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/15/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Cellular therapy is a promising tool for improving the outcome of organ transplantation. Various cell types with different immunoregulatory and regenerative properties may find application for specific transplant rejection or injury-related indications. The current era is crucial for the development of cellular therapies. Preclinical models have demonstrated the feasibility of efficacious cell therapy in transplantation, early clinical trials have shown safety of several of these therapies, and the first steps towards efficacy studies in humans have been made. In this review, we address the current state of the art of cellular therapies in clinical transplantation and discuss monitoring tools and endpoints for these studies.
Collapse
Affiliation(s)
- Martin J. Hoogduijn
- Nephrology and TransplantationDepartment of Internal MedicineErasmus University Medical CenterErasmus Medical CenterRotterdamThe Netherlands
| | - Fadi Issa
- Transplantation Research and Immunology GroupNuffield Department of Surgical SciencesJohn Radcliffe HospitalUniversity of OxfordOxfordUK
| | | | - Marlies E. J. Reinders
- Nephrology and TransplantationDepartment of Internal MedicineErasmus University Medical CenterErasmus Medical CenterRotterdamThe Netherlands
| |
Collapse
|
12
|
van Overbeeke E, Michelsen S, Toumi M, Stevens H, Trusheim M, Huys I, Simoens S. Market access of gene therapies across Europe, USA, and Canada: challenges, trends, and solutions. Drug Discov Today 2020; 26:399-415. [PMID: 33242695 DOI: 10.1016/j.drudis.2020.11.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/21/2020] [Accepted: 11/19/2020] [Indexed: 01/19/2023]
Abstract
This review can inform gene therapy developers on challenges that can be encountered when seeking market access. Moreover, it provides an overview of trends among challenges and potential solutions.
Collapse
Affiliation(s)
- Eline van Overbeeke
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium.
| | - Sissel Michelsen
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium; Healthcare Management Centre, Vlerick Business School, Reep 1, 9000 Ghent, Belgium
| | - Mondher Toumi
- Public Health Department, Aix Marseille University, 27 bd Jean Moulin, Marseille, France
| | - Hilde Stevens
- Institute for Interdisciplinary Innovation in Healthcare (I(3)h), Université libre de Bruxelles, Route de Lennik 808, Brussels, Belgium
| | - Mark Trusheim
- Massachusetts Institute of Technology, 100 Main Street, Cambridge, MA 02139, USA
| | - Isabelle Huys
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium
| | - Steven Simoens
- Clinical Pharmacology and Pharmacotherapy, University of Leuven, Herestraat 49 Box 521, 3000 Leuven, Belgium
| |
Collapse
|
13
|
Fritsche E, Volk HD, Reinke P, Abou-El-Enein M. Toward an Optimized Process for Clinical Manufacturing of CAR-Treg Cell Therapy. Trends Biotechnol 2020; 38:1099-1112. [DOI: 10.1016/j.tibtech.2019.12.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/21/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023]
|
14
|
Abstract
Head and neck structures govern the vital functions of breathing and swallowing. Additionally, these structures facilitate our sense of self through vocal communication, hearing, facial animation, and physical appearance. Loss of these functions can lead to loss of life or greatly affect quality of life. Regenerative medicine is a rapidly developing field that aims to repair or replace damaged cells, tissues, and organs. Although the field is largely in its nascence, regenerative medicine holds promise for improving on conventional treatments for head and neck disorders or providing therapies where no current standard exists. This review presents milestones in the research of regenerative medicine in head and neck surgery.
Collapse
Affiliation(s)
- Michael J McPhail
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Jeffrey R Janus
- Department of Otolaryngology - Head and Neck Surgery, Mayo Clinic Florida, Jacksonville, FL, USA
| | - David G Lott
- Head and Neck Regenerative Medicine Laboratory, Mayo Clinic Arizona, Scottsdale, AZ, USA
- Department of Otolaryngology - Head and Neck Surgery, Mayo Clinic Arizona, Phoenix, AZ, USA
| |
Collapse
|
15
|
Vollrath I, Mathaes R, Sediq AS, Jere D, Jörg S, Huwyler J, Mahler HC. Subvisible Particulate Contamination in Cell Therapy Products—Can We Distinguish? J Pharm Sci 2020; 109:216-219. [DOI: 10.1016/j.xphs.2019.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/30/2019] [Accepted: 09/06/2019] [Indexed: 12/31/2022]
|
16
|
Bailey SR, Maus MV. Gene editing for immune cell therapies. Nat Biotechnol 2019; 37:1425-1434. [DOI: 10.1038/s41587-019-0137-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 04/22/2019] [Indexed: 02/06/2023]
|
17
|
Quinn C, Young C, Thomas J, Trusheim M. Estimating the Clinical Pipeline of Cell and Gene Therapies and Their Potential Economic Impact on the US Healthcare System. VALUE IN HEALTH : THE JOURNAL OF THE INTERNATIONAL SOCIETY FOR PHARMACOECONOMICS AND OUTCOMES RESEARCH 2019; 22:621-626. [PMID: 31198178 DOI: 10.1016/j.jval.2019.03.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 02/05/2019] [Accepted: 03/28/2019] [Indexed: 06/09/2023]
Abstract
OBJECTIVES To estimate, at the indication level, durable gene and cellular therapy new product launches in the United States through 2030, and the number of treated patients. METHODS A statistical analysis of clinical trials pipeline data and disease incidence and prevalence was conducted to estimate the impact of new cell and gene therapies. We used Citeline's® Pharmaprojects® database to estimate the rates and timing of new product launches, on the basis of the phase of development, duration in phase, and probability of progression. Disease incidence and prevalence data were combined with estimates of market adoption to project the size of reimbursed patient populations. RESULTS We project that about 350 000 patients will have been treated with 30 to 60 products by 2030. About half the launches are expected to be in B-cell (CD-19) lymphomas and leukemias. CONCLUSIONS Cell and gene therapies promise durable clinical benefit from a single treatment course. High upfront reimbursement for these products means that the total costs could exceed what the healthcare system can manage. This creates a need for precision financing solutions and new reimbursement models that can ensure appropriate patient access to needed treatments, increase affordability for payers, and sustain private investment in innovation.
Collapse
Affiliation(s)
- Casey Quinn
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
| | - Colin Young
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Jonathan Thomas
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mark Trusheim
- Center for Biomedical Innovation, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Bandeiras C, Cabral JM, Finkelstein SN, Ferreira FC. Modeling biological and economic uncertainty on cell therapy manufacturing: the choice of culture media supplementation. Regen Med 2018; 13:917-933. [PMID: 30488770 DOI: 10.2217/rme-2018-0034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIM To evaluate the cost-effectiveness of autologous cell therapy manufacturing in xeno-free conditions. MATERIALS & METHODS Published data on the isolation and expansion of mesenchymal stem/stromal cells introduced donor, multipassage and culture media variability on cell yields and process times on adherent culture flasks to drive cost simulation of a scale-out campaign of 1000 doses of 75 million cells each in a 400 square meter Good Manufacturing Practices facility. RESULTS & CONCLUSION Passage numbers in the expansion step are strongly associated with isolation cell yield and drive cost increases per donor of $1970 and 2802 for fetal bovine serum and human platelet lysate. Human platelet lysate decreases passage numbers and process costs in 94.5 and 97% of donors through lower facility and labor costs. Cost savings are maintained with full equipment depreciation and higher numbers of cells per dose, highlighting the number of cells per passage step as the key cost driver.
Collapse
Affiliation(s)
- Cátia Bandeiras
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal.,Institute for Data, Systems & Society, Massachusetts Institute of Technology, 50 Ames Street, Cambridge MA 02139, USA.,Division of Clinical Informatics, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston MA 02215, USA
| | - Joaquim Ms Cabral
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal
| | - Stan N Finkelstein
- Institute for Data, Systems & Society, Massachusetts Institute of Technology, 50 Ames Street, Cambridge MA 02139, USA.,Division of Clinical Informatics, Department of Medicine, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston MA 02215, USA
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB - Institute for Bioengineering & Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisboa, Portugal.,The Discoveries Centre for Regenerative & Precision Medicine, Lisbon Campus, Universidade de Lisboa, Portugal
| |
Collapse
|
19
|
Migliori E, Chang M, Muranski P. Restoring antiviral immunity with adoptive transfer of ex-vivo generated T cells. Curr Opin Hematol 2018; 25:486-493. [PMID: 30281036 DOI: 10.1097/moh.0000000000000461] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Latent viruses such as cytomegalovirus (CMV), Epstein-Barr virus (EBV) and adenovirus (ADV) often reactivate in immunocompromised patients, contributing to poor clinical outcomes. A rapid reconstitution of antiviral responses via adoptive transfer of virus-specific T cells (VSTs) can prevent or eradicate even refractory infections. Here, we evaluate this strategy and the associated methodological, manufacturing and clinical advances. RECENT FINDINGS From the early pioneering but cumbersome efforts to isolate CMV-specific T cell clones, new approaches and techniques have been developed to provide quicker, safer and broader-aimed ex-vivo antigen-specific cells. New manufacturing strategies, such as the use of G-Rex flasks or 'priming' with a library of overlapping viral peptides, allow for culturing greater numbers of cells that could be patient-specific or stored in cell banks for off-the-shelf applications. Rapid isolation of T cells using major histocompatibility complex tetramer or cytokine capture approaches, or genetic reprogramming of cells to target viral antigens can accelerate the generation of potent cellular products. SUMMARY Advances in the ex-vivo generation of VSTs in academic medical centres and as off-the-shelf blood bank-based or commercially produced reagents are likely to result in broader accessibility and possible manufacturing cost reduction of these cell products, and will open new therapeutic prospects for vulnerable and critically ill immunocompromised patients.
Collapse
Affiliation(s)
- Edoardo Migliori
- Columbia Center for Translational Immunology (CCTI), Division of Hematology/Oncology, Columbia University Medical Center, New York, New York, USA
| | | | | |
Collapse
|
20
|
Teraa M, Gremmels H, Wijnand JGJ, Verhaar MC. Cell Therapy for Chronic Limb-Threatening Ischemia: Current Evidence and Future Directions. Stem Cells Transl Med 2018; 7:842-846. [PMID: 30070050 PMCID: PMC6265636 DOI: 10.1002/sctm.18-0025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 06/16/2018] [Accepted: 07/03/2018] [Indexed: 12/19/2022] Open
Abstract
Cell‐based therapies have gained interest as a potential treatment method in cardiovascular disease in the past two decades, peripheral artery disease amongst others. Initial pre‐clinical and small pilot clinical studies showed promising effects of cell therapy in peripheral artery disease and chronic limb‐threatening ischemia in particular. However, these promising results were not corroborated in larger high quality blinded randomized trials. This has led to a shift of the field towards more sophisticated cell products, especially mesenchymal stromal cells. Mesenchymal stromal cells have some important benefits, making these cells ideal for regenerative medicine, e.g., potential for allogeneic application, loss of disease‐mediated cell dysfunction, reduced production costs, off‐the‐shelf availability. Future high quality and large clinical studies have to prove the efficacy of mesenchymal stromal cells in the treatment of peripheral artery disease. Stem Cells Translational Medicine2018;7:842–846
Collapse
Affiliation(s)
- Martin Teraa
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Hendrik Gremmels
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joep G J Wijnand
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands.,Department of Vascular Surgery, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
21
|
Hampson G, Towse A, Pearson SD, Dreitlein WB, Henshall C. Gene therapy: evidence, value and affordability in the US health care system. J Comp Eff Res 2017; 7:15-28. [PMID: 29144165 DOI: 10.2217/cer-2017-0068] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIMS To explore the challenges presented by gene therapies, discuss potential solutions, and present policy recommendations. METHODS A review of the literature and series of expert interviews were conducted and discussed at a Policy Forum convened by The Institute for Clinical and Economic Review (ICER). The Policy Forum was attended by independent experts and senior representatives from 20 payer organizations and life sciences companies. RESULTS Three categories of challenges are identified: evidence generation, assessing value and affordability. Possible solutions and policy recommendations are presented for each of the three main categories of challenges. CONCLUSIONS Gene therapies present exciting opportunities, but also pose major challenges. Dialogue between manufacturers and payers around the issues and possible solutions is crucial.
Collapse
Affiliation(s)
| | | | | | | | - Chris Henshall
- Office of Health Economics, London, UK.,Independent Consultant, London, UK
| |
Collapse
|
22
|
Morrow D, Ussi A, Migliaccio G. Addressing Pressing Needs in the Development of Advanced Therapies. Front Bioeng Biotechnol 2017; 5:55. [PMID: 28993805 PMCID: PMC5622199 DOI: 10.3389/fbioe.2017.00055] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/06/2017] [Indexed: 12/16/2022] Open
Abstract
The commercial development of advanced therapy medicinal products (ATMPs) represents great opportunity for therapeutic innovation but is beset by many challenges for its developers. Although the ATMP field continues to progress at a rapid pace, evidenced by the increasing number of clinical trials conducted over the past few years, several factors continue to complicate the introduction of ATMPs as a curative treatment for multiple disease types, by blocking their translational pathway from research to the patient. While several recent publications (Trounson and McDonald, 2015; Abou-El-Enein et al., 2016a,b) as well as an Innovative Medicines Initiative consultation (IMI, 2016) this year have highlighted the major gaps in ATMP development, with manufacturing, regulatory, and reimbursement issues at the forefront, there remains to be formulated a coherent strategy to address these by bringing the relevant stakeholders to a single forum, whose task it would be to design and execute a delta plan to alleviate the most pressing bottlenecks. This article focuses on two of the most urgent areas in need of attention in ATMP development, namely manufacturing and reimbursement, and promotes the concept of innovation-dedicated research infrastructures to support a multi-sector approach for ensuring the successful development, entry, and ensuing survival of ATMPs in the healthcare market.
Collapse
Affiliation(s)
- David Morrow
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, Netherlands
| | - Anton Ussi
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, Netherlands
| | - Giovanni Migliaccio
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, Netherlands
| |
Collapse
|
23
|
Elsanhoury A, Sanzenbacher R, Reinke P, Abou-El-Enein M. Accelerating Patients' Access to Advanced Therapies in the EU. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:15-19. [PMID: 28971109 PMCID: PMC5609878 DOI: 10.1016/j.omtm.2017.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
- Ahmed Elsanhoury
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Germany
| | - Ralf Sanzenbacher
- Division of Medical Biotechnology, Paul-Ehrlich-Institut-Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Straße 51-59, Langen, 63225, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Germany.,Department of Nephrology and Internal Intensive Care, Charité - Universitätsmedizin Berlin, Germany
| | - Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Germany.,Department of Nephrology and Internal Intensive Care, Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
24
|
Davies BM, Smith J, Rikabi S, Wartolowska K, Morrey M, French A, MacLaren R, Williams D, Bure K, Pinedo-Villanueva R, Mathur A, Birchall M, Snyder E, Atala A, Reeve B, Brindley D. A quantitative, multi-national and multi-stakeholder assessment of barriers to the adoption of cell therapies. J Tissue Eng 2017; 8:2041731417724413. [PMID: 28835816 PMCID: PMC5557158 DOI: 10.1177/2041731417724413] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 07/14/2017] [Indexed: 01/20/2023] Open
Abstract
Cellular therapies, such as stem cell-based treatments, have been widely researched and numerous products and treatments have been developed. Despite this, there has been relatively limited use of these technologies in the healthcare sector. This study sought to investigate the perceived barriers to this more widespread adoption. An anonymous online questionnaire was developed, based on the findings of a pilot study. This was distributed to an audience of clinicians, researchers and commercial experts in 13 countries. The results were analysed for all respondents, and also sub-grouped by geographical region, and by profession of respondents. The results of the study showed that the most significant barrier was manufacturing, with other factors such as efficacy, regulation and cost-effectiveness being identified by the different groups. This study further demonstrates the need for these important issues to be addressed during the development of cellular therapies to enable more widespread adoption of these treatments.
Collapse
Affiliation(s)
- Benjamin M Davies
- Division of Trauma and Orthopaedic Surgery, Department of Surgery, University of Cambridge, Cambridge, UK.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.,The UCL-Oxford Centre for the Advancement of Sustainable Medical Innovation, University of Oxford, Oxford, UK
| | - James Smith
- The UCL-Oxford Centre for the Advancement of Sustainable Medical Innovation, University of Oxford, Oxford, UK
| | - Sarah Rikabi
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Karolina Wartolowska
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Mark Morrey
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Anna French
- The UCL-Oxford Centre for the Advancement of Sustainable Medical Innovation, University of Oxford, Oxford, UK
| | - Robert MacLaren
- Nuffield Laboratory of Ophthalmology, University of Oxford, Oxford, UK
| | - David Williams
- Centre for Biological Engineering, The Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Kim Bure
- Sartorius Stedim, Göttingen, Germany
| | - Rafael Pinedo-Villanueva
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK.,MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Anthony Mathur
- NIHR Cardiovascular Biomedical Research Unit, London Chest Hospital, London, UK.,Department of Cardiology, Barts Health NHS Trust, London, UK
| | - Martin Birchall
- Royal National Throat, Nose, and Ear Hospital, University College London, London, UK
| | - Evan Snyder
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Brock Reeve
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - David Brindley
- The UCL-Oxford Centre for the Advancement of Sustainable Medical Innovation, University of Oxford, Oxford, UK.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Paediatrics, University of Oxford, Oxford, UK.,Saïd Business School, University of Oxford, Oxford, UK.,Centre for Behavioural Medicine, UCL School of Pharmacy, University College London, London, UK
| |
Collapse
|
25
|
Abstract
While advanced therapy medicinal products offer great clinical promise, most EU-approved products have not achieved satisfactory commercial performance. Here we highlight a number of issues that prevent current products from obtaining commercial success and pitfalls that developers must overcome in future product development.
Collapse
|
26
|
Tran R, Myers DR, Denning G, Shields JE, Lytle AM, Alrowais H, Qiu Y, Sakurai Y, Li WC, Brand O, Le Doux JM, Spencer HT, Doering CB, Lam WA. Microfluidic Transduction Harnesses Mass Transport Principles to Enhance Gene Transfer Efficiency. Mol Ther 2017; 25:2372-2382. [PMID: 28780274 PMCID: PMC5628863 DOI: 10.1016/j.ymthe.2017.07.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 07/01/2017] [Accepted: 07/02/2017] [Indexed: 12/24/2022] Open
Abstract
Ex vivo gene therapy using lentiviral vectors (LVs) is a proven approach to treat and potentially cure many hematologic disorders and malignancies but remains stymied by cumbersome, cost-prohibitive, and scale-limited production processes that cannot meet the demands of current clinical protocols for widespread clinical utilization. However, limitations in LV manufacture coupled with inefficient transduction protocols requiring significant excess amounts of vector currently limit widespread implementation. Herein, we describe a microfluidic, mass transport-based approach that overcomes the diffusion limitations of current transduction platforms to enhance LV gene transfer kinetics and efficiency. This novel ex vivo LV transduction platform is flexible in design, easy to use, scalable, and compatible with standard cell transduction reagents and LV preparations. Using hematopoietic cell lines, primary human T cells, primary hematopoietic stem and progenitor cells (HSPCs) of both murine (Sca-1+) and human (CD34+) origin, microfluidic transduction using clinically processed LVs occurs up to 5-fold faster and requires as little as one-twentieth of LV. As an in vivo validation of the microfluidic-based transduction technology, HSPC gene therapy was performed in hemophilia A mice using limiting amounts of LV. Compared to the standard static well-based transduction protocols, only animals transplanted with microfluidic-transduced cells displayed clotting levels restored to normal.
Collapse
Affiliation(s)
- Reginald Tran
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - David R Myers
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | | | - Jordan E Shields
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Allison M Lytle
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Hommood Alrowais
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Yongzhi Qiu
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Yumiko Sakurai
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - William C Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - Oliver Brand
- School of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Joseph M Le Doux
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA
| | - H Trent Spencer
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, GA 30322, USA
| | - Christopher B Doering
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Graduate Program in Molecular and Systems Pharmacology, Laney Graduate School, Emory University, Atlanta, GA 30322, USA.
| | - Wilbur A Lam
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, Aflac Cancer Center and Blood Disorders Service of Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA 30322, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA 30332, USA.
| |
Collapse
|
27
|
Integration of a CD19 CAR into the TCR Alpha Chain Locus Streamlines Production of Allogeneic Gene-Edited CAR T Cells. Mol Ther 2017; 25:949-961. [PMID: 28237835 PMCID: PMC5383629 DOI: 10.1016/j.ymthe.2017.02.005] [Citation(s) in RCA: 208] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/05/2017] [Accepted: 02/06/2017] [Indexed: 11/25/2022] Open
Abstract
Adoptive cellular therapy using chimeric antigen receptor (CAR) T cell therapies have produced significant objective responses in patients with CD19+ hematological malignancies, including durable complete responses. Although the majority of clinical trials to date have used autologous patient cells as the starting material to generate CAR T cells, this strategy poses significant manufacturing challenges and, for some patients, may not be feasible because of their advanced disease state or difficulty with manufacturing suitable numbers of CAR T cells. Alternatively, T cells from a healthy donor can be used to produce an allogeneic CAR T therapy, provided the cells are rendered incapable of eliciting graft versus host disease (GvHD). One approach to the production of these cells is gene editing to eliminate expression of the endogenous T cell receptor (TCR). Here we report a streamlined strategy for generating allogeneic CAR T cells by targeting the insertion of a CAR transgene directly into the native TCR locus using an engineered homing endonuclease and an AAV donor template. We demonstrate that anti-CD19 CAR T cells produced in this manner do not express the endogenous TCR, exhibit potent effector functions in vitro, and mediate clearance of CD19+ tumors in an in vivo mouse model.
Collapse
|
28
|
The path to successful commercialization of cell and gene therapies: empowering patient advocates. Cytotherapy 2016; 19:293-298. [PMID: 27956199 DOI: 10.1016/j.jcyt.2016.10.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 10/28/2016] [Accepted: 10/31/2016] [Indexed: 12/31/2022]
Abstract
Often, novel gene and cell therapies provide hope for many people living with incurable diseases. To facilitate and accelerate a successful regulatory approval and commercialization path for effective, safe and affordable cell and gene therapies, the involvement of patient advocacy groups (PAGs) should be considered early in the development process. This report provides a thorough overview of the various roles PAGs play in the clinical translation of cell and gene therapies and how they can bring about positive changes in the regulatory process, infrastructure improvements and market stability.
Collapse
|
29
|
Abou-El-Enein M, Duda GN, Gruskin EA, Grainger DW. Strategies for Derisking Translational Processes for Biomedical Technologies. Trends Biotechnol 2016; 35:100-108. [PMID: 27499276 DOI: 10.1016/j.tibtech.2016.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/18/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022]
Abstract
Inefficient translational processes for technology-oriented biomedical research have led to some prominent and frequent failures in the development of many leading drug candidates, several designated investigational drugs, and some medical devices, as well as documented patient harm and postmarket product withdrawals. Derisking this process, particularly in the early stages, should increase translational efficiency and streamline resource utilization, especially in an academic setting. In this opinion article, we identify a 12-step guideline for reducing risks typically associated with translating medical technologies as they move toward prototypes, preclinical proof of concept, and possible clinical testing. Integrating the described 12-step process should prove valuable for improving how early-stage academic biomedical concepts are cultivated, culled, and manicured toward intended clinical applications.
Collapse
Affiliation(s)
- Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany.
| | - Georg N Duda
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; Julius Wolff Institute (JWI), Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | | | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84112, USA; Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|