1
|
Kong Y, Li J, Zhao X, Wu Y, Chen L. CAR-T cell therapy: developments, challenges and expanded applications from cancer to autoimmunity. Front Immunol 2025; 15:1519671. [PMID: 39850899 PMCID: PMC11754230 DOI: 10.3389/fimmu.2024.1519671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Chimeric Antigen Receptor (CAR)-T cell therapy has rapidly emerged as a groundbreaking approach in cancer treatment, particularly for hematologic malignancies. However, the application of CAR-T cell therapy in solid tumors remains challenging. This review summarized the development of CAR-T technologies, emphasized the challenges and solutions in CAR-T cell therapy for solid tumors. Also, key innovations were discussed including specialized CAR-T, combination therapies and the novel use of CAR-Treg, CAR-NK and CAR-M cells. Besides, CAR-based cell therapy have extended its reach beyond oncology to autoimmune disorders. We reviewed preclinical experiments and clinical trials involving CAR-T, Car-Treg and CAAR-T cell therapies in various autoimmune diseases. By highlighting these cutting-edge developments, this review underscores the transformative potential of CAR technologies in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Yanwei Wu
- School of Medicine, Shanghai University, Shanghai, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
2
|
Grace Tetteh M, Gupta S, Kumar M, Trollman H, Salonitis K, Jagtap S. Pharma 4.0: A deep dive top management commitment to successful Lean 4.0 implementation in Ghanaian pharma manufacturing sector. Heliyon 2024; 10:e36677. [PMID: 39296213 PMCID: PMC11408067 DOI: 10.1016/j.heliyon.2024.e36677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/09/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024] Open
Abstract
The primary aim of this study is to assess the significance of top management commitment in the context of Lean 4.0 implementation within the pharmaceutical manufacturing industry in Ghana. The study seeks to understand and evaluate the overall effectiveness and achievements associated with adopting Lean 4.0. Employing a positivist mindset, the research utilizes an explanatory quantitative research design and a survey technique. Data collected from 181 employees of pharmaceutical companies in Ghana undergo analysis using SmartPLS (version 4) and IBM SPSS version 26. The study employs a combination of descriptive statistics to summarise data characteristics and inferential statistics to test various hypotheses related to Lean 4.0 adoption. The analysis reveals that the successful integration of lean methods and Industry 4.0 technologies requires meticulous management. Simultaneously, individual implementations of lean principles and Industry 4.0 technologies positively impact business performance. Surprisingly, the study does not observe a substantial positive influence of Lean 4.0 on corporate performance, suggesting that immediate improvements in efficiency or profitability may not result from the adoption of this framework. This research contributes to the field by highlighting the need for careful management in integrating lean methods and Industry 4.0 technologies. It also emphasizes the positive impacts of lean principles and Industry 4.0 technology on business performance. The unexpected finding regarding the lack of immediate improvements in corporate efficiency or profitability with Lean 4.0 adoption prompts considerations of initial implementation challenges or the organization's need for time to adapt to this integrated approach.
Collapse
Affiliation(s)
- Michelle Grace Tetteh
- Sustainable Manufacturing Systems Centre, Cranfield University, Cranfield, MK43 0AL, UK
| | - Sumit Gupta
- Department of Mechanical Engineering, A.S.E.T., Amity University, Uttar Pradesh, Noida, 201313, India
| | - Mukesh Kumar
- National Institute of Technology Patna, Patna, 800005, India
| | - Hana Trollman
- School of Business, University of Leicester, Leicester, LE2 1RQ, UK
| | | | - Sandeep Jagtap
- Sustainable Manufacturing Systems Centre, Cranfield University, Cranfield, MK43 0AL, UK
- Department of Industrial Management and Logistics, Faculty of Engineering, Lund University, Lund, Sweden
| |
Collapse
|
3
|
Zhen J, Li X, Yu H, Du B. High-density lipoprotein mimetic nano-therapeutics targeting monocytes and macrophages for improved cardiovascular care: a comprehensive review. J Nanobiotechnology 2024; 22:263. [PMID: 38760755 PMCID: PMC11100215 DOI: 10.1186/s12951-024-02529-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/03/2024] [Indexed: 05/19/2024] Open
Abstract
The prevalence of cardiovascular diseases continues to be a challenge for global health, necessitating innovative solutions. The potential of high-density lipoprotein (HDL) mimetic nanotherapeutics in the context of cardiovascular disease and the intricate mechanisms underlying the interactions between monocyte-derived cells and HDL mimetic showing their impact on inflammation, cellular lipid metabolism, and the progression of atherosclerotic plaque. Preclinical studies have demonstrated that HDL mimetic nanotherapeutics can regulate monocyte recruitment and macrophage polarization towards an anti-inflammatory phenotype, suggesting their potential to impede the progression of atherosclerosis. The challenges and opportunities associated with the clinical application of HDL mimetic nanotherapeutics, emphasize the need for additional research to gain a better understanding of the precise molecular pathways and long-term effects of these nanotherapeutics on monocytes and macrophages to maximize their therapeutic efficacy. Furthermore, the use of nanotechnology in the treatment of cardiovascular diseases highlights the potential of nanoparticles for targeted treatments. Moreover, the concept of theranostics combines therapy and diagnosis to create a selective platform for the conversion of traditional therapeutic medications into specialized and customized treatments. The multifaceted contributions of HDL to cardiovascular and metabolic health via highlight its potential to improve plaque stability and avert atherosclerosis-related problems. There is a need for further research to maximize the therapeutic efficacy of HDL mimetic nanotherapeutics and to develop targeted treatment approaches to prevent atherosclerosis. This review provides a comprehensive overview of the potential of nanotherapeutics in the treatment of cardiovascular diseases, emphasizing the need for innovative solutions to address the challenges posed by cardiovascular diseases.
Collapse
Affiliation(s)
- Juan Zhen
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiangjun Li
- School of Pharmaceutical Science, Jilin University, Changchun, 130021, China
| | - Haitao Yu
- The First Hospital of Jilin University, Changchun, 130021, China
| | - Bing Du
- The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
4
|
Shokati A, Naser Moghadasi A, Ghashghaei A, Sahraian MA, Chahardouli B, Mousavi SA, Ai J, Nikbakht M. Good manufacturing practices production of human placental derived mesenchymal stem cells for therapeutic applications: focus on multiple sclerosis. Mol Biol Rep 2024; 51:460. [PMID: 38551770 DOI: 10.1007/s11033-024-09372-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Among neurological diseases, multiple sclerosis (MS) affects mostly young adults and can cause long-term disability. While most medications with approval from regulatory agencies are very effective in treating MS disease, they are unable to repair the tissue damage found in the central nervous system (CNS). Consequently, Cell-based therapy particularly using mesenchymal stem/stromal cells (MSCs), holds promise for neuroprotection and tissue repair in MS treatment. Furthermore, placenta-derived MSCs (PLMSCs) have shown the potential to treat MS due to their abundance, noninvasive isolation from discarded tissues, no ethical problems, anti-inflammatory, and reparative properties. Accordingly, good manufacturing practices (GMPs) plays a crucial part in clinical SCs manufacturing. The purpose of our article is to discuss GMP-grade PLMSC protocols for treating MS as well as other clinical applications. METHODS AND RESULTS Placental tissue obtained of a healthy donor during the caesarean delivery and PLMSCs isolated by GMP standards. Flow cytometry was used to assess the expression of the CD markers CD34, CD105, CD90, and CD73 in the MSCs and the mesodermal differentiation ability was evaluated. Furthermore, Genetic evaluation of PLMSCs was done by G-banded karyotyping and revealed no chromosomal instability. In spite of the anatomical origin of the starting material, PLMSCs using this method of culture were maternal in origin. CONCLUSIONS We hope that our protocol for clinical manufacturing of PLMSCs according to GMP standards will assist researchers in isolating MSCs from placental tissue for clinical and pre-clinical applications.
Collapse
Affiliation(s)
- Ameneh Shokati
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Abdorreza Naser Moghadasi
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Andisheh Ghashghaei
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Sahraian
- Multiple Sclerosis Research Center, Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Bahram Chahardouli
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Asadollah Mousavi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohsen Nikbakht
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Research Institute for Oncology, Hematology and Cell Therapy, Shariati Hospital, Tehran University of Medical Sciences, Kargar Shomali Street, P. O. Box.: 1411713131, Tehran, Iran.
| |
Collapse
|
5
|
Sall IM, Flaviu TA. Plant and mammalian-derived extracellular vesicles: a new therapeutic approach for the future. Front Bioeng Biotechnol 2023; 11:1215650. [PMID: 37781539 PMCID: PMC10534050 DOI: 10.3389/fbioe.2023.1215650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 08/16/2023] [Indexed: 10/03/2023] Open
Abstract
Background: In recent years, extracellular vesicles have been recognized as important mediators of intercellular communication through the transfer of active biomolecules (proteins, lipids, and nucleic acids) across the plant and animal kingdoms and have considerable roles in several physiological and pathological mechanisms, showing great promise as new therapeutic strategies for a variety of pathologies. Methods: In this study, we carefully reviewed the numerous articles published over the last few decades on the general knowledge of extracellular vesicles, their application in the therapy of various pathologies, and their prospects as an approach for the future. Results: The recent discovery and characterization of extracellular vesicles (EVs) of diverse origins and biogenesis have altered the current paradigm of intercellular communication, opening up new diagnostic and therapeutic perspectives. Research into these EVs released by plant and mammalian cells has revealed their involvement in a number of physiological and pathological mechanisms, such as embryonic development, immune response, tissue regeneration, and cancer. They are also being studied as potential biomarkers for disease diagnosis and vectors for drug delivery. Conclusion: Nanovesicles represent powerful tools for intercellular communication and the transfer of bioactive molecules. Their molecular composition and functions can vary according to their origin (plant and mammalian), so their formation, composition, and biological roles open the way to therapeutic applications in a variety of pathologies, which is arousing growing interest in the scientific community. Clinical Trial Registration: ClinicalTrials.gov identifier: NCT03608631.
Collapse
Affiliation(s)
| | - Tabaran Alexandru Flaviu
- Department of Anatomic Pathology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca, Romania
| |
Collapse
|
6
|
Shah M, Krull A, Odonnell L, de Lima MJ, Bezerra E. Promises and challenges of a decentralized CAR T-cell manufacturing model. FRONTIERS IN TRANSPLANTATION 2023; 2:1238535. [PMID: 38993860 PMCID: PMC11235344 DOI: 10.3389/frtra.2023.1238535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 07/13/2024]
Abstract
Autologous chimeric antigen receptor-modified T-cell (CAR T) products have demonstrated un-precedent efficacy in treating many relapsed/refractory B-cell and plasma cell malignancies, leading to multiple commercial products now in routine clinical use. These positive responses to CAR T therapy have spurred biotech and big pharma companies to evaluate innovative production methods to increase patient access while maintaining adequate quality control and profitability. Autologous cellular therapies are, by definition, manufactured as single patient batches, and demand has soared for manufacturing facilities compliant with current Good Manufacturing Practice (cGMP) regulations. The use of a centralized production model is straining finite resources even in developed countries in North America and the European Union, and patient access is not feasible for most of the developing world. The idea of having a more uniform availability of these cell therapy products promoted the concept of point-of-care (POC) manufacturing or decentralized in-house production. While this strategy can potentially decrease the cost of manufacturing, the challenge comes in maintaining the same quality as currently available centrally manufactured products due to the lack of standardized manufacturing techniques amongst institutions. However, academic medical institutions and biotech companies alike have forged ahead innovating and adopting new technologies to launch clinical trials of CAR T products produced exclusively in-house. Here we discuss POC production of CAR T products.
Collapse
Affiliation(s)
- Manan Shah
- Department of Hematology, the James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, United States
| | - Ashley Krull
- Department of Cell Therapy Manufacturing and Engineering, the James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, United States
| | - Lynn Odonnell
- Department of Hematology, Cellular Therapy Lab, the James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, United States
| | - Marcos J. de Lima
- Department of Hematology, The James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, United States
| | - Evandro Bezerra
- Department of Hematology, The James Cancer Hospital and Solove Research Institute, Ohio State University, Columbus, OH, United States
| |
Collapse
|
7
|
Ciccocioppo R, Guadalajara H, Astori G, Carlino G, García-Olmo D. Misconceptions, hurdles and recommendations regarding the use of mesenchymal stem/stromal cells in perianal Crohn disease. Cytotherapy 2023; 25:230-234. [PMID: 36543715 DOI: 10.1016/j.jcyt.2022.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 10/14/2022] [Accepted: 11/26/2022] [Indexed: 12/24/2022]
Affiliation(s)
- Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy.
| | - Hector Guadalajara
- Division of Surgery and Cell Therapy Unit, Institute for Health Research, Jiménez Díaz Foundation University Hospital, Madrid, Spain
| | - Giuseppe Astori
- Laboratory of Advanced Cellular Therapies, Hematology Unit, Vicenza Hospital, Vicenza, Italy
| | - Giorgio Carlino
- Gastroenterology Unit, Department of Medicine, Azienda Ospedaliera Universitaria Integrata Policlinico G.B. Rossi and University of Verona, Verona, Italy
| | - Damián García-Olmo
- Division of Surgery and Cell Therapy Unit, Institute for Health Research, Jiménez Díaz Foundation University Hospital, Madrid, Spain
| |
Collapse
|
8
|
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
|
9
|
Syzdykova L, Zauatbayeva G, Keyer V, Ramanculov Y, Arsienko R, Shustov AV. Process for production of chimeric antigen receptor-transducing lentivirus particles using infection with replicon particles containing self-replicating RNAs. Biochem Eng J 2023. [DOI: 10.1016/j.bej.2023.108814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
10
|
Towards sustainability and affordability of expensive cell and gene therapies? Applying a cost-based pricing model to estimate prices for Libmeldy and Zolgensma. Cytotherapy 2022; 24:1245-1258. [PMID: 36216697 DOI: 10.1016/j.jcyt.2022.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 09/01/2022] [Accepted: 09/01/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND AIMS Drug prices are regarded as one of the most influential factors in determining accessibility and affordability to novel therapies. Cell and gene therapies such as OTL-200 (brand name: Libmeldy) and AVXS-101 (brand name: Zolgensma) with (expected) list prices of 3.0 million EUR and 1.9 million EUR per treatment, respectively, spark a global debate on the affordability of such therapies. The aim of this study was to use a recently published cost-based pricing model to calculate prices for cell and gene therapies, with OTL-200 and AVXS-101 as case study examples. METHODS Using the pricing model proposed by Uyl-de Groot and Löwenberg, we estimated a price for both therapies. We searched the literature and online public sources to estimate (i) research and development (R&D) expenses adjusted for risk of failure and cost of capital, (ii) the eligible patient population and (iii) costs of drug manufacturing to calculate a base-case price for OTL-200 and AVXS-101. All model input parameters were varied in a stepwise, deterministic sensitivity analysis and scenario analyses to assess their impact on the calculated prices. RESULTS Prices for OTL-200 and AVXS-101 were estimated at 1 048 138 EUR and 380 444 EUR per treatment, respectively. In deterministic sensitivity analyses, varying R&D estimates had the greatest impact on the price for OTL-200, whereas for AVXS-101, changes in the profit margin changed the calculated price substantially. Highest prices in scenario analyses were achieved when assuming the lowest number of patients for OTL-200 and highest R&D expenses for AVXS-101. The lowest R&D expenses scenario resulted in lowest prices for either therapy. CONCLUSIONS Our results show that, using the proposed model, prices for both OTL-200 and AVXS-101 lie substantially below the currently (proposed) list prices for both therapies. Nevertheless, the uncertainty of the used model input parameters is considerable, which translates in a wide range of estimated prices. This is mainly because of a lack of transparency from pharmaceutical companies regarding R&D expenses and the costs of drug manufacturing. Simultaneously, the disease indications for both therapies remain heavily understudied in terms of their epidemiological profile. Despite the considerable variation in the estimated prices, our results may support the public debate on value-based and cost-based pricing models, and on "fair" drug prices in general.
Collapse
|
11
|
Rezaie J, Feghhi M, Etemadi T. A review on exosomes application in clinical trials: perspective, questions, and challenges. Cell Commun Signal 2022; 20:145. [PMID: 36123730 PMCID: PMC9483361 DOI: 10.1186/s12964-022-00959-4] [Citation(s) in RCA: 250] [Impact Index Per Article: 83.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/16/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Exosomes are progressively known as significant mediators of cell-to-cell communication. They convey active biomolecules to target cells and have vital functions in several physiological and pathological processes, and show substantial promise as novel treatment strategies for diseases. METHODS In this review study, we studied numerous articles over the past two decades published on application of exosomes in different diseases as well as on perspective and challenges in this field. RESULTS The main clinical application of exosomes are using them as a biomarker, cell-free therapeutic agents, drug delivery carriers, basic analysis for exosome kinetics, and cancer vaccine. Different exosomes from human or plant sources are utilized in various clinical trials. Most researchers used exosomes from the circulatory system for biomarker experiments. Mesenchymal stem cells (MSCs) and dendritic cells (DCs) are two widely held cell sources for exosome use. MSCs-derived exosomes are commonly used for inflammation treatment and drug delivery, while DCs-exosomes are used to induce inflammation response in cancer patients. However, the clinical application of exosomes faces various questions and challenges. In addition, translation of exosome-based clinical trials is required to conform to specific good manufacturing practices (GMP). In this review, we summarize exosomes in the clinical trials according to the type of application and disease. We also address the main questions and challenges regarding exosome kinetics and clinical applications. CONCLUSIONS Exosomes are promising platforms for treatment of many diseases in clinical trials. This exciting field is developing hastily, understanding of the underlying mechanisms that direct the various observed roles of exosomes remains far from complete and needs further multidisciplinary research in working with these small vesicles. Video Abstract.
Collapse
Affiliation(s)
- Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St, Ershad Blvd., P.O. BoX: 1138, Urmia, 57147, Iran.
| | - Maryam Feghhi
- Institute of Molecular Biophysics, Florida State University, Florida, USA
| | - Tahereh Etemadi
- Department of Biology, Faculty of Science, Arak University, Arak, Iran
| |
Collapse
|
12
|
Lee E, Shah D, Porteus M, Wright JF, Bacchetta R. Design of experiments as a decision tool for cell therapy manufacturing. Cytotherapy 2022; 24:590-596. [PMID: 35227602 DOI: 10.1016/j.jcyt.2022.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND AIMS Cell therapies are costlier to manufacture than small molecules and protein therapeutics because they require multiple manipulations and are often produced in an autologous manner. Strategies to lower the cost of goods to produce a cell therapy could make a significant impact on its total cost. METHODS Borrowing from the field of bioprocess development, the authors took a design of experiments (DoE)-based approach to understanding the manufacture of a cell therapy product in pre-clinical development, analyzing main cost factors in the production process. The cells used for these studies were autologous CD4+ T lymphocytes gene-edited using CRISPR/Cas9 and recombinant adeno-associated virus (AAV) to restore normal FOXP3 gene expression as a prospective investigational product for patients with immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. RESULTS Using gene editing efficiency as the response variable, an initial screen was conducted for other variables that could influence the editing frequency. The multiplicity of infection (MOI) of AAV and amount of single guide RNA (sgRNA) were the significant factors used for the optimization step to generate a response contour plot. Cost analysis was done for multiple points in the design space to find cost drivers that could be reduced. For the range of values tested (50 000-750 000 vg/cell AAV and 0.8-4 μg sgRNA), editing with the highest MOI and sgRNA yielded the best gene editing frequency. However, cost analysis showed the optimal solution was gene editing at 193 000 vg/cell AAV and 1.78 μg sgRNA. CONCLUSIONS The authors used DoE to define key factors affecting the gene editing process for a potential investigational therapeutic, providing a novel and faster data-based approach to understanding factors driving complex biological processes. This approach could be applied in process development and aid in achieving more robust strategies for the manufacture of cellular therapeutics.
Collapse
Affiliation(s)
- Esmond Lee
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | | | - Matthew Porteus
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA; Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - J Fraser Wright
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA; Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rosa Bacchetta
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA; Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
13
|
Shin H, Kim E. Analysis of GMP for marketing authorization of ATMPs: comparison in the US, the EU, Japan and South Korea. Regen Med 2022; 17:283-297. [PMID: 35232285 DOI: 10.2217/rme-2021-0153] [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: This study compared regulatory systems of competent authorities related to GMP for marketing authorization of advanced therapy medicinal products (ATMPs). Methods: Dossiers for GMP and regulations and guidelines for facilities and equipment were analyzed using gap analysis. The risk-based approach (RBA) and GMP inspection were evaluated with regulations and guidelines. Results: The dossier was similar for the competent authorities. However, whereas a site master file is required in the EU, Japan and South Korea, the US requires only a biologics license application. The regulations and guidelines of facilities and equipment emphasized preventing contamination. There are differences among the competent authorities in GMP inspection and RBAs. Conclusion: Differences among the competent authorities in the marketing authorization process related to GMP for ATMPs should be considered.
Collapse
Affiliation(s)
- Hocheol Shin
- Department of Pharmaceutical Industry, Chung-Ang University, Seoul, 06974, South Korea
| | - Eunyoung Kim
- Department of Pharmaceutical Industry, Chung-Ang University, Seoul, 06974, South Korea.,Clinical Data Analysis, Evidence-Based Clinical Research Laboratory, Departments of Health Science & Clinical Pharmacy, College of Pharmacy, Chung-Ang University, Seoul, 06974, South Korea
| |
Collapse
|
14
|
Hole G, Hole AS, McFalone-Shaw I. Digitalization in pharmaceutical industry: What to focus on under the digital implementation process? Int J Pharm X 2021; 3:100095. [PMID: 34712948 PMCID: PMC8528719 DOI: 10.1016/j.ijpx.2021.100095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 11/22/2022] Open
Abstract
Digitalization of any manufacture industry is a key step in any progress of the production process. The process of digitalization includes both increased use of robotics, automatization solutions and computerization, thereby allowing to reduce costs, to improve efficiency and productivity, and to be flexible to changes. Pharmaceutical Industry (PI) has however been resistant to digitalization, mainly due to fair experience and complexity of the entailed development and manufacture processes. Nevertheless, there is a clear need to digitalize PI as the demand in both traditional and new drugs is constantly growing. Contract Development Manufacture Organizations (CDMOs) have a special digitalizing challenge. Digitalization of PI, and CDMO precisely, should be tightly related to the main aspects of Good Manufacture Practice (GMP), and, to succeed in PI digitalizing requires constant focus on GMP. Close collaboration with constantly changing stakeholders is another important factor which should be in focus during digitalization of CDMO. This paper represents an overview over the main aspects of CDMO digitalization and discusses both the opportunities and challenges of the process, focusing on the practical solutions for successive digital implementation.
Collapse
Key Words
- AIDS, Acquired Immune Deficiency Syndrome
- CDMO, Contract Development and Manufacturing Organization
- Contract development manufacture organization
- Digitalization
- EMA, European Medicines Agency
- EU, European Union
- FDA, Food and Drug Administration
- GMP, Good Manufacturing Practice
- ITA., International Trade Administration
- MHRA, Medicines and Healthcare Products Regulatory Agency
- PAI, Pre-Approval Inspections
- PI, Pharmaceutical Industry
- Pharmaceutical industry
- Process improvements
- TDM, Traditional Drug Manufacturing
- USD, United States Dollars
Collapse
Affiliation(s)
- Glenn Hole
- Molde University College, Molde and Procuratio Consulting, Drammen, Norway
| | | | - Ian McFalone-Shaw
- Molde University College, Molde and Procuratio Consulting, Drammen, Norway
| |
Collapse
|
15
|
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: 115] [Impact Index Per Article: 28.8] [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
|
16
|
Lam C, Meinert E, Yang A, Cui Z. Comparison between centralized and decentralized supply chains of autologous chimeric antigen receptor T-cell therapies: a UK case study based on discrete event simulation. Cytotherapy 2021; 23:433-451. [PMID: 33674239 DOI: 10.1016/j.jcyt.2020.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/21/2020] [Accepted: 08/16/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND AIMS Decentralized, or distributed, manufacturing that takes place close to the point of care has been a manufacturing paradigm of heightened interest within the cell therapy domain because of the product's being living cell material as well as the need for a highly monitored and temperature-controlled supply chain that has the potential to benefit from close proximity between manufacturing and application. METHODS To compare the operational feasibility and cost implications of manufacturing autologous chimeric antigen receptor T (CAR T)-cell products between centralized and decentralized schemes, a discrete event simulation model was built using ExtendSIM 9 for simulating the patient-to-patient supply chain, from the collection of patient cells to the final administration of CAR T therapy in hospitals. Simulations were carried out for hypothetical systems in the UK using three demand levels-low (100 patients per annum), anticipated (200 patients per annum) and high (500 patients per annum)-to assess resource allocation, cost per treatment and system resilience to demand changes and to quantify the risks of mix-ups within the supply chain for the delivery of CAR T treatments. RESULTS The simulation results show that although centralized manufacturing offers better economies of scale, individual facilities in a decentralized system can spread facility costs across a greater number of treatments and better utilize resources at high demand levels (annual demand of 500 patients), allowing for an overall more comparable cost per treatment. In general, raw material and consumable costs have been shown to be one of the greatest cost drivers, and genetic modification-associated costs have been shown to account for over one third of raw material and consumable costs. Turnaround time per treatment for the decentralized scheme is shown to be consistently lower than its centralized counterpart, as there is no need for product freeze-thaw, packaging and transportation, although the time savings is shown to be insignificant in the UK case study because of its rather compact geographical setting with well-established transportation networks. In both schemes, sterility testing lies on the critical path for treatment delivery and is shown to be critical for treatment turnaround time reduction. CONCLUSIONS Considering both cost and treatment turnaround time, point-of-care manufacturing within the UK does not show great advantages over centralized manufacturing. However, further simulations using this model can be used to understand the feasibility of decentralized manufacturing in a larger geographical setting.
Collapse
Affiliation(s)
- Ching Lam
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK
| | - Edward Meinert
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Zhanfeng Cui
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, UK.
| |
Collapse
|
17
|
Comisel RM, Kara B, Fiesser FH, Farid SS. Lentiviral vector bioprocess economics for cell and gene therapy commercialization. Biochem Eng J 2021. [DOI: 10.1016/j.bej.2020.107868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Estimation of manufacturing development costs of cell-based therapies: a feasibility study. Cytotherapy 2021; 23:730-739. [PMID: 33593688 DOI: 10.1016/j.jcyt.2020.12.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 11/23/2022]
Abstract
BACKGROUND AIMS Cell-based therapies (CBTs) provide opportunities to treat rare and high-burden diseases. Manufacturing development of these innovative products is said to be complex and costly. However, little research is available providing insight into resource use and cost drivers. Therefore, this study aimed to assess the feasibility of estimating the cost of manufacturing development of two cell-based therapy case studies using a CBT cost framework specifically designed for small-scale cell-based therapies. METHODS A retrospective costing study was conducted in which the cost of developing an adoptive immunotherapy of Epstein-Barr virus-specific cytotoxic T lymphocytes (CTLs) and a pluripotent stem cell (PSC) master cell bank was estimated. Manufacturing development was defined as products advancing from technology readiness level 3 to 6. The study was conducted in a Scottish facility. Development steps were recreated via developer focus groups. Data were collected from facility administrative and financial records and developer interviews. RESULTS Application of the manufacturing cost framework to retrospectively estimate the manufacturing design cost of two case studies in one Scottish facility appeared feasible. Manufacturing development cost was estimated at £1,201,016 for CTLs and £494,456 for PSCs. Most costs were accrued in the facility domain (56% and 51%), followed by personnel (20% and 32%), materials (19% and 15%) and equipment (4% and 2%). CONCLUSIONS Based on this study, it seems feasible to retrospectively estimate resources consumed in manufacturing development of cell-based therapies. This fosters inclusion of cost in the formulation and dissemination of best practices to facilitate early and sustainable patient access and inform future cost-conscious manufacturing design decisions.
Collapse
|
19
|
Roemhild A, Otto NM, Moll G, Abou-El-Enein M, Kaiser D, Bold G, Schachtner T, Choi M, Oellinger R, Landwehr-Kenzel S, Juerchott K, Sawitzki B, Giesler C, Sefrin A, Beier C, Wagner DL, Schlickeiser S, Streitz M, Schmueck-Henneresse M, Amini L, Stervbo U, Babel N, Volk HD, Reinke P. Regulatory T cells for minimising immune suppression in kidney transplantation: phase I/IIa clinical trial. BMJ 2020; 371:m3734. [PMID: 33087345 PMCID: PMC7576328 DOI: 10.1136/bmj.m3734] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To assess whether reshaping of the immune balance by infusion of autologous natural regulatory T cells (nTregs) in patients after kidney transplantation is safe, feasible, and enables the tapering of lifelong high dose immunosuppression, with its limited efficacy, adverse effects, and high direct and indirect costs, along with addressing several key challenges of nTreg treatment, such as easy and robust manufacturing, danger of over immunosuppression, interaction with standard care drugs, and functional stability in an inflammatory environment in a useful proof-of-concept disease model. DESIGN Investigator initiated, monocentre, nTreg dose escalation, phase I/IIa clinical trial (ONEnTreg13). SETTING Charité-University Hospital, Berlin, Germany, within the ONE study consortium (funded by the European Union). PARTICIPANTS Recipients of living donor kidney transplant (ONEnTreg13, n=11) and corresponding reference group trial (ONErgt11-CHA, n=9). INTERVENTIONS CD4+ CD25+ FoxP3+ nTreg products were given seven days after kidney transplantation as one intravenous dose of 0.5, 1.0, or 2.5-3.0×106 cells/kg body weight, with subsequent stepwise tapering of triple immunosuppression to low dose tacrolimus monotherapy until week 48. MAIN OUTCOME MEASURES The primary clinical and safety endpoints were assessed by a composite endpoint at week 60 with further three year follow-up. The assessment included incidence of biopsy confirmed acute rejection, assessment of nTreg infusion related adverse effects, and signs of over immunosuppression. Secondary endpoints addressed allograft functions. Accompanying research included a comprehensive exploratory biomarker portfolio. RESULTS For all patients, nTreg products with sufficient yield, purity, and functionality could be generated from 40-50 mL of peripheral blood taken two weeks before kidney transplantation. None of the three nTreg dose escalation groups had dose limiting toxicity. The nTreg and reference groups had 100% three year allograft survival and similar clinical and safety profiles. Stable monotherapy immunosuppression was achieved in eight of 11 (73%) patients receiving nTregs, while the reference group remained on standard dual or triple drug immunosuppression (P=0.002). Mechanistically, the activation of conventional T cells was reduced and nTregs shifted in vivo from a polyclonal to an oligoclonal T cell receptor repertoire. CONCLUSIONS The application of autologous nTregs was safe and feasible even in patients who had a kidney transplant and were immunosuppressed. These results warrant further evaluation of Treg efficacy and serve as the basis for the development of next generation nTreg approaches in transplantation and any immunopathologies. TRIAL REGISTRATION NCT02371434 (ONEnTreg13) and EudraCT:2011-004301-24 (ONErgt11).
Collapse
Affiliation(s)
- Andy Roemhild
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Natalie Maureen Otto
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Guido Moll
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mohamed Abou-El-Enein
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Daniel Kaiser
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Gantuja Bold
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Schachtner
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Oellinger
- Department of Abdominal and Transplant Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sybille Landwehr-Kenzel
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Karsten Juerchott
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Cordula Giesler
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anett Sefrin
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Carola Beier
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Dimitrios Laurin Wagner
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Stephan Schlickeiser
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Streitz
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schmueck-Henneresse
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Leila Amini
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Ulrik Stervbo
- Medical Department 1, University hospitals of the Ruhr University of Bochum, Herne, Germany
| | - Nina Babel
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Medical Department 1, University hospitals of the Ruhr University of Bochum, Herne, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
20
|
Iancu EM, Kandalaft LE. Challenges and advantages of cell therapy manufacturing under Good Manufacturing Practices within the hospital setting. Curr Opin Biotechnol 2020; 65:233-241. [DOI: 10.1016/j.copbio.2020.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 01/06/2023]
|
21
|
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]
|
22
|
What does cell therapy manufacturing cost? A framework and methodology to facilitate academic and other small-scale cell therapy manufacturing costings. Cytotherapy 2020; 22:388-397. [DOI: 10.1016/j.jcyt.2020.03.432] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 12/26/2022]
|
23
|
Abstract
Schwann cells as glial cells in the peripheral nervous system can participate in neurons protection and forming myelin. Additionally, they are important for nerve pulse conduction supporting along axons. On the other hand, it was demonstrated that they are promising cells for the treatment of demyelinating disorders and also central nervous system damages. Herein, for therapeutic application, Schwann cells should be manufactured based on good manufacturing practice standards to achieve safe and effective clinical products. In this respect, the current chapter tries to introduce a standard protocol for manufacturing of human GMP-compliant Schwann cells for clinical application.
Collapse
|
24
|
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]
|
25
|
Ebrahimi-Barough S, Ai J, Payab M, Alavi-Moghadam S, Shokati A, Aghayan HR, Larijani B, Arjmand B. Standard Operating Procedure for the Good Manufacturing Practice-Compliant Production of Human Endometrial Stem Cells for Multiple Sclerosis. Methods Mol Biol 2020; 2286:199-212. [PMID: 32504294 DOI: 10.1007/7651_2020_281] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is the most common cause of neurological diseases. Although, there are some effective medications with regulatory approval for treating MS, they are only partially effective and cannot promote repairing of tissue damage directly which occurs in the central nervous system. Therefore, there is an essential need to develop novel therapeutic approaches for neuroprotection or repairing damaged tissue in MS. Accordingly, cell-based therapies as a novel therapeutic strategy have opened a new horizon in treatment of MS. Each setting in cell therapy has potential benefits. Human endometrial stem cells as an invaluable source for cell therapy have introduced treatment for MS. In this respect, good manufacturing practice (GMP) has a pivotal role in clinical production of stem cells. This chapter tries to describe the protocol of GMP-grade endometrial stem cells for treatment of MS.
Collapse
Affiliation(s)
- Somayeh Ebrahimi-Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Moloud Payab
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ameneh Shokati
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran. .,Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
26
|
A multiscale simulation framework for the manufacturing facility and supply chain of autologous cell therapies. Cytotherapy 2019; 21:1081-1093. [DOI: 10.1016/j.jcyt.2019.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 06/02/2019] [Accepted: 07/15/2019] [Indexed: 02/02/2023]
|
27
|
Bak A, Friis KP, Wu Y, Ho RJ. Translating Cell and Gene Biopharmaceutical Products for Health and Market Impact. Product Scaling From Clinical to Marketplace: Lessons Learned and Future Outlook. J Pharm Sci 2019; 108:3169-3175. [DOI: 10.1016/j.xphs.2019.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 01/28/2023]
|
28
|
Ambekar RS, Kandasubramanian B. Progress in the Advancement of Porous Biopolymer Scaffold: Tissue Engineering Application. Ind Eng Chem Res 2019. [DOI: 10.1021/acs.iecr.8b05334] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Rushikesh S. Ambekar
- Rapid Prototype & Electrospinning Lab, Department of Metallurgical and Materials Engineering, DIAT (DU), Ministry of Defence, Girinagar, Pune 411025, India
| | - Balasubramanian Kandasubramanian
- Rapid Prototype & Electrospinning Lab, Department of Metallurgical and Materials Engineering, DIAT (DU), Ministry of Defence, Girinagar, Pune 411025, India
| |
Collapse
|
29
|
Kurtz A, Elsallab M, Sanzenbacher R, Abou-El-Enein M. Linking Scattered Stem Cell-Based Data to Advance Therapeutic Development. Trends Mol Med 2019; 25:8-19. [DOI: 10.1016/j.molmed.2018.10.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 02/07/2023]
|
30
|
Challenges in Advanced Therapy Medicinal Product Development: A Survey among Companies in Europe. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:121-130. [PMID: 30456217 PMCID: PMC6234262 DOI: 10.1016/j.omtm.2018.10.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/01/2018] [Indexed: 01/13/2023]
Abstract
Advanced therapy medicinal products (ATMPs) hold promise as treatments for previously untreatable and high-burden diseases. Expectations are high and active company pipelines are observed, yet only 10 market authorizations were approved in Europe. Our aim was to identify challenges experienced in European ATMP clinical development by companies. A survey-based cohort study was conducted among commercial ATMP developers. Respondents shared challenges experienced during various development phases, as well as developer and product characteristics. Descriptions of challenges were grouped in domains (clinical, financial, human resource management, regulatory, scientific, technical, other) and further categorized using thematic content analysis. A descriptive analysis was performed. We invited 271 commercial ATMP developers, of which 68 responded providing 243 challenges. Of products in development, 72% were in early clinical development and 40% were gene therapies. Most developers were small- or medium-sized enterprises (65%). The most often mentioned challenges were related to country-specific requirements (16%), manufacturing (15%), and clinical trial design (8%). The European ATMP field is still in its early stages, and developers experience challenges on many levels. Challenges are multifactorial and a mix of ATMP-specific and generic development aspects, such as new and orphan indications, novel technologies, and inexperience, adding complexity to development efforts.
Collapse
|
31
|
Bauer G, Elsallab M, Abou-El-Enein M. Concise Review: A Comprehensive Analysis of Reported Adverse Events in Patients Receiving Unproven Stem Cell-Based Interventions. Stem Cells Transl Med 2018; 7:676-685. [PMID: 30063299 PMCID: PMC6127222 DOI: 10.1002/sctm.17-0282] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 02/06/2023] Open
Abstract
The promise of stem cell (SC) therapies to restore functions of damaged tissues and organs brings enormous hope to patients, their families, loved ones, and caregivers. However, limits may exist for which indications SC therapies might be useful, efficacious, and safe. Applications of innovative therapies within regulatory boundaries and within the framework of controlled clinical trials are the norm in the scientific and medical community; such a system minimizes patient risk by setting a clear and acceptable safety and efficacy profile for new therapeutics before marketing authorization. This careful clinical validation approach often takes time, which patients suffering from terminal or debilitating diseases do not have. Not validated, unproven stem cell interventions (SCI) that promise a working treatment or cure for severe diseases have therefore found their way into the patient community, and providers of such treatments often take advantage of the public's willingness to pay large amounts of money for the misguided hope of a reliable recovery from their illnesses. We conducted a review of scientific publications, clinical case reports, and mass media publications to assess the reported cases and safety incidents associated with unproven SCI. The review also analyzes the main factors that were identified as contributing to the emergence and global rise of the “stem cell tourism” phenomenon. stemcellstranslationalmedicine2018;1–10
Collapse
Affiliation(s)
- Gerhard Bauer
- University of California Davis, Institute For Regenerative Cures (IRC), Sacramento, California, USA
| | - Magdi Elsallab
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charite' - Universitatsmedizin Berlin, Berlin, Germany
| | - Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charite' - Universitatsmedizin Berlin, Berlin, Germany
| |
Collapse
|
32
|
Viganò M, Giordano R, Lazzari L. Challenges of running a GMP facility for regenerative medicine in a public hospital. Regen Med 2017; 12:803-813. [DOI: 10.2217/rme-2017-0051] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Advanced therapy medicinal products represent a new generation of medicinal products for regenerative medicine. Since the implementation of the EU regulation for this innovative class of drugs, the academic and hospital institutions have played a central role in their development and manufacture. For these institutions that are not familiar with the industrial context, being in compliance with the pharmaceutical standards is extremely challenging. This report describes how we dealt with some specific issues during our hospital-based GMP experience. Furthermore, we identify as a future perspective the consistent stimulating contribution that a public entity can ensure for advanced therapy medicinal product development and licensing.
Collapse
Affiliation(s)
- Mariele Viganò
- Cell Factory, Laboratory of Regenerative Medicine, Department of Services & Preventive Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Rosaria Giordano
- Cell Factory, Laboratory of Regenerative Medicine, Department of Services & Preventive Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
| | - Lorenza Lazzari
- Cell Factory, Laboratory of Regenerative Medicine, Department of Services & Preventive Medicine, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milano, Italy
| |
Collapse
|
33
|
A roadmap for cost-of-goods planning to guide economic production of cell therapy products. Cytotherapy 2017; 19:1383-1391. [PMID: 28935190 DOI: 10.1016/j.jcyt.2017.06.009] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/29/2017] [Indexed: 12/31/2022]
Abstract
Cell therapy products are frequently developed and produced without incorporating cost considerations into process development, contributing to prohibitively costly products. Herein we contextualize individual process development decisions within a broad framework for cost-efficient therapeutic manufacturing. This roadmap guides the analysis of cost of goods (COG) arising from tissue procurement, material acquisition, facility operation, production, and storage. We present the specific COG considerations related to each of these elements as identified through a 2013 International Society for Cellular Therapy COG survey, highlighting the differences between autologous and allogeneic products. Planning and accounting for COG at each step in the production process could reduce costs, allowing for more affordable market pricing to improve the long-term viability of the cell therapy product and facilitate broader patient access to novel and transformative cell therapies.
Collapse
|
34
|
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
|
35
|
Evaluation of 2 Purification Methods for Isolation of Human Adipose-Derived Stem Cells Based on Red Blood Cell Lysis With Ammonium Chloride and Hypotonic Sodium Chloride Solution. Ann Plast Surg 2017; 78:83-90. [DOI: 10.1097/sap.0000000000000953] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
36
|
Abou-El-Enein M, Volk HD, Reinke P. Clinical Development of Cell Therapies: Setting the Stage for Academic Success. Clin Pharmacol Ther 2016; 101:35-38. [PMID: 27709611 DOI: 10.1002/cpt.523] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 09/19/2016] [Accepted: 09/21/2016] [Indexed: 01/05/2023]
Abstract
Cellular therapies have potential to treat a wide range of diseases with autologous immunotherapies showing unprecedented therapeutic promise in clinical trials. Such therapies are mainly developed by academic researchers applying small-scale production, targeting rare and unmet medical needs. Here, we highlight the clinical translation of immunotherapy product in an academic setting, which may serve as a success model for early academic development of cell-based therapeutics.
Collapse
Affiliation(s)
- M Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - H-D Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - P Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
37
|
Abou-El-Enein M, Bauer G, Medcalf N, Volk HD, Reinke P. Putting a price tag on novel autologous cellular therapies. Cytotherapy 2016; 18:1056-1061. [PMID: 27288308 DOI: 10.1016/j.jcyt.2016.05.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 05/05/2016] [Accepted: 05/09/2016] [Indexed: 12/24/2022]
Abstract
Cell therapies, especially autologous therapies, pose significant challenges to researchers who wish to move from small, probably academic, methods of manufacture to full commercial scale. There is a dearth of reliable information about the costs of operation, and this makes it difficult to predict with confidence the investment needed to translate the innovations to the clinic, other than as small-scale, clinician-led prescriptions. Here, we provide an example of the results of a cost model that takes into account the fixed and variable costs of manufacture of one such therapy. We also highlight the different factors that influence the product final pricing strategy. Our findings illustrate the need for cooperative and collective action by the research community in pre-competitive research to generate the operational models that are much needed to increase confidence in process development for these advanced products.
Collapse
Affiliation(s)
- Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité-University Medicine, Campus Virchow, Berlin, Germany.
| | - Gerhard Bauer
- Institute for Regenerative Cures, University of California Davis, Sacramento, CA, USA
| | - Nicholas Medcalf
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough, UK
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Campus Virchow, Berlin, Germany; Institute of Medical Immunology, Charité University Medicine, Campus Virchow, Berlin, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies, Charité University Medicine, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité-University Medicine, Campus Virchow, Berlin, Germany
| |
Collapse
|
38
|
Development of advanced therapies in Italy: Management models and sustainability in six Italian cell factories. Cytotherapy 2016; 18:481-6. [DOI: 10.1016/j.jcyt.2016.01.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 12/20/2015] [Accepted: 01/03/2016] [Indexed: 11/22/2022]
|
39
|
|
40
|
Olender E, Brubaker S, Uhrynowska-Tyszkiewicz I, Wojtowicz A, Kaminski A. Autologous osteoblast transplantation, an innovative method of bone defect treatment: role of a tissue and cell bank in the process. Transplant Proc 2015; 46:2867-72. [PMID: 25380938 DOI: 10.1016/j.transproceed.2014.09.071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
BACKGROUND The idea of cell treatment of various diseases and medical conditions has become very popular. Some procedures are well established, as is autologous chondrocyte implantation, whereas others are still in the process of early development, laboratory experiments, and some clinical trials. METHODS This report is devoted to an example of an emerging cell treatment: bone augmentation with the use of autologous cells and its legal and technical background. Various requirements set by law must be met by tissue banks performing cell seeding of grafts. In Europe, the requirements are described in directives 2004/23/EC, 2006/17/EC, 2006/86/EC, and in the regulation 2007/1394/EC. RESULTS Revitalization of biostatic allografts gives new, promising tools for creation of functional parts of organs; brings the methodology used in tissue banks closer to tissue engineering; places the enterprise in the mainstream of advanced biotechnology; allows the full potential of tissue allografts; and opens a new, large area for clinical and laboratory research. Cell and tissue processing also have a financial impact on the treatment: it produces additional expenditures. CONCLUSIONS Clinical effectiveness will be the most decisive factor of whether this innovative treatment will be applied in a particular type of medical condition. From a tissue establishment perspective, the most important issue is to develop a procedure that ensures safety for the patient in graft quality terms.
Collapse
Affiliation(s)
- E Olender
- Department of Transplantology and Central Tissue Bank, Medical University of Warsaw, and National Centre for Tissue and Cell Banking, Warsaw, Poland.
| | - S Brubaker
- American Association of Tissue Banks, McLean, Virginia
| | - I Uhrynowska-Tyszkiewicz
- Department of Transplantology and Central Tissue Bank, Medical University of Warsaw, and National Centre for Tissue and Cell Banking, Warsaw, Poland
| | - A Wojtowicz
- Department of Dental Surgery, Medical University of Warsaw, Warsaw, Poland
| | - A Kaminski
- Department of Transplantology and Central Tissue Bank, Medical University of Warsaw, and National Centre for Tissue and Cell Banking, Warsaw, Poland
| |
Collapse
|
41
|
Roemhild A, Reinke P. Virus-specific T-cell therapy in solid organ transplantation. Transpl Int 2015; 29:515-26. [PMID: 26284570 DOI: 10.1111/tri.12659] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 04/07/2015] [Accepted: 08/12/2015] [Indexed: 12/12/2022]
Abstract
This article reviews the current state of T-cell therapy as therapeutic option for virus-associated diseases against the background of the most common viral complications and their standard treatment regimens after SOT. The available data of clinical T-cell trials in SOT are summarized. References to the hematopoietic stem cell transplantation are made if applicable data in SOT are not available and their content was considered likewise valid for cell therapy in SOT. Moreover, aspects of different manufacturing approaches including beneficial product characteristics and the importance of GMP compliance are addressed.
Collapse
Affiliation(s)
- Andy Roemhild
- Department of Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- Department of Nephrology and Internal Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
42
|
|
43
|
Ducret M, Fabre H, Degoul O, Atzeni G, McGuckin C, Forraz N, Alliot-Licht B, Mallein-Gerin F, Perrier-Groult E, Farges JC. Manufacturing of dental pulp cell-based products from human third molars: current strategies and future investigations. Front Physiol 2015; 6:213. [PMID: 26300779 PMCID: PMC4526817 DOI: 10.3389/fphys.2015.00213] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 07/16/2015] [Indexed: 01/01/2023] Open
Abstract
In recent years, mesenchymal cell-based products have been developed to improve surgical therapies aimed at repairing human tissues. In this context, the tooth has recently emerged as a valuable source of stem/progenitor cells for regenerating orofacial tissues, with easy access to pulp tissue and high differentiation potential of dental pulp mesenchymal cells. International guidelines now recommend the use of standardized procedures for cell isolation, storage and expansion in culture to ensure optimal reproducibility, efficacy and safety when cells are used for clinical application. However, most dental pulp cell-based medicinal products manufacturing procedures may not be fully satisfactory since they could alter the cells biological properties and the quality of derived products. Cell isolation, enrichment and cryopreservation procedures combined to long-term expansion in culture media containing xeno- and allogeneic components are known to affect cell phenotype, viability, proliferation and differentiation capacities. This article focuses on current manufacturing strategies of dental pulp cell-based medicinal products and proposes a new protocol to improve efficiency, reproducibility and safety of these strategies.
Collapse
Affiliation(s)
- Maxime Ducret
- Laboratoire de Biologie Tissulaire et Ingénierie thérapeutique, UMR5305 Centre National de la Recherche Scientifique/Université Claude Bernard Lyon 1 Lyon, France ; Faculté d'Odontologie, Université de Lyon, Université Claude Bernard Lyon 1 Lyon, France ; Hospices Civils de Lyon, Service de Consultations et Traitements Dentaires Lyon, France
| | - Hugo Fabre
- Laboratoire de Biologie Tissulaire et Ingénierie thérapeutique, UMR5305 Centre National de la Recherche Scientifique/Université Claude Bernard Lyon 1 Lyon, France
| | - Olivier Degoul
- CTI-BIOTECH, Cell Therapy Research Institute Meyzieu, France
| | | | - Colin McGuckin
- CTI-BIOTECH, Cell Therapy Research Institute Meyzieu, France
| | - Nico Forraz
- CTI-BIOTECH, Cell Therapy Research Institute Meyzieu, France
| | - Brigitte Alliot-Licht
- Faculté d'Odontologie, Institut National de la Santé et de la Recherche Médicale UMR1064, Centre de Recherche en Transplantation et Immunologie, Université de Nantes Nantes, France
| | - Frédéric Mallein-Gerin
- Laboratoire de Biologie Tissulaire et Ingénierie thérapeutique, UMR5305 Centre National de la Recherche Scientifique/Université Claude Bernard Lyon 1 Lyon, France
| | - Emeline Perrier-Groult
- Laboratoire de Biologie Tissulaire et Ingénierie thérapeutique, UMR5305 Centre National de la Recherche Scientifique/Université Claude Bernard Lyon 1 Lyon, France
| | - Jean-Christophe Farges
- Laboratoire de Biologie Tissulaire et Ingénierie thérapeutique, UMR5305 Centre National de la Recherche Scientifique/Université Claude Bernard Lyon 1 Lyon, France ; Faculté d'Odontologie, Université de Lyon, Université Claude Bernard Lyon 1 Lyon, France ; Hospices Civils de Lyon, Service de Consultations et Traitements Dentaires Lyon, France
| |
Collapse
|
44
|
Cary WA, Hori CN, Pham MT, Nacey CA, McGee JL, Hamou M, Berman RF, Bauer G, Nolta JA, Waldau B. Efficient Generation of Induced Pluripotent Stem and Neural Progenitor Cells From Acutely Harvested Dura Mater Obtained During Ventriculoperitoneal Shunt Surgery. World Neurosurg 2015; 84:1256-66.e1. [PMID: 26074438 DOI: 10.1016/j.wneu.2015.05.076] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/29/2015] [Accepted: 05/30/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND The dura mater can be easily biopsied during most cranial neurosurgical operations. We describe a protocol that allows for robust generation of induced pluripotent stem cells (iPSCs) and neural progenitors from acutely harvested dura mater. OBJECTIVE To generate iPSCs and neural progenitor cells from dura mater obtained during ventriculoperitoneal shunt surgery. METHODS Dura was obtained during ventriculoperitoneal shunt surgery for normal pressure hydrocephalus from a 60-year-old patient with severe cognitive impairment. Fibroblasts were isolated from the dural matrix and transduced with nonintegrating Sendai virus for iPSC induction. A subset of successfully generated iPSC clones underwent immunocytochemical analysis, teratoma assay, karyotyping, and targeted neural differentiation. RESULTS Eleven iPSC clones were obtained from the transduction of an estimated 600,000 dural fibroblasts after 3 passages. Three clones underwent immunocytochemical analysis and were shown to express the transcription factors OCT-4, SOX2, and the embryonic cell markers SSEA-4, TRA-1-60, and Nanog. Two clones were tested for pluripotency and formed teratomas at the injection site in immunodeficient mice. Three clones underwent chromosomal analysis and were found to have a normal metaphase spread and karyotype. One clone underwent targeted neural differentiation and formed neural rosettes as well as TuJ1/SOX1-positive neural progenitor cells. CONCLUSIONS IPSCs and neural progenitor cells can be efficiently derived from the dura of patients who need to undergo cranial neurosurgical operations. IPSCs were obtained with a nonintegrating virus and exhibited a normal karyotype, making them candidates for future autotransplantation after targeted differentiation to treat functional deficits.
Collapse
Affiliation(s)
- Whitney Ann Cary
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Courtney Namiko Hori
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Missy Trananh Pham
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Catherine Ann Nacey
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Jeannine Logan McGee
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Mattan Hamou
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Robert F Berman
- Department of Neurosurgery, UC Davis Medical Center, Sacramento, California, USA
| | - Gerhard Bauer
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Jan A Nolta
- Institute for Regenerative Cures (IRC), Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA
| | - Ben Waldau
- Department of Neurosurgery, UC Davis Medical Center, Sacramento, California, USA; UC Davis Stem Cell Program, Sacramento, California, USA.
| |
Collapse
|
45
|
Abou-El-Enein M, Bauer G, Reinke P, Renner M, Schneider CK. A roadmap toward clinical translation of genetically-modified stem cells for treatment of HIV. Trends Mol Med 2014; 20:632-42. [PMID: 25262540 DOI: 10.1016/j.molmed.2014.08.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 08/25/2014] [Accepted: 08/26/2014] [Indexed: 12/21/2022]
Abstract
During the past decade, successful gene therapies for immunodeficiencies were finally brought to the clinic. This was accomplished through new gene therapy vectors and improved procedures for genetic modification of autologous hematopoietic stem cells. For HIV, autologous hematopoietic stem cell (HSC) gene therapy with 'anti-HIV genes' promises a functional cure for the disease. However, to develop such a therapy and translate it into a clinical application is rather challenging. The risks and benefits of such a therapy have to be understood, and regulatory hurdles need to be overcome. In this joint paper by academic researchers and regulators, we are, therefore, outlining a high level roadmap for the early stage development of HSC gene therapy as a potential functional cure for HIV.
Collapse
Affiliation(s)
- Mohamed Abou-El-Enein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany.
| | - Gerhard Bauer
- University of California Davis, Institute For Regenerative Cures (IRC) Sacramento, CA, USA
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité University Medicine Berlin, Campus Virchow, Berlin, Germany; Department of Nephrology and Internal Intensive Care, Charité University Medicine Berlin, Campus Virchow, Berlin, Germany
| | - Matthias Renner
- Paul-Ehrlich-Institut, Paul-Ehrlich-Str. 51-59, D-63225 Langen, Germany
| | - Christian K Schneider
- Formerly Committee for Advanced Therapies, European Medicines Agency, 7, Westferry Circus E14 4HB, London, UK; Danish Health and Medicines Authority, Axel Heides Gade 1, 2300 Copenhagen, Denmark; Twincore Centre for Experimental and Clinical Infection Research, Feodor-Lynen-Straße 730625 Hannover, Germany
| |
Collapse
|
46
|
Erben RG, Silva-Lima B, Reischl I, Steinhoff G, Tiedemann G, Dalemans W, Vos A, Janssen RTA, Le Blanc K, van Osch GJVM, Luyten FP. White paper on how to go forward with cell-based advanced therapies in Europe. Tissue Eng Part A 2014; 20:2549-54. [PMID: 24749762 DOI: 10.1089/ten.tea.2013.0589] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The current White paper summarizes the discussions and exchange of experiences during the first European Interdisciplinary Summit on Cell-Based ATMPs held in Vienna, Austria, May 02-03, 2013. The meeting was supported by the Research Networking Programme REMEDIC (regenerative medicine) funded by the European Science Foundation and by the British Medical Research Council. To improve the competitiveness of Europe in the field of cell-based Advanced Medicinal Therapy Products (ATMPs), the following key issues were identified during the meeting: removal of national hurdles in the European Union, harmonization of national and subnational differences in Hospital Exemption rules, improved treatment algorithms for reimbursement, better knowledge on the mode of action, predictive preclinical efficacy and safety testing, need for innovative systems for preclinical testing, appropriate product characterization, manufacturing with cost of goods in mind, and appropriate design of clinical trials.
Collapse
Affiliation(s)
- Reinhold G Erben
- 1 Department of Biomedical Sciences, University of Veterinary Medicine , Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lowest numbers of primary CD8(+) T cells can reconstitute protective immunity upon adoptive immunotherapy. Blood 2014; 124:628-37. [PMID: 24855206 DOI: 10.1182/blood-2013-12-547349] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are threatened by potentially lethal viral manifestations like cytomegalovirus (CMV) reactivation. Because the success of today's virostatic treatment is limited by side effects and resistance development, adoptive transfer of virus-specific memory T cells derived from the stem cell donor has been proposed as an alternative therapeutic strategy. In this context, dose minimization of adoptively transferred T cells might be warranted for the avoidance of graft-versus-host disease (GVHD), in particular in prophylactic settings after T-cell-depleting allo-HSCT protocols. To establish a lower limit for successful adoptive T-cell therapy, we conducted low-dose CD8(+) T-cell transfers in the well-established murine Listeria monocytogenes (L.m.) infection model. Major histocompatibility complex-Streptamer-enriched antigen-specific CD62L(hi) but not CD62L(lo) CD8(+) memory T cells proliferated, differentiated, and protected against L.m. infections after prophylactic application. Even progenies derived from a single CD62L(hi) L.m.-specific CD8(+) T cell could be protective against bacterial challenge. In analogy, low-dose transfers of Streptamer-enriched human CMV-specific CD8(+) T cells into allo-HSCT recipients led to strong pathogen-specific T-cell expansion in a compassionate-use setting. In summary, low-dose adoptive T-cell transfer (ACT) could be a promising strategy, particularly for prophylactic treatment of infectious complications after allo-HSCT.
Collapse
|