1
|
Tseng CY, Wang K, Lin LH, Zhang C, White CC, Wang B. Advancing autologous CAR T-cell therapy through real-time patient health data integration: a simulation-based approach. Cytotherapy 2024; 26:1152-1162. [PMID: 38795115 DOI: 10.1016/j.jcyt.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 05/27/2024]
Abstract
Autologous chimeric antigen receptor T-cell therapy presents promising treatment outcomes for various cancers. However, its potential is restrained by unique supply chain challenges, including dynamic patient health conditions and extended turnaround time. These challenges often lead to missed optimal treatment windows, impeding the effective delivery of life-saving treatments. This article presents SimPAC (simulation-based decision support for Patient-centric manufacturing of autologous cell therapies). SimPAC is designed to model and incorporate real-time patient health conditions into the supply chain decisions of autologous chimeric antigen receptor T-cell therapy. SimPAC integrates system dynamics and agent-based simulation techniques, facilitating the adaptation of manufacturing processes and production schedules based on real-time patient health conditions. SimPAC can model various patient disease progressions using parametric functions, nonparametric functions, or tabular data. Additionally, SimPAC offers easy configuration options to model various cell therapy supply chains. We provide two case studies to demonstrate the capabilities of SimPAC and highlight the benefits of patient-centric manufacturing, including improved survival rates and potential economic advantages. However, while the benefits are significant, our study also emphasizes the importance of balancing improved patient outcomes, economic viability and ethical considerations in the context of personalized medicine. SimPAC can be used to explore applications of this approach to diverse therapeutic contexts and supply chain configurations.
Collapse
Affiliation(s)
- Chin-Yuan Tseng
- H. Milton Stewart School of Industrial & Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Kan Wang
- H. Milton Stewart School of Industrial & Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, Georgia, USA.
| | - Li-Hsiang Lin
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, USA
| | - Chuck Zhang
- H. Milton Stewart School of Industrial & Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Chelsea C White
- H. Milton Stewart School of Industrial & Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Ben Wang
- H. Milton Stewart School of Industrial & Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
2
|
Stemer G, Mittermayr T, Schnell-Inderst P, Wild C. Costs, challenges and opportunities of decentralised chimeric antigen receptor T-cell production: a literature review and clinical experts' interviews. Eur J Hosp Pharm 2024:ejhpharm-2024-004130. [PMID: 39209448 DOI: 10.1136/ejhpharm-2024-004130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
The objectives were to summarise the evidence and clinical experts' views comparing the use of decentralised produced chimeric antigen receptor (CAR) T-cell therapies versus commercially available products, regarding drug costs, time to finalised product and other reported advantages, disadvantages, challenges and facilitators. A literature review according to the PRISMA guidelines was conducted in Medline, Embase and Trip databases. Publications were included if they reported information on cost estimates, time to finalised products and other outcomes of interest of a decentralised CAR T-cell production strategy. A structured interview guide was developed and used for qualitative expert interviews. Five experts were purposively selected, and interviews were either conducted face-to-face or online, and recorded for the purpose of transcription. Transcripts were analysed and categories and codes extracted. Reporting is based on the COREQ checklist for reporting qualitative research. Costs of decentralised produced CAR T-cells appear to be lower by a factor two to 14, compared with commercial products. But there is high uncertainty about this estimate, because it is unclear whether cost components included are comparable and due to the heterogeneity of the studies. The most commonly reported advantages were proximity to patients and decreased product risks and costs, whereas the continuing dependency on centrally manufactured reagents and specific characteristics of 'fresh' CAR T-cells are reported as disadvantages. Compliance with regulatory requirements is mentioned as the biggest challenge. The availability of closed-system production devices is reported as one main facilitator, as are clear commitment, secured financing and knowledge transfer from already experienced centres. Apparent cost differences open a field for healthcare decision-makers to discuss and justify investment costs for implementation of a complementing decentralised production programme and to realise other associated benefits of such a strategy, such as flexibility, patient proximity and expanding patient access.
Collapse
Affiliation(s)
- Gunar Stemer
- Pharmacy Department, University Hospital Vienna, Vienna, Austria
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT TIROL - University for Health Sciences and Technology, Hall in Tirol, Austria
| | | | - Petra Schnell-Inderst
- Institute of Public Health, Medical Decision Making and Health Technology Assessment, UMIT TIROL - University for Health Sciences and Technology, Hall in Tirol, Austria
| | - Claudia Wild
- Austrian Institute for Health Technology Assessment, Vienna, Austria
| |
Collapse
|
3
|
Triantafyllou N, Sarkis M, Krassakopoulou A, Shah N, Papathanasiou MM, Kontoravdi C. Uncertainty quantification for gene delivery methods: A roadmap for pDNA manufacturing from phase I clinical trials to commercialization. Biotechnol J 2024; 19:e2300103. [PMID: 37797343 DOI: 10.1002/biot.202300103] [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: 03/06/2023] [Revised: 07/01/2023] [Accepted: 09/28/2023] [Indexed: 10/07/2023]
Abstract
The fast-growing interest in cell and gene therapy (C>) products has led to a growing demand for the production of plasmid DNA (pDNA) and viral vectors for clinical and commercial use. Manufacturers, regulators, and suppliers need to develop strategies for establishing robust and agile supply chains in the otherwise empirical field of C>. A model-based methodology that has great potential to support the wider adoption of C> is presented, by ensuring efficient timelines, scalability, and cost-effectiveness in the production of key raw materials. Specifically, key process and economic parameters are identified for (1) the production of pDNA for the forward-looking scenario of non-viral-based Chimeric Antigen Receptor (CAR) T-cell therapies from clinical (200 doses) to commercial (40,000 doses) scale and (2) the commercial (40,000 doses) production of pDNA and lentiviral vectors for the current state-of-the-art viral vector-based CAR T-cell therapies. By applying a systematic global sensitivity analysis, we quantify uncertainty in the manufacturing process and apportion it to key process and economic parameters, highlighting cost drivers and limitations that steer decision-making. The results underline the cost-efficiency and operational flexibility of non-viral-based therapies in the overall C> supply chain, as well as the importance of economies-of-scale in the production of pDNA.
Collapse
Affiliation(s)
- Niki Triantafyllou
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Miriam Sarkis
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | | | - Nilay Shah
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Maria M Papathanasiou
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Cleo Kontoravdi
- The Sargent Centre for Process Systems Engineering, Imperial College London, London, UK
- Department of Chemical Engineering, Imperial College London, London, UK
| |
Collapse
|
4
|
Wang K, Tseng CY, Li Z, White C, Wang B, Levine BL, Fesnak AD. A simulation-based comparison of centralized and point-of-care supply chain strategies for autologous cell therapy. Cytotherapy 2023; 25:1370-1379. [PMID: 37747394 DOI: 10.1016/j.jcyt.2023.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND AIMS The selection between centralized and point-of-care (POC) manufacturing supply-chain network design is a crucial consideration in the autologous cell therapy (AuCT) industry, as each approach offers its advantages and disadvantages. METHODS This study uses a simulation-based approach to compare and examine the two strategies using the supply chain for chimeric antigen receptor T-cell therapy manufacturing as an exemplar. When does it make sense to use one manufacturing strategy over another? Currently, major manufacturers in the AuCT industry use centralized supply-chain strategies predominantly in practice. The simulation results explain the reasons for this choice. To enhance the competitiveness of the POC strategy, two operation rules are proposed and tested with the simulation. The study uses key performance indicators such as cost, fulfillment time, service level, and resource utilization to provide generic guidelines based on the findings. RESULTS The results have revealed that (i) the centralized supply-chain strategy has a significant advantage at current demand levels of a few thousand products per year; (ii) "optimal capacity" exists for the POC strategy that minimizes the cost of goods and (iii) allowing part-time labor and order transshipment can significantly increase the competitiveness of the POC strategy. CONCLUSIONS This study may be useful in helping commercial manufacturers make informed decisions about their manufacturing approach to enhance their competitiveness in the market and to ensure a high level of patient benefit.
Collapse
Affiliation(s)
- Kan Wang
- Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, Georgia, USA.
| | - Chin-Yuan Tseng
- Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, Georgia, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA.
| | - Zhaowei Li
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Chip White
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Ben Wang
- Georgia Tech Manufacturing Institute, Georgia Institute of Technology, Atlanta, Georgia, USA; H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA; School of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrew D Fesnak
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
5
|
Elsallab M, Maus MV. Expanding access to CAR T cell therapies through local manufacturing. Nat Biotechnol 2023; 41:1698-1708. [PMID: 37884746 DOI: 10.1038/s41587-023-01981-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 09/05/2023] [Indexed: 10/28/2023]
Abstract
Chimeric antigen receptor (CAR) T cells are changing the therapeutic landscape for hematological malignancies. To date, all six CAR T cell products approved by the US Food and Drug Administration (FDA) are autologous and centrally manufactured. As the numbers of approved products and indications continue to grow, new strategies to increase cell-manufacturing capacity are urgently needed to ensure patient access. Distributed manufacturing at the point of care or at other local manufacturing sites would go a long way toward meeting the rising demand. To ensure successful implementation, it is imperative to harness novel technologies to achieve uniform product quality across geographically dispersed facilities. This includes the use of automated cell-production systems, in-line sensors and process simulation for enhanced quality control and efficient supply chain management. A comprehensive effort to understand the critical quality attributes of CAR T cells would enable better definition of widely attainable release criteria. To supplement oversight by national regulatory agencies, we recommend expansion of the role of accreditation bodies. Moreover, regulatory standards may need to be amended to accommodate the unique characteristics of distributed manufacturing models.
Collapse
Affiliation(s)
- Magdi Elsallab
- Harvard-MIT Center for Regulatory Science, Harvard Medical School, Boston, MA, USA
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
6
|
Delgadillo J, Kerkelä E, Waters A, Akker EVD, Lechanteur C, Baudoux E, Gardiner N, De Vos J, Vives J. A management model in blood, tissue and cell establishments to ensure rapid and sustainable patient access to advanced therapy medicinal products in Europe. Cytotherapy 2023; 25:1259-1264. [PMID: 37737767 DOI: 10.1016/j.jcyt.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 09/23/2023]
Abstract
Blood, tissue and cell establishments (BTCs) stand out in the management of donor selection, procurement and processing of all types of substances of human origin (SoHO). In the last decades, the framework created around BTCs, including hospitals and national health system networks, and their links to research, development and innovation organizations and agencies have spurred their involvement in the study of groundbreaking advanced therapy medicinal products (ATMP). To further improve strategic synergies in the development of ATMPs, it will be required to promote intra- and inter-European collaborations by creating an international network involving BTCs and major stakeholders (i.e., research organizations, hospitals, universities, patient associations, public agencies). This vision is already shared with the European Blood Alliance, the association of non-profit blood establishments, with 26 member states throughout the European Union and European Free Trade Association states. Herein we present and analyze the "BTC for ATMP Development And Manufacture" (BADAM) model, an ethically responsible business model based on the values and missions of BTCs and their commitment to health equity, patient access and education (based on voluntary donation of SoHO to address unmet clinical needs, while contributing to training professionals and scientific literacy of our Society).
Collapse
Affiliation(s)
- Joaquín Delgadillo
- Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain; Transfusion Medicine Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Vantaa, Finland
| | - Allison Waters
- Irish Blood Transfusion Service, National Blood Centre, Dublin, Ireland
| | - Emile van den Akker
- Department of Hematopoiesis and Sanquin Research, Landsteiner Laboratory, Department of Molecular Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Chantal Lechanteur
- University of Liège, Laboratory of Cell and Gene Therapy LTCG, Liège, Belgium
| | - Etienne Baudoux
- University of Liège, Laboratory of Cell and Gene Therapy LTCG, Liège, Belgium
| | - Nicola Gardiner
- Cryobiology Laboratory Stem Cell Facility, St. James's Hospital, Dublin, Ireland
| | - John De Vos
- Département d'ingénierie Cellulaire et Tissulaire, Unité de Thérapie Cellulaire, Hôpital Saint-Eloi, Montpellier, France
| | - Joaquim Vives
- Banc de Sang i Teixits (BST), Edifici Dr. Frederic Duran i Jordà, Barcelona, Spain; Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
7
|
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
|
8
|
Orentas RJ, Dropulić B, de Lima M. Place of care manufacturing of chimeric antigen receptor cells: opportunities and challenges. Semin Hematol 2023; 60:20-24. [PMID: 37080706 DOI: 10.1053/j.seminhematol.2023.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/19/2023] [Indexed: 01/26/2023]
Abstract
The landscape of therapeutic options for B cell malignancies has fundamentally changed with regulatory and marketing approval of chimeric antigen receptor (CAR)-engineered T cell products. The cell types used for CAR-T production, the length of time of manufacture, the stimulation matrix, and the nature of the gene vector used to transduce human T cells all are significant variables that require adequate quality control before infusion. Having approved products available to clinicians using a centralized production paradigm has not stopped innovation in investigator-initiated trials. Moreover, the high costs of the commercial products have been a significant wake-up call to those concerned about rising costs in health care, and the ability of developing nations, and nations with managed care systems to support these costs. Place-of-care manufacturing is a clear alternative to the approved products created in a centralized manufacturing approach. It is supported by continued technological innovation and the willingness of clinicians to develop new ways to decrease costs and make these curative therapies equitably available.
Collapse
|
9
|
Doglio M, Alexander T, Del Papa N, Snowden JA, Greco R. New insights in systemic lupus erythematosus: From regulatory T cells to CAR-T-cell strategies. J Allergy Clin Immunol 2022; 150:1289-1301. [PMID: 36137815 DOI: 10.1016/j.jaci.2022.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/11/2022] [Accepted: 08/02/2022] [Indexed: 12/14/2022]
Abstract
Systemic lupus erythematous is a heterogeneous autoimmune disease with potentially multiorgan damage. Its complex etiopathogenesis involves genetic, environmental, and hormonal factors, leading to a loss of self-tolerance with autoantibody production and immune complex formation. Given the relevance of autoreactive B lymphocytes, several therapeutic approaches have been made targeting these cells. However, the disease remains incurable, reflecting an unmet need for effective strategies. Novel therapeutic concepts have been investigated to provide more specific and sustainable disease modification compared with continued immunosuppression. Autologous hematopoietic stem cell transplantation has already provided the proof-of-concept that immunodepletion can lead to durable treatment-free remissions, albeit with significant treatment-related toxicity. In the future, chimeric antigen receptor-T-cell therapies, for example, CD19 chimeric antigen receptor-T, may provide a more effective lymphodepletion and with less toxicity than autologous hematopoietic stem cell transplantation. An emerging field is to enhance immune tolerance by exploiting the suppressive capacities of regulatory T cells, which are dysfunctional in patients with systemic lupus erythematous, and thus resemble promising candidates for adoptive cell therapy. Different approaches have been developed in this area, from polyclonal to genetically engineered regulatory T cells. In this article, we discuss the current evidence and future directions of cellular therapies for the treatment of systemic lupus erythematous, including hematopoietic stem cell transplantation and advanced regulatory T-cell-based cellular therapies.
Collapse
Affiliation(s)
- Matteo Doglio
- Experimental Hematology Unit, Department of Immunology Transplantations and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Vita-Salute University, Milan, Italy; Unit of Pediatric Immunohematology, San Raffaele Hospital, Milan, Italy
| | - Tobias Alexander
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, Berlin, Germany; Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany.
| | | | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals Foundation NHS Trust, Sheffield, United Kingdom
| | - Raffaella Greco
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Mila, Italy.
| |
Collapse
|
10
|
Martin R, Lei R, Zeng Y, Zhu J, Chang H, Ye H, Cui Z. Membrane Applications in Autologous Cell Therapy. MEMBRANES 2022; 12:1182. [PMID: 36557091 PMCID: PMC9788437 DOI: 10.3390/membranes12121182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 06/17/2023]
Abstract
Stem cell and cell therapies, particularly autologous cell therapies, are becoming a common practice. However, in order for these technologies to achieve wide-scale clinical application, the prohibitively high cost associated with these therapies must be addressed through creative engineering. Membranes can be a disruptive technology to reshape the bioprocessing and manufacture of cellular products and significantly reduce the cost of autologous cell therapies. Examples of successful membrane applications include expansions of CAR-T cells, various human stem cells, and production of extracellular vesicles (EVs) using hollow fibre membrane bioreactors. Novel membranes with tailored functions and surface properties and novel membrane modules that can accommodate the changing needs for surface area and transport properties are to be developed to fulfil this key role.
Collapse
Affiliation(s)
- Risto Martin
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
| | - Rui Lei
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
| | - Yida Zeng
- Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou 215123, China
| | - Jiachen Zhu
- Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou 215123, China
| | - Hong Chang
- Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou 215123, China
| | - Hua Ye
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
- Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou 215123, China
| | - Zhanfeng Cui
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford OX3 7DQ, UK
- Oxford Suzhou Centre for Advanced Research (OSCAR), University of Oxford, Suzhou 215123, China
| |
Collapse
|
11
|
Gagelmann N, Sureda A, Montoto S, Murray J, Bolaños N, Kenyon M, Beksac M, Schönland S, Hayden P, Scheurer H, Morgan K, Garderet L, McLornan DP, Ruggeri A. Access to and affordability of CAR T-cell therapy in multiple myeloma: an EBMT position paper. Lancet Haematol 2022; 9:e786-e795. [PMID: 36174641 DOI: 10.1016/s2352-3026(22)00226-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/21/2022] [Accepted: 07/08/2022] [Indexed: 06/16/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a promising immunotherapeutic approach in the treatment of multiple myeloma, and the recent approval of the first two CAR T-cell products could result in improved outcomes. However, it remains a complex and expensive technology, which poses challenges to health-care systems and society in general, especially in times of crises. This potentially accelerates pre-existing inequalities as access to CAR T-cell therapy varies, both between countries, depending on the level of economic development, and within countries, due to structural disparities in access to quality health care-a parameter strongly correlated with socioeconomic status, ethnicity, and lifestyle. Here, we identify two important issues: affordability and access to CAR T-cell treatment. This consensus statement from clinical investigators, clinicians, nurses, and patients from the European Society for Blood and Marrow Transplantation (EBMT) proposes solutions as part of an innovative collaborative strategy to make CAR T-cell therapy accessible to all patients with multiple myeloma.
Collapse
Affiliation(s)
- Nico Gagelmann
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Anna Sureda
- Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain
| | - Silvia Montoto
- St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - John Murray
- The Christie NHS Foundation Trust, Manchester, UK
| | | | - Michelle Kenyon
- Department of Haematology, King's College Hospital NHS Foundation Trust, London, UK
| | - Meral Beksac
- Department of Hematology, Ankara University, Ankara, Turkey
| | - Stefan Schönland
- Medical Department V, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Hayden
- Department of Hematology, Trinity College Dublin, St James's Hospital, Dublin, Ireland
| | | | | | - Laurent Garderet
- Service d'Hématologie et Thérapie Cellulaire, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Donal P McLornan
- Department of Haematology and Stem Cell Transplantation, University College Hospital, London, UK
| | - Annalisa Ruggeri
- Hematology and BMT Unit, San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
12
|
Otegbeye F. Bench at Bedside Models Facilitate CAR-T Cell Supply Chain. Transplant Cell Ther 2022; 28:221-222. [PMID: 35523472 DOI: 10.1016/j.jtct.2022.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Folashade Otegbeye
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, WA 98109.
| |
Collapse
|
13
|
Improving cell viability using counterflow centrifugal elutriation. Cytotherapy 2022; 24:650-658. [DOI: 10.1016/j.jcyt.2022.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/15/2022] [Accepted: 01/24/2022] [Indexed: 11/19/2022]
|
14
|
Lam C, Meinert E, Yang A, Cui Z. Impact of fast-track regulatory designations on strategic commercialization decisions for autologous cell therapies. Regen Med 2022; 17:155-174. [PMID: 35073729 DOI: 10.2217/rme-2021-0061] [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
Background: Regulatory authorities around the world have introduced incentives to improve the speed-to-market of innovative therapies. Aim & methods: To better understand the capacity and portfolio planning decisions of autologous cell therapies and particularly the impact of fast-tracking designations, this paper describes a mixed-integer linear programming approach for the optimization of capacity investment and portfolio selection decisions to maximize the net present value of a candidate portfolio of therapies under different regulatory programs. Results: The illustrative example shows that fast-track designations allow a 25% earlier breakeven, 42-86% higher net present value over a 20-year horizon with earlier upfront capital and reduce the portfolio's sensitivity to uncertainties. Conclusion: Fast-track designations are effective in providing commercialization incentives, but high capital risks given the compressed timeline should be better considered.
Collapse
Affiliation(s)
- Ching Lam
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, OX3 7DQ, UK
| | - Edward Meinert
- Department of Paediatrics, University of Oxford, Oxford, OX3 9DU, UK
| | - Aidong Yang
- Department of Engineering Science, University of Oxford, Oxford, OX1 3PJ, UK
| | - Zhanfeng Cui
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, OX3 7DQ, UK
| |
Collapse
|
15
|
Cell therapy products are revolutionizing multiple therapeutic areas; to maintain this pace of innovation, manufacturing solutions must adapt. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021. [DOI: 10.1016/j.cobme.2021.100340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Vázquez-Serrano JI, Peimbert-García RE, Cárdenas-Barrón LE. Discrete-Event Simulation Modeling in Healthcare: A Comprehensive Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:12262. [PMID: 34832016 PMCID: PMC8625660 DOI: 10.3390/ijerph182212262] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 11/26/2022]
Abstract
Discrete-event simulation (DES) is a stochastic modeling approach widely used to address dynamic and complex systems, such as healthcare. In this review, academic databases were systematically searched to identify 231 papers focused on DES modeling in healthcare. These studies were sorted by year, approach, healthcare setting, outcome, provenance, and software use. Among the surveys, conceptual/theoretical studies, reviews, and case studies, it was found that almost two-thirds of the theoretical articles discuss models that include DES along with other analytical techniques, such as optimization and lean/six sigma, and one-third of the applications were carried out in more than one healthcare setting, with emergency departments being the most popular. Moreover, half of the applications seek to improve time- and efficiency-related metrics, and one-third of all papers use hybrid models. Finally, the most popular DES software is Arena and Simul8. Overall, there is an increasing trend towards using DES in healthcare to address issues at an operational level, yet less than 10% of DES applications present actual implementations following the modeling stage. Thus, future research should focus on the implementation of the models to assess their impact on healthcare processes, patients, and, possibly, their clinical value. Other areas are DES studies that emphasize their methodological formulation, as well as the development of frameworks for hybrid models.
Collapse
Affiliation(s)
- Jesús Isaac Vázquez-Serrano
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Northeast Nuevo Leon, Mexico; (J.I.V.-S.); (L.E.C.-B.)
| | - Rodrigo E. Peimbert-García
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Northeast Nuevo Leon, Mexico; (J.I.V.-S.); (L.E.C.-B.)
- School of Engineering, Macquarie University, Sydney, NSW 2109, Australia
| | - Leopoldo Eduardo Cárdenas-Barrón
- School of Engineering and Sciences, Tecnologico de Monterrey, Monterrey 64849, Northeast Nuevo Leon, Mexico; (J.I.V.-S.); (L.E.C.-B.)
| |
Collapse
|
17
|
Saini KS, Svane IM, Juan M, Barlesi F, André F. Manufacture of adoptive cell therapies at academic cancer centers: scientific, safety and regulatory challenges. Ann Oncol 2021; 33:6-12. [PMID: 34655734 DOI: 10.1016/j.annonc.2021.09.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/27/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- K S Saini
- Labcorp Drug Development Inc., Princeton, USA
| | - I M Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - M Juan
- Department of Immunology, Hospital Clinic, IDIBAPS, Immunotherapy Platform Hospital Sant Joan de Déu, Universidad de Barcelona, Barcelona, Spain
| | - F Barlesi
- Department of Medical Oncology, Institut Gustave Roussy, Villejuif, France; Aix Marseille University, CNRS, INSERM, CRCM, Marseille, France
| | - F André
- Institut Gustave Roussy, INSERM UMR981, Université Paris Saclay, Paris, France.
| |
Collapse
|