1
|
Abdo L, Batista-Silva LR, Bonamino MH. Cost-effective strategies for CAR-T cell therapy manufacturing. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200980. [PMID: 40291594 PMCID: PMC12022644 DOI: 10.1016/j.omton.2025.200980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
CAR-T cell therapy has revolutionized cancer treatment, with approvals for conditions like acute B-leukemia, large B cell lymphoma (LBCL), follicular lymphoma (FL), mantle cell lymphoma (MCL), and multiple myeloma. However, its high costs limit accessibility. Key factors driving these costs include the need for personalized, autologous treatments, transportation to specialized facilities, reliance on viral vectors requiring advanced laboratories, and lengthy cell expansion processes. To address these challenges, alternative strategies aim to simplify and reduce production complexity. Non-viral vectors, such as Sleeping Beauty, piggyBac, and CRISPR, delivered via nanoparticles or electroporation, present promising solutions. These methods could streamline manufacturing, eliminate the need for viral vectors, and reduce associated costs. Furthermore, shortening cell expansion periods and optimizing protocols could significantly accelerate production. An emerging approach involves using genetically edited T cells from healthy donors to create universal CAR-T products capable of treating multiple patients. Finally, decentralized point-of-care (POC) manufacturing of CAR-T cells minimize logistical expenses, eliminating the need for complex infrastructure, and enabling localized production closer to patients. This innovative strategy holds potential for broadening access and reducing costs, representing a step toward democratizing CAR-T therapy. Combined, these advances could make this groundbreaking treatment more feasible for healthcare systems worldwide.
Collapse
Affiliation(s)
- Luiza Abdo
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
| | - Leonardo Ribeiro Batista-Silva
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
| | - Martín Hernán Bonamino
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
- Vice-Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
2
|
Melocchi A, Schmittlein B, Sadhu S, Nayak S, Lares A, Uboldi M, Zema L, di Robilant BN, Feldman SA, Esensten JH. Automated manufacturing of cell therapies. J Control Release 2025; 381:113561. [PMID: 39993639 DOI: 10.1016/j.jconrel.2025.02.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Advanced therapy medicinal products (ATMPs), particularly genetically engineered cell-based therapies, are a major class of drugs with several high-profile Food and Drug Administration (FDA) approvals in the past decade. However, the high cost and limited production capacity of these drugs remain a barrier to access. These costs are primarily due to the complex manufacturing processes (often a single batch for a single patient), which increases personnel and facility expenses, and the challenges associated with tech-transfer from research and development stages to clinical-stage production. In order to scale up and scale out in a cost-effective way, automated solutions capable of multi-step manufacturing have been developed in academia and industry. The aim of the present article is to summarize the design approaches and key features of current multi-step automated systems for cell therapy manufacturing. For each system described in the literature, we will discuss different aspects in detail such as cell specificity, modularity, processing models, manufacturing locations, and integrated quality control. Our analysis highlights that developers need to balance competing needs in an environment where the biological, business, and technological factors are constantly evolving. Thus, designing engineering solutions that align with the pharmaceutical end-user is essential. Adopting a risk-based approach grounded in published data is the most effective strategy to evaluate existing and emerging automated systems.
Collapse
Affiliation(s)
- Alice Melocchi
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy; Multiply Labs, San Francisco, CA, USA.
| | | | | | | | | | - Marco Uboldi
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | - Lucia Zema
- Sezione di Tecnologia e Legislazione Farmaceutiche "M. E. Sangalli", Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Milano, Italy
| | | | - Steven A Feldman
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jonathan H Esensten
- Advanced Biotherapy Center (ABC), Sheba Medical Center, Tel Hashomer, Israel
| |
Collapse
|
3
|
Dobruskin M, Toner G, Kander R. Cryopreservation practices in clinical and preclinical iPSC-based cell therapies: Current challenges and future directions. Biotechnol Prog 2025:e70031. [PMID: 40171754 DOI: 10.1002/btpr.70031] [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: 08/20/2024] [Revised: 11/22/2024] [Accepted: 03/24/2025] [Indexed: 04/04/2025]
Abstract
Induced pluripotent stem cells (iPSCs) offer significant therapeutic potential, but cryopreservation challenges, particularly the reliance on cytotoxic Dimethyl Sulfoxide (Me2SO), hinder their clinical application. This review examines current cryopreservation practices in clinical and preclinical iPSC-based therapies, highlighting the consistent use of Me2SO and the logistical challenges of post-thaw processing. The findings underscore the urgent need for alternative cryopreservation techniques to ensure the safety and efficacy of off-the-shelf iPSC therapies.
Collapse
Affiliation(s)
- Michael Dobruskin
- Jefferson Institute for Bioprocessing, Thomas Jefferson University, Spring House, Pennsylvania, USA
| | - Geoffrey Toner
- Jefferson Institute for Bioprocessing, Thomas Jefferson University, Spring House, Pennsylvania, USA
| | - Ronald Kander
- Jefferson Institute for Bioprocessing, Thomas Jefferson University, Spring House, Pennsylvania, USA
| |
Collapse
|
4
|
Kunitskaya A, Piret JM. Impacts of transient exposure of human T cells to low oxygen, temperature, pH and nutrient levels relevant to bioprocessing for cell therapy applications. Cytotherapy 2025; 27:522-533. [PMID: 39891634 DOI: 10.1016/j.jcyt.2025.01.002] [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: 10/11/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND T-cell therapy advances have stimulated the development of bioprocesses to address the specialized needs of cell therapy manufacturing. During concentrated cell washing, the cells are frequently exposed to transiently reduced oxygen, temperature, pH, and nutrient levels. Longer durations of these conditions can be caused by process deviations or, if they are not harmful, be used to ease the scheduling of process stages during experiments as well as manufacturing. METHODS To avoid unpredictable impacts on T-cell quality during bioprocessing, we measured the influences of such environmental exposures generated by settling 250 million activated human T cells per mL, for up to 6 h at temperatures from 4 to 37°C. RESULTS The measured glucose concentration decreased to as low as 0.5 mM and the pH to 6, while lactate increased up to 55 mM. The concentrated cell conditions at 37°C resulted in by far the greatest losses in viable cell numbers with, on average, only 58% and 41% of the cells recovered after 3 and 6 h, respectively. Likewise, their subsequent cell expansion cultures were substantially reduced even after only 3 h of exposure, and with decreased percentages of central memory T cells and increased percentages of effector memory and effector T cells. Although under similar environmental conditions at room temperatures, the negative impacts of high cell concentrations were greatly diminished for up to 3 h. At 4°C the transient conditions were less extreme, and the cells well maintained for 6 h. CONCLUSIONS Overall, when developing processes and devices for T-cell therapy manufacturing that involve concentrated cells, the results of this study indicate that more practically feasible room temperatures can be used for up to 3 h to obtain high viable cell recoveries whereas lower temperatures such as 4°C should be used if there is a need for more prolonged concentrated T-cell conditions.
Collapse
Affiliation(s)
- Alina Kunitskaya
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; The School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - James M Piret
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada; The School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada; Department of Chemical and Biological Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
5
|
Mousavi S, Khazaee-Nasirabadi MH, Seyedmehdi MS, Bazi A, Mirzaee Khalilabadi R. Natural killer cells: a new promising source for developing chimeric antigen receptor anti-cancer cells in hematological malignancies. Leuk Lymphoma 2025; 66:594-616. [PMID: 39656564 DOI: 10.1080/10428194.2024.2438802] [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: 06/10/2024] [Revised: 11/18/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
In recent times, the application of CAR-T cell treatment has significantly progressed, showing auspicious treatment outcomes in hematologic malignancies. However, along with these advances, certain limitations and challenges hurdle the widespread utilization of this technology. Recently, CAR-NK cells have gained attention in cancer treatment, as this approach has an important advantage over CART therapy (i.e. no need for HLA matching) for targeting foreign cells. This review aims to explore the benefits of CAR NK cell therapy, and generation strategies, as well as the challenges and limitations hindering the application of CAR NK cells in experimental studies and trials on hematologic malignancies.
Collapse
Affiliation(s)
- Shahrzad Mousavi
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Maryam Sadat Seyedmehdi
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Islamic Republic of Iran
| | - Ali Bazi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Roohollah Mirzaee Khalilabadi
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
Szarzynski A, Spadiut O, Reisbeck M, Jobst G, Paterson RL, Kamenskaya A, Gateau E, Lesch HP, Henry L, Kozma B. CGT 4.0: a distant dream or inevitable future? Smart process automation is critical to make efficient scalability of CGT manufacturing a reality. Front Bioeng Biotechnol 2025; 13:1563878. [PMID: 40177621 PMCID: PMC11962537 DOI: 10.3389/fbioe.2025.1563878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 03/07/2025] [Indexed: 04/05/2025] Open
Affiliation(s)
- Aleksander Szarzynski
- Research Unit of Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| | - Oliver Spadiut
- Research Unit of Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| | | | | | - Rachel L. Paterson
- Stemmatters Biotecnologia e Medicina Regenerativa SA, Guimarães, Portugal
| | | | | | | | | | - Bence Kozma
- Research Unit of Biochemical Engineering, Institute of Chemical, Environmental and Bioscience Engineering, TU Wien, Vienna, Austria
| |
Collapse
|
7
|
Labara Tirado J, Herdean A, Ralph PJ. The need for smart microalgal bioprospecting. NATURAL PRODUCTS AND BIOPROSPECTING 2025; 15:7. [PMID: 39815030 PMCID: PMC11735771 DOI: 10.1007/s13659-024-00487-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/03/2024] [Indexed: 01/18/2025]
Abstract
Microalgae's adaptability and resilience to Earth's diverse environments have evolved these photosynthetic microorganisms into a biotechnological source of industrially relevant physiological functions and biometabolites. Despite this, microalgae-based industries only exploit a handful of species. This lack of biodiversity hinders the expansion of the microalgal industry. Microalgal bioprospecting, searching for novel biological algal resources with new properties, remains a low throughput and time-consuming endeavour due to inefficient workflows that rely on non-selective sampling, monoalgal culture status and outdated, non-standardized characterization techniques. This review will highlight the importance of microalgal bioprospecting and critically explore commonly employed methodologies. We will also explore current advances driving the next generation of smart algal bioprospecting focusing on novel workflows and transdisciplinary methodologies with the potential to enable high-throughput microalgal biodiscoveries. Images adapted from (Addicted04 in Wikipedia File: Australia on the globe (Australia centered).svg. 2014.; Jin et al. in ACS Appl Bio Mater 4:5080-5089, 2021; Kim et al. in Microchim Acta 189:88, 2022; Tony et al. in Lab on a Chip 15, 19:3810-3810; Thermo Fisher Scientific INC. in CTS Rotea Brochure).
Collapse
Affiliation(s)
- Joan Labara Tirado
- Faculty of Science, Climate Change Cluster (C3), Algal Biotechnology & Biosystems, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Andrei Herdean
- Faculty of Science, Climate Change Cluster (C3), Algal Biotechnology & Biosystems, University of Technology Sydney, Sydney, NSW, 2007, Australia.
| | - Peter J Ralph
- Faculty of Science, Climate Change Cluster (C3), Algal Biotechnology & Biosystems, University of Technology Sydney, Sydney, NSW, 2007, Australia
| |
Collapse
|
8
|
Dobruskin M, Toner G, Kander R. Optimizing cryopreservation strategies for scalable cell therapies: A comprehensive review with insights from iPSC-derived therapies. Biotechnol Prog 2024; 40:e3504. [PMID: 39268839 DOI: 10.1002/btpr.3504] [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: 07/29/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
Abstract
Off-the-shelf cell therapies hold significant curative potential for conditions, such as Parkinson's disease and heart failure. However, these therapies face unique cryopreservation challenges, especially when novel routes of administration, such as intracerebral or epicardial injection, require cryopreservation media that are safe for direct post-thaw administration. Current practices often involve post-thaw washing to remove dimethyl sulfoxide (Me2SO), a cytotoxic cryoprotective agent, which complicates the development and clinical translation of off-the-shelf therapies. To overcome these obstacles, there is a critical need to explore Me2SO-free cryopreservation methods. While such methods typically yield suboptimal post-thaw viability with conventional slow-freeze protocols, optimizing freezing profiles offers a promising strategy to enhance their performance. This comprehensive review examines the latest advancements in cryopreservation techniques across various cell therapy platforms, with a specific case study of iPSC-derived therapies used to illustrate the scalability challenges. By identifying key thermodynamic and biochemical phenomena that occur during freezing, this review aims to identify cell-type independent approaches to improve the efficiency and efficacy of cryopreservation strategies, thereby supporting the widespread adoption and clinical success of off-the-shelf cell therapies.
Collapse
Affiliation(s)
- Michael Dobruskin
- Jefferson Institute for Bioprocessing, Thomas Jefferson University, Spring House, Pennsylvania, USA
| | - Geoffrey Toner
- Jefferson Institute for Bioprocessing, Thomas Jefferson University, Spring House, Pennsylvania, USA
| | - Ronald Kander
- Jefferson Institute for Bioprocessing, Thomas Jefferson University, Spring House, Pennsylvania, USA
| |
Collapse
|
9
|
Matsueda S, Chen L, Li H, Yao H, Yu F. Recent clinical researches and technological development in TIL therapy. Cancer Immunol Immunother 2024; 73:232. [PMID: 39264449 PMCID: PMC11393248 DOI: 10.1007/s00262-024-03793-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 07/29/2024] [Indexed: 09/13/2024]
Abstract
Tumor-infiltrating lymphocyte (TIL) therapy represents a groundbreaking advancement in the solid cancer treatment, offering new hope to patients and their families with high response rates and long overall survival. TIL therapy involves extracting immune cells from a patient's tumor tissue, expanding them ex vivo, and infusing them back into the patient to target and eliminate cancer cells. This revolutionary approach harnesses the power of the immune system to combat cancers, ushering in a new era of T cell-based therapies along with CAR-T and TCR-therapies. In this comprehensive review, we aim to elucidate the remarkable potential of TIL therapy by delving into recent advancements in basic and clinical researches. We highlight on the evolving landscape of TIL therapy as a prominent immunotherapeutic strategy, its multifaceted applications, and the promising outcomes. Additionally, we explore the future horizons of TIL therapy, next-generation TILs, and combination therapy, to overcome the limitations and improve clinical efficacy of TIL therapy.
Collapse
Affiliation(s)
- Satoko Matsueda
- Fresh Wind Biotechnologies USA Inc, 4502 Riverstone Blvd, STE1104, Missouri City, TX, 77459, USA.
| | - Lei Chen
- Department of Neurosurgery, Tianjin Fifth Central Hospital, Tianjin, 300450, China
| | - Hongmei Li
- Department of Oncology, Qingdao University Medical School, Qinddao, 266003, China
| | - Hui Yao
- Fresh Wind Biotechnologies USA Inc, 4502 Riverstone Blvd, STE1104, Missouri City, TX, 77459, USA
| | - Fuli Yu
- Fresh Wind Biotechnologies USA Inc, 4502 Riverstone Blvd, STE1104, Missouri City, TX, 77459, USA
| |
Collapse
|
10
|
Killela P, Herrity K, Frontier L, Horton R, Kurtzberg J, Van’t Hof W. Mitigation of supply chain challenges in cell therapy manufacturing: perspectives from the cord blood alliance. Stem Cells Transl Med 2024; 13:843-847. [PMID: 39169677 PMCID: PMC11386209 DOI: 10.1093/stcltm/szae048] [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: 03/29/2024] [Accepted: 06/15/2024] [Indexed: 08/23/2024] Open
Abstract
Cellular therapies rely on highly specialized supply chains that often depend on single source providers. Public cord blood banks (CBB) manufacturing the first cell therapy to be highly regulated by the FDA and related international agencies are a prime example of being subject to this phenomenon. In addition to banking unrelated donor cord blood units for transplantation, CBBs also source and characterize starting materials for supply to allogeneic cell therapy developers that often employ customized technologies offered by just a small number of manufacturers. As such, these supply chains are especially sensitive to even minor changes which often result in potential major impacts. Regulations can shape supply chain efficiencies, both directly via the definition of restricted technology and process requirements and indirectly by steering strategic business decisions of critical supply or service providers. We present 3 current supply chain issues with different root causes that are swaying efficiencies in cord blood banking and beyond. Specifically, the shortage of Hespan, a common supplement used in cord blood processing, the decision by the provider to stop supporting medical device marking of the Sepax system broadly used in cord blood banking, and a new European ruling on phasing out plasticizers that are critical for providing flexibility to cord blood collection bags, are all threatening downstream supply chain issues for the biologics field. We discuss overcoming these hurdles through the prism of unified mitigation strategies, defined, and implemented by multi-factorial teams and stakeholders, to negotiate resolutions with providers and regulators alike.
Collapse
Affiliation(s)
- Patrick Killela
- Carolinas Cord Blood Bank, Duke University, Durham, NC, United States
- Office of Regulatory Affairs and Quality, School of Medicine, Duke University, Durham, NC, United States
| | | | | | | | - Joanne Kurtzberg
- Carolinas Cord Blood Bank, Duke University, Durham, NC, United States
- Cord Blood Association, Washington, DC, United States
| | - Wouter Van’t Hof
- Cord Blood Association, Washington, DC, United States
- Cleveland Cord Blood Center, Cleveland, OH, United States
| |
Collapse
|
11
|
Hu T, Kumar AR, Luo Y, Tay A. Automating CAR-T Transfection with Micro and Nano-Technologies. SMALL METHODS 2024; 8:e2301300. [PMID: 38054597 DOI: 10.1002/smtd.202301300] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/15/2023] [Indexed: 12/07/2023]
Abstract
Cancer poses a significant health challenge, with traditional treatments like surgery, radiotherapy, and chemotherapy often lacking in cell specificity and long-term curative potential. Chimeric antigen receptor T cell (CAR-T) therapy,utilizing genetically engineered T cells to target cancer cells, is a promising alternative. However, its high cost limits widespread application. CAR-T manufacturing process encompasses three stages: cell isolation and activation, transfection, and expansion.While the first and last stages have straightforward, commercially available automation technologies, the transfection stage lags behind. Current automated transfection relies on viral vectors or bulk electroporation, which have drawbacks such as limited cargo capacity and significant cell disturbance. Conversely, micro and nano-tool methods offer higher throughput and cargo flexibility, yet their automation remains underexplored.In this perspective, the progress in micro and nano-engineering tools for CAR-T transfection followed by a discussion to automate them is described. It is anticipated that this work can inspire the community working on micro and nano transfection techniques to examine how their protocols can be automated to align with the growing interest in automating CAR-T manufacturing.
Collapse
Affiliation(s)
- Tianmu Hu
- Engineering Science Programme, National University of Singapore, Singapore, 117575, Singapore
| | - Arun Rk Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yikai Luo
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Tissue Engineering Programme, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
12
|
Dias J, Garcia J, Agliardi G, Roddie C. CAR-T cell manufacturing landscape-Lessons from the past decade and considerations for early clinical development. Mol Ther Methods Clin Dev 2024; 32:101250. [PMID: 38737799 PMCID: PMC11088187 DOI: 10.1016/j.omtm.2024.101250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
CAR-T cell therapies have consolidated their position over the last decade as an effective alternative to conventional chemotherapies for the treatment of a number of hematological malignancies. With an exponential increase in the number of commercial therapies and hundreds of phase 1 trials exploring CAR-T cell efficacy in different settings (including autoimmunity and solid tumors), demand for manufacturing capabilities in recent years has considerably increased. In this review, we explore the current landscape of CAR-T cell manufacturing and discuss some of the challenges limiting production capacity worldwide. We describe the latest technical developments in GMP production platform design to facilitate the delivery of a range of increasingly complex CAR-T cell products, and the challenges associated with translation of new scientific developments into clinical products for patients. We explore all aspects of the manufacturing process, namely early development, manufacturing technology, quality control, and the requirements for industrial scaling. Finally, we discuss the challenges faced as a small academic team, responsible for the delivery of a high number of innovative products to patients. We describe our experience in the setup of an effective bench-to-clinic pipeline, with a streamlined workflow, for implementation of a diverse portfolio of phase 1 trials.
Collapse
Affiliation(s)
- Juliana Dias
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital NHS Foundation Trust, London NW3 2QG, UK
- Research Department of Haematology, Cancer Institute, University College London, London WC1E 6DD, UK
| | - John Garcia
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital NHS Foundation Trust, London NW3 2QG, UK
- Research Department of Haematology, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Giulia Agliardi
- Centre for Cell, Gene and Tissue Therapeutics, Royal Free Hospital NHS Foundation Trust, London NW3 2QG, UK
- Research Department of Haematology, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Claire Roddie
- Research Department of Haematology, Cancer Institute, University College London, London WC1E 6DD, UK
| |
Collapse
|
13
|
Huso T, Buban K, Van Denakker TA, Haddaway K, Smetana H, Marshall C, Rai H, Ness PM, Bloch EM, Tobian AAR, Crowe EP. Reevaluation of the medical necessity of washed red blood cell transfusion in chronically transfused adults. Transfusion 2024; 64:216-222. [PMID: 38130071 DOI: 10.1111/trf.17690] [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: 11/18/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Washing red blood cell (RBC) units mitigates severe allergic transfusion reactions. However, washing reduces the time to expiration and the effective dose. Automated washing is time- and labor-intensive. A shortage of cell processor tubing sets prompted review of medical necessity for washed RBC for patients previously thought to require washing. STUDY DESIGN AND METHODS A single-center, retrospective study investigated discontinuing wash RBC protocols in chronically transfused adults. In select patients with prior requirements for washing, due to a history of allergic transfusion reactions, trials of unwashed transfusions were performed. Patient demographic, clinical, laboratory, and transfusion data were compiled. The per-unit washing cost was the sum of the tubing set, saline, and technical labor costs. RESULTS Fifteen patients (median age 34 years interquartile range [IQR] 23-53 years, 46.7% female) were evaluated. These patients had been transfused with a median of 531 washed RBC units (IQR 244-1066) per patient over 12 years (IQR 5-18 years), most commonly for recurrent, non-severe allergic reactions. There were no transfusion reactions with unwashed RBCs aside from one patient with one episode of pruritus and another with recurrent pruritus, which was typical even with washed RBC. We decreased the mean number of washed RBC units per month by 72.9% (104 ± 10 vs. 28.2 ± 25.2; p < .0001) and saved US $100.25 per RBC unit. CONCLUSION Washing of RBCs may be safely reconsidered in chronically transfused patients without a history of anaphylaxis. Washing should be implemented judiciously due to potential lack of necessity and logistical/operational challenges.
Collapse
Affiliation(s)
- Tait Huso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristen Buban
- Division of Transfusion Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Tayler A Van Denakker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Pathology and Laboratory Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kathy Haddaway
- Division of Transfusion Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Heather Smetana
- Division of Transfusion Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Christi Marshall
- Division of Transfusion Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Herleen Rai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Paul M Ness
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Elizabeth P Crowe
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
14
|
Motallebnejad P, Kantardjieff A, Cichocki F, Azarin SM, Hu WS. Process engineering of natural killer cell-based immunotherapy. Trends Biotechnol 2023; 41:1314-1326. [PMID: 37142447 PMCID: PMC10523923 DOI: 10.1016/j.tibtech.2023.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 05/06/2023]
Abstract
Cell therapy offers the potential for curative treatment of cancers. Although T cells have been the predominantly used cell type, natural killer (NK) cells have attracted great attention owing to their ability to kill cancer cells and because they are naturally suitable for allogeneic applications. Upon stimulation by cytokines or activation by a target cell, NK cells proliferate and expand their population. These cytotoxic NK cells can be cryopreserved and used as an off-the-shelf medicine. The production process for NK cells thus differs from that of autologous cell therapies. We briefly outline key biological features of NK cells, review the manufacturing technologies for protein biologics, and discuss their adaptation for developing robust NK cell biomanufacturing processes.
Collapse
Affiliation(s)
- Pedram Motallebnejad
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Samira M Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
15
|
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
|
16
|
A Multi-Stage Bioprocess for the Expansion of Rodent Skin-Derived Schwann Cells in Computer-Controlled Bioreactors. Int J Mol Sci 2023; 24:ijms24065152. [PMID: 36982227 PMCID: PMC10049355 DOI: 10.3390/ijms24065152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/03/2023] [Accepted: 02/14/2023] [Indexed: 03/11/2023] Open
Abstract
Regenerative therapies for the treatment of peripheral nerve and spinal cord injuries can require hundreds of millions of autologous cells. Current treatments involve the harvest of Schwann cells (SCs) from nerves; however, this is an invasive procedure. Therefore, a promising alternative is using skin-derived Schwann cells (Sk-SCs), in which between 3–5 million cells can be harvested from a standard skin biopsy. However, traditional static planar culture is still inefficient at expanding cells to clinically relevant numbers. As a result, bioreactors can be used to develop reproducible bioprocesses for the large-scale expansion of therapeutic cells. Here, we present a proof-of-concept SC manufacturing bioprocess using rat Sk-SCs. With this integrated process, we were able to simulate a feasible bioprocess, taking into consideration the harvest and shipment of cells to a production facility, the generation of the final cell product, and the cryopreservation and shipment of cells back to the clinic and patient. This process started with 3 million cells and inoculated and expanded them to over 200 million cells in 6 days. Following the harvest and post-harvest cryopreservation and thaw, we were able to maintain 150 million viable cells that exhibited a characteristic Schwann cell phenotype throughout each step of the process. This process led to a 50-fold expansion, producing a clinically relevant number of cells in a 500 mL bioreactor in just 1 week, which is a dramatic improvement over current methods of expansion.
Collapse
|
17
|
Lamers-Kok N, Panella D, Georgoudaki AM, Liu H, Özkazanc D, Kučerová L, Duru AD, Spanholtz J, Raimo M. Natural killer cells in clinical development as non-engineered, engineered, and combination therapies. J Hematol Oncol 2022; 15:164. [DOI: 10.1186/s13045-022-01382-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/26/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractNatural killer (NK) cells are unique immune effectors able to kill cancer cells by direct recognition of surface ligands, without prior sensitization. Allogeneic NK transfer is a highly valuable treatment option for cancer and has recently emerged with hundreds of clinical trials paving the way to finally achieve market authorization. Advantages of NK cell therapies include the use of allogenic cell sources, off-the-shelf availability, and no risk of graft-versus-host disease (GvHD). Allogeneic NK cell therapies have reached the clinical stage as ex vivo expanded and differentiated non-engineered cells, as chimeric antigen receptor (CAR)-engineered or CD16-engineered products, or as combination therapies with antibodies, priming agents, and other drugs. This review summarizes the recent clinical status of allogeneic NK cell-based therapies for the treatment of hematological and solid tumors, discussing the main characteristics of the different cell sources used for NK product development, their use in cell manufacturing processes, the engineering methods and strategies adopted for genetically modified products, and the chosen approaches for combination therapies. A comparative analysis between NK-based non-engineered, engineered, and combination therapies is presented, examining the choices made by product developers regarding the NK cell source and the targeted tumor indications, for both solid and hematological cancers. Clinical trial outcomes are discussed and, when available, assessed in comparison with preclinical data. Regulatory challenges for product approval are reviewed, highlighting the lack of specificity of requirements and standardization between products. Additionally, the competitive landscape and business field is presented. This review offers a comprehensive overview of the effort driven by biotech and pharmaceutical companies and by academic centers to bring NK cell therapies to pivotal clinical trial stages and to market authorization.
Collapse
|
18
|
Mfarrej B, Vicari O, Ouffai S, Malenfant C, Granata A, Thevenet S, Chabannon C, Lemarié C, Calmels B. Sepax-2 cell processing device: a study assessing reproducibility of concentrating thawed hematopoietic progenitor cells. J Transl Med 2022; 20:503. [PMID: 36329460 PMCID: PMC9632571 DOI: 10.1186/s12967-022-03703-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
Background Autologous hematopoietic progenitor cell (HPC) transplantation is currently the standard of care for a fraction of patients with newly diagnosed myelomas and relapsed or refractory lymphomas. After high-dose chemotherapy, cryopreserved HPC are either infused directly after bedside thawing or washed and concentrated before infusion. We previously reported on the comparability of washing/concentrating HPC post-thaw vs. infusion without manipulation in terms of hematopoietic engraftment, yet settled for the prior favoring cell debris and DMSO removal. For almost two decades, automation of this critical step of washing/concentrating cells has been feasible. As part of continuous process verification, we aim to evaluate reproducibility of this procedure by assessing intra-batch and inter-batch variability upon concentration of thawed HPC products using the Sepax 2 S-100 cell separation system. Methods Autologous HPC collected from the same patient were thawed and washed either in two batches processed within a 3-4 h interval and immediately infused on the same day (intra-batch, n = 45), or in two batches on different days (inter-batch, n = 49) for those patients requiring 2 or more high-dose chemotherapy cycles. Quality attributes assessed were CD34+ cell recovery, viability and CD45+ viability; CFU assay was only performed for allogeneic grafts. Results Intra-batch and inter-batch median CD34+ cell recovery was comparable (75% vs. 73% and 77% vs. 77%, respectively). Similarly, intra-batch and inter-batch median CD45+ cell viability was comparable (79% vs. 80% and 79% vs. 78%, respectively). Bland-Altman analysis describing agreement between batches per patient revealed a bias close to 0%. Additionally, lower HPC recoveries noted in batch 1 were noted as well in batch 2, regardless of the CD34+ cell dose before cryopreservation, both intra- and inter-batch, suggesting that the quality of the collected product plays an important role in downstream recovery. Intrinsic (high mature and immature granulocyte content) and extrinsic (delay between apheresis and cryopreservation) variables of the collected product resulted in a significantly lower CD45+ viability and CD34+ cell recovery upon thawing/washing. Conclusions Automated post-thaw HPC concentration provides reproducible cell recoveries and viabilities between different batches. Implications of this work go beyond HPC to concentrate cell suspension/products during manufacturing of cell and gene therapy products. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-022-03703-1.
Collapse
Affiliation(s)
- Bechara Mfarrej
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| | - Olivier Vicari
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| | - Sarah Ouffai
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| | - Carine Malenfant
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| | - Angela Granata
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| | - Sophie Thevenet
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| | - Christian Chabannon
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France ,grid.5399.60000 0001 2176 4817Aix-Marseille Université Faculté des Sciences Médicales et Paramédicales, Marseille, France
| | - Claude Lemarié
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| | - Boris Calmels
- grid.418443.e0000 0004 0598 4440Centre de Thérapie Cellulaire, Institut Paoli-Calmettes, Marseille, France ,Module Biothérapies du Centre d’Investigation Clinique de Marseille, AP-HM, Aix- Marseille Université, Institut Paoli-Calmettes, CBT-1409 Inserm, Marseille, France
| |
Collapse
|
19
|
Mawji I, Roberts EL, Dang T, Abraham B, Kallos MS. Challenges and Opportunities in Downstream Separation Processes for Mesenchymal Stromal Cells Cultured in Microcarrier-based Stirred Suspension Bioreactors. Biotechnol Bioeng 2022; 119:3062-3078. [PMID: 35962467 DOI: 10.1002/bit.28210] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/27/2022] [Accepted: 08/11/2022] [Indexed: 11/08/2022]
Abstract
Mesenchymal stromal cells (MSC) are a promising platform for regenerative medicine applications because of their multi-lineage differentiation abilities and ease of collection, isolation, and growth ex-vivo. To meet the demand for clinical applications, large scale manufacturing will be required using three-dimension culture platforms in vessels such as stirred suspension bioreactors. As MSCs are an adherent cell type, microcarriers are added to the culture to increase the available surface area for attachment and growth. Although extensive research has been performed on efficiently culturing MSCs using microcarriers, challenges persist in downstream processing including harvesting, filtration, and volume reduction which all play a critical role for the translation of cell therapies to the clinic. The objective of this review is to assess the current state of downstream technologies available for microcarrier-based MSC cultures. This includes a review of current research within the three stages: harvesting, filtration, and volume reduction. Using this information, a downstream process for MSCs is proposed which can be applied for a wide range of applications. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Inaara Mawji
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Erin L Roberts
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Tiffany Dang
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Brett Abraham
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Biomedical Engineering, Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
20
|
Li M, Morse B, Kassim S. Development and clinical translation considerations for the next wave of gene modified hematopoietic stem and progenitor cells therapies. Expert Opin Biol Ther 2022; 22:1177-1191. [PMID: 35833356 DOI: 10.1080/14712598.2022.2101361] [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: 11/04/2022]
Abstract
INTRODUCTION Consistent and reliable manufacture of gene modified hematopoietic stem and progenitor cell (HPSC) therapies will be of the utmost importance as they become more mainstream and address larger populations. Robust development campaigns will be needed to ensure that these products will be delivered to patients with the highest quality standards. AREAS COVERED Through publicly available manuscripts, press releases, and news articles - this review touches on aspects related to HSPC therapy, development, and manufacturing. EXPERT OPINION Recent advances in genome modification technology coupled with the longstanding clinical success of HSPCs warrants great optimism for the next generation of engineered HSPC-based therapies. Treatments for some diseases that have thus far been intractable now appear within reach. Reproducible manufacturing will be of critical importance in delivering these therapies but will be challenging due to the need for bespoke materials and methods in combination with the lack of off-the-shelf solutions. Continued progress in the field will manifest in the form of industrialization which currently requires attention and resources directed toward the custom reagents, a focus on closed and automated processes, and safer and more precise genome modification technologies that will enable broader, faster, and safer access to these life-changing therapies.
Collapse
Affiliation(s)
| | - Brent Morse
- Dark Horse Consulting Group, Walnut Creek, CA, USA
| | | |
Collapse
|
21
|
Lu X, Ai Y. Automatic Microfluidic Cell Wash Platform for Purifying Cells in Suspension: Puriogen. Anal Chem 2022; 94:9424-9433. [PMID: 35658406 DOI: 10.1021/acs.analchem.2c01616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cell wash is an essential cell sample preparation procedure to eliminate or minimize interfering substances for various subsequent cell analyses. The commonly used cell wash method is centrifugation which separates cells from other biomolecules in a solution with manual pipetting and has many drawbacks such as being labor-intensive and time-consuming with substantial cell loss and cell clumping. To overcome these issues, a centrifuge-free and automatic cell wash platform for cell purity generation, termed Puriogen, has been developed in this work. Compared with other developed products such as AcouWash, Puriogen can process samples with a high throughput of above 500 μL/min. Puriogen utilizes a uniquely designed inertial microfluidic device to complete the automatic cell wash procedure. One single-cell wash procedure with the Puriogen platform can remove more than 90% ambient proteins and nucleic acids from the cell sample. It can also remove most of the residual fluorescent dye after cell staining to significantly reduce the background signals for subsequent cell analysis. By removing the dead cell debris, it can increase the live cell percentage in the sample by 2-fold. Moreover, the percentage of single-cell population is also increased by 20% because of further disassociation of small-cell aggregates (e.g., doublets and triplets) into singlets. To freely adjust cell concentrations, the Puriogen platform can concentrate cells 5 times in a single flow-through process. The presented Puriogen cell wash solution has broad applications in cell preparation with its excellent simplicity in operation and wash efficiency, especially in single-cell sequencing.
Collapse
Affiliation(s)
- Xiaoguang Lu
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| | - Ye Ai
- Pillar of Engineering Product Development, Singapore University of Technology and Design, Singapore 487372, Singapore
| |
Collapse
|
22
|
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]
|