Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 59
2.
Nature ; 614(7949): 635-648, 2023 02.
Article En | MEDLINE | ID: mdl-36813894

The remarkable clinical activity of chimeric antigen receptor (CAR) therapies in B cell and plasma cell malignancies has validated the use of this therapeutic class for liquid cancers, but resistance and limited access remain as barriers to broader application. Here we review the immunobiology and design principles of current prototype CARs and present emerging platforms that are anticipated to drive future clinical advances. The field is witnessing a rapid expansion of next-generation CAR immune cell technologies designed to enhance efficacy, safety and access. Substantial progress has been made in augmenting immune cell fitness, activating endogenous immunity, arming cells to resist suppression via the tumour microenvironment and developing approaches to modulate antigen density thresholds. Increasingly sophisticated multispecific, logic-gated and regulatable CARs display the potential to overcome resistance and increase safety. Early signs of progress with stealth, virus-free and in vivo gene delivery platforms provide potential paths for reduced costs and increased access of cell therapies in the future. The continuing clinical success of CAR T cells in liquid cancers is driving the development of increasingly sophisticated immune cell therapies that are poised to translate to treatments for solid cancers and non-malignant diseases in the coming years.


Immunotherapy, Adoptive , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Genetic Therapy/methods , Genetic Therapy/trends , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Immunotherapy, Adoptive/trends , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes/immunology , Tumor Microenvironment , B-Lymphocytes/pathology
3.
Nature ; 615(7951): 315-322, 2023 03.
Article En | MEDLINE | ID: mdl-36755094

Further advances in cell engineering are needed to increase the efficacy of chimeric antigen receptor (CAR) and other T cell-based therapies1-5. As T cell differentiation and functional states are associated with distinct epigenetic profiles6,7, we hypothesized that epigenetic programming may provide a means to improve CAR T cell performance. Targeting the gene that encodes the epigenetic regulator ten-eleven translocation 2 (TET2)8 presents an interesting opportunity as its loss may enhance T cell memory9,10, albeit not cause malignancy9,11,12. Here we show that disruption of TET2 enhances T cell-mediated tumour rejection in leukaemia and prostate cancer models. However, loss of TET2 also enables antigen-independent CAR T cell clonal expansions that may eventually result in prominent systemic tissue infiltration. These clonal proliferations require biallelic TET2 disruption and sustained expression of the AP-1 factor BATF3 to drive a MYC-dependent proliferative program. This proliferative state is associated with reduced effector function that differs from both canonical T cell memory13,14 and exhaustion15,16 states, and is prone to the acquisition of secondary somatic mutations, establishing TET2 as a guardian against BATF3-induced CAR T cell proliferation and ensuing genomic instability. Our findings illustrate the potential of epigenetic programming to enhance T cell immunity but highlight the risk of unleashing unchecked proliferative responses.


Basic-Leucine Zipper Transcription Factors , Cell Proliferation , DNA-Binding Proteins , Dioxygenases , Immunotherapy, Adoptive , Lymphocyte Activation , Receptors, Chimeric Antigen , T-Lymphocytes , Humans , Male , Cell Differentiation/genetics , Dioxygenases/metabolism , DNA-Binding Proteins/metabolism , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Leukemia/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Epigenesis, Genetic , Immunologic Memory , Basic-Leucine Zipper Transcription Factors/metabolism
5.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article En | MEDLINE | ID: mdl-35055086

Chimeric-antigen-receptor (CAR)-T-cell therapy is already widely used to treat patients who are relapsed or refractory to chemotherapy, antibodies, or stem-cell transplantation. Multiple myeloma still constitutes an incurable disease. CAR-T-cell therapy that targets BCMA (B-cell maturation antigen) is currently revolutionizing the treatment of those patients. To monitor and improve treatment outcomes, methods to detect CAR-T cells in human peripheral blood are highly desirable. In this study, three different detection reagents for staining BCMA-CAR-T cells by flow cytometry were compared. Moreover, a quantitative polymerase chain reaction (qPCR) to detect BCMA-CAR-T cells was established. By applying a cell-titration experiment of BCMA-CAR-T cells, both methods were compared head-to-head. In flow-cytometric analysis, the detection reagents used in this study could all detect BCMA-CAR-T cells at a similar level. The results of false-positive background staining differed as follows (standard deviation): the BCMA-detection reagent used on the control revealed a background staining of 0.04% (±0.02%), for the PE-labeled human BCMA peptide it was 0.25% (±0.06%) and for the polyclonal anti-human IgG antibody it was 7.2% (±9.2%). The ability to detect BCMA-CAR-T cells down to a concentration of 0.4% was similar for qPCR and flow cytometry. The qPCR could detect even lower concentrations (0.02-0.01%). In summary, BCMA-CAR-T-cell monitoring can be reliably performed by both flow cytometry and qPCR. In flow cytometry, reagents with low background staining should be preferred.


B-Cell Maturation Antigen/metabolism , Flow Cytometry , Polymerase Chain Reaction , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/metabolism , B-Cell Maturation Antigen/genetics , Biomarkers , Flow Cytometry/methods , Flow Cytometry/standards , Humans , Immunophenotyping , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Polymerase Chain Reaction/methods , Polymerase Chain Reaction/standards , Real-Time Polymerase Chain Reaction , Receptors, Chimeric Antigen/genetics , Reproducibility of Results , Sensitivity and Specificity , T-Lymphocytes/immunology
6.
Int J Mol Sci ; 22(19)2021 Oct 07.
Article En | MEDLINE | ID: mdl-34639168

Over the last decade remarkable progress has been made in enhancing the efficacy of CAR T therapies. However, the clinical benefits are still limited, especially in solid tumors. Even in hematological settings, patients that respond to CAR T therapies remain at risk of relapsing due to several factors including poor T-cell expansion and lack of long-term persistence after adoptive transfer. This issue is even more evident in solid tumors, as the tumor microenvironment negatively influences the survival, infiltration, and activity of T-cells. Limited persistence remains a significant hindrance to the development of effective CAR T therapies due to several determinants, which are encountered from the cell manufacturing step and onwards. CAR design and ex vivo manipulation, including culture conditions, may play a pivotal role. Moreover, previous chemotherapy and lymphodepleting treatments may play a relevant role. In this review, the main causes for decreased persistence of CAR T-cells in patients will be discussed, focusing on the molecular mechanisms underlying T-cell exhaustion. The approaches taken so far to overcome these limitations and to create exhaustion-resistant T-cells will be described. We will also examine the knowledge gained from several key clinical trials and highlight the molecular mechanisms determining T-cell stemness, as promoting stemness may represent an attractive approach to improve T-cell therapies.


Immunotherapy, Adoptive/standards , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Animals , Humans , Neoplasms/immunology
7.
Cancer Cell ; 39(7): 885-888, 2021 07 12.
Article En | MEDLINE | ID: mdl-34256903

CAR T cell therapy successes are challenged by several mechanisms of resistance including the development of dysfunctional states such as exhaustion. The features of CAR T cell exhaustion, its role in limiting the efficacy of CAR T therapy in both liquid and solid malignancies, and potential strategies to overcome it are discussed.


Drug Resistance, Neoplasm , Immunotherapy, Adoptive/standards , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , Humans , Immunotherapy, Adoptive/methods , Neoplasms/immunology , Tumor Microenvironment
8.
Bull Cancer ; 108(12S): S20-S25, 2021 Dec.
Article Fr | MEDLINE | ID: mdl-34215432

The SARS-CoV-2 (COVID-19) pandemic has rapidly impacted cell therapy activities across the globe. Not only was this, unexpected event, a threat to patients who had previously received hematopoietic cell transplantation or other cell therapy such as CAR-T cells, but also, it was responsible for a disruption of cell therapy activities due to the danger of the virus and to the lack of solid scientific data on the management of patients and donors. The Francophone Society of Bone Marrow Transplantation and Cellular Therapy (SFGM-TC) devoted a workshop to issue useful recommendations in such an unexpected event in order to harmonize the actions of all the actors involved in cellular therapy programs so that we can collectively face, in the future, the challenges that could threaten our patients. This work is not specifically dedicated to the SARS-CoV-2 outbreak, but the latter has been used as a concrete example of an unexpected event to build up our recommendations.


Bone Marrow Transplantation/standards , COVID-19/epidemiology , Cell Transplantation/standards , Pandemics , Cryopreservation , Health Services Accessibility , Hematopoietic Stem Cell Transplantation/standards , Humans , Immunotherapy, Adoptive/standards , Receptors, Chimeric Antigen/therapeutic use , Societies, Medical , Tissue Donors
9.
Clin Toxicol (Phila) ; 59(5): 376-385, 2021 May.
Article En | MEDLINE | ID: mdl-33576256

INTRODUCTION: New cancer treatments with immunotherapy have led to unique toxicities affecting cancer patients. As cancer-related visits to the emergency department increase, the emergency physician and the medical toxicologist should be aware of immunotherapy-related toxicities. In this review we discuss immune related adverse events (irAEs) from chimeric antigen receptor T (CAR-T) cell therapy and immune checkpoint inhibitors (ICI). DISCUSSION: While presentation of the irAEs may mimic common conditions, it is important to recognize them as they may be life-threatening. A thorough history and examination of the patient, including their cancer treatment history in the past year is crucial. Conditions such as cytokine release syndrome (CRS) and immune effector cell associated neurotoxicity syndrome (ICANS), which can occur after CAR-T treatment, can progress rapidly to a fatal outcome if not recognized and managed in a timely manner. ICI can affect any organ system and irAEs may present like a typical autoimmune disease of the affected organ. While most of the irAEs we discuss in this review will benefit from treatment with glucocorticoids, it is important to know the grade of the condition, as it will determine the treatment dose, route and further management considerations. CONCLUSION: Patients experiencing irAEs from ICI and CAR-T can present with subtle symptoms that can rapidly progress if not recognized early. The emergency physician and the medical toxicologist should keep in mind these toxicities and the patient's oncologic history to adequately recognize and manage these conditions.


Antidotes/therapeutic use , Emergency Medical Services/standards , Immune Checkpoint Inhibitors/toxicity , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/standards , Neoplasms/drug therapy , Neurotoxicity Syndromes/drug therapy , Practice Guidelines as Topic , Adult , Aged , Aged, 80 and over , Female , Humans , Immunologic Factors/therapeutic use , Immunologic Factors/toxicity , Male , Middle Aged , Neoplasms/immunology , Neurotoxicity Syndromes/etiology , United States
10.
Bull Cancer ; 107(12S): S193-S201, 2020 Dec.
Article Fr | MEDLINE | ID: mdl-33187683

CAR T-cells are anti-cancer immunocellular therapy drugs that involve reprogramming the patient's T-cells using a transgene encoding a chimeric antigen receptor (CAR). Although CAR T-cells are cellular therapies, the organization for manufacturing and delivering these medicinal products is in many ways different from the one for hematopoietic cell grafts or donor lymphocyte infusions. The implementation of this innovative therapy is recent and requires close coordination between clinical teams, the therapeutic apheresis unit, the cell therapy unit, the pharmaceutical laboratory, and pharmacy. Apart from the regulatory texts, which are regularly modified, and the specific requirements of each pharmaceutical laboratory, there is currently no guide to help the centers initiating their activity and there is no specific indicator to assess the quality of the CAR T-cell activity in each center. The purpose of the current harmonization workshop is to clarify the regulatory prerequisites warranted for a center to have a CAR T-cell activity and to propose recommendations for implementing quality tools, in particular indicators, and allowing their sharing.


Immunotherapy, Adoptive/standards , Quality Assurance, Health Care , Receptors, Chimeric Antigen , Accreditation , Congresses as Topic/organization & administration , France , Health Personnel/education , Humans , Immunotherapy, Adoptive/legislation & jurisprudence , Societies, Medical
11.
Cancer Immunol Immunother ; 69(7): 1165-1175, 2020 Jul.
Article En | MEDLINE | ID: mdl-32130452

Chimeric antigen receptor T cell (CAR-T) therapy is a novel approved treatment for hematological malignancies, still under development for solid tumors. Here, we use a rate equation-based mathematical model to discover regimens and schedules that maintain efficacy while potentially reducing toxicity by decreasing the amount of CAR-T infused. Tested on an in vivo murine model of spontaneous breast cancer, we show that our mathematical model accurately recapitulates in vivo tumor growth results achieved in the previous experiments. Moreover, we use the mathematical model to predict results of new therapy schedules and successfully prospectively validated these predictions in the in vivo. We conclude that using one tenth and even one percent of a full CAR-T dose used in preclinical trials can achieve efficacious results similar to full dose treatment.


Disease Models, Animal , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Mammary Neoplasms, Experimental/therapy , Models, Theoretical , Receptors, Antigen, T-Cell/immunology , Animals , Female , Mammary Neoplasms, Experimental/immunology , Mice
12.
Cancer Immunol Res ; 8(6): 743-755, 2020 06.
Article En | MEDLINE | ID: mdl-32209638

T-cell receptor (TCR)-modified T-cell gene therapy can target a variety of extracellular and intracellular tumor-associated antigens, yet has had little clinical success. A potential explanation for limited antitumor efficacy is a lack of T-cell activation in vivo We postulated that expression of proinflammatory cytokines in TCR-modified T cells would activate T cells and enhance antitumor efficacy. We demonstrate that expression of interleukin 18 (IL18) in tumor-directed TCR-modified T cells provides a superior proinflammatory signal than expression of interleukin 12 (IL12). Tumor-targeted T cells secreting IL18 promote persistent and functional effector T cells and a proinflammatory tumor microenvironment. Together, these effects augmented overall survival of mice in the pmel-1 syngeneic tumor model. When combined with sublethal irradiation, IL18-secreting pmel-1 T cells were able to eradicate tumors, whereas IL12-secreting pmel-1 T cells caused toxicity in mice through excessive cytokine secretion. In another xenograft tumor model, IL18 secretion enhanced the persistence and antitumor efficacy of NY-ESO-1-reactive TCR-modified human T cells as well as overall survival of tumor-bearing mice. These results demonstrate a rationale for optimizing the efficacy of TCR-modified T-cell cancer therapy through expression of IL18.See related commentary by Wijewarnasuriya et al., p. 732.


CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/standards , Interleukin-12/metabolism , Interleukin-18/metabolism , Lymphocyte Activation/immunology , Melanoma/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Humans , Immunotherapy, Adoptive/methods , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Knockout , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Farm. hosp ; 44(1): 26-31, ene.-feb. 2020. tab
Article Es | IBECS | ID: ibc-187489

El desarrollo y la comercialización de medicamentos de terapia celular con células T con receptor de antígeno quimérico (CAR-T) suponen un nuevo reto para la farmacia hospitalaria en España. El objetivo de este artículo es revisar los aspectos clave de estos medicamentos y describir el papel del farmacéutico oncohematológico dentro del equipo clínico multidisciplinar en las diferentes fases del proceso transversal que implica el tratamiento con medicamentos CAR-T, desde la indicación hasta el seguimiento a corto y largo plazo de los pacientes tratados con este tipo de terapias, con una importante mención al manejo de sus principales efectos adversos. La terapia tipo CAR-T ofrece al farmacéutico hospitalario la oportunidad de trabajar en estrecha colaboración con el resto de los profesionales clínicos implicados en el proceso, permitiendo su contribución en el desarrollo de procedimientos, guías de práctica clínica de abordaje global y estableciendo puntos de partida para afrontar tratamientos futuros de complejidad similar e incluso mejorar procesos base anteriormente establecidos


The development and commercialization of cell therapy drugs with chimeric antigen receptor T cells (CAR-T) represent a new challenge for Spain's hospital pharmacy. The aim of this article is to review the key aspects of these medicines and to describe the oncohematological pharmacist's role within the multidisciplinary clinical team. This includes the different phases in the transversal process that involves a therapy with CAR-T medicines, ranging from indication to short and long term follow-up of patients treated with this type of therapy, and emphasizing on the management of its main adverse effects. CAR-T therapy offers the hospital pharmacist the opportunity to work closely with the rest of the clinical professionals involved in the process, allowing their contribution to the development of procedures, clinical practice guidelines of global approach, and establishing starting points when facing future therapies of similar complexity -and even improving previously established basic processes


Humans , Pharmacists/organization & administration , Pharmacists/standards , Immunotherapy, Adoptive/methods , Pharmacy Service, Hospital , Immunotherapy, Adoptive/standards , Medical Device Legislation/standards , Pharmacovigilance
14.
Bull Cancer ; 107(1S): S85-S93, 2020 Jan.
Article Fr | MEDLINE | ID: mdl-31547937

The extraordinary and unexpected success of cellular immunotherapy using genetically engineered T-cells to express a chimeric antigen receptor (CAR) targeting CD19, in the treatment of refractory or relapsing B-hematological malignancies, has provided a real therapeutic hope. Indeed, remission rates reach more than 80 % in patients at a stage, without any other possibilities of treatment, notably in the child's acute lymphoblastic leukemia. These results, initially resulting from academic research, led to Food and Drug accreditation for market access of two innovative autologous therapy drugs, Kimryah® and Yescarta®. Based on the impressive clinical results, mainly so far in hematological malignancies (LAL, MM, LBDGC, etc.), the development of several types of cells expressing a CAR receptor suggests a wide range of future applications, particularly in the field of solid tumors. However, while the development of CAR-T cells now appears to be in the hands of private pharmaceuticals companies, the logistical constraints, the cryopreservation and the very high cost of these personalized medicines may ultimately limit their use. The development of academic productions by CAR-T cells could bypass some of these disadvantages. The strong innovation capacity of healthcare institutions associated with research units allows them to identify the ideal tumor target and efficient performing cells. Thus, authorized production platforms could allow for shorter administration times and reasonable production costs for national health systems. The aim of this workshop is to identify the requirements for the academic production of CAR-T cells, while respecting the research standards useful to establish proof of concept, but also at the preclinical development stage, leading in fine to the manufacture, through an authorized pharmaceutical establishment, of the innovative therapy drug, and in accordance with Good Manufacturing Practice (GMP). The ultimate goal is to make these innovative and high-performance medicines available to as many patients as possible.


Immunotherapy, Adoptive/standards , Academic Medical Centers , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Cell Culture Techniques , Cell Line , Drug Industry , Europe , France , Genetic Vectors/genetics , Humans , Immunotherapy, Adoptive/economics , Immunotherapy, Adoptive/methods , Lentivirus/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Cell Antigen Receptor Specificity , Transgenes
16.
Methods Mol Biol ; 2086: 1-10, 2020.
Article En | MEDLINE | ID: mdl-31707664

Immunotherapy has been growing in the past decade as a therapeutic alternative for cancer treatment. In this chapter, we deal with CAR-T cells, genetically engineered autologous T cells to express a chimeric receptor specific for an antigen expressed on tumor cell surface. While this type of personalized therapy is revolutionizing cancer treatment, especially B cell malignancies, it has some challenging limitations. Here, we discuss the basic immunological and technological aspects of CAR-T cell therapy, the limitations that have compromised its efficacy and safety, and the current proposed strategies to overcome these limitations, thereby allowing for greater therapeutic application of CAR-T cells.


Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antigens, Neoplasm , Cellular Microenvironment/genetics , Cellular Microenvironment/immunology , Genetic Engineering , Humans , Immunomodulation , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Immunotherapy, Adoptive/trends , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Research Design
17.
Methods Mol Biol ; 2086: 139-150, 2020.
Article En | MEDLINE | ID: mdl-31707673

Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the immunotherapy field with high rate complete responses especially for hematological diseases. Despite the diversity of tumor specific-antigens, the manufacturing process is consistent and involves multiple steps, including selection of T cells, activation, genetic modification, and in vitro expansion. Among these complex manufacturing phases, the choice of culture system to generate a high number of functional cells needs to be evaluated and optimized. Flasks, bags, and rocking motion bioreactor are the most used platforms for CAR-T cell expansion in the current clinical trials but are far from being standardized. New processing options are available and a systematic effort seeking automation, standardization and the increase of production scale, would certainly help to bring the costs down and ultimately democratize this personalized therapy. In this review, we describe different cell expansion platforms available as well as the quality control requirements for clinical-grade production.


Bioreactors , Cell Culture Techniques , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/metabolism , Cell Culture Techniques/standards , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Quality Control , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Workflow
18.
Methods Mol Biol ; 2086: 151-163, 2020.
Article En | MEDLINE | ID: mdl-31707674

Cell expansion is typically a long and labor-intensive step in CAR-T cell manufacture. The Xuri Cell Expansion System (CES) W25 semiautomates this step while functionally closing the process. Cells for autologous or allogeneic cell therapies are cultured inside a single-use Xuri Cellbag™ bioreactor. Wave-induced agitation, performed by a rocking Base Unit, transfers gas and mixes the culture. The integral UNICORN™ software allows customization of culture conditions and media perfusion schedules. Culture volumes can range from 300 mL to 25 L, making the Xuri CES W25 system suitable for both scale-up and scale-out manufacturing processes. CAR-T cell therapies have been successfully generated using the Xuri CES W25 system, which reduces manual labor compared with static culturing methods. This chapter details how to initiate a culture, install the Xuri CES W25, and install a 2 L Cellbag bioreactor. Protocols on inoculation, monitoring, and sampling are also outlined in this chapter.


Bioreactors , Cell Culture Techniques , Immunotherapy, Adoptive , T-Lymphocytes , Automation, Laboratory , Bioreactors/standards , Cell Count , Cell Culture Techniques/instrumentation , Cell Culture Techniques/standards , Cell Survival , Culture Media , Genetic Therapy/methods , Genetic Therapy/standards , Humans , Immunophenotyping , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Methods Mol Biol ; 2086: 165-177, 2020.
Article En | MEDLINE | ID: mdl-31707675

The G-Rex cell culture platform is based on a gas-permeable membrane technology that provides numerous advantages over other systems. Conventional bioreactor platform technologies developed for large scale mammalian cell expansion are typically constrained by the mechanics of delivering oxygen to an expanding cell population. These systems often utilize complex mechanisms to enhance oxygen delivery, such as stirring, rocking, or perfusion, which adds to expense and increases their overall risk of failure. On the other hand, G-Rex gas-permeable membrane-based bioreactors provide a more physiologic environment and avoid the risk and cost associated with more complex systems. The result is a more robust, interacting cell population established through unlimited oxygen and nutrients that are available on demand. By removing the need to actively deliver oxygen, these bioreactors can hold larger medium volumes (more nutrients) which allows the cells to reach a maximum density without complexity or need for media exchange. This platform approach is scaled to meet the needs of research through commercial production with a direct, linear correlation between small and large devices. In the G-Rex platform, examples of cell expansion (9-14 day duration) include; CAR-T cells, which have atypical harvest density of 20-30 × 106/cm2 (or 2-3 × 109 cells in a 100 cm2 device); NK cells, which have a typical harvest density of 20-30 × 106/cm2 (or 2-3 × 109 cells in a 100 cm2 device) and numerous other cell types that proliferate without the need for intervention or complex processes normally associated with large scale culture. Here we describe the methods and concepts used to optimize expansion of various cell types in the static G-Rex bioreactor platform.


Bioreactors , Cell Culture Techniques , Immunotherapy, Adoptive , T-Lymphocytes , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Culture Techniques/standards , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Methods Mol Biol ; 2086: 181-194, 2020.
Article En | MEDLINE | ID: mdl-31707676

Lentiviral vectors are being used in a growing number of clinical applications, including T cell immunotherapy for cancer. As this new technology moves forward, a safety concern is the inadvertent recombination and subsequent development of a replication-competent lentivirus (RCL) during the manufacture of the vector material. To assess this risk, regulators have required screening of T cell products infused into patients for RCL. Since vector particles have many of the proteins and nucleotide sequences found in RCL, a biologic assay has proven the most sensitive method for RCL detection. As regulators have required screening of up to 108 cells per T cell product, this method described a procedure for assessing RCL contamination of large-volume T cell products.


Genetic Vectors/genetics , Immunotherapy, Adoptive/standards , Lentivirus/genetics , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes , Transduction, Genetic , Biological Assay , Cell Line , Clinical Trials as Topic , Humans , Immunotherapy, Adoptive/methods , Neoplasms/pathology , Quality Assurance, Health Care , Quality Control , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Virus Replication
...