Abstract
Whether as a cure or bridge to transplant, chimeric antigen receptor (CAR)-T cell therapies have shown dramatic outcomes for the treatment of hematologic malignancies, and particularly relapsed/refractory B cell leukemia and lymphoma. However, these therapies are not effective for all patients, and are not without toxicities. The challenge now is to optimize these products and their manufacture. The manufacturing process is complex and subject to numerous variabilities at each step. These variabilities can affect the critical quality attributes of the final product, and this can ultimately impact clinical outcomes. This review will focus on optimizing the manufacturing variables that can impact the safety, purity, potency, consistency and durability of CAR-T cells.
Keywords: CAR-T cells, manufacturing, critical quality attributes
Introduction
The FDA approval of two chimeric antigen receptor (CAR)-T cell products in 2017 marked a historic moment for the field of cellular therapy as the first gene therapies for the treatment of cancer were made available in the United States. Since then, CAR-T cell clinical trials have rapidly expanded with targets for both solid tumors and hematologic malignancies.1 Initial findings of these clinical trials have been promising.2–4 Optimizing these therapies, however, remains a challenge as seen with the variable efficacy, persistence, and adverse effects of these cells.
Manufacturing CAR-T cells commonly occurs over 7–12 days and begins with an apheresis collection, followed by target cell selection and enrichment, CAR gene transfer, and cell expansion. At each step in manufacturing, several seemingly trivial details can impact the critical quality attributes of the final product, including safety, purity, potency, consistency, and durability. Safety includes not only negative sterility testing, but also producing a product lacking oncogenic or viral replicative potential; purity describes a product with a high concentration of CAR-T cells and minimal contaminating cells; consistency includes producing products that meet lot release criteria time after time; potency is characterized by a product capable of eliminating tumor cells; and durability is indicated by a product that persists in circulation and maintains its anti-tumor effects. At most steps during manufacture, one or more of these parameters may be compromised, but careful selection of methods and procedures may minimize these vulnerabilities. This review intends to address some of the manufacturing variables that can affect the critical quality attributes of CAR-T cells and efforts to optimize these attributes.
Apheresis Cell Collection
The starting material for CAR-T cell products is typically obtained through a nonmobilized autologous mononuclear cell apheresis collection. Even before the apheresis procedure begins, however, there are a number of patient factors that may be able to predict compromised quantity or quality of CAR-T cells in the final product, including certain disease types, advanced age, prior therapy, and peripheral blood characteristics.5,6 (Table 1) In comparison with healthy donors, patients referred for CAR-T cell therapy have often been previously treated with chemo- or radiation therapy, or have peripheral blood cell parameters that may complicate collections, such as lymphopenia or high peripheral disease burden.
Table 1.
Sources of Variation during CAR-T Cell Manufacturing
Manufacturing Phase | Variable |
---|---|
Collection | Patient characteristics (autologous products) |
Peripheral blood cell composition | |
Apheresis procedure and collection efficiency | |
Cell enrichment | Method of cell enrichment |
Quantity of contaminating cells | |
Lymphocyte phenotypes | |
Cell stimulation | Method of cell stimulation |
Duration of cell stimulation | |
Presence of insoluble reagents | |
Gene transfer | CAR construct design |
Method of gene delivery | |
Timing of transduction | |
Use of transduction enhancers | |
Vector envelope (viral vectors) | |
Cell expansion | Cytokine selection |
Culture systems | |
Duration of expansion | |
Cryopreservation | Cryoprotectant type and concentration |
Method and rate of freezing |
Tuazon, et al. examined variables associated with low lymphocyte collection efficiencies for patients participating in anti-CD19 CAR-T cell trials, and reported reduced collection efficiencies (< 40%) in patients with advanced age, higher pre-apheresis platelet counts, and B-lymphoblastic leukemia (B-ALL), as compared to non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL).5 While this study and others have expressed feasibility in obtaining adequate quantities of lymphocytes for CAR-T cell manufacture, insufficient collections or cell expansion after collection does occur.7,8 In a study of children with neuroblastoma, manufacturing failures due to insufficient cell growth of CD8+ cells were reported for two children who had received metaiodobenzylguanidine (MIBG) therapy within 8 weeks of apheresis, prompting a mandatory 12-week washout period prior to collection for future patients who had undergone this therapy.6
Several others have also investigated the impact of the differential state of T cells on CAR-T cell products, with most agreeing that having a higher proportion of naïve T cells phenotypes in the peripheral blood or starting material is associated with greater expansion and more sustained remissions, and accordingly, a more potent and durable product.9 Hoffman and colleagues evaluated apheresis products from untreated CLL patients compared to healthy donors and found that CLL patients had expectedly higher numbers of circulating malignant B cells that may have contributed to reduced expansion of naïve T cells and increased expression of exhaustion markers.10 Another study retrospectively analyzed cellular material for patients who ultimately suffered CD19 CAR-T cell treatment failures, and identified that the associated apheresis material contained an increased frequency of LAG-3+/TNF-αlo CD8+ T cells.11 In addition, these products demonstrated rapid expression of functional exhaustion markers following adoptive transfer. Apheresis material containing higher frequencies of CD27+CD45RO–CD8+ T cells were also found to be associated with more sustained remissions for patients with CLL treated with CD19 CAR-T cells.12 Likewise, CD8+ T cells in CLL have been found to have impaired mitochondrial biogenesis, which may affect expansion and persistence of the infused product.13
While apheresis procedures are being increasingly utilized to procure lymphocytes for CAR-T cell manufacture, the parameters needed to optimize these autologous collections have not been well established. Collection efficiencies for mononuclear cells may vary between collection devices, with one study in pediatric populations reporting improved efficiencies on the Spectra Optia than with the COBE Spectra.8 As with hematopoietic progenitor cell collections, processing volumes needed to obtain sufficient cells may also vary between patients. However, if collection targets are known, the duration of procedures can be tailored to meet manufacturing needs, particularly for patients with low peripheral blood counts.14 To avoid some of these disease-related collection and cell quality challenges, there are ongoing studies utilizing healthy donor allogeneic material to produce CAR-T cells, though risks associated with graft-versus-host disease and T-cell rejection first need to be addressed.15,16 The use of allogeneic donor cells allows for the manufacture of CAR T cells using only apheresis products meeting predetermined criteria. Allogeneic donor CAR-T cells can be made in large quantities and cryopreserved allowing for the establishment of an inventory of CAR-T cells that meet all critical quality attributes.
Starting Material Cell Enrichment
Following the apheresis procedure, a cell selection or enrichment step is undertaken to further isolate target cells. Contaminating red blood cells (RBCs), monocytes, and neutrophils in the starting material may adversely affect T cell expansion in culture as well as final CAR-T cell product characteristics. Notably, increased quantities of monocytes have been shown to be associated with reduced T cell expansion, while excess neutrophils may be associated with reduced transduction efficiency.17,18 Hence, as there is limited technical flexibility during the apheresis procedure to fully avoid the collection of undesired cells, employing a cell selection or enrichment step is a critical part of the manufacturing process.
The methods used for cell enrichment can be variable, however, this variability can permit flexibility during manufacturing, as techniques can be tailored to meet clinical needs and cost constraints. The available methods can be broadly characterized as either a method to eliminate contaminating cells, or one to specifically select for T cells. GMP-compliant cell elimination techniques include density gradient separation, counter-flow elutriation, and flask adherence techniques. Density gradient separation utilizes a lymphocyte separation medium, such as Ficoll, during centrifugation to separate RBCs and granulocytes from peripheral blood mononuclear cells (PBMCs), platelets, and plasma. Counter flow elutriation separates cells based on size, and to a lesser extent density, and has been successfully used to generate CAR-T cells.19 Cell separation can also be achieved through flask adherence techniques, which exploit the adherent properties of myeloid cells to plastic to permit isolation of lymphocytes in T flasks.18
In terms of cell selection techniques, investigators have used various antibody-fluorochrome conjugates, antibody-magnetic bead conjugates, as well as Streptamer-based isolation methods.20,21 These methods separate cells based on cell surface antigen expression and are capable of not only selecting for T cells, but also T cell subsets. Through cell selection techniques, Turtle, et al. produced anti-CD19 CAR-T cells at a fixed 1:1 ratio of CD4:CD8 T cells in order to provide a more consistent final product.22 Even without altering ratios, the use of CD4/CD8 selection during CAR-T cell manufacture has been found to yield more consistent transduction efficiencies, though the clinical impact of this remains to be determined as this may be associated with greater toxicities.23
Though cell elimination techniques are cost-effective and have been successfully utilized to manufacture CAR-T cells, they result in greater contamination with unwanted cells as compared to more expensive cell selection techniques. While no single cell enrichment method has been shown to be superior to others, certain populations and disease states may require purer cellular material be used for beginning the manufacturing process. Likely the most compelling case for ensuring a pure T cell population was made in 2018 when it was reported that the unintentional transduction of a single leukemic B cell during anti-CD19 CAR T cell manufacture led to not only resistance to CAR-T cell therapy by masking the CD19 epitope, but also the iatrogenic creation of a CAR-B cell form of leukemia.24
Cell Stimulation/Activation
Following cell selection or enrichment, polyclonal, antigen-independent T cell stimulation is used to facilitate T cell expansion so that T cell doses sufficient to treat patients can be obtained. With the expanding field of adoptive T cell therapies, GMP-grade reagents are rapidly becoming available to support clinical trials. There are now a number of different types of stimulatory reagents, but further research is needed to determine the differences between them and the downstream effects that they may induce. For example, while some stimulatory reagents may result in greater cell expansion than others, this could also result in more differentiated T cell phenotypes, and accordingly, a less durable product.25
Cell-based and bead-based artificial antigen presenting cells (aAPCs) have been tested for the manufacture of adoptive cell therapies, and can effectively simulate the T cell activation effects of innate antigen presenting cells.26 Antibody-based methods of stimulation are also commonly used, and require activation through both CD3 as well as a costimulatory signal in order to generate substantial expansion. These reagents may be soluble or utilize magnetic beads that require removal prior to cell infusion. Some of the GMP-grade activation reagents available at this time include: soluble anti-CD3 monoclonal antibody (clone OKT3), CTS™ Dynabeads™ CD3/CD28 (ThermoFisher), MACS® GMP T Cell TransACT™ CD3/CD28 (Miltenyi Biotec), and Cloudz™ T Cell Activation CD3/CD28 (R&D Systems).
The number of days that T cells are activated also needs to be carefully considered. At our institution, we have found that CAR-T cells manufactured on the CliniMACS Prodigy® (Miltenyi Biotec) demonstrated reduced viability, fold expansion, CD3%, and transduction efficiency when cell stimulation was extended to 5 days from 3 days (unpublished results). In addition, these cells demonstrated reduced Polyfunctional Strength Indexes (PSI™) as measured by technology from IsoPlexis, suggesting that they may have reduced immune programs that may affect the potency of the product.
CAR Gene Transfer
Variability during CAR-T cell manufacturing is also introduced during the gene transfer phase, and the method used may impact the critical quality attributes of the final product. Gene delivery may be accomplished through viral or non-viral systems, of which the former is highly efficient, though carries a higher risk of insertional mutagenesis.27 Clinical CAR-T cell trials have largely utilized viral transduction-based methods, which include the use of γ-retroviral and lentiviral vectors. γ-retroviral vectors were the first viral vectors used in manufacture CAR-T cells, and benefit from high transduction efficiencies. Lentiviral vectors, on the other hand, have the advantage that they are capable of transducing both dividing and non-dividing cells. Lentiviral vectors may also carry a lower risk of insertional mutagenesis based on integration patterns.28
GMP-compliant viral vector production, however, is expensive, and there is currently manufacturing under-capacity. More accessible alternatives to viral vector-based transduction are available, though still being tested in clinical trials. Some of the methods being explored include electroporation of mRNA or via transposase systems, as well as targeted insertion strategies such as zinc finger nucleases, transcription activator-like effector nucleases (TALENs), and CRISPR/Cas9.29–31 Studies have used the latter approach to concurrently and strategically disrupt the T cell receptor alpha constant (TRAC) gene encoding T cell receptors, which could support the manufacture of allogeneic CAR-T cells, as the risks of graft versus host disease are alleviated.16,32 Ultimately, whether CAR-T products manufactured with these non-viral techniques will be equally safe and effective remains to be determined.
When viral vectors are used to produce CAR-T cells, transduction related variables include the timing of transduction and the use of transduction enhancers. Vector transduction can be performed during or a few days after cell stimulation, and the transduction step can be performed more than once. Methods to enhance vector interaction with T cells have also been used, and include spinoculation, also known as centrifugal inoculation, which has been shown to improve transduction efficiency.33 Spinoculation, however, also carries the risk of bag breakage, and in some cases, can be circumvented as a step in manufacturing without compromising transduction efficiency.34 Chemical enhancers include polybrene and recombinant fibronectin (RetroNectin, Takara Bio), and are used to promote the co-localization of viral particles and T cells.34,35
There is also significant variability with the CAR construct, which is typically composed of an extracellular target-binding domain, a hinge region, a transmembrane domain, and one or more intracellular domains. As CARs have developed, the intracellular domains have distinguished CARs as first (CD3ζ only), second (one costimulatory domain and CD3ζ), or third generation (more than one costimulatory domain and CD3ζ). Each of these domains are designed with a purpose, with variabilities conferring both advantages and disadvantages, and together, the design of the construct can heavily impact the potency and durability of the final product.
The target-binding domain is designed to recognize tumor cells, and can be derived from both human and non-human sources.36 The use of humanized constructs reduces the risk of development of an immune response directed toward the CAR-T cells. The ideal CAR-T cell target is present on tumor cells and absent from normal tissue, so that on-target, off-tumor toxicities can be reduced or avoided. The hinge region provides stability and enables flexibility for the construct. The transmembrane domain anchors the CAR to the cell membrane, and enables proper CAR T cell signaling and expression. Lastly, variability is also introduced with the costimulatory domain, which provides a second signal necessary for durable CAR-T cell activation. The two types of costimulatory molecules most commonly used include CD28 and 4–1BB, where the former results in more rapid T cell activation but shorter T cell persistence than the later.37 These characteristics can be exploited to maximally tailor the design of the CAR to the disease being treated.36
Cell Expansion
The cell expansion phase is another step where optimization is necessary to meet targeted doses for treatment. Effective CAR-T cell products may be produced from a relatively small amount of starting leukocytes, as the therapeutic cells may be expanded in culture after the gene transfer phase. Variabilities in the expansion phase include reagents used in the culture media, duration of culture, and choice of bioreactor or culture platform.
GMP-grade T cell culture media are available to support clinical CAR-T cell trials, and are typically supplemented with cytokines to enhance ex vivo CAR-T cell proliferation. Type 1 cytokines, and particularly members of the γ-chain family of cytokines, play critical roles in the survival, proliferation, and differentiation of T cells.38 IL-2 has been the most common cytokine used for CAR-T cell manufacture and can induce robust T cell expansion, though potentially at the expense of differentiation and loss of the naïve T cell phenotype.39 As a product composed of terminally-differentiated lymphocytes may have compromised potency and durability, future clinical studies are needed to determine whether the use of other γ-chain cytokines can improve CAR-T cell efficacy.
In addition, the duration of expansion needs to be carefully considered, as while extended cultures may produce more T cells, these could likewise be more differentiated and have less tumor killing capacity. For example, Kaartinen, et al. found that by limiting the length of expansion and by adjusting the concentration of IL-2 used, the number of early memory T cells could be increased, which could enhance in vivo performance.40 In a murine xenograft model of B-ALL, Ghassemi, et al. showed that anti-CD19 CAR-T cells harvested at earlier time points demonstrated robust leukemic control even at lower doses, as compared to cells harvested after extended culture.41 Hence, cytokine types and doses as well as culture duration should be adjusted based on protocol needs.
Further, while traditional cell culture relied on flask systems, closed-system bags are becoming increasingly used to manufacture CAR-T cells and other immunotherapies.42 In addition, partially- and fully-automated systems such as the CliniMACS Prodigy and the Cocoon™ (Lonza) are now available or being tested, and are capable of performing all functions of CAR-T cell manufacturing from cell selection, activation, gene transduction, expansion, and final harvest in a scalable, GMP-grade fashion. The shift to closed system processing and the incorporation of single-use disposable materials can also effectively reduce the risks of microbial contamination to provide a safer product, while automation can potentially promote a more consistent product. In addition, the choice of bioreactor is in itself a variability during manufacturing, as are the reagents associated with a given culture platform, and this can ultimately affect the critical quality attributes of the final product.
Cryopreservation and Thaw
Cell cryopreservation is an optional step during CAR-T cell manufacturing that can permit flexibility in scheduling patient infusions as well as time to complete extended quality control tests required for product release. This step may be performed prior to culture initiation and/or after the final product has been manufactured. From a quality perspective, this step is critical, as suboptimal cryopreservation can result in cell loss, impaired viability and altered cell phenotype and function. Accordingly, a validated process for this step is essential for clinical CAR-T cell manufacture.
Variables during this step include the type and concentration of cryoprotectant and additives in the freezing medium, the method of freezing, and the storage conditions. While most cryopreservation techniques utilize 5 – 10% dimethyl sulfoxide (DMSO) as a cryoprotectant, DMSO-free cryoprotectants are also being tested.43,44 Post thaw recovery of cells may also be influenced by the method of freezing, with controlled-rate freezing representing an essential process to cool cells and minimize cell damage from ice crystals or cytotoxic solutes.45 Following controlled rate freezing, cells are typically stored at <−150°C under temperature-monitored conditions. Lastly, automated devices to aliquot cells and reagents in preparation for cryopreservation are also becoming available, and may be able to provide more consistent products.
Following cryopreservation, PBMCs may be thawed to initiate culture, or final CAR-T cell products may be thawed for infusion. A number of variables may also be introduced at this step, and include the method, duration, and rate of thaw.46 While water baths set at 37°C have been most commonly used to thaw cell therapy products, dry thawing systems that reduce water-borne contamination risks are also available, and may aid in supplying a safer product.
Given the potential variabilities seen during cryopreservation and thaw, the techniques used require optimization as well in order to maximize cell recovery. At our facility, we systematically examined the clinical impact of cryopreserving the starting material of PBMCs as well as the final CAR-T cell products.47 Cryopreserved starting material did lose viability when thawed and placed in culture, particularly around two days into the process. However, all cultured products eventually recovered and expanded to a sufficient degree to meet clinical doses, and final CAR-T cells demonstrated no differences in viable cell recovery, transduction efficiency, %CD3 positivity, and CD4:CD8 ratios before and after cryopreservation. In addition, gene expression profiling performed in this study identified distinct differences between products that had or had not been cryopreserved, where products that had been cryopreserved demonstrated greater expression of apoptotic pathways, and less expression of T cell receptor signaling pathways. Ultimately, however, cryopreservation was not found to impact clinical outcomes including peak CAR-T cell levels or CAR-T cell persistence as measured in vivo. However, additional studies are needed in this regard.
Release Testing and Product Characterization
During and following CAR-T cell manufacturing, various in-process and quality control tests are performed to characterize the product and ensure its safety. CAR-T cell products are released under a certificate of analysis which features the minimum specifications required for several critical quality attributes, including safety, purity, and potency. (Table 2) Release testing is typically performed according to regulatory agency-approved assays, but where this is not possible, assays developed in-house may be used, provided that they are validated to prove assay integrity. As there are a number of different ways to test any given item, release testing represents another source of variation during the manufacturing process.
Table 2.
Measures of Critical Quality Attributes
Critical Quality | Measure | Method of Measurement | |
---|---|---|---|
Attribute | |||
Safety | Sterility testing | Bacterial and fungal culture Gram stain | |
Mycoplasma | PCR or culture | ||
RCL/RCR | PCR or culture | ||
Vector copy number | PCR | ||
Bead content (insoluble material) | Microscopy | ||
Purity | T cell content | Flow cytometry | |
CAR-T cell content | Flow cytometry | ||
Contaminating cell content1 | Cell count Flow cytometry | ||
Potency | CAR-T cell content | Flow cytometry PCR | |
In vitro cytotoxicity | Cytotoxicity assays | ||
Cytokine secretion | Cytokine release assays | ||
Durability | CAR-T cell persistence | Flow cytometry PCR | |
In vivo CAR-T cell expansion | Flow cytometry PCR | ||
Consistency | Measures of consistency includes all of the above for safety, purity, potency, and durability, but are applied across different lots of CAR-T cell products. |
Abbreviations: PCR: polymerase chain reaction; RCL/RCR: Replication competent lentivirus/Replication competent retrovirus
Contaminating cells include red blood cells, circulating malignant cells, and non-T cell leukocytes.
Measures of safety include testing for microbial contamination, including bacterial and fungal sterility as well as mycoplasma, which can contaminate cell cultures and be difficult to isolate from routine bacterial testing. When viral vectors are used, safety testing also includes testing for replication competent viruses and vector copy number. Flow cytometry can be utilized to assess the purity of a CAR-T cell product, including the proportion of cells positive for T cell surface markers such as CD3, CD4, and CD8, as well as markers of the CAR. In diseases where tumor cells may circulate in the periphery, flow cytometry may also be used as a safety measure to ensure that these cells have been eliminated from the final product. Viability testing is also performed to assess the proportion of viable cells before or possibility after cryopreservation, however, minimum viability specifications for clinical efficacy remain to be determined.48
Conclusion
CAR-T cells as immunotherapies herald an exciting era, evolving as the fourth pillar in cancer treatment along with chemotherapy, radiation and surgery. Nevertheless, standardizing manufacture of these nuanced “living agents” is a challenge, as variables may be introduced during each step in the process. Critical quality attributes of CAR-T cells are currently being assessed by a set of parameters similar across various studies, however, continued research and evaluation is needed to fine-tune these standards and their requirements. Alongside the major strides in automation and closed-system manufacturing solutions, it is critical to develop standardized assays for more robust testing of CAR-T cell functionality, in order to deliver better-characterized products to patients.
Notably, clearly-defined measures of CAR-T cell potency capable of predicting clinical outcomes do not yet exist. Potency may be assessed in vivo after the cells have been infused, such as by CAR-T cell expansion or tumor burden reduction, or through in vitro bioassays before the cells are released – though the latter may not correlate with clinical efficacy.49 Measurements of transgene expression may serve as a surrogate marker of CAR-T cell activity, and potency has also been assessed by testing interferon-γ secretion or through cytotoxicity assays. Newer potency assays are also being evaluated to assess T cell gene expression and exhaustion states, T cell metabolic activity, and T cell immune programs.50–52 As more is understood on how these measures correlate with clinical outcomes, these findings may be useful to further optimize manufacturing processes.
Footnotes
Declaration of Competing Interest
None.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Appelbaum JS, Pinto N, Orentas RJ. Promising Chimeric Antigen Receptors for Non-B-Cell Hematological Malignancies, Pediatric Solid Tumors, and Carcinomas In: Lee DW, Shah NN, eds. Chimeric Antigen Receptor T-Cell Therapies for Cancer. Cambridge, MA: Elsevier; 2020:137–63. [Google Scholar]
- 2.Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in Adult Relapsed or Refractory Diffuse Large B-Cell Lymphoma. N Engl J Med 2019;380:45–56. [DOI] [PubMed] [Google Scholar]
- 3.Brudno JN, Maric I, Hartman SD, et al. T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma. J Clin Oncol 2018;36:2267–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Fry TJ, Shah NN, Orentas RJ, et al. CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nature medicine 2018;24:20–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tuazon SA, Li A, Gooley T, et al. Factors affecting lymphocyte collection efficiency for the manufacture of chimeric antigen receptor T cells in adults with B-cell malignancies. Transfusion 2019;59:1773–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ceppi F, Rivers J, Annesley C, et al. Lymphocyte apheresis for chimeric antigen receptor T-cell manufacturing in children and young adults with leukemia and neuroblastoma. Transfusion 2018;58:1414–20. [DOI] [PubMed] [Google Scholar]
- 7.Allen ES, Stroncek DF, Ren J, et al. Autologous lymphapheresis for the production of chimeric antigen receptor T cells. Transfusion 2017;57:1133–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Even-Or E, Di Mola M, Ali M, et al. Optimizing autologous nonmobilized mononuclear cell collections for cellular therapy in pediatric patients with high-risk leukemia. Transfusion 2017;57:1536–42. [DOI] [PubMed] [Google Scholar]
- 9.Klaver Y, van Steenbergen SC, Sleijfer S, Debets R, Lamers CH. T Cell Maturation Stage Prior to and During GMP Processing Informs on CAR T Cell Expansion in Patients. Frontiers in immunology 2016;7:648. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Hoffmann JM, Schubert ML, Wang L, et al. Differences in Expansion Potential of Naive Chimeric Antigen Receptor T Cells from Healthy Donors and Untreated Chronic Lymphocytic Leukemia Patients. Frontiers in immunology 2017;8:1956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Finney OC, Brakke HM, Rawlings-Rhea S, et al. CD19 CAR T cell product and disease attributes predict leukemia remission durability. The Journal of clinical investigation 2019;129:2123–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Fraietta JA, Lacey SF, Orlando EJ, et al. Determinants of response and resistance to CD19 chimeric antigen receptor (CAR) T cell therapy of chronic lymphocytic leukemia. Nature medicine 2018;24:563–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.van Bruggen JAC, Martens AWJ, Fraietta JA, et al. Chronic lymphocytic leukemia cells impair mitochondrial fitness in CD8(+) T cells and impede CAR T-cell efficacy. Blood 2019;134:44–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chen J, Goss C, Avecilla ST, et al. Evaluation of peripheral blood mononuclear cell collection by leukapheresis. Transfusion 2019;59:1765–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Depil S, Duchateau P, Grupp SA, Mufti G, Poirot L. ‘Off-the-shelf’ allogeneic CAR T cells: development and challenges. Nat Rev Drug Discov 2020. [DOI] [PubMed] [Google Scholar]
- 16.Poirot L, Philip B, Schiffer-Mannioui C, et al. Multiplex Genome-Edited T-cell Manufacturing Platform for “Off-the-Shelf” Adoptive T-cell Immunotherapies. Cancer Res 2015;75:3853–64. [DOI] [PubMed] [Google Scholar]
- 17.Elavia N, Panch SR, McManus A, et al. Effects of starting cellular material composition on chimeric antigen receptor T-cell expansion and characteristics. Transfusion 2019;59:1755–64. 18. [DOI] [PubMed] [Google Scholar]
- 18.Stroncek DF, Ren J, Lee DW, et al. Myeloid cells in peripheral blood mononuclear cell concentrates inhibit the expansion of chimeric antigen receptor T cells. Cytotherapy 2016;18:893–901. 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Stroncek DF, Lee DW, Ren J, et al. Elutriated lymphocytes for manufacturing chimeric antigen receptor T cells. Journal of translational medicine 2017;15:59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Panch SR, Reddy OL, Li K, et al. Robust Selections of Various Hematopoietic Cell Fractions on the CliniMACS Plus Instrument. Clinical Hematology International 2019;1:161–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.van Loenen MM, de Boer R, van Liempt E, et al. A Good Manufacturing Practice procedure to engineer donor virus-specific T cells into potent anti-leukemic effector cells. Haematologica 2014;99:759–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. The Journal of clinical investigation 2016;126:2123–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Shah NN, Highfill SL, Shalabi H, et al. CD4/CD8 T-Cell Selection Affects Chimeric Antigen Receptor (CAR) T-Cell Potency and Toxicity: Updated Results From a Phase I Anti-CD22 CAR T-Cell Trial. J Clin Oncol 2020:JCO1903279. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ruella M, Xu J, Barrett DM, et al. Induction of resistance to chimeric antigen receptor T cell therapy by transduction of a single leukemic B cell. Nature medicine 2018;24:1499–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Li Y, Kurlander RJ. Comparison of anti-CD3 and anti-CD28-coated beads with soluble anti-CD3 for expanding human T cells: differing impact on CD8 T cell phenotype and responsiveness to restimulation. Journal of translational medicine 2010;8:104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Neal LR, Bailey SR, Wyatt MM, et al. The Basics of Artificial Antigen Presenting Cells in T Cell-Based Cancer Immunotherapies. J Immunol Res Ther 2017;2:68–79. [PMC free article] [PubMed] [Google Scholar]
- 27.Rothe M, Modlich U, Schambach A. Biosafety challenges for use of lentiviral vectors in gene therapy. Curr Gene Ther 2013;13:453–68. [DOI] [PubMed] [Google Scholar]
- 28.Vannucci L, Lai M, Chiuppesi F, Ceccherini-Nelli L, Pistello M. Viral vectors: a look back and ahead on gene transfer technology. New Microbiol 2013;36:1–22. [PubMed] [Google Scholar]
- 29.Kebriaei P, Singh H, Huls MH, et al. Phase I trials using Sleeping Beauty to generate CD19-specific CAR T cells. The Journal of clinical investigation 2016;126:3363–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Beatty GL, Haas AR, Maus MV, et al. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer immunology research 2014;2:112–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Zhao J, Lin Q, Song Y, Liu D. Universal CARs, universal T cells, and universal CAR T cells. J Hematol Oncol 2018;11:132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Eyquem J, Mansilla-Soto J, Giavridis T, et al. Targeting a CAR to the TRAC locus with CRISPR/Cas9 enhances tumour rejection. Nature 2017;543:113–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Quintas-Cardama A, Yeh RK, Hollyman D, et al. Multifactorial optimization of gammaretroviral gene transfer into human T lymphocytes for clinical application. Hum Gene Ther 2007;18:1253–60. 34. [DOI] [PubMed] [Google Scholar]
- 34.Tumaini B, Lee DW, Lin T, et al. Simplified process for the production of anti-CD19-CARengineered T cells. Cytotherapy 2013;15:1406–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Sommermeyer D, Hudecek M, Kosasih PL, et al. Chimeric antigen receptor-modified T cells derived from defined CD8+ and CD4+ subsets confer superior antitumor reactivity in vivo. Leukemia 2016;30:492–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Guedan S, Calderon H, Posey AD Jr., Maus MV. Engineering and Design of Chimeric Antigen Receptors. Molecular therapy Methods & clinical development 2019;12:145–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Milone MC, Fish JD, Carpenito C, et al. Chimeric receptors containing CD137 signal transduction domains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Mol Ther 2009;17:1453–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Rochman Y, Spolski R, Leonard WJ. New insights into the regulation of T cells by gamma(c) family cytokines. Nat Rev Immunol 2009;9:480–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Dwyer CJ, Knochelmann HM, Smith AS, et al. Fueling Cancer Immunotherapy With Common Gamma Chain Cytokines. Frontiers in immunology 2019;10:263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Kaartinen T, Luostarinen A, Maliniemi P, et al. Low interleukin-2 concentration favors generation of early memory T cells over effector phenotypes during chimeric antigen receptor T-cell expansion. Cytotherapy 2017;19:689–702. [DOI] [PubMed] [Google Scholar]
- 41.Ghassemi S, Nunez-Cruz S, O’Connor RS, et al. Reducing Ex Vivo Culture Improves the Antileukemic Activity of Chimeric Antigen Receptor (CAR) T Cells. Cancer immunology research 2018;6:1100–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Fekete N, Beland AV, Campbell K, Clark SL, Hoesli CA. Bags versus flasks: a comparison of cell culture systems for the production of dendritic cell-based immunotherapies. Transfusion 2018;58:1800–13. [DOI] [PubMed] [Google Scholar]
- 43.Weng L, Beauchesne PR. Dimethyl sulfoxide-free cryopreservation for cell therapy: A review. Cryobiology 2020. [DOI] [PubMed] [Google Scholar]
- 44.Pi CH, Hornberger K, Dosa P, Hubel A. Understanding the freezing responses of T cells and other subsets of human peripheral blood mononuclear cells using DSMO-free cryoprotectants. Cytotherapy 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Li R, Johnson R, Yu G, McKenna DH, Hubel A. Preservation of cell-based immunotherapies for clinical trials. Cytotherapy 2019;21:943–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Baust JM, Campbell LH, Harbell JW. Best practices for cryopreserving, thawing, recovering, and assessing cells. In Vitro Cell Dev Biol Anim 2017;53:855–71. [DOI] [PubMed] [Google Scholar]
- 47.Panch SR, Srivastava SK, Elavia N, et al. Effect of Cryopreservation on Autologous Chimeric Antigen Receptor T Cell Characteristics. Mol Ther 2019;27:1275–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Chong EA, Levine BL, Grupp SA, et al. CAR T cell viability release testing and clinical outcomes: is there a lower limit? Blood 2019;134:1873–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Guidance for Industry: Potency Tests for Cellular and Gene Therapy Products US Department of Health and Human Services, Food and Drug Administration, Center for Biologics Evaluation and Research, 2011. (Accessed April 13, 2020, at https://www.fda.gov/media/79856/download.)
- 50.Rossi J, Paczkowski P, Shen YW, et al. Preinfusion polyfunctional anti-CD19 chimeric antigen receptor T cells are associated with clinical outcomes in NHL. Blood 2018;132:804–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Cesano A nCounter® PanCancer Immune Profiling Panel (NanoString Technologies, Inc., Seattle, WA). Journal for immunotherapy of cancer 2015;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kawalekar OU, O’Connor RS, Fraietta JA, et al. Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. Immunity 2016;44:380–90. [DOI] [PubMed] [Google Scholar]