Abstract
Based on laboratory and clinical research findings and investments in immunotherapy by many institutions in academia, government funded laboratories, and industry, there is tremendous and deserved excitement in the field of cell and gene therapy. In particular, understanding of immune mediated control of cancer has created opportunities to develop new forms of therapies based on engineered T cells. Unlike conventional drugs or biologics, the source material for these new therapies is collected from the patient or donor. The next step is commonly either enrichment to deplete unwanted cells, or methods to positively select T cells prior to polyclonal expansion or antigen specific expansion. As the first generation of engineered T cell therapies have demonstrated proof of concept, the next stages of development will require the integration of automated technologies to enable more consistent manufacturing and the ability to produce therapies for more patients.
Keywords: cellular therapy, immunotherapy, gene therapy, T-lymphocytes, leukemia, lymphoma
Introduction
Engineered T cell therapies are entering a new stage of therapeutic development. From the early beginnings in the 1980’s and early 1990’s, the adoptive transfer of viral specific T cells was investigated in clinical trials (1–4). These manufacturing strategies, while novel, were laborious and time consuming, often taking as long as 6 months to manufacture each cell product (2). These cell products also involved the use of antigen presenting cells such as fibroblasts which had to be infected by live CMV, and then used to present antigen to the T cells. Nearly twenty-five years later, cellular therapy has emerged as one of the most promising treatments of cancer and other disorders. Manufacturing strategies and technologies have also advanced. Not only can virus-specific T cells be manufactured in 10 days and targeting 6 viruses, but T cells can now be manufactured from cord blood (5–7), from naïve T cells (8) to target leukemia (9, 10) and other cancers as well – and with apparent clinical benefit (9, 11). The description of chimeric antigen receptors to redirect T cell specificity (12, 13), led to early clinical trials in HIV and cancer that demonstrated the feasibility of this approach, but showed limited or no demonstration of clinical activity (14–16). These early trials relied on previous generations of manufacturing processes and adaptation of legacy equipment. Promising clinical trials and easily modifiable platforms have sparked an enormous interest and investment in the field (17–19) resulting in a massive increase in the number of clinical trials of engineered T cells around the world. Logistic and regulatory challenges accompany the move into later phase multi-center and global clinical trials (20). Simultaneously, a deepening understanding of T cell biology and enhanced methods of genetic manipulation have brought within sight the vision of engineered T cells as therapeutic autonomous vehicles. The manufacturing process thus continues to evolve incorporating new science and new technologies (Figure 1). The next generation cell therapy manufacturing will enjoy the benefits of instruments designed with cell therapy applications in mind- offering improved standardization, automation and closed systems for each unit of processing in the manufacturing cycle. These advances are critical to providing wider access to patients as cell therapies move forward on the path to commercialization.
Figure 1.

Engineered T Cell Manufacturing Lifecycle - T cells can be obtained from a number of tissues depending on the application. While whole blood is most convenient, leukapheresis products contain much larger numbers of cells. Both cord blood and healthy donors may be a source for T cell product manufacture in the context of a hematopoietic stem cell transplant. Tumor infiltrating lymphocytes are already enriched for specificity. Examples of methods for isolation and enrichment, T cell activation, gene delivery, and culture provide flexibility dependent on the application. With variables at each processing step, the resulting final product derived from the same cell source will differ in cell number, phenotype, antigen specificity, gene expression level, and potency. The ultimate benefit to multiple available methods and technologies at each step is the opportunity for innovation and therapeutic development.
Collection
Apheresis mononuclear cell (MNC) collection is a common source of cellular material for cellular immunotherapy manufacturing. During MNC collection, the patient’s whole blood enters the apheresis instrument, is anticoagulated and then is separated by centrifugal force. The MNC layer, which includes T lymphocytes as well as B lymphocytes and monocytes, is identified and removed as the remaining material is simultaneously returned to the patient in real time. As extracorporeal blood volumes are small, large blood volumes may be processed and billions of leukocytes may be obtained. Several instruments are available for this purpose including the COBE Spectra and Spectra Optia (TerumoBCT) as well as the Amicus (Fenwal). While each instrument offers a unique set of advantages and disadvantages, comparison of the COBE Spectra and Spectra Optia illustrates the general direction in which the field of cell therapy manufacturing is moving. Compared to the COBE Spectra, the Spectra Optia incorporates a system to automatically detect the cell interface and adjust flow rates accordingly. The Spectra Optia also includes a feature to allow collection of a subset of the buffy coat layer. Both features highlight the goal of cell therapy equipment manufacturers to provide automated instruments that produce high purity MNC products.
Often, MNC product total cell yield and purity are competing interests. The MNC layer itself contains multiple cell types, some of which may not be desirable for a given application. Further, non-MNC contaminants can also inhibit downstream manufacturing. Collecting adequate lymphocyte yields and purities for cell manufacturing from patients with B cell malignancies can be challenging. These patients frequently have low circulating absolute lymphocyte counts. To improve yield, more total blood volume can be processed as the patient tolerates. However, if the MNC layer is small at the time of collection due to low absolute lymphocyte count (ALC), simply increasing the volume processed may increase total cell yield, but produce a low purity product. Without compensatory adjustments, fluctuations in blood flow through the instrument change the location of the target cell layer and alter the contents collected. When dealing with a narrow MNC layer, this is particularly noticeable. Non-MNC cell types (red blood cells, granulocytes) are frequently collected during MNC collection from patients with low ALC counts due to difficulty obtaining or maintaining the proper interface during collection. Granulocytic cells may inhibit ex vivo T cell expansion by a number of mechanisms(21). Within the MNC layer itself, non-target lymphocytes (eg. B cells, circulating lymphoid tumor cells) may add to product impurity. Residual contaminant tumor cells inhibit T cell activation and expansion, and monocytes interfere with activation (22). Further complicating matters, required cell yield and purity depends on downstream manufacturing capabilities. For example, a protocol in which ex vivo expansion is robust may prioritize purity over yield, whereas a protocol that includes extensive purification may value yield over purity. Given this complexity, to ensure the best change of manufacturing success, a collection approach should be tailored to each protocol’s needs and clearly communicated to the collection site.
While MNC collection is the primary source of starting material for cell therapies, it is not the sole source. Cell therapies have been generated from tumor infiltrating lymphocytes obtained by surgical resection, from mobilized hematopoietic stem cell products, bone marrow, cord blood, and even whole blood (23, 24). Each of these sources offers a unique cost/benefit profile with respect to yield and purity. It is important to note that many current challenges in MNC collection of B cell malignancy patients also limit use of alternative sources of cellular material. In addition, it has now been demonstrated that cytotoxic chemotherapy commonly used in B cell malignancies can impair the ability of benign T cells to expand ex vivo (25). This suggests treating a patient with conventional lymphotoxic therapies may limit the ability to successfully manufacture a cell therapy from that patient if cells are collected before T lymphocyte counts and function are reconstituted. Nevertheless, whole blood is useful in instances where an allogeneic donor is used, such as after hematopoietic stem cell transplantation, where tumor-associated antigen tumor antigen associated (TAA)-specific T cells in the case of leukemia,(9, 10) or Epstein-Barr Virus (EBV)-specific T cells in the case of EBV+ lymphoma,(26–28) can be expanded from the donor and transferred to the recipient. As more is learned about the long-term consequences of these treatments on benign immune cells, collection timing can be optimized and the likelihood of manufacturing success improved.
Enrichment
Post collection T cell enrichment can be achieved through several selection or depletion methods. Depending on the target cell population and the relative frequency of contaminant cell types, enrichment may be performed through density-based, size- and density-based, or immunophenotype-based techniques. Density gradients, such as Ficoll can further deplete granulocytes and red blood cells from lymphocytes and monocytes. While Ficoll has traditionally been performed in centrifuge tubes, an open and laborious method, a large scale, closed system Ficoll protocol has been reported making this approach more feasible in the Good Manufacturing Practices (GMP) setting (29, 30). A density gradient alone, however, cannot distinguish lymphocyte and monocyte fractions. Alternative separation techniques that rely on both size and density, such as elutriation, do allow for isolation of a lymphocyte fraction from a monocyte-rich product (31, 32). Closed system elutriators are commercially available and widely used in cell therapy applications (Elutra, TerumoBCT; Elutriation System, Beckman Coulter). Elutriation, however, cannot separate subsets of lymphocytes such as tumor lymphocytes or T regulatory cells that decrease efficiency of downstream manufacturing.
Separation by cell surface immunophenotype offers the highest resolution of subset isolation. Monoclonal antibodies fused to magnetic particles or fluorophores may be used to distinguish between target and non-target cells. If marking target cells, the bead-coated cells can be retained in a magnetic field or sorted by flow cytometry while non-desirable and unmarked cell types are removed. Alternatively, antibody-bead/fluorophore coated non-target cells can be negatively selected. The closed and semi-automated CliniMACS system (Miltenyi Biotec) is a magnetic bead based separation platform in which a specific immunophenotypic subset can be positively or negatively selected. Flow cytometric cell sorters can be setup for clinical grade cell sorting (33, 34). While density gradients, elutriation and magnetic bead separation have been in use in the clinical setting for years, novel cell therapy applications have driven continued effort to improve the standardization, automation and closed nature of these systems.
The optimal immunophenotype of an effective B cell directed cell therapy is under investigation. Because this remains largely unknown, the active ingredient in these therapies is somewhat ill-defined. For example, engineered T cell therapies are often defined as CD3+ cells that carry the transgenic modification. However, many infused products contain a variable number of CD4+, CD8+ and memory T cell subsets. Because the optimal immune subset for each potential therapeutic application is still under investigation, the best cell subset to initiate cultures and variables among patients is also unknown.
Manufacture and reinfusion of defined CD4 and CD8 subsets has been shown to generate potent anti-tumor responses (35), however it is not clear that a particular CD4 to CD8 ratio consistently enhances potency over bulk T cells. Memory T cell subsets appear to correlate with clinical response and can be promoted in culture (25, 36, 37). Better understanding of in vivo effector and memory responses of these cells will continue to drive manufacturing protocol development to skew cultures toward production of potent and persistent anti-tumor cells.
Expansion
To achieve a clinically relevant dose of cells, many cell therapy protocols require ex vivo expansion. Cell therapy culture expansion may occur in static culture or dynamic culture. Static culture may occur in flasks or gas permeable culture bags. These systems allow for culture of small and large volumes and are easily scalable. Many bag systems include docking for heat-sealed sterile tubing, which allows for the system to remain closed through sampling, media exchange or addition of reagents. Some bags are also qualified to be cryopreserved, simplifying materials needed for harvest and cryopreservation procedures. For flasks and bags, gas exchange only occurs at the liquid-air interface. The G-Rex device, which allows gas transfer through a gas-permeable membrane on the bottom of the device, permits the addition of cultures with larger volumes since the gas diffusion does not occur through the top of the culture as in most flasks and tissue culture plates.(38) The use of the G-Rex device has allowed for more rapid expansion and increased cell yield in many processes that tend to be laborious.(7, 8) One disadvantage of the original G-Rex is that the device has a large, open top, which increases the potential for contamination. WilsonWolf, the manufacturer, has recently released a closed version of the device, which could help lead the translation of these devices into clinical manufacturing – particularly in later phase clinical studies.
Dynamic bioreactors address both of these potential limitations. Dynamic bioreactors such as the WAVE Bioreactor System (GE) gently agitate cells and media in a bag while simultaneously applying heat. Here too, the bioreactor systems frequently include sterile docking to maintain a closed system during media exchange or sampling. Newer systems record performance and quality data to streamline auditing. While bioreactors improve gas exchange and even distribution of culture components, such systems add capital cost and can be challenging to scale across a production line. Installation and ongoing maintenance can be expensive. Preclinical collaborators rarely have access to these instruments, placing additional demands on clinical manufacturing validation.
The Miltenyi Prodigy combines the cell selection capabilities of the CliniMACS with the dynamic bioreactor technology available in other bioreactor systems.(40) The closed nature of this device, along with its automation, make this instrument appealing to programs or companies which have limited space, staff, and experience and are also appealing to industry for many of the same reasons, along with their ability to standardize procedures. For example, because the Prodigy can select subpopulations of cells based on the expression of surface markers,(41) T cells recognizing EBV antigens could theoretically be selected based on secretion of interferon gamma and then used as a treatment for lymphoma such as post-transplant lymphoproliferative disease (PTLD).(42) CAR T cell manufacturing may also be adapted to the Prodigy (43). Significant capital cost outlay and ongoing maintenance are also considerations with this device that are balanced with savings in labor.
The use of dynamic bioreactors and selection of cell populations is feasible for chimeric antigen receptor (CAR) T cell therapies and for selecting and infusing specific cell populations. In contrast, the expansion of TAA-specific T cells such as those targeting the leukemia antigens Preferentially Expressed Antigen of Melanoma (PRAME), Wilm’s Tumor Antigen (WT-1), and Survivin, involves the expansion of rare cell populations, likely derived from the naïve T cell population, and priming of T cells followed by repeated stimulations of expanded T cells with antigen-presenting cells such as dendritic cells. Because of the small cell volume and complexities associated with these expansions, the use of closed bioreactor systems has proven challenging, and a more rapid process will be required before it is widely implemented.
Whether culture of T cells occurs in dynamic bioreactors or static culture vessels, a critical determinant of cell number, efficiency of gene delivery, and potency is the culture media formulation. Studies showing the importance of retaining a central memory or stem cell memory, rather than an effector memory, phenotype (44, 45) have provided rationale for the addition of cytokines such as IL-7 and IL-15 to culture media (46–48). Another approach to achieve the end goal of enhanced engraftment and potency has been to pharmacologically inhibit the kinase Akt (49, 50). Method of T cell stimulation can also enhance potency and expand a polyclonal, rather than oligoclonal population. This is particularly evident in chronic lymphoid leukemia (CLL), where there is an intrinsic defect in the immune synapse that results in impaired signaling and T cell function that can be reversed by stimulation with anti-CD3/anti-CD28 antibody coated beads or by lenolidomide (51, 52). Ibrutinib is a recently approved therapy for CLL that targets Bruton’s Kinase (BTK). Although some patients progress on this therapy, the drug has a significant effect in restoring T cell function in patients with CLL and Mantle Cell Lymphoma (53, 54). This property of Ibrutinib is now being exploited in a clinical trial of CAR T cells (NCT02640209). Cell-based artificial antigen presenting cells can be tuned to express various combinations of stimulatory ligands that preferentially expand a selectable array of T cell subsets (55, 56). Serum free media and shorter culture times are additional enhancements to cell processing that have recently been reported (57). Ultimately, regardless of the technique, ex vivo expansion aims to increase the number of total cells for infusion. However, it is important to note that total cell number in a dose is not always a good predictor of potency and thus in vivo response. In vivo activation and proliferation vary from patient to patient due to T cell intrinsic and tumor intrinsic factors. Further, split dosing of more cells may improve safety while retaining efficacy (58). With better definition of the effective cell type, total doses will be reduced and robust ex vivo expansion will become less critical.
Genetic modification and editing
Efficient, GMP-compliant, ex vivo modification of T cells can be achieved by several mechanisms. Broadly speaking, T cells can be modified to express novel proteins or to ablate expression of endogenous proteins. In addition, such modifications can be transient or permanent. The appropriate strategy for introduction or ablation of targets is protocol dependent. Next generation T cell protocols are often multiplexed, introducing and/or ablating in a complementary fashion.
The most common T cell modification strategies introduce novel constructs into T cells by viral transduction or electroporation of DNA or RNA. Early work with retroviral transduction proved that efficient T transduction could be achieved; however, safety concerns were raised in light of oncogenesis observed in human retroviral based gene therapy trials (59). Lentiviral vectors have a potentially decreased risk of insertional oncogenesis compared to retroviral vectors, and do not require cell division for transduction. Sensitization of T cells to lentiviral transduction can be achieved by TCR-mediated activation or exposure to IL-2 and IL-7, among other methods (60, 61). Alternatively, DNA or RNA may be electroporated into cells. Electroporation transiently disrupts lipid bilayers, allowing passage of extracellular components into the cytosol. Electroporation of mRNA can lead to efficient, but transient expression of CARs (62). Electroporation of a DNA transposon with transposase can generate integration and stable expression of CARs (63).
Targeted gene disruption in T cells can be achieved through protein-endonuclease or RNA-endonuclease methods. Zinc Finger Nucleases (ZFN) and Transcription Activator-like Effector Nucleases (TALENs) share amino acid based targeting of specific DNA sequences. Amino acid arrays can be assembled to target sequences within the genomic DNA and when coupled with an endonuclease, double stranded breaks (DSB) can be generated at those loci. During DSB repair, bases may be lost or added, functionally disrupting the targeted locus. ZFNs and TALENs have been used to disrupt a number of endogenous proteins during ex vivo T cell modification (64, 65). ZFNs were the first gene editing method to be tested in humans (66). The RNA based Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 system can also be used to target disruption of genomic DNA. CRISPR-Cas9, unlike ZFN and TALEN systems, use RNA to confer specify, however the ultimate goal of DNA disruption at the target locus is shared. CRISPR-Cas9 has the added advantage in that generation of GMP grade RNA is easier and more economical than creating customized, GMP-compliant protein or amino acid arrays. All gene disruption techniques can be used in combination with viral transduction to generate CAR T cells with additional modifications (67, 68).
Conclusion: Competing visions of next-gen cell therapy manufacturing
The field of cell therapy manufacturing for the treatment of B cell malignancies is aligned on many goals. It is universally recognized that standardized, automated and closed systems are needed to increase manufacturing capacity and scale out these treatments. Next generation equipment that meets these needs will find an enthusiastic and expanding market. While there is widespread agreement on the needs, the path to achieving these goals is more controversial.
In the future state, manufacturing may occur at centralized locations or be distributed to small scale laboratories, much like the model established by hematopoietic stem cell transplantation over the past 60 years. Central manufacturing supports greater standardization, quality oversight and cost savings through economy of scale and minimization of idle capacity. However, smaller manufacturing operations distributed closer to translational investigators, such as in university medical centers, support more collaborative efforts to accelerate new therapies to clinical manufacturing and, in some cases, alleviate the need to cryopreserve cells. These approaches are not mutually exclusive and could coexist with novel therapies developed in smaller academic GMP labs and more mature products being manufacturing at scale in large centralized facilities.
Manufacturing protocols may employ modular or integrated systems. Modular systems allow the developer to optimize across several platforms and tailor a given manufacturing approach to the specific product specifications. While this requires a greater level of expertise during development, each individual component of the manufacturing cycle can be selected and optimized for a given process. Fully integrated protocols, however, allow for better automation, streamlining the process and standardization. Such standardization is critical to make these therapies commercially attractive and improve widespread availability. Manufacturing process design will also depend on the choice of manufacturing location. Centralized manufacturing makes control over and standardization of manufacturing easier. On the other hand, logistics of material and product transport add costs and challenges. Ultimately, as some therapies move toward FDA approval, costs will be reconsidered in the setting of potential reimbursement. Though unknown, one hopes that compensation for these therapies will consider the potential systemic cost savings of potentially curative cell therapies.
Table 1.
Instrumentation used in the manufacture of cellular therapy products
| Instrument | Process | Company | Advantages | Limitations |
|---|---|---|---|---|
| Optia | Apheresis Collection | Terumo BCT | Automated detection of interface | Limited in-process modification to collection |
| Spectra | Apheresis Collection | Terumo BCT | Vast prior experience | Soon to be no longer supported by manufacturer Yields/purities may be user dependent |
| Amicus | Apheresis Collection | Fenwal | Highly automated processing | Less frequent opportunity for in-process adjustments |
| Elutra | Enrichment | Terumo BCT | Closed system Distinguishes lymphocytes from monocytes | Cannot distinguish lymphocyte subsets or some tumor types |
| CliniMACS | Cell Selection | Miltenyi Biotec | Closed system Specific selection of cell populations* | Manual, lengthy procedure Expensive |
| SEPAX | Enrichment | GE Healthcare | Enrichment of mononuclear fractions Closed system | Limited processing volume |
| WAVE | Cell Expansion | GE Healthcare | Closed system Ability to control multiple variables | Footprint, Modified CD4+/CD8+ T cell ratios, Shear force damage(39) |
| Prodigy | Cell Expansion/Cell Selection | Miltenyi Biotec | Closed system Modular Contains centrifuge, Ability to enrich | Expensive, Not feasible for all processes |
| Quantum | Cell Expansion | Terumo BCT | Closed, automated | Limited reports on its use, Large, one size reactor |
| G-Rex | Cell Expansion | Wilson Wolf | Simple, Low cost design Effective | Potential for contamination, Limited sizes – small scale |
Food and Drug Administration (FDA) approval as a Humanitarian Use Device for specific indications
Acknowledgments
P.J.H. is supported by Children’s Cancer Foundation’s Next Gen Award; B.L.L. is supported by National Institutes of Health grant 1RO1CA165206 and P30-CA016520-35. The authors would like to acknowledge the inspiration and support from our patients and our professional colleagues.
The University of Pennsylvania has entered into a partnership with Novartis for the development of chimeric antigen receptors. This partnership is managed in accordance with the University of Pennsylvania’s Conflict of Interest Policy.
Bruce L. Levine reports grants from Novartis, during the conduct of the study; personal fees from GE Healthcare, outside the submitted work; In addition, Dr. Levine has a patent 8,906,682 with royalties paid to University of Pennsylvania, a patent 8,916,381 with royalties paid to University of Pennsylvania, a patent 8,911,993 with royalties paid to University of Pennsylvania, a patent 9,102,761 with royalties paid to University of Pennsylvania, a patent 9,102,760 with royalties paid to University of Pennsylvania, a patent 9,161,971 with royalties paid to University of Pennsylvania, a patent 9,101,584 with royalties paid to University of Pennsylvania, a patent 9,464,140 with royalties paid to University of Pennsylvania, a patent 9,518,123 with royalties paid to University of Pennsylvania, a patent 9,481,728 with royalties paid to University of Pennsylvania, a patent 9,328,156 with royalties paid to University of Pennsylvania, a patent 9,499,629 with royalties paid to University of Pennsylvania, a patent 9,540,445 with royalties paid to University of Pennsylvania, and a patent 9,572,836 with royalties paid to University of Pennsylvania.
Footnotes
Conflict of Interest
Andrew D. Fesnak and Patrick J. Hanley each declare no potential conflicts of interest.
Bibliography
- 1.Greenberg P, Goodrich J, Riddell S. Adoptive immunotherapy of human cytomegalovirus infection: potential role in protection from disease progression. Transplant Proc. 1991;23(3 Suppl 3):97–101. Epub 1991/06/01. [PubMed] [Google Scholar]
- 2.Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science. 1992;257(5067):238–41. doi: 10.1126/science.1352912. Epub 1992/07/10. [DOI] [PubMed] [Google Scholar]
- 3.Riddell SR, Greenberg PD, Overell RW, Loughran TP, Gilbert MJ, Lupton SD, Agosti J, Scheeler S, Coombs RW, Corey L. Phase I study of cellular adoptive immunotherapy using genetically modified CD8+ HIV-specific T cells for HIV seropositive patients undergoing allogeneic bone marrow transplant. The Fred Hutchinson Cancer Research Center and the University of Washington School of Medicine, Department of Medicine, Division of Oncology. Hum Gene Ther. 1992;3(3):319–38. doi: 10.1089/hum.1992.3.3-319. Epub 1992/06/01. [DOI] [PubMed] [Google Scholar]
- 4.Rooney CM, Smith CA, Ng CY, Loftin S, Li C, Krance RA, Brenner MK, Heslop HE. Use of gene-modified virus-specific T lymphocytes to control Epstein-Barr-virus-related lymphoproliferation. Lancet. 1995;345(8941):9–13. doi: 10.1016/s0140-6736(95)91150-2. Epub 1995/01/07. [DOI] [PubMed] [Google Scholar]
- 5.Hanley PJ, Bollard CM, Brunstein CG. Adoptive immunotherapy with the use of regulatory T cells and virus-specific T cells derived from cord blood. Cytotherapy. 2015;17(6):749–55. doi: 10.1016/j.jcyt.2014.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Hanley PJ, Cruz CR, Savoldo B, Leen AM, Stanojevic M, Khalil M, Decker W, Molldrem JJ, Liu H, Gee AP, Rooney CM, Heslop HE, Dotti G, Brenner MK, Shpall EJ, Bollard CM. Functionally active virus-specific T cells that target CMV, adenovirus, and EBV can be expanded from naive T-cell populations in cord blood and will target a range of viral epitopes. Blood. 2009;114(9):1958–67. doi: 10.1182/blood-2009-03-213256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Hanley PJ, Lam S, Shpall EJ, Bollard CM. Expanding cytotoxic T lymphocytes from umbilical cord blood that target cytomegalovirus, Epstein-Barr virus, and adenovirus. J Vis Exp. 2012;(63):e3627. doi: 10.3791/3627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hanley PJ, Melenhorst JJ, Nikiforow S, Scheinberg P, Blaney JW, Demmler-Harrison G, Cruz CR, Lam S, Krance RA, Leung KS, Martinez CA, Liu H, Douek DC, Heslop HE, Rooney CM, Shpall EJ, Barrett AJ, Rodgers JR, Bollard CM. CMV-specific T cells generated from naive T cells recognize atypical epitopes and may be protective in vivo. Sci Transl Med. 2015;7(285):285ra63. doi: 10.1126/scitranslmed.aaa2546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, Schmitt TM, Duerkopp N, Roberts IM, Pogosov GL, Ho WY, Ochsenreither S, Wolfl M, Bar M, Radich JP, Yee C, Greenberg PD. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med. 2013;5(174):174ra27. doi: 10.1126/scitranslmed.3004916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Weber G, Gerdemann U, Caruana I, Savoldo B, Hensel NF, Rabin KR, Shpall EJ, Melenhorst JJ, Leen AM, Barrett AJ, Bollard CM. Generation of multi-leukemia antigen-specific T cells to enhance the graft-versus-leukemia effect after allogeneic stem cell transplant. Leukemia. 2013;27(7):1538–47. doi: 10.1038/leu.2013.66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Williams KM, Grant M, Ismail M, Hoq F, Martin-Manso M, Hoover J, Mintz K, Namata A, Williams E, Barese C, Albihani S, Cruz CR, Lang H, Hanley P, Gottschalk S, McCurdy S, Jones R, Bollard C. Complete remissions post infusion of multiple tumor antigen specific T cells for the treatment of high risk leukemia and lymphoma patients after HCT. Cytotherapy. 2017;19(5, Supplement):e3. https://doi.org/10.1016/j.jcyt.2017.03.013. [Google Scholar]
- 12.Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules as functional receptors with antibody-type specificity. Proc Natl Acad Sci U S A. 1989;86(24):10024–8. doi: 10.1073/pnas.86.24.10024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kuwana Y, Asakura Y, Utsunomiya N, Nakanishi M, Arata Y, Itoh S, Nagase F, Kurosawa Y. Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions. Biochem Biophys Res Commun. 1987;149(3):960–8. doi: 10.1016/0006-291x(87)90502-x. Epub 1987/12/31. [DOI] [PubMed] [Google Scholar]
- 14.Hege KM, Bergsland EK, Fisher GA, Nemunaitis JJ, Warren RS, McArthur JG, Lin AA, Schlom J, June CH, Sherwin SA. Safety, tumor trafficking and immunogenicity of chimeric antigen receptor (CAR)-T cells specific for TAG-72 in colorectal cancer. J Immunother Cancer. 2017;5:22. doi: 10.1186/s40425-017-0222-9. Epub 2017/03/28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Mitsuyasu RT, Anton PA, Deeks SG, Scadden DT, Connick E, Downs MT, Bakker A, Roberts MR, June CH, Jalali S, Lin AA, Pennathur-Das R, Hege KM. Prolonged survival and tissue trafficking following adoptive transfer of CD4zeta gene-modified autologous CD4(+) and CD8(+) T cells in human immunodeficiency virus-infected subjects. Blood. 2000;96(3):785–93. [PubMed] [Google Scholar]
- 16.Walker RE, Bechtel CM, Natarajan V, Baseler M, Hege KM, Metcalf JA, Stevens R, Hazen A, Blaese RM, Chen CC, Leitman SF, Palensky J, Wittes J, Davey RT, Jr, Falloon J, Polis MA, Kovacs JA, Broad DF, Levine BL, Roberts MR, Masur H, Lane HC. Long-term in vivo survival of receptor-modified syngeneic T cells in patients with human immunodeficiency virus infection. Blood. 2000;96(2):467–74. [PubMed] [Google Scholar]
- 17.Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3(95):95ra73. doi: 10.1126/scitranslmed.3002842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson M, Phan GQ, Hughes MS, Sherry RM, Yang JC, Kammula US, Devillier L, Carpenter R, Nathan DA, Morgan RA, Laurencot C, Rosenberg SA. B-cell depletion and remissions of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19 chimeric-antigen-receptor-transduced T cells. Blood. 2012;119(12):2709–20. doi: 10.1182/blood-2011-10-384388. Epub 2011/12/14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365(8):725–33. doi: 10.1056/NEJMoa1103849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Levine BL, Miskin J, Wonnacott K, Keir C. Global Manufacturing of CAR T Cell Therapy. Mol Ther Methods Clin Dev. 2017;4:92–101. doi: 10.1016/j.omtm.2016.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Pillay J, Tak T, Kamp VM, Koenderman L. Immune suppression by neutrophils and granulocytic myeloid-derived suppressor cells: similarities and differences. Cell Mol Life Sci. 2013;70(20):3813–27. doi: 10.1007/s00018-013-1286-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Stroncek DF, Lee DW, Ren J, Sabatino M, Highfill S, Khuu H, Shah NN, Kaplan RN, Fry TJ, Mackall CL. Elutriated lymphocytes for manufacturing chimeric antigen receptor T cells. J Transl Med. 2017;15(1):59. doi: 10.1186/s12967-017-1160-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hexner EO, Luger SM, Reshef R, Jeschke GR, Mangan JK, Frey NV, Frank DM, Richman LP, Vonderheide RH, Aqui NA, Rosenbach M, Zhang Y, Chew A, Loren AW, Stadtmauer EA, Levine BL, June CH, Emerson SG, Porter DL. Infusion of CD3/CD28 costimulated umbilical cord blood T cells at the time of single umbilical cord blood transplantation may enhance engraftment. Am J Hematol. 2016;91(5):453–60. doi: 10.1002/ajh.24303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Borrello I, Noonan KA. Marrow-Infiltrating Lymphocytes - Role in Biology and Cancer Therapy. Front Immunol. 2016;7:112. doi: 10.3389/fimmu.2016.00112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Singh N, Perazzelli J, Grupp SA, Barrett DM. Early memory phenotypes drive T cell proliferation in patients with pediatric malignancies. Sci Transl Med. 2016;8(320):320ra3. doi: 10.1126/scitranslmed.aad5222. Demonstrates treatment-related expansion defects of benign T cells in patients with acute lymphoblastic leukemia, suggesting that T cell manufacturing from these patients may be limited by prior treatment. [DOI] [PubMed] [Google Scholar]
- 26.Barth MJ, Chu Y, Hanley PJ, Cairo MS. Immunotherapeutic approaches for the treatment of childhood, adolescent and young adult non-Hodgkin lymphoma. British journal of haematology. 2016;173(4):597–616. doi: 10.1111/bjh.14078. Epub 2016/04/12. [DOI] [PubMed] [Google Scholar]
- 27.Bollard CM, Gottschalk S, Torrano V, Diouf O, Ku S, Hazrat Y, Carrum G, Ramos C, Fayad L, Shpall EJ, Pro B, Liu H, Wu MF, Lee D, Sheehan AM, Zu Y, Gee AP, Brenner MK, Heslop HE, Rooney CM. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2014;32(8):798–808. doi: 10.1200/JCO.2013.51.5304. Epub 2013/12/18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Heslop HE, Slobod KS, Pule MA, Hale GA, Rousseau A, Smith CA, Bollard CM, Liu H, Wu MF, Rochester RJ, Amrolia PJ, Hurwitz JL, Brenner MK, Rooney CM. Long-term outcome of EBV-specific T-cell infusions to prevent or treat EBV-related lymphoproliferative disease in transplant recipients. Blood. 2010;115(5):925–35. doi: 10.1182/blood-2009-08-239186. Epub 2009/11/03. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Janssen WE, Ribickas A, Meyer LV, Smilee RC. Large-scale Ficoll gradient separations using a commercially available, effectively closed, system. Cytotherapy. 2010;12(3):418–24. doi: 10.3109/14653240903479663. [DOI] [PubMed] [Google Scholar]
- 30.Kaur I, Zulovich JM, Gonzalez M, McGee KM, Ponweera N, Thandi D, Alvarez EF, Annandale K, Flagge F, Jr, Rezvani K, Shpall E. Comparison of two methodologies for the enrichment of mononuclear cells from thawed cord blood products: The automated Sepax system versus the manual Ficoll method. Cytotherapy. 2017;19(3):433–9. doi: 10.1016/j.jcyt.2016.11.010. [DOI] [PubMed] [Google Scholar]
- 31.Powell DJ, Jr, Brennan AL, Zheng Z, Huynh H, Cotte J, Levine BL. Efficient clinical-scale enrichment of lymphocytes for use in adoptive immunotherapy using a modified counterflow centrifugal elutriation program. Cytotherapy. 2009;11(7):923–35. doi: 10.3109/14653240903188921. [DOI] [PubMed] [Google Scholar]
- 32.Stroncek DF, Fellowes V, Pham C, Khuu H, Fowler DH, Wood LV, Sabatino M. Counter-flow elutriation of clinical peripheral blood mononuclear cell concentrates for the production of dendritic and T cell therapies. J Transl Med. 2014;12:241. doi: 10.1186/s12967-014-0241-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Pollack SM, Jones RL, Farrar EA, Lai IP, Lee SM, Cao J, Pillarisetty VG, Hoch BL, Gullett A, Bleakley M, Conrad EU, 3rd, Eary JF, Shibuya KC, Warren EH, Carstens JN, Heimfeld S, Riddell SR, Yee C. Tetramer guided, cell sorter assisted production of clinical grade autologous NY-ESO-1 specific CD8(+) T cells. J Immunother Cancer. 2014;2(1):36. doi: 10.1186/s40425-014-0036-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, Herold KC, Lares A, Lee MR, Li K, Liu W, Long SA, Masiello LM, Nguyen V, Putnam AL, Rieck M, Sayre PH, Tang Q. Type 1 diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med. 2015;7(315):315ra189. doi: 10.1126/scitranslmed.aad4134. Demonstrates the feasibility of using FACS cell sorting to isolate therapeutic cell subsets (Tregs in this case) for clinical use. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, Sommermeyer D, Melville K, Pender B, Budiarto TM, Robinson E, Steevens NN, Chaney C, Soma L, Chen X, Yeung C, Wood B, Li D, Cao J, Heimfeld S, Jensen MC, Riddell SR, Maloney DG. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest. 2016;126(6):2123–38. doi: 10.1172/JCI85309. Demonstrates a novel approach toward standardizing starting cellular material for therapeutic cell manufacturing and the associated improvement of clinical outcomes. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Fraietta JA, Lacey SF, Wilcox NS, Bedoya F, Chen F, Orlando E, Brogdon JL, Hwang WT, Frey N, Young RM, Pequignot E, Ambrose DE, Levine BL, Bitter H, Porter DL, Xu J, June CH, Melenhorst JJ. Biomarkers of Response to Anti-CD19 Chimeric Antigen Receptor (CAR) T-Cell Therapy in Patients with Chronic Lymphocytic Leukemia. Blood. 2016;128:22. Demonstrates the ability to predict response to CD19 specific CAR T cells using biomarker signatures. [Google Scholar]
- 37.Hurton LV, Singh H, Najjar AM, Switzer KC, Mi T, Maiti S, Olivares S, Rabinovich B, Huls H, Forget MA, Datar V, Kebriaei P, Lee DA, Champlin RE, Cooper LJ. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc Natl Acad Sci U S A. 2016;113(48):E7788–E97. doi: 10.1073/pnas.1610544113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Vera JF, Brenner LJ, Gerdemann U, Ngo MC, Sili U, Liu H, Wilson J, Dotti G, Heslop HE, Leen AM, Rooney CM. Accelerated production of antigen-specific T cells for preclinical and clinical applications using gas-permeable rapid expansion cultureware (G-Rex) J Immunother. 2010;33(3):305–15. doi: 10.1097/CJI.0b013e3181c0c3cb. This publication demonstrates the capability of the Grex device in augmenting cell expansion. It compares the manufacturing process of EBV-specific T cells expanded in 24-well plates and the Grex and evaluates criteria such as production hours, number of flasks, and final yield. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Somerville RP, Devillier L, Parkhurst MR, Rosenberg SA, Dudley ME. Clinical scale rapid expansion of lymphocytes for adoptive cell transfer therapy in the WAVE(R) bioreactor. Journal of translational medicine. 2012;10:69. doi: 10.1186/1479-5876-10-69. Epub 2012/04/06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Bunos M, Hummer C, Wingenfeld E, Sorg N, Pfirrmann V, Bader P, Seifried E, Bonig H. Automated isolation of primary antigen-specific T cells from donor lymphocyte concentrates: results of a feasibility exercise. Vox sanguinis. 2015;109(4):387–93. doi: 10.1111/vox.12291. Epub 2015/05/09. [DOI] [PubMed] [Google Scholar]
- 41.Kumaresan P, Figliola M, Moyes JS, Huls MH, Tewari P, Shpall EJ, Champlin R, Cooper LJ. Automated Cell Enrichment of Cytomegalovirus-specific T cells for Clinical Applications using the Cytokine-capture System. J Vis Exp. 2015;(104) doi: 10.3791/52808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bollard CM. Improving T-cell therapy for epstein-barr virus lymphoproliferative disorders. Journal of clinical oncology: official journal of the American Society of Clinical Oncology. 2013;31(1):5–7. doi: 10.1200/JCO.2012.43.5784. Epub 2012/11/22. [DOI] [PubMed] [Google Scholar]
- 43.Mock U, Nickolay L, Philip B, Cheung GW, Zhan H, Johnston IC, Kaiser AD, Peggs K, Pule M, Thrasher AJ, Qasim W. Automated manufacturing of chimeric antigen receptor T cells for adoptive immunotherapy using CliniMACS prodigy. Cytotherapy. 2016;18(8):1002–11. doi: 10.1016/j.jcyt.2016.05.009. Epub 2016/07/06. [DOI] [PubMed] [Google Scholar]
- 44.Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, Riddell SR. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates. J Clin Invest. 2008;118(1):294–305. doi: 10.1172/jci32103. Epub 2007/12/07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang X, Popplewell LL, Wagner JR, Naranjo A, Blanchard MS, Mott MR, Norris AP, Wong CW, Urak RZ, Chang WC, Khaled SK, Siddiqi T, Budde LE, Xu J, Chang B, Gidwaney N, Thomas SH, Cooper LJ, Riddell SR, Brown CE, Jensen MC, Forman SJ. Phase I studies of central-memory-derived CD19 CAR T cell therapy following autologous HSCT in patients with B-cell NHL. Blood. 2016 doi: 10.1182/blood-2015-12-686725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Xu Y, Zhang M, Ramos CA, Durett A, Liu E, Dakhova O, Liu H, Creighton CJ, Gee AP, Heslop HE, Rooney CM, Savoldo B, Dotti G. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood. 2014;123(24):3750–9. doi: 10.1182/blood-2014-01-552174. Epub 2014/05/02. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Perna SK, De Angelis B, Pagliara D, Hasan ST, Zhang L, Mahendravada A, Heslop HE, Brenner MK, Rooney CM, Dotti G, Savoldo B. Interleukin 15 provides relief to CTLs from regulatory T cell-mediated inhibition: implications for adoptive T cell-based therapies for lymphoma. Clin Cancer Res. 2013;19(1):106–17. doi: 10.1158/1078-0432.ccr-12-2143. Epub 2012/11/15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Perna SK, Pagliara D, Mahendravada A, Liu H, Brenner MK, Savoldo B, Dotti G. Interleukin-7 mediates selective expansion of tumor-redirected cytotoxic T lymphocytes (CTLs) without enhancement of regulatory T-cell inhibition. Clin Cancer Res. 2014;20(1):131–9. doi: 10.1158/1078-0432.ccr-13-1016. Epub 2013/10/08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Crompton JG, Sukumar M, Roychoudhuri R, Clever D, Gros A, Eil RL, Tran E, Hanada K, Yu Z, Palmer DC, Kerkar SP, Michalek RD, Upham T, Leonardi A, Acquavella N, Wang E, Marincola FM, Gattinoni L, Muranski P, Sundrud MS, Klebanoff CA, Rosenberg SA, Fearon DT, Restifo NP. Akt inhibition enhances expansion of potent tumor-specific lymphocytes with memory cell characteristics. Cancer Res. 2015;75(2):296–305. doi: 10.1158/0008-5472.CAN-14-2277. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.van der Waart AB, van de Weem NM, Maas F, Kramer CS, Kester MG, Falkenburg JH, Schaap N, Jansen JH, van der Voort R, Gattinoni L, Hobo W, Dolstra H. Inhibition of Akt signaling promotes the generation of superior tumor-reactive T cells for adoptive immunotherapy. Blood. 2014;124(23):3490–500. doi: 10.1182/blood-2014-05-578583. Epub 2014/10/23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bonyhadi M, Frohlich M, Rasmussen A, Ferrand C, Grosmaire L, Robinet E, Leis J, Maziarz RT, Tiberghien P, Berenson RJ. In vitro engagement of CD3 and CD28 corrects T cell defects in chronic lymphocytic leukemia. J Immunol. 2005;174(4):2366–75. doi: 10.4049/jimmunol.174.4.2366. Epub 2005/02/09. [DOI] [PubMed] [Google Scholar]
- 52.Ramsay AG, Johnson AJ, Lee AM, Gorgun G, Le Dieu R, Blum W, Byrd JC, Gribben JG. Chronic lymphocytic leukemia T cells show impaired immunological synapse formation that can be reversed with an immunomodulating drug. J Clin Invest. 2008;118(7):2427–37. doi: 10.1172/jci35017. Epub 2008/06/14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM, Lacey SF, Melenhorst JJ, McGettigan SE, Cook DR, Zhang C, Xu J, Do P, Hulitt J, Kudchodkar SB, Cogdill AP, Gill S, Porter DL, Woyach JA, Long M, Johnson AJ, Maddocks K, Muthusamy N, Levine BL, June CH, Byrd JC, Maus MV. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood. 2016;127(9):1117–27. doi: 10.1182/blood-2015-11-679134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ruella M, Kenderian SS, Shestova O, Fraietta JA, Qayyum S, Zhang Q, Maus MV, Liu X, Nunez-Cruz S, Klichinsky M, Kawalekar OU, Milone M, Lacey SF, Mato A, Schuster SJ, Kalos M, June CH, Gill S, Wasik MA. The Addition of the BTK Inhibitor Ibrutinib to Anti-CD19 Chimeric Antigen Receptor T Cells (CART19) Improves Responses against Mantle Cell Lymphoma. Clin Cancer Res. 2016;22(11):2684–96. doi: 10.1158/1078-0432.ccr-15-1527. Epub 2016/01/29. [DOI] [PubMed] [Google Scholar]
- 55.Paulos CM, Carpenito C, Plesa G, Suhoski MM, Varela-Rohena A, Golovina TN, Carroll RG, Riley JL, June CH. The inducible costimulator (ICOS) is critical for the development of human T(H)17 cells. Sci Transl Med. 2010;2(55):55ra78. doi: 10.1126/scitranslmed.3000448. Epub 2010/10/29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Suhoski MM, Golovina TN, Aqui NA, Tai VC, Varela-Rohena A, Milone MC, Carroll RG, Riley JL, June CH. Engineering artificial antigen-presenting cells to express a diverse array of co-stimulatory molecules. Mol Ther. 2007;15(5):981–8. doi: 10.1038/mt.sj.6300134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lu TL, Pugach O, Somerville R, Rosenberg SA, Kochendefer JN, Better M, Feldman SA. A Rapid Cell Expansion Process for Production of Engineered Autologous CAR-T Cell Therapies. Human gene therapy methods. 2016;27(6):209–18. doi: 10.1089/hgtb.2016.120. Epub 2016/11/30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Frey NV, Shaw PA, Hexner EO, Gill S, Marcucci K, Luger SM, Mangan JK, Grupp SA, Maude SL, Ericson S. Optimizing chimeric antigen receptor (CAR) T cell therapy for adult patients with relapsed or refractory (r/r) acute lymphoblastic leukemia (ALL) American Society of Clinical Oncology; 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hacein-Bey-Abina S, Garrigue A, Wang GP, Soulier J, Lim A, Morillon E, Clappier E, Caccavelli L, Delabesse E, Beldjord K, Asnafi V, MacIntyre E, Dal Cortivo L, Radford I, Brousse N, Sigaux F, Moshous D, Hauer J, Borkhardt A, Belohradsky BH, Wintergerst U, Velez MC, Leiva L, Sorensen R, Wulffraat N, Blanche S, Bushman FD, Fischer A, Cavazzana-Calvo M. Insertional oncogenesis in 4 patients after retrovirus-mediated gene therapy of SCID-X1. J Clin Invest. 2008;118(9):3132–42. doi: 10.1172/JCI35700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Frecha C, Levy C, Cosset FL, Verhoeyen E. Advances in the field of lentivector-based transduction of T and B lymphocytes for gene therapy. Mol Ther. 2010;18(10):1748–57. doi: 10.1038/mt.2010.178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Verhoeyen E, Dardalhon V, Ducrey-Rundquist O, Trono D, Taylor N, Cosset FL. IL-7 surface-engineered lentiviral vectors promote survival and efficient gene transfer in resting primary T lymphocytes. Blood. 2003;101(6):2167–74. doi: 10.1182/blood-2002-07-2224. [DOI] [PubMed] [Google Scholar]
- 62.Zhao Y, Zheng Z, Cohen CJ, Gattinoni L, Palmer DC, Restifo NP, Rosenberg SA, Morgan RA. High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation. Mol Ther. 2006;13(1):151–9. doi: 10.1016/j.ymthe.2005.07.688. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Deniger DC, Yu J, Huls MH, Figliola MJ, Mi T, Maiti SN, Widhopf GF, 2nd, Hurton LV, Thokala R, Singh H, Olivares S, Champlin RE, Wierda WG, Kipps TJ, Cooper LJ. Sleeping Beauty Transposition of Chimeric Antigen Receptors Targeting Receptor Tyrosine Kinase-Like Orphan Receptor-1 (ROR1) into Diverse Memory T-Cell Populations. PLoS One. 2015;10(6):e0128151. doi: 10.1371/journal.pone.0128151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Berdien B, Mock U, Atanackovic D, Fehse B. TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer. Gene Ther. 2014;21(6):539–48. doi: 10.1038/gt.2014.26. [DOI] [PubMed] [Google Scholar]
- 65.Perez EE, Wang J, Miller JC, Jouvenot Y, Kim KA, Liu O, Wang N, Lee G, Bartsevich VV, Lee YL, Guschin DY, Rupniewski I, Waite AJ, Carpenito C, Carroll RG, Orange JS, Urnov FD, Rebar EJ, Ando D, Gregory PD, Riley JL, Holmes MC, June CH. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nat Biotechnol. 2008;26(7):808–16. doi: 10.1038/nbt1410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Tebas P, Stein D, Tang WW, Frank I, Wang SQ, Lee G, Spratt SK, Surosky RT, Giedlin MA, Nichol G, Holmes MC, Gregory PD, Ando DG, Kalos M, Collman RG, Binder-Scholl G, Plesa G, Hwang WT, Levine BL, June CH. Gene editing of CCR5 in autologous CD4 T cells of persons infected with HIV. N Engl J Med. 2014;370(10):901–10. doi: 10.1056/NEJMoa1300662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Poirot L, Philip B, Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, Potrel P, Bas C, Lemaire L, Galetto R, Lebuhotel C, Eyquem J, Cheung GW, Duclert A, Gouble A, Arnould S, Peggs K, Pule M, Scharenberg AM, Smith J. Multiplex Genome-Edited T-cell Manufacturing Platform for “Off-the-Shelf” Adoptive T-cell Immunotherapies. Cancer Res. 2015;75(18):3853–64. doi: 10.1158/0008-5472.CAN-14-3321. [DOI] [PubMed] [Google Scholar]
- 68.Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y. Multiplex Cripsr/Cas9 Genome Editing to Generate Potent Universal CART and PD1-Deficient Cells Against Leukemia. Blood. 2015;126(23):4280. [Google Scholar]
