scholarly journals OPTIMIZATION OF A PROCESS FOR HIGH-YIELD LENTIVIRAL VECTOR PRODUCTION APPLIED TO CAR-T CELL GENERATION

2020 ◽  
Vol 42 ◽  
pp. 421-422
Author(s):  
D.M.C. Fantacini ◽  
S.C.G. Lima ◽  
H. Brand ◽  
L.C. Batista ◽  
R. Cunha ◽  
...  
Author(s):  
Filippos T. Charitidis ◽  
Elham Adabi ◽  
Frederic B. Thalheimer ◽  
Colin Clarke ◽  
Christian J. Buchholz

2019 ◽  
Vol 12 (1) ◽  
Author(s):  
Yuan Mao ◽  
Weifei Fan ◽  
Hao Hu ◽  
Louqian Zhang ◽  
Jerod Michel ◽  
...  

Abstract Background Cancer/testis antigens (CTAs) are a special type of tumor antigen and are believed to act as potential targets for cancer immunotherapy. Methods In this study, we first screened a rational CTA MAGE-A1 for lung adenocarcinoma (LUAD) and explored the detailed characteristics of MAGE-A1 in LUAD development through a series of phenotypic experiments. Then, we developed a novel MAGE-A1-CAR-T cell (mCART) using lentiviral vector based on our previous MAGE-A1-scFv. The anti-tumor effects of this mCART were finally investigated in vitro and in vivo. Results The results showed striking malignant behaviors of MAGE-A1 in LUAD development, which further validated the rationality of MAGE-A1 as an appropriate target for LUAD treatment. Then, the innovative mCART was successfully constructed, and mCART displayed encouraging tumor-inhibitory efficacy in LUAD cells and xenografts. Conclusions Taken together, our data suggest that MAGE-A1 is a promising candidate marker for LUAD therapy and the MAGE-A1-specific CAR-T cell immunotherapy may be an effective strategy for the treatment of MAGE-A1-positive LUAD.


2020 ◽  
Vol 42 (2) ◽  
pp. 150-158 ◽  
Author(s):  
Virgínia Picanço-Castro ◽  
Pablo Diego Moço ◽  
Amanda Mizukami ◽  
Leticia Delfini Vaz ◽  
Marcelo de Souza Fernandes Pereira ◽  
...  

Bioengineered ◽  
2021 ◽  
Vol 12 (1) ◽  
pp. 2095-2105
Author(s):  
Qu-Lai Tang ◽  
Li-Xing Gu ◽  
Yao Xu ◽  
Xing-Hua Liao ◽  
Yong Zhou ◽  
...  

2020 ◽  
Vol 17 ◽  
pp. 359-368 ◽  
Author(s):  
David C. Bishop ◽  
Lisa Caproni ◽  
Kavitha Gowrishankar ◽  
Michal Legiewicz ◽  
Kinga Karbowniczek ◽  
...  

2022 ◽  
Vol 12 ◽  
Author(s):  
Zhicai Lin ◽  
Xiangzhen Liu ◽  
Tao Liu ◽  
Haixia Gao ◽  
Sitong Wang ◽  
...  

Nonviral transposon piggyBac (PB) and lentiviral (LV) vectors have been used to deliver chimeric antigen receptor (CAR) to T cells. To understand the differences in the effects of PB and LV on CAR T-cell functions, a CAR targeting CD19 was cloned into PB and LV vectors, and the resulting pbCAR and lvCAR were delivered to T cells to generate CD19pbCAR and CD19lvCAR T cells. Both CD19CAR T-cell types were strongly cytotoxic and secreted high IFN-γ levels when incubated with Raji cells. TNF-α increased in CD19pbCAR T cells, whereas IL-10 increased in CD19lvCAR T cells. CD19pbCAR and CD19lvCAR T cells showed similar strong anti-tumor activity in Raji cell-induced mouse models, slightly reducing mouse weight while enhancing mouse survival. High, but not low or moderate, concentrations of CD19pbCAR T cells significantly inhibited Raji cell-induced tumor growth in vivo. These CD19pbCAR T cells were distributed mostly in mesenteric lymph nodes, bone marrow of the femur, spleen, kidneys, and lungs, specifically accumulating at CD19-rich sites and CD19-positive tumors, with CAR copy number being increased on day 7. These results indicate that pbCAR has its specific activities and functions in pbCAR T cells, making it a valuable tool for CAR T-cell immunotherapy.


Oncotarget ◽  
2017 ◽  
Vol 8 (10) ◽  
pp. 17002-17011 ◽  
Author(s):  
Jiangtao Ren ◽  
Xuhua Zhang ◽  
Xiaojun Liu ◽  
Chongyun Fang ◽  
Shuguang Jiang ◽  
...  
Keyword(s):  
T Cell ◽  

2021 ◽  
Vol 2021 ◽  
pp. 1-9
Author(s):  
Viktor Lukjanov ◽  
Irena Koutná ◽  
Pavel Šimara

Chimeric antigen receptor T-cells (CAR T-cells) represent a novel and promising approach in cancer immunotherapy. According to the World Health Organization (WHO), the number of oncological patients is steadily growing in developed countries despite immense progress in oncological treatments, and the prognosis of individual patients is still relatively poor. Exceptional results have been recorded for CAR T-cell therapy in patients suffering from B-cell malignancies. This success opens up the possibility of using the same approach for other types of cancers. To date, the most common method for CAR T-cell generation is the use of viral vectors. However, dealing with virus-derived vectors brings possible obstacles in the CAR T-cell manufacturing process owing to strict regulations and high cost demands. Alternative approaches may facilitate further development and the transfer of the method to clinical practice. The most promising substitutes for virus-derived vectors are transposon-derived vectors, most commonly sleeping beauty, which offer great coding capability and a safe integration profile while maintaining a relatively low production cost. This review is aimed at summarizing the state of the art of nonviral approaches in CAR T-cell generation, with a unique perspective on the conditions in clinical applications and current Good Manufacturing Practice. If CAR T-cell therapy is to be routinely used in medical practice, the manufacturing cost and complexity need to be as low as possible, and transposon-based vectors seem to meet these criteria better than viral-based vectors.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3243-3243
Author(s):  
Graham Lilley ◽  
Alden Ladd ◽  
Daniel Cossette ◽  
Laura Viggiano ◽  
Gregory Hopkins ◽  
...  

Abstract T cells engineered with chimeric antigen receptors (CAR) specific to CD19 have caused rapid and durable clinical responses in ~90% of patients with acute lymphoblastic leukemia. These data support the development of additional CAR T cell products for the treatment of other hematological malignancies. Recently, B cell maturation antigen (BCMA) expression has been proposed as a marker for identification of malignant plasma cells in patients with multiple myeloma (MM). Nearly all MM and some non-Hodgkin's lymphoma tumor cells express BCMA, while normal tissue expression is restricted to plasma cells and a subset of mature B cells. Therefore, BCMA is an attractive CAR T cell target to treat patients with MM and some B cell lymphomas. To this end, using lentiviral vector technology, we successfully generated CAR T cells specific to BCMA that exhibit potent anti-tumor activity to both multiple myeloma and Burkitt's lymphoma in animal models. Manufacture of CAR T cells for individual patient treatment requires the establishment of a robust and reproducible process - since variability in manufacturing could impact the potency of each cell product. To begin to understand the parameters of the manufacturing process that might contribute to the activity of the final product, we first tested the impact of lentiviral vector (LVV) multiplicity of infection (MOI) on CAR T cell phenotype and function. Using a broad range of MOIs (0.625 to 40) across multiple independent PBMC donors we observed no differences in population doubling or cell size throughout the ~10 day manufacturing process, irrespective of the MOI used. As expected, the number of anti-BCMA CAR expressing cells, the level of CAR expression per cell and the average vector copy number (VCN) in the cell product increased proportionally with MOI. Similarly, T cell function, as determined by an IFNg cytokine release assay in response to BCMA-expressing K562 target cells, was also correlated with the LVV MOI. Notably, increased IFNg expression was readily observable at MOIs as low as 1.25 and reached a plateau with T cells generated using an MOI of 20 or more - highlighting the sensitivity of this functional assay. Analogous data demonstrating MOI dependent in vitro killing activity were obtained using a BCMA-expressing tumor cell cytotoxicity assay. Varying the LVV MOI used during transduction simultaneously alters both the amount of anti-BCMA CAR molecules expressed per cell as well as the number of T cells in the cell product that express anti-BCMA CAR. To evaluate each variable in isolation we generated T cell products containing the same frequency of anti-BCMA CAR T cells (26 ± 4% CAR+ T cells) but different levels of anti-BCMA expression per cell by diluting T cell products made with MOIs from 5 to 40 with donor-matched untransduced cells. While these populations had markedly different levels of CAR surface expression per cell (based on anti-BCMA CAR MFI levels measured by flow cytometry) both low and high expressing anti-BCMA CAR T cell products exhibited identical levels of cytotoxicity against BCMA-expressing tumor cells. These data suggest it is the number of CAR expressing cells that is the critical driver of higher functional activity (perhaps due to the efficiency of LVV mediated anti-BCMA CAR expression per transduced cell). Finally, using this information the variability in manufacturing of anti-BCMA CAR T cells was evaluated across 11 independent normal PBMC donors. All 11 products demonstrated very similar properties with respect to cell growth (population doublings, cell volume), and VCN. Importantly, using our standard MOI we obtained a consistent and high level of anti-BCMA CAR expressing T cells that resulted in robust IFNg cytokine release when co-cultured with BCMA-expression cells. Together, our data highlight the frequency of anti-BCMA CAR T cells per cell product as a key parameter for anti-tumor activity in vitro. Moreover, these data suggest that our LVV driven T cell engineering process can reproducibly generate robust anti-BCMA CAR expressing T cell products in a donor independent manner. A phase I clinical trial to evaluate this technology as a cell-based gene therapy for MM is under development. Disclosures Lilley: bluebird bio, Inc: Employment, Equity Ownership. Ladd:bluebird bio, Inc: Employment, Equity Ownership. Cossette:bluebird bio, Inc: Employment, Equity Ownership. Viggiano:bluebird bio, Inc: Employment, Equity Ownership. Hopkins:bluebird bio, Inc: Employment, Equity Ownership. Evans:bluebird bio, Inc: Employment, Equity Ownership. Li:bluebird bio, Inc: Employment, Equity Ownership. Latimer:bluebird bio: Employment, Equity Ownership. Miller:bluebird bio: Employment, Equity Ownership. Kuczewski:bluebird bio: Employment, Equity Ownership. Bakeman:bluebird bio, Inc: Employment, Equity Ownership. MacLeod:bluebird bio, Inc: Employment, Equity Ownership. Friedman:bluebird bio: Employment, Equity Ownership. Maier:bluebird bio, Inc: Employment, Equity Ownership. Paglia:bluebird bio, Inc: Employment, Equity Ownership. Morgan:bluebird bio: Employment, Equity Ownership. Angelino:bluebird bio, Inc: Employment, Equity Ownership.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 477-477
Author(s):  
Abigail Culshaw ◽  
Frederick Arce Vargas ◽  
Gerardo Santiago Toledo ◽  
Claire Roddie ◽  
Paul Shaughnessy ◽  
...  

Abstract INTRODUCTION We have previously described AUTO1, a CD19 CAR with a fast off-rate binding domain, designed to reduce CAR T-cell immune toxicity and improve engraftment. Clinical testing in two academic studies in relapsed/refractory (r/r) paediatric [NCT02443831; CARPALL] and adult B-ALL, B-NHL and B-CLL [NCT02935257; ALLCAR19] confirmed the intended function of the receptor, with low levels of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) and long-term engraftment of CAR T-cell 1,2. Based on data in adult B-ALL, we initiated a phase Ib/II registration study in r/r adult B-ALL [NCT04404660; FELIX]. To facilitate this study and future commercialization, industrialization of the manufacturing process was required. METHODS & RESULTS The original process in the academic studies was based on the Miltenyi CliniMACS Prodigy T cell transduction process. Leukapheresis was performed at the same site as manufacture. T cells were isolated from pheresis by CD4/CD8 positive selection and seeded onto the Prodigy to be activated using the Miltenyi CD3/CD28 targeting activating reagent, TransAct. The following day, transduction was carried out using a lentiviral vector. Cells were cryopreserved after an expansion phase of up to day 10 of the process. To facilitate industrialization of the AUTO1 manufacture in the multi-center, multi-regional FELIX study, we first explored the use of cryopreserved pheresis (81.3% median viability pre-selection (range 71.9 - 94.3), 1.0 days median doubling time (range 0.9 - 1.5) and 47.6% median CD19 CAR expression (range 19.1-62.1)). We concluded that optimal manufacture includes the use of fresh pheresis and the initiation of manufacture within 72 hours (99.0% median viability pre-selection (range 92.5 - 99.7), 1.3 days median doubling time (range 1.1 - 2.1) and 69.3% median CD19 CAR expression (range 22.9-86.2)). To further simplify the process, we explored removal of the pre-selection step. Full-scale runs using starting material from 4 healthy donors were conducted to compare CD4/CD8 selected with unselected cells. On the day following activation, selected cells displayed a higher percentage of viable cells, defined as cPARP-FVS780- (median: 76.1%, range: 84.5-66.4) as compared to unselected cells (median: 52.2%, range: 43.6-59.0). In addition, selected cells demonstrated a median of 23-fold expansion (range: 20.0 - 29.1) compared to a 13.3-fold expansion for unselected cells (range 6.1-17.4). Median transduction efficiencies of viable CAR+ T-cells were 53.9% (range: 43.2-56.9) and 78.0% (range: 64.5-81.1) in selected and unselected cells, respectively. CD4/8 pre-selection was determined to be a critical part of the process. A comparison of phenotype between 18 batches manufactured using the academic process and 5 batches produced from fresh material using the industrial process was carried out. No significant differences, as determined by 2-way ANOVA, were observed between the percentage of CAR+ CD3+ cells, the memory phenotype (% TSC/naive, % TCM, % TEM and % TEMRA) and the percentage expression of PD1 (figure 1). The CD4/CD8 ratio was also comparable between products of the two processes. Data from the initial 6 fresh in patients show that engraftment in the FELIX study is consistent with ALLCAR19 engraftment results. Additional patients, updated clinical data and longer follow-up will be presented at the conference. CONCLUSION Industrialization of an autologous Miltenyi CAR T process is feasible, leading to a comparable product to that manufactured in an academic setting. We have now opened the pivotal multi-center phase II part of the FELIX study in r/r adult B-ALL patients. REFERENCES Ghorashian S et al. (2019) Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat Med, 25(9):1408-1414.Roddie C et al. (2021) Durable responses and low toxicity after fast off-rate CD19 CAR-T therapy in adults with relapsed/ refractory B-ALL. J Clin Oncol, in press Figure 1. Comparison of phenotype between 18 CAR T cell batches manufactured using the academic process and 5 batches produced using the industrial process. Boxes represent median, 25th and 75th percentiles and whiskers represent minimum and maximum. Figure 1 Figure 1. Disclosures Culshaw: Autolus Ltd.: Current Employment. Arce Vargas: Autolus Ltd.: Current Employment. Santiago Toledo: Autolus Ltd.: Current Employment. Roddie: Novartis: Consultancy; Celgene: Consultancy, Speakers Bureau; Gilead: Consultancy, Speakers Bureau. Shaughnessy: BMS: Honoraria, Speakers Bureau; Sanofi: Honoraria, Speakers Bureau; Kite: Honoraria, Speakers Bureau. Cerec: Autolus Ltd.: Current Employment. Duffy: Autolus Ltd.: Current Employment. Perna: Autolus Ltd.: Current Employment. Brugger: Autolus Ltd.: Current Employment. Merges: Autolus Ltd.: Current Employment. Pule: Autolus Ltd: Current Employment.


Sign in / Sign up

Export Citation Format

Share Document