Development of a closed system process for purifying naive CD8+ cells, culturing and transducing with a CD19/22 chimeric antigen receptor (CAR) to produce a clinical T memory stem cell product directed against B cell malignancies

Cytotherapy ◽  
2020 ◽  
Vol 22 (5) ◽  
pp. S143-S144
Author(s):  
V. Fellowes ◽  
Y. Cai ◽  
E. Rodriguez-Mesa ◽  
P. Grandinetti ◽  
J. Jin ◽  
...  
Cytotherapy ◽  
2019 ◽  
Vol 21 (7) ◽  
pp. 769-781 ◽  
Author(s):  
Jun-Xia Cao ◽  
Wei-Jian Gao ◽  
Jia You ◽  
Li-Hua Wu ◽  
Jin-Long Liu ◽  
...  

2013 ◽  
Vol 31 (15_suppl) ◽  
pp. TPS10072-TPS10072
Author(s):  
Stephan A. Grupp ◽  
David L. Porter ◽  
Bruce Levine ◽  
Michael Kalos ◽  
Christine Strait ◽  
...  

TPS10072 Background: Outcomes remain poor for patients (pts) with relapsed or refractory (r/r) B-cell malignancies such as acute lymphoblastic leukemia (ALL) and chronic lymphocytic leukemia (CLL). CD19 is an attractive therapeutic target because it is widely expressed on normal and malignant B cells throughout B-cell maturation but not on pluripotent stem cells or non–B-cell tissues. We have developed chimeric antigen receptor T cells to target CD19+ cells (CART-19 or CTL019). This approach involves patient-derived T cells that are genetically modified via lentiviral transduction to express a CD19 antigen recognition domain attached to intracellular signaling domains (TCRζ and 4-1BB) that mediate T-cell activation. A recent study in CLL showed that CART-19 therapy had potent activity with responses in 5/8 evaluable pts (3 CR, 2 PR). Pts achieving CR (two > 24 months and one > 5 months) remain in CR with detectable CART-19 cells (Porter et al. ASH 2012). Here we describe a study of CART-19 therapy in pediatric pts with r/r leukemia and lymphoma (NCT01626495). Methods: Pts eligible for this single-arm, open-label study are aged 1 to 24 years with r/r CD19+ B-cell malignancies, without a transplant option or having relapsed after allogeneic stem cell transplantation (ASCT). Pts will undergo leukapheresis to obtain T cells, which will be stimulated, expanded, and transduced ex vivo to express the chimeric antigen receptor. Pts may receive lymphodepleting chemotherapy prior to CART-19 infusion. Study objectives: determine the safety and feasibility of administering CART-19 therapy to pediatric pts, assess duration of in vivo survival of CART-19 cells, and measure antitumor response. There are 2 cohorts in the study: Cohort 1 includes pts who have not undergone ASCT and are not currently eligible for a transplant, and cohort 2 includes pts who relapsed after ASCT. Cells are collected from the recipient, not the donor, which allows for this approach to be used in pts s/p cord blood transplant, and which we hypothesize will reduce the risk of GVHD as a result of toleration of the T cells in the recipient. To date, 7 pts have been enrolled. Clinical trial information: NCT01626495.


2020 ◽  
Vol 8 (Suppl 2) ◽  
pp. A54.1-A54
Author(s):  
N Klein-González ◽  
EA González-Navarro ◽  
A Bartoló-Ibars ◽  
V Ortiz-Maldonado ◽  
M Torrebadell ◽  
...  

BackgroundChimeric Antigen Receptor (CAR)-T cells directed against CD19 have induced high rates of response in patients with relapsed/refractory (R/R) B-cell malignancies. Two CD19-targeting constructs have been approved by the FDA and EMA (Yescarta ®, Kymriah ®) for B lymphoblastic leukemia (B-ALL) and aggressive lymphoma. Despite deep remissions, there are still major challenges and disparate data are reported about the immunogenicity induced by CART-cell therapy. On May/2017, the Spanish Agency of Medicines approved our first clinical trial (clinicaltrials.gov NCT03144583) with a fully academic CART-19.Materials and MethodsEligibility criteria included R/R B-ALL (adult and pediatric), non-Hodgkin’s lymphoma (NHL) and chronic lymphocytic leukemia(CLL) who failed standard therapy. The primary objective of the study was safety and secondary objectives were response rate and its duration. The humoral anti-CART response was assessed by a (cell-based) fluorescence assay to detect human anti-murine antibodies (HAMA) in patients sera. Assessment was conducted at different time points: 1) at baseline (pre-dose), 2) on day 14 after the administration of ARI-0001 cells, 3) on day 28, 4) on day 100, and 5) every 3 months thereafter.ResultsForty-seven patients (37 adults/10 pediatrics) received ARI-0001 cells. Thirty-eight patients had a diagnosis of R/R B-ALL (28 adults and 10 children); all but 5 had relapsed after allogeneic hematopoietic stem cell transplant (HCT). Seven patients had a diagnosis of NHL, four of them (57%) had relapsed after HCT, and 2 patients had a diagnosis of CLL (2). Median age was 27 years (3–68). After conditioning with fludarabine (90 mg/m2) and cyclophosphamide (900 mg/m2), a total dose of 0.5–5 x106 ARI-0001 cells/kg was infused. Autologous T-cells from peripheral blood were expanded and transduced with a lentivirus to express a CAR with a single-chain variable fragment (scFv) with anti-CD19 specificity, conjugated with the co-stimulatory regions 4-1BB and CD3z. The scFv was originated from a mouse monoclonal antibody A3B1. Twenty-five per cent of the patients tested positive for the presence of anti-CAR antibodies, all of them post-dose, in contrast to previous data reported on Kymriah® with a significant presence of pre-dose anti-murine CAR19 antibody. Of these 12 patients, 8 patients presented with a weak, and 4 patients with a strong presence of HAMA. The last 4 patients had lost the effectiveness of the CART- therapy at that time point, reflected by simultaneous B-cell recovery in the periphery. Moreover, three of them received a second dose of CART-19, which did not revert the relapse.ConclusionsTo conclude, these data suggest the importance of the immunogenicity induced by CART-cell therapies. Immune monitoring should include the assessment of humoral response, especially before considering a second dose after relapse.Disclosure InformationN. Klein-González: None. E.A. González-Navarro: None. A. Bartoló-Ibars: None. V. Ortiz-Maldonado: None. M. Torrebadell: None. M. Castellà: None. D. Benítez: None. M. Caballero-Baños: None. R. Cabezón: None. M. Español: None. T. Baumann: None. E. Giné: None. P. Castro: None. J. Esteve: None. J. Yagüe: None. S. Rives: None. Á. Álvaro Urbano-Ispizua: None. J. Delgado: None. M. Juan: None.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3949-3949
Author(s):  
Barbara Savoldo ◽  
Carlos A. Ramos ◽  
Catherine M. Bollard ◽  
Enli Liu ◽  
Martha Mims ◽  
...  

Abstract Abstract 3949 B cell lymphomas have been effectively treated by immunotherapy, including monoclonal antibodies (MAbs) and adoptive T cell transfer. To extend this success, investigators have genetically modified T cells to express a B cell specific antibody incorporated in an artificial chimeric antigen receptor (CAR), essentially combining antibody and cell-based approaches. Early B cell directed CARs combined the antigen binding domains of the variable regions of a CD19 or CD20 MAb (scFv) with the CD3ζ endodomain of the TCR/CD3 complex. Although such CARs confer potent cytotoxic function to T cells, initial clinical trials showed that T cells modified to express CARs engineered in this way had limited in vivo persistence, apparently receiving insufficient costimulation following CAR engagement. To overcome the above limitations, a multiplicity of costimulatory endodomains, including CD28, 41BB or OX40, have been incorporated into the CAR molecule. Because of patient heterogeneity, it has proved difficult to draw definitive conclusions about the relative expansion, persistence and effectiveness of cells with each modification, so that their comparative value in human subjects remains speculative. We therefore designed a phase I, dose escalation clinical trial in which patients with refractory/relapsed B cell malignancies were simultaneously infused with two autologous T cell products. Both express a CAR with an identical CD19-specific exodomain, but one CAR also has a CD28-ζ endodomain while the other expresses only a ζ endodomain. With this study design, each patient acts as a “self-control”, allowing us to directly discover the consequences of CD28 costimulation for the fate of the T cells in vivo. We enrolled two patients at each of the three cell dose levels (1st level = 2×107/m2 of each product; 2nd level = 1×108/m2; 3rd level = 2×108/m2). End points of the study were safety, persistence of each of the two generations of CAR-modified T cells, and assessment of antitumor activity. T cell products were generated by activation of autologous PBMC with immobilized OKT3 and gene modified with retroviral vectors encoding either CAR.19ζ or CAR.19-28ζ. After transduction, T cells were expanded ex vivo for a median of 14 days (range 6–18) in the presence of IL-2. CAR expression was 42%±18% and 49%±16% for CAR.19ζ and CAR.19-28ζ, respectively. This corresponded to 51,246±16,795 and 18,283±9,484 transgene copy numbers/μg DNA, respectively, as measured by Q-PCR. Products contained both CD8+ cells (CAR.19ζ = 49%±22%, CAR.19-28ζ = 48%±22%). Few naïve T cells were present in either transduced population (CD45RA+ = 6%±5% and 6%±6%, respectively), and memory T cells predominated in both (CD45R0+CD62L+ = 50%±24% and 47%±66%, respectively). Both T cell components equally and specifically targeted CD19+ tumors in vitro as assessed by 51Cr release assays (specific lysis was 53%±10% for CAR.19ζ and 65%±19% for CAR.19-28ζ at a 20:1 E:T ratio). All infusions were well tolerated in all patients. Persistence of CAR+ T cell was assessed in peripheral blood by Q-PCR assays specific for CAR.19ζ and CAR.19-28ζ. Molecular signals for CAR.19-28ζ began at a low level after infusion, but progressively increased (7 to 63 fold) to peak at 1–2 weeks post infusion, before declining to background levels over the ensuing 8 to 13 weeks. By contrast, molecular signals corresponding to CAR.19ζ+ cells were barely detectable after infusion, showed no expansion, and rapidly disappeared. Currently 4 patients are evaluable for disease response; 2 had stable disease for up to 6 months and 2 had progressive disease. Hence, infusion of both CAR.19ζ and CD19-28ζ T cells has been safe at current doses. Direct comparison of each cell product in individual patients indicates that inclusion of the CD28 costimulatory endodomain (2nd generation CAR) enhances expansion and persistence. Nonetheless, both the limited expansion and persistence and the modest clinical effects suggest that additional modifications will need to be made to CAR endodomains to optimize the benefits of this therapy. We suggest our approach will allow these modifications to be evaluated systematically and directly even in small-scale clinical studies. Disclosures: Off Label Use: T cell products in studies conducted under INDs.


Sign in / Sign up

Export Citation Format

Share Document