scholarly journals Point-Of-Care CAR T-Cell Production (ARI-0001) Using a Closed Semi-automatic Bioreactor: Experience From an Academic Phase I Clinical Trial

2020 ◽  
Vol 11 ◽  
Author(s):  
Maria Castella ◽  
Miguel Caballero-Baños ◽  
Valentín Ortiz-Maldonado ◽  
Europa Azucena González-Navarro ◽  
Guillermo Suñé ◽  
...  
2017 ◽  
Vol 35 (15_suppl) ◽  
pp. TPS7568-TPS7568 ◽  
Author(s):  
Jae Hong Park ◽  
Isabelle Riviere ◽  
Xiuyan Wang ◽  
Brigitte Senechal ◽  
Yvette Bernal ◽  
...  

TPS7568 Background: Despite the recent progress in the therapy of CLL with BTK, PI3Kδ, and BCL2 inhibitors, CLL remains incurable and patients with high-risk disease features (i.e. del17p, complex karyotype) and patients whose disease progress after treatment with the above targeted agents continue to have extremely poor prognosis. CD19-specific chimeric antigen receptor (CAR) T cell therapy with various 2nd generation CARs (19-28z or 19-41BBz) have demonstrated anti-tumor efficacy in CLL but the complete response (CR) rates in CLL have been suboptimal (20-45%) compared to CR rates in ALL (80-90%). The suboptimal activity of the current 2nd generation CAR T cells can be due to the inhibitory tumor microenvironment (TM) of CLL. We believe one approach to over the hostile TM is through the use of CD19-CAR T cells further modified to express a second costimulatory ligand, 4-1BBL. A binding of 4-1BBL to its cognate receptor enhances T cell proliferation, IL-2 secretion, and survival and cytolytic activity of the T cells compared to 19-28z. 19-41BBz and 1928BBz (Zhao Z et al. Cancer Cell 2015;28:415-428). Methods: This phase I dose escalating trial is a single-center clinical trial (MSKCC) to study the safety and efficacy of autologous EGFRt/19-28z/4-1BBL+ CAR T cells in patients with relapsed CLL. Given the concern for potential systemic toxicity the vector includes a "safety switch" in the form of a gene for the expression of truncated form of human epidermal growth factor receptor (EGFRt). Patients with relapsed CLL are eligible for the trial. Patients will receive conditioning chemotherapy of cyclophosphamide followed by escalating doses of CAR T cells (1x105 – 3x106 CAR T cells/kg). The primary endpoint is safety and maximum tolerated doses of the CAR T ells. Secondary objectives include response assessment by iwCLL criteria. The comprehensive treatment algorithms for CRS and neurotoxicity are based on our CAR T cell experience in other studies. The study will begin enrollment in February 2017 and enroll up to 30 patients. Clinical trial information: Pending.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii113-ii113
Author(s):  
Linchun Jin ◽  
Haipeng Tao ◽  
Meng Na ◽  
Aida Karachi ◽  
Hector Mendez-Gomez ◽  
...  

Abstract BACKGROUND Chimeric antigen receptor (CAR) modified T cell therapy is a promising treatment strategy for cancer therapy. We developed IL-8 receptor-linked CD70CAR (8R-70CAR), aiming to enhance T-cell intratumoral trafficking and persistence, guided by IL-8-secreting tumors. The preclinical results show that 8R-70CAR T cells induce complete tumor regression and long-lasting antitumor immunity, which necessitate further clinical development of this technology. OBJECTIVE To assess the toxicity of 8R-70CAR T cells, evaluate in vitro tumor recognition and expansion of the 8R-70CAR T cells, and develop clinically compliant SOPs for the CAR T cell manufacture. METHODS The m8R-70CAR (mouse), h8R-70CAR (human), and control CAR were constructed. Toxicology: C57BL/6J mice were intravenously injected with or without 2x106 m8R-70CAR T cells after total body irradiation (5Gy). The body weight, clinical signs, hematology, serum chemistry, serum cytokines, gross pathology, and histopathology of 12 vital organs were evaluated. Tumor recognition: T cells derived from GBM patients were transduced with h8R-70CAR and then co-cultured with CD70+ glioblastoma. INF-g production was measured. Ex vivo expansion: The G-Rex system was utilized for CAR T cell production, and the expansion of h8R-70CAR T cells was assessed by cell count. RESULTS No CAR T cell-related toxicity was observed in mouse study. In addition to a robust antitumor reactivity, the h8R-70CAR T cells present an auto-stimulative property that elicits greater ex vivo expansion compared to other CAR T cells tested. An approximately 1200-fold expansion was achieved during the 2 weeks CAR T cell production without additional stimulation, which allows production of the CAR T cells on a clinical trial scale using < 100mL of whole blood. CONCLUSION The 8R-70CAR T cells are effective, auto-stimulative, and safe. These properties are fundamental for successful clinical applications in cancer treatment. A phase I trial using the 8R-70CAR T cells in newly diagnosed GBM will be initiated soon.


2016 ◽  
Vol 39 (3) ◽  
pp. 140-148 ◽  
Author(s):  
Hyeon-Seok Eom ◽  
Beom K. Choi ◽  
Youngjoo Lee ◽  
Hyewon Lee ◽  
Tak Yun ◽  
...  

2020 ◽  
Vol 38 (17) ◽  
pp. 1938-1950 ◽  
Author(s):  
Nirali N. Shah ◽  
Steven L. Highfill ◽  
Haneen Shalabi ◽  
Bonnie Yates ◽  
Jianjian Jin ◽  
...  

PURPOSE Patients with B-cell acute lymphoblastic leukemia who experience relapse after or are resistant to CD19-targeted immunotherapies have limited treatment options. Targeting CD22, an alternative B-cell antigen, represents an alternate strategy. We report outcomes on the largest patient cohort treated with CD22 chimeric antigen receptor (CAR) T cells. PATIENTS AND METHODS We conducted a single-center, phase I, 3 + 3 dose-escalation trial with a large expansion cohort that tested CD22-targeted CAR T cells for children and young adults with relapsed/refractory CD22+ malignancies. Primary objectives were to assess the safety, toxicity, and feasibility. Secondary objectives included efficacy, CD22 CAR T-cell persistence, and cytokine profiling. RESULTS Fifty-eight participants were infused; 51 (87.9%) after prior CD19-targeted therapy. Cytokine release syndrome occurred in 50 participants (86.2%) and was grade 1-2 in 45 (90%). Symptoms of neurotoxicity were minimal and transient. Hemophagocytic lymphohistiocytosis–like manifestations were seen in 19/58 (32.8%) of subjects, prompting utilization of anakinra. CD4/CD8 T-cell selection of the apheresis product improved CAR T-cell manufacturing feasibility as well as heightened inflammatory toxicities, leading to dose de-escalation. The complete remission rate was 70%. The median overall survival was 13.4 months (95% CI, 7.7 to 20.3 months). Among those who achieved a complete response, the median relapse-free survival was 6.0 months (95% CI, 4.1 to 6.5 months). Thirteen participants proceeded to stem-cell transplantation. CONCLUSION In the largest experience of CD22 CAR T-cells to our knowledge, we provide novel information on the impact of manufacturing changes on clinical outcomes and report on unique CD22 CAR T-cell toxicities and toxicity mitigation strategies. The remission induction rate supports further development of CD22 CAR T cells as a therapeutic option in patients resistant to CD19-targeted immunotherapy.


2018 ◽  
Vol 31 (2) ◽  
pp. 126-134 ◽  
Author(s):  
Adrian P. Gee
Keyword(s):  
T Cell ◽  

2020 ◽  
Vol 23 ◽  
pp. S83
Author(s):  
T. Ran ◽  
P. Schmidt ◽  
S.B. Eichmüller ◽  
M. Schlander

2020 ◽  
Vol 148 (2) ◽  
pp. 516-517
Author(s):  
Tao Ran ◽  
Stefan B. Eichmüller ◽  
Patrick Schmidt ◽  
Michael Schlander
Keyword(s):  
T Cell ◽  

Sign in / Sign up

Export Citation Format

Share Document