scholarly journals The Safety and Efficacy of a CRISPR/Cas9-Engineered Universal CAR-T Cell Product (CTA101) in Patients with Relapsed/Refractory B-Cell Acute Lymphoblastic Leukemia

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 52-52 ◽  
Author(s):  
Yongxian Hu ◽  
Yali Zhou ◽  
Mingming Zhang ◽  
Wengang Ge ◽  
Yi Li ◽  
...  

Introduction Although autologous CAR-T therapy targeting CD19+ B-cell has proven efficacy in hematological malignancies. The challenges such as undesirable waiting time, possible manufacturing failures and high cost, remain to be solved. In addition, antigen-loss/downregulation leads to treatment failure after single-target CAR-T therapy. To tackle these issues, we developed a universal CD19/CD22-targeting CAR-T cell with the TRAC region and CD52 gene disrupted using CRISPR/Cas9 technology. Pre-clinical data demonstrated the safety of CRISPR gene editing and the anti-leukemia ability of CTA101. Method Pts with CD19+ or CD22+ r/r B-ALL received a single dose of CTA101 with traditional 3+3 dose escalation (dose level: 1, 3 and 6×106 CAR+ T cells/kg). Prior to CTA101 infusion, pts received pre-conditioning chemotherapy regimen consisting of cyclophosphamide, fludarabine and alemtuzumab. The primary endpoint is the frequency of DLTs and AEs and secondary endpoints including ORR, OS, cellular PK, etc. (NCT04227015) Results Characteristics of CTA101 CTA101 was manufactured by electroporation of ribonucleoprotein complexes (RNP) comprising Cas9 loaded with sgRNA targeting TRAC and CD52 followed by lentiviral transduction of the CAR transgene. All products were expanded above 100-folds after 10-12 days manufacturing (Fig.1a). CAR expression was detected by FCM staining, ranging from 40~80% (Fig.1b). The frequency of editing as determined by flow cytometry was consistently above 85% (88%-97%) for TRAC and above 65% (68%-83%) for CD52 (Fig.1c, 1d). The CD19/CD22 dual targeting universal CAR-T cells showed potent antigen specific cytotoxicity, and mitigated tumor antigen escape (Fig.1e). Whole genome sequencing was carried out to assess the on-target and off-target editing events. While most mutations were on target, a few off-target mutations were identified, which needed further investigation (Fig.2). QPCR assays identified no detectable rearrangements occurred with the simultaneous editing of four loci: TRAC and CD52 (TRAC-CD52) during the manufacture process. Residual Cas9 protein was quantified during the manufacturing process, showing declining levels that were 0.195 fg/cell (detection limit of sensitive ELISA) in the final product. Clinical result As of August 5th 2020, 8 adult pts were screened, and 6 adult pts were enrolled. All of 6 patients received one infusion of CTA101 (DL1: 3 pts; DL2: 3 pts), with no more than 8 days between enrollment and infusion. The median age of 6 pts was 49 years (range, 26 to 56), the median prior lines of therapies were 5 (range, 2 to 8), and the median marrow blast percentage was 52% (range, 1 to 82). 3 pts had hyperleukocytosis prior to enrollment with difficulties in apheresis for autologous CAR-T, 1 relapsed after autologous CD22 CAR-T, and 3 had high-risk genomic lesions (Table 1). All 6 pts were available for evaluation of safety and efficacy. No DLTs, GvHD, ICANS and death occurred to date. All pts experienced CRS (3 G1, 2 G2, 1 G3), and the G3 CRS recovered within 7 days with one dose of tocilizumab and glucocorticoids. Other common AEs were prolonged cytopenia (3 G3), virus reactivation/infections (CMV 1 G2, 2 G3; ADV 1 G1), bacterial pneumonia (1 G3), and fungal sepsis (1 G3). The majority of events recovered to date. No replication competent lentivirus (RCL) has been detected. On D28 after CTA101 infusion, 5/6 (83.3%) pts achieved CR/CRi, and 5/5 (100%) pts achieved MRD-. With a median follow-up of 85 days (range, 53 to 202), 4/5 pts remained MRD-, 1/5 had MRD+ CR (Fig. 3). Only 1 pt received HSCT in remission on D60. Expansion had been observed in all pts and peaked from D10 to D14 (Fig.4). The correlation among dose level, donor difference, tumor burden and degree of expansion could not be identified. The median duration of persistence was 42 days. The lowest peak expansion was detected by qPCR in the pt without response and whose T lymphocyte recovered within D7, while FCM did not detect CTA101 expansion in the pt. Conclusions CTA101 demonstrates manageable safety profile and deep response of high MRD- CR/CRi rate. No GvHD and CRISPR/Cas9 genome editing associated AEs occurred. Early data of cellular PK and efficacy illustrate that CRISPR gene editing does not curtail the expansion and anti-leukemia capacity of CAR-T. Updated data, including long term gene editing-related AEs, will be presented after further follow-up. Disclosures Zhou: Nanjing Bioheng Biotech Co., Ltd: Current Employment. Ge:Nanjing Bioheng Biotech Co., Ltd: Current Employment. Han:Nanjing Bioheng Biotech Co., Ltd: Current equity holder in private company. Wang:Nanjing Bioheng Biotech Co., Ltd: Current Employment. Zhang:Nanjing Bioheng Biotech Co., Ltd: Current Employment. Ren:Nanjing Bioheng Biotech Co., Ltd: Current equity holder in private company.

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 4187-4187 ◽  
Author(s):  
Zixun Yan ◽  
Wen Wang ◽  
Zhong Zheng ◽  
Ming Hao ◽  
Su Yang ◽  
...  

Abstract Introduction JWCAR029 is a novel CD19-directed 4-1BB stimulated chimeric antigen receptor T (CAR-T) cell type, which is different from JWCAR017 with independent production of CD4 and CD8 T cells and transfusion in non-fixed ratio. We conducted a single arm, open-label, dose escalation Phase I trial of JWCAR029 in relapsed and refractory B-cell non-Hodgkin lymphoma (NCT03355859). Methods From January to July 2018, 10 patients have been enrolled in this trial, including eight diffused large B cell lymphoma (DLBCL) and two MALT lymphoma, with median age of 47 years (range 32 to 59 years). All the patients received immunochemotherapy as induction and more than two lines of salvage treatment. Two patients received bridging chemotherapy after T-cell collection due to rapid tumor progression, followed by re-evaluation before CAR-T cell infusion. Lymphodepletion preconditioning was accomplished by fludarabine 25mg/m2/d and cyclophosphamide 250mg/m2/d on Day-4 to D-2, followed by CAR-T cell infusion on Day0. JWCAR029 was administrated as a single infusion in escalation dose levels, from 2.5×107 CAR-T cells (dose level 1, DL1) to 5.0×107 CAR-T cells (dose level 2, DL2) and to 1.0×108 CAR-T cells (dose level 3, DL3) according to mTPI-2 algorithm. Circulating blood count, serum biochemistry, and coagulation status were follow-up after infusion. Cytokines were assessed on a Luminex platform. Tumor evaluation was performed on Day 29 by PET-CT. PK data were detected by flow cytometry and real-time quantitative polymerase chain reaction system. All the adverse events were recorded. The study was approved by the Shanghai Rui Jin Hospital Review Board with informed consent obtained in accordance with the Declaration of Helsinki. Results The demographic characteristics of the patients were demonstrated in Table 1. Among six evaluable patients (3 of DL1 and 3 of DL2), the ORR was 100% on Day 29, including four complete remission and 2 partial remission. Cytokine release syndrome (CRS) was 100% in Gr 1, with main symptoms as fever (<39.0 degrees), fatigue, and muscle soreness. No neurotoxicity was observed. Four of the six patients with fever >38.0 degrees used prophylactic IL-6 Inhibitor (8mg/kg, ACTEMRA, two patients administered twice). No patients received steroids. The CRS showed no difference between dose level groups (p>0.99). Adverse effects included leukopenia (Gr 3-4: 83.3%, Gr 1-2: 16.7%), hypofibrinogenemia (Gr 1: 16.7%, Gr 2-4: 0%), liver dysfunction (Gr 1: 33.3%, Gr 2-4: 0%), elevated CRP (Gr 1: 83.3%, Gr 2-4: 0%), ferritin (Gr 1-2: 83.3%, Gr 2-4: 0%), or IL-6 (Gr 1-2:100%, Gr 3-4: 0%, Table 2). Conclusion Although long-term follow-up was needed, the preliminary data of six patients in this trial have demonstrated high response rates and safety of JWCAR029 in treating relapsed and refractory B-cell non-Hodgkin lymphoma. Disclosures Hao: JW Therapeutics: Employment, Equity Ownership.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. SCI-37-SCI-37 ◽  
Author(s):  
James N. Kochenderfer

Multiple myeloma (MM) is a usually incurable malignancy of plasma cells. While the therapy of MM has improved greatly in the past 15 years, therapies with novel mechanisms of action are needed for MM. Allogeneic stem cell transplantation has been shown to have a potent anti-myeloma effect, and allogeneic donor lymphocyte infusions can cause remissions of MM. These results from allogeneic transplantation show that MM can be vulnerable to cellular immunotherapies, but allogeneic transplants have substantial rates of mortality and morbidity. Anti-CD19 CAR T cells have been shown to have powerful activity against B-cell malignancies. The success of anti-CD19 CAR T cells against B-cell malignancies has motivated investigators to develop genetically-modified T-cell therapies for MM. CD19 has been targeted as a therapy for multiple myeloma. A clinical trial of anti-CD19 CAR T cells for MM is underway. Part of the rationale for targeting CD19 is that CD19 might be expressed on a myeloma stem cell, which might be a mature B cell. The NY-ESO antigen has been targeted by human-leukocyte-antigen-restricted T cells in a clinical trial enrolling MM patients. B-cell maturation antigen (BCMA) is expressed by most cases of MM. We conducted the first-in-humans clinical trial of CAR T cells targeting BCMA at the National Cancer Institute. T cells expressing the CAR used in this work (CAR-BCMA) specifically recognized BCMA-expressing cells. Twelve patients received CAR-BCMA T cells on this dose-escalation trial. Among the 6 patients treated on the lowest two dose levels, limited anti-myeloma activity and mild toxicity occurred. On the third dose level, one patient obtained a very good partial remission. Two patients were treated on the fourth dose level of 9x106 CAR+T cells/kg bodyweight. Before treatment, the first patient on the fourth dose level had chemotherapy-resistant MM making up 90% of bone marrow cells. After treatment, bone marrow plasma cells became undetectable by flow cytometry, and the patient's MM entered a stringent complete remission that lasted for 17 weeks before relapse. The second patient on the fourth dose level had chemotherapy-resistant MM with 80% bone marrow plasma cells before treatment. Twenty-eight weeks after this patient received CAR-BCMA T-cells, bone marrow plasma cells were undetectable by flow cytometry, and the serum monoclonal protein had decreased by >95%. Both patients treated on the fourth dose level had toxicity consistent with cytokine-release syndrome including fever, hypotension, and dyspnea. Both patients also had prolonged cytopenias. In summary, our findings demonstrated strong anti-myeloma activity of CAR-BCMA T cells. One of the best attributes of the CAR T-cell field is that there are multiple avenues for improving CAR T-cell therapies. New CAR designs are being tested. Any part of the CAR might be improved including development of new fully-human single chain variable fragments (scFv) for the antigen-recognition component of the CAR, testing different hinge and transmembrane domains, and defining the optimal costimulatory moieties. Another avenue for improving CAR T-cell therapies is improving T-cell culture methods. Optimizing clinical application of CAR T cells, especially enhancing toxicity management, is another important avenue of improving CAR T-cell therapies. Finally, identifying new CAR target antigens is a critically important area of CAR research. In summary, genetically-modified T cells hold great promise to make a profound improvement in the therapy of multiple myeloma. Disclosures Kochenderfer: bluebird bio: Patents & Royalties, Research Funding; Kite Pharma: Patents & Royalties, Research Funding.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 1730-1730
Author(s):  
Ying Zhang ◽  
Jiaqi Li ◽  
Xiangping Zong ◽  
Jin Zhou ◽  
Sixun Jia ◽  
...  

Abstract Objective: Despite the remarkable success of chimeric antigen receptor modified T (CAR-T) cell therapy for refractory or relapsed B cell non-Hodgkin lymphoma (R/R B-NHL), high rates of treatment failure and relapse after CAR-T cell therapy are considerable obstacles to overcome. Preclinical models have demonstrated that anti-PD-1 antibody is an attractive option following CAR-T therapy to reverse T cell exhaustion. Thus, we investigated their combination in R/R B-NHL. Methods: We performed a prospective, single-arm study of CAR-T cell combined with anti-PD-1 antibody treatment in R/R B-NHL (NCT04539444). Anti-PD-1 antibody was administrated on day 1 after patients received sequential infusion of anti-CD19 and anti-CD22 second-generation CAR-T cells, and the efficacy and safety of the combination treatment were evaluated. Results: From August 1, 2020 to June 30, 2021, a total of 11 patients were enrolled and completed at least 3 months follow-up. The median follow-up time is 5.8 months. Overall response was achieved in 9 of 11 patients (81.8%), and the complete response (CR) was achieved in 8 of 11 patients (72.7%). All 8 patients achieving CR still sustained remission at the last follow-up. The progression-free survival (PFS) and overall survival (OS) rates at 6 months were 80.8% and 100.0%, respectively. Cytokine release syndrome (CRS) occurred in only 4 patients (all were grade 1), and no neurotoxicity were observed. Conclusion: This study suggests that CAR-T cells combined with anti-PD-1 antibody elicit a safe and durable response in R/R B-NHL. Keywords: chimeric antigen receptor modified T cell, anti-PD-1 antibody, CD19/CD22, refractory or relapsed B cell non-Hodgkin lymphoma Disclosures No relevant conflicts of interest to declare. OffLabel Disclosure: We use the T cells were transduced with a lentivirus encoding the CD19-4-1BB-CD3 z and CD22-4-1BB-CD3 ztransgene to produce CAR-T cells. The main purpose of our study is to improve the response rate in patients with R/R B-NHL.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 2876-2876 ◽  
Author(s):  
Zhitao Ying ◽  
Pengpeng Xu ◽  
Li Wang ◽  
Shu Cheng ◽  
Wen Wu ◽  
...  

Introduction JWCAR029 is a CD19-directed 4-1BB chimeric antigen receptor (CAR) T cell product with a 4-1BB costimulatory domain, of which CD4 and CD8 CAR T cells are produced together and transfused in non-fixed ratio. We conducted a single arm, open-label, dose escalation Phase I trial of JWCAR029 in relapsed and refractory B-cell non-Hodgkin lymphoma (NCT03344367 and NCT03355859). Methods Eligible pts had confirmed B-cell NHL with R/R disease after ≥2 prior lines of therapy. All subjects received lymphodepleting chemotherapy prior to receiving JWCAR029. After lymphodepleting chemotherapy, JWCAR029 was administrated as a single infusion in escalating dose levels, from 25×106 CAR T cells (dose level 1, DL1), 50×106 CAR T cells (dose level 2, DL2), 100×106 CAR T cells (dose level 3, DL3) to 150×106 CAR T cells (dose level 4, DL4) according to mTPI-2 algorithm. Circulating blood counts, serum biochemistry, coagulation status, and cytokines were followed up after infusion. Cytokines were assessed on a Luminex platform. Tumor evaluation was evaluated per the Lugano criteria by PET-CT (Cheson, 2014) and safety and disease status was followed at approximately 1, 3, 6, 9, 12, 18 and 24 months after receiving JWCAR029. PK was measured by flow cytometry and real-time quantitative polymerase chain reaction system. All the adverse events were recorded for 24 months after infusion. The study was approved by Beijing Cancer Hospital and Shanghai Rui Jin Hospital Review Board with informed consent obtained in accordance with the Declaration of Helsinki. Results As of July 5, 2019, 44 patients were screened and 32 patients were enrolled and received treatment in two study sites in China. Twenty nine patients are evaluable and have been followed for at least 6 months: 20 diffuse large B cell lymphoma (DLBCL) and 9 follicular lymphoma, mantle cell lymphoma and extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue lymphoma. Median age was 52 years (range 29 to 68 years). The demographic characteristics of the patients are shown in Table 1. All patients received immunochemotherapy as induction and a median of four lines of salvage treatment (range 2 to 7). Eleven (34%) patients received bridging chemotherapy after T cell collection due to rapid tumor progression, followed by re-evaluation before CAR T cell infusion. Lymphodepletion consisted of fludarabine 25mg/m2/d and cyclophosphamide 250mg/m2/d on Day -4 to Day -2, followed by CAR T cell infusion on Day 0. Median time to peak CAR+ T cell expansion was 11 (8-15) days. No DLTs were reported. There were no treatment-related deaths. Seventeen patients (53.1%) reported cytokine release syndrome (CRS) with 16 grade 1 or 2 (50%) and 1 (3.1%) grade 3. No grade 4 or 5 CRS was observed. Main symptoms were fever (>39.0 degrees), fatigue, and muscle soreness. The rate of CRS was similar across dose level groups. Grade 1 and 2 neurotoxicity was observed in 5 patients (15.6%). No grade ≥3 neurotoxicity was reported. Most common adverse events (frequency >20%) included leukopenia (Gr 3-4: 21.9%, Gr 1-2: 43.8%), lymphopenia (Gr 1-2: 21.9%, Gr 3-4: 21.9%), neutropenia (Gr 1-2: 37.5%, Gr 3-4: 28.2%), thrombocytopenia (Gr 1-2: 21.9%, Gr 3-4: 3.1%), pyrexia (Gr 1-2: 21.9%) and immunoglobulins decreased (Gr 1: 28.1%). Among all 29 efficacy-evaluable patients (6 of DL1, 6 of DL2, 8 of DL3 and 9 of DL4), the best ORR was 89.7%; 85% for DLBCL patients. ORR/CRR of all evaluable patients at 1, 3 and 6 months were 86.2%/65.5%, 69%/62.1% and 58.6%/55.2%, respectively, and for the 20 DLBCL patients the ORR/CRR was 80%/60%, 55%/55%, and 45%/45%, respectively (Table 2). Conclusion Although longer follow-up is needed, the data from 29 evaluable patients in this Phase I trial have demonstrated high response rates and a favorable safety profile of JWCAR029 in relapsed and refractory B-cell non-Hodgkin lymphoma. A Ph II trial that further assess safety and efficacy of JWCAR029 in DLBCL and FL patients has been initiated and is open for enrollment. Disclosures Wang: JW therapeutics (Shanghai) Co., Ltd: Employment, Equity Ownership. Hao:JW therapeutics (Shanghai) Co., Ltd: Employment, Equity Ownership. Yang:JW therapeutics (Shanghai) Co., Ltd: Employment, Equity Ownership. Lam:JW therapeutics (Shanghai) Co., Ltd: Employment, Equity Ownership. Li:JW therapeutics (Shanghai) Co., Ltd: Employment, Equity Ownership. Zheng:JW therapeutics (Shanghai) Co., Ltd: Employment, Equity Ownership.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 3010-3010 ◽  
Author(s):  
Jesus G. Berdeja ◽  
Yi Lin ◽  
Noopur S. Raje ◽  
David Samuel DiCapua Siegel ◽  
Nikhil C. Munshi ◽  
...  

3010 Background: To test the safety and efficacy of the CAR T cell modality in relapsed/refractory multiple myeloma (MM), we have designed a second-generation CAR construct targeting B cell maturation antigen (BCMA) to redirect T cells to MM. bb2121 consists of autologous T cells transduced with a lentiviral vector encoding a novel CAR incorporating an anti-BCMA scFv, a 4-1BB costimulatory motif and a CD3-zeta T cell activation domain. We will report updated safety and efficacy following initial results (Berdeja et al, ENA 2016). Methods: CRB-401 (NCT02658929) is a multi-center phase 1 dose escalation trial of bb2121 in patients with relapsed and/or refractory MM who have received ≥ 3 prior regimens, including a proteasome inhibitor and an immunomodulatory agent, or are double-refractory, and have ≥ 50% BCMA expression on plasma cells. Peripheral blood mononuclear cells are collected via leukapheresis. Patients undergo lymphodepletion with Flu (30 mg/m2) / Cy (300 mg/m2) daily for 3 days then receive 1 infusion of bb2121. The study follows a standard 3+3 design with planned dose levels of 5, 15, 45, 80 and 120 x 107 CAR+ T cells. Results: As of November 18, 2016, 11 patients had been infused with bb2121 in the first 4 dose cohorts, and 9 patients had reached at least 1 month of follow-up. As of data cut-off, no dose-limiting toxicities and no > Grade 2 neurotoxicities or cytokine release syndrome (CRS) had been observed. Grade 1-2 CRS had been reported in 8/11 (73%) treated patients. All patients treated with doses of 15.0 x 107or higher remained on study and the overall response rate (ORR) in the 6 evaluable patients at these doses was 100%, including 2 sCRs and 2 MRD-negative responses (1 sC, 1 VGPR). CAR+ T cell expansion has been demonstrated consistently. An additional 6 months of follow up on previously reported results and initial data from an additional ~10 patients will be presented. Conclusions: bb2121 shows promising efficacy at dose levels above 5 x 107 CAR+ T cells, including 2 sCRs and ongoing clinical responses at 6 months, with mild and manageable CRS to date. These initial data support the potential of CAR T therapy with bb2121 as a new treatment paradigm in MM. Study sponsored by bluebird bio. Clinical trial information: NCT02658929.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 53-54
Author(s):  
Robbie G. Majzner ◽  
Matthew J. Frank ◽  
Christopher Mount ◽  
Aidan Tousley ◽  
David M. Kurtz ◽  
...  

CD19 CAR T cells have revolutionized the treatment of relapsed and refractory (R/R) large B cell lymphomas (LBCL), mediating durable complete responses in approximately 40-50% of patients. Besides a loss or decrease in CD19 expression, no studies have identified tumor specific factors driving inherent or acquired resistance to CAR T cells in LBCL. Mutations in and loss of expression of LFA-3 (CD58) have been described in approximately 20% of cases of LBCL. As the ligand for CD2 on T cells, CD58 provides costimulation to T cells and CD58 loss or mutation has been linked to immune resistance in LBCL. We evaluated CD58 status in fifty-one R/R LBCL patients treated at Stanford with commercial axicabtagene ciloleucel (axi-cel) through immunohistochemistry (IHC) on tumor biopsy samples and/or deep sequencing of circulating tumor DNA by CAPP-Seq. We identified 12/51 (24%) patients with a CD58 aberration (lack of expression by IHC or mutation by CAPP-Seq). Progression-free survival (PFS) was significantly decreased in patients with a CD58 aberration (median PFS for CD58 aberration 3 months vs. not reached for CD58 intact, p&lt;0.0001). In fact, only 1/12 patients with a CD58 alteration achieved a durable, complete response to axi-cel, while the remaining 11 patients progressed, most commonly after a period of initial response. Partial responses were more common among patients with CD58 aberrations (58% for CD58 aberration vs 10% for CD58 intact, p&lt;0.001), and complete responses were less common (25% for CD58 aberration vs 82% for CD58 intact, p&lt;0.0001). To probe the biology of CAR T cell responses towards tumors lacking functional CD58, we generated a CD58 knockout Nalm6 model. CD19.CD28.ζ, CD19.4-1BB.ζ, and CD22.4-1BB.ζ CAR T cells demonstrated significantly reduced cytokine production and cytolytic activity in response to CD58 KO vs wildtype (WT) tumor cells. Additionally, while mice inoculated with WT Nalm6 and treated with any of the three CARs demonstrate complete responses and prolonged leukemia-free survival, mice inoculated with CD58KO Nalm6 demonstrated only partial responses, eventual tumor progression, and death from leukemia. CD2, the T cell ligand for CD58, plays both an adhesive role and a costimulatory role in T cells. CD2 knockout resulted in significantly reduced cytokine production after CAR stimulation. Re-expression of only the CD2 extracellular domain did not rescue CAR function, indicating that CD2 signaling is essential for full CAR activation. Additionally, when we stimulated CD19 CAR T cells with anti-idiotype antibody (CAR stimulation), soluble CD58 (CD2 stimulation), or both, we observed significantly enhanced phosphorylation of both CD3ζ and ERK by western blot in CAR T cells stimulated through both the CAR and CD2. Phosphorylation analysis by mass spectrometry revealed that CD2 stimulation enhances phosphorylation of proximal signaling molecules in the TCR pathway (LCK, LAT, CD3ε among others) and also mediators of actin-cytoskeletal rearrangement in CAR T cells, consistent with effects in natural T cell responses. To overcome CD58 loss in LBCL, we generated second- and third-generation CAR T cell constructs integrating CD2 costimulatory domains within the CAR molecule. While these cis constructs demonstrated increased potency against CD58KO cells in vitro, they were unable to ultimately overcome CD58 loss in vivo. However, when CARs were co-expressed with an additional CD2 receptor in trans, they mediated significant anti-tumor activity in vivo, overcoming CD58 knockout in tumor cells. In conclusion, we have identified that CD58 status is an important biomarker for durable response to CAR T cells in LBCL. We modeled the biologic basis for this finding and generated CAR T cells capable of overcoming CD58 loss in B cell malignancies. CD58 mutations have been reported in many cancers, including multiple myeloma and colon cancer, and are likely to play a role in immune evasion for CAR T cells as they are developed for additional histologies. These data provide rationale for investigating CD58 status for patients receiving CAR based therapeutics and devising next generation CARs capable of overcoming this newly discovered mechanism of resistance. Disclosures Majzner: Xyphos Biopharma: Consultancy; Zai Lab: Consultancy; Lyell Immunopharma: Consultancy; GammaDelta Therapeutics: Membership on an entity's Board of Directors or advisory committees; Aprotum Group: Consultancy; Illumina Radiopharmaceuticals: Consultancy. Kurtz:Roche: Consultancy; Genentech: Consultancy; Foresight Diagnostics: Other: Ownership. Sotillo:Lyell Immunopharma: Consultancy, Other: Consultancy. Alizadeh:Janssen: Consultancy; Genentech: Consultancy; Pharmacyclics: Consultancy; Chugai: Consultancy; Celgene: Consultancy; Gilead: Consultancy; Roche: Consultancy; Pfizer: Research Funding. Miklos:Miltenyi Biotec: Research Funding; Janssen: Consultancy, Other: Travel support; Pharmacyclics: Consultancy, Other: Travel support, Patents & Royalties, Research Funding; Novartis: Consultancy, Other: Travel support, Research Funding; Allogene Therapeutics Inc.: Research Funding; Juno-Celgene-Bristol-Myers Squibb: Consultancy, Other: Travel support, Research Funding; Kite-Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees, Other: Travel support, Research Funding; Adaptive Biotech: Consultancy, Other: Travel support, Research Funding. Mackall:BMS: Consultancy; Allogene: Current equity holder in publicly-traded company; Apricity Health: Consultancy, Current equity holder in private company; Nektar Therapeutics: Consultancy; NeoImmune Tech: Consultancy; Lyell Immunopharma: Consultancy, Current equity holder in private company.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 681-681 ◽  
Author(s):  
Natalie S Grover ◽  
Steven I Park ◽  
Anastasia Ivanova ◽  
Paul Eldridge ◽  
Kathryn McKay ◽  
...  

Abstract Background: Infusion of chimeric antigen receptor modified T cells targeting the CD30 molecule and encoding the CD28 endodomain (CD30.CAR-Ts) in the absence of lymphodepleting chemotherapy has been shown to be safe with responses seen in patients (pts) with relapsed/refractory (r/r) CD30+ lymphomas (Ramos et al., JCI 2017). We present here the results of a phase 1b/2 trial of CD30.CAR-Ts administered after lymphodepleting chemotherapy in pts with r/r CD30+ Hodgkin (HL) and Non-Hodgkin lymphoma (NHL). Methods: The primary objective of the phase 1b portion of the study was to determine the recommended phase 2 dose level (RP2DL) of CD30.CAR-Ts using a standard 3+3 design. Two dose levels were tested: 1 x 108 CAR-Ts/m2 (DL1) and 2 x 108 CAR-Ts/m2 (DL2). For lymphodepletion, the first 8 pts (including the first 3 pts treated at DL1) received 2 days of bendamustine (benda) 90 mg/m2, while the 10 remaining pts received 3 days of benda 70 mg/m2 and fludarabine (flu) 30 mg/m2, except for one pt who received only 1 day of benda and flu due to possible benda toxicity. Inclusion criteria were age ≥ 18 years, CD30+ disease, and r/r HL or NHL having failed ≥2 prior therapies. Results: At the time of data cut off (7/1/2018), 18 pts with a median age of 40.5 years (range: 23-70) had received CD30.CAR-Ts and undergone response assessment. Sixteen pts had HL, 1 had enteropathy associated T cell lymphoma, and 1 had Sezary syndrome. All pts were heavily pre-treated with a median of 8.5 prior therapies (range: 4-17). All pts had received prior brentuximab vedotin and 13 had prior checkpoint inhibitor therapy. Fourteen pts had prior autologous stem cell transplant (SCT) and 7 had prior allogeneic SCT. Treatment was well tolerated with no dose limiting toxicities; as a consequence, the highest dose level of CAR-T cells (2 x 108 CAR-Ts/m2) was given as the RP2DL. Three pts developed cytokine release syndrome (CRS) (grade 1: 2 pts and grade 2: 1 pt). Grade 1 CRS resolved spontaneously, while the pt with grade 2 CRS responded to tocilizumab. No neurotoxicity was observed. Out of the 18 pts, 4 were in a complete response (CR) before infusion due to bridging chemotherapy and remained disease free at 6 wk follow up. Two of these pts have since relapsed with PFS of 3.8 months and 11.9 months while the other 2 pts are still in CR after 1 year of follow up. The 14 pts with evidence of disease pre-lymphodepletion were included in efficacy analysis. Of these 14 pts, 6 had a CR (43%, all in the benda/flu cohort), 1 had partial response (7%), 2 had stable disease (14%) and 5 had progressive disease (35%) at disease assessment. No responses occurred in the 3 pts treated at DL1. At median follow up of 138 days, the median PFS was 129 days. The median PFS for the 3 evaluable pts who received benda at DL1 was 55 days vs 172 days for the 9 pts who received benda/flu at DL2 (p = 0.039). The median PFS for the 2 evaluable pts at DL2 who received benda lymphodepletion was 85.5 days but this was not included in the comparison due to small sample size. Two out of 14 evaluable pts remain in CR at 1 year. Using PCR on peripheral blood, CD30.CAR-Ts were found to be increased in the circulation of all pts, peaking at wk 2 post infusion, with increasing CAR-T cells in pts receiving greater number of CAR-T cells or more robust lymphodepletion (3.4x103 ± 2.9x103 copies/ug of DNA for DL1-beda vs. 61x103 ± 41x103 for DL2-benda vs. 59x103 ± 22x103 for benda/flu). These differences were confirmed by flow cytometry (CD3+CAR+ cells = 13%±9% for DL1-benda vs 21%±10% for DL2-benda vs 35%±8% for benda/flu). Persistence was also related to dose level and lymphodepletion (0.06x103 ± 0.01x103 vs 0.44x103 ± 0.41x103 vs 28x103 ± 15x103/ug of DNA at wk 4 for DL1-benda, DL2-benda, and benda/flu, respectively). Although both lymphodepletion regimens reduced the lymphocyte counts, only the combination of benda/flu was found to have a significant increase in IL-15 and IL-7 cytokines (13 fold, p<0.01 and 3 fold, p=0.016, respectively) that was sustained for 2 wks post infusion. Conclusions: We show that CD30.CAR-Ts combined with lymphodepletion with benda/ flu are safe and at 2 x 108 CAR T cells/m2 demonstrate excellent antitumor activity for pts with r/r CD30+ lymphomas. We also find that the addition of flu is critical for enhancing cytokines important for T cell growth and persistence. Finally, we demonstrate a significant PFS advantage in pts with r/r CD30+ lymphoma who received the highest dose level combined with benda and flu. Disclosures Grover: Seattle Genetics: Consultancy. Park:G1 Therapeutics: Consultancy; Gilead: Speakers Bureau; Seattle Genetics: Research Funding; Teva: Research Funding; Rafael Pharma: Consultancy; BMS: Consultancy; BMS: Research Funding; Seattle Genetics: Speakers Bureau; Teva: Consultancy; Takeda: Research Funding. Dittus:Seattle Genetics: Consultancy. Serody:Merck: Research Funding.


Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 806-806
Author(s):  
Sara Ghorashian ◽  
Anne Marijn Kramer ◽  
Sarah Jayne Albon ◽  
Gary Wright ◽  
Fernanda Castro ◽  
...  

Abstract Introduction: Published studies of CD19 CAR T cells have shown unprecedented response rates in ALL but with a 23-27% incidence of severe Cytokine Release Syndrome (CRS) and 27-50% incidence of severe neurotoxicity which may limit broader application. We developed a novel second generation CD19CAR (CAT-41BBz CAR) with a lower affinity and faster off-rate but equivalent on-rate than the FMC63-41BBz CAR (Kd 116 nM vs 0.9 nM, T1/2 10s vs 1260s) utilised in CTL019 currently under consideration by the FDA. Pre-clinical studies indicated T-cells transduced with CAT-41BBz mediate enhanced tumor clearance and show increased expansion in an NSG-NALM6 stress test model (Kramer et al., submitted). We here report interim results from a multi-centre, Phase I clinical study of autologous CAT-41BBz CAR T cells as therapy for high risk/relapsed paediatric ALL, CARPALL (NCT02443831) demonstrating efficacy with an excellent safety profile. Methods: Autologous T cells were activated with anti-CD3/CD28 beads, transduced with a SIN lentiviral vector encoding CAT-41BBz CAR and expanded for 4 days prior to magnetic bead removal and cryopreservation. Transduction efficiency was assessed using an anti-idiotype antibody. Serum levels of cytokines associated with CRS were measured using cytometric bead array. All patients received lymphodepletion with fludarabine 150 mg/m2 + cyclophosphamide 1.5g/m2 followed by a single infusion of CAR T cells at a dose of 1x106 CAR+ T cells. Patients were monitored for the presence of CAR T cells in the blood by flow cytometry and by qPCR for the 41BBz junctional region, as well as circulating B cell count monthly for 6 months and then 6 weekly to 1 year. Disease status was assessed in the bone marrow morphologically, by IgH qPCR, as well as by flow cytometric assessment of MRD at the same time-points to establish durability of responses as a stand-alone therapy. The primary end-points were incidence of grade 3-5 toxicity related to CAR T cells within 30 days and the proportion of patients achieving molecular remission. Results: We have enrolled 10 patients and treated 8 to date. Six of 8 had relapsed post myeloablative SCT. The median disease burden prior to lymphodepletion was 9% blasts (ranging from molecular CR to 74% blasts, Table 1). It was possible to generate a product meeting release criteria in all but 1 patient (90% feasibility). Median transduction efficiency was 18.1% (range 6.7 to 76.3%). All treated patients received the anticipated dose of 1x106 CAR T cells/kg. Cytokine release syndrome occurred in all patients (grade 1 n=4, grade 2 n=4), but to date none have developed ≥ grade 3 CRS, required ICU admission or therapy with Tocilizumab. CRS was associated with modest elevations of IL-6, IFN-γ and IL-10 and resolved spontaneously in all. Grade 2 neurotoxicity was observed in 3 patients and resolved spontaneously, but no severe (≥grade 3) neurotoxicity was seen. Five patients had prolonged grade 4 neutropenia lasting &gt; 30 days but this resolved in all by 2 months. Only 1 patient experienced significant infective complications in the context of pre-existing poor marrow reserve following allogeneic SCT. 6/7 (86%) evaluable patients achieved molecular remission at a median of 30 days post infusion (range 30-60 days, Table 1). One patient did not respond and died of CD19+ disease progression. At a median follow-up of 5.9 months (range 28-328 days), 4/7 evaluable patients remain in flow MRD negative remission of whom 3 show no evidence of molecular MRD at 1, 7.5 and 9 months. Two patients relapsed with CD19- disease at 3 and 4 months post infusion: 1 of these remains alive with disease at 11 months and the other died of disease progression. Reflecting our pre-clinical data with CAT-41BBz CAR, we have seen excellent CAR T cell expansion (median 65459 copies/µg DNA at 1 month, range 609 to 230112) and persistence at up to 11 months post-infusion (Figure 1). All 7 evaluable patients have ongoing CAR T cell persistence detectable by both flow and qPCR as well as ongoing B cell aplasia at last follow-up. Conclusions: These interim results with a novel low affinity CD19 CAR show similar remission rates to those reported by US studies in paediatric ALL with an improved safety profile. No severe (grade ≥3) CRS or neurotoxicity has occurred to date despite high tumour burden in 4 patients. Excellent CAR T cell expansion has been documented, as well as long duration of CAR T cell persistence and associated B cell aplasia. Disclosures Ghorashian: UCL: Patents & Royalties: UCL Business. Kramer: UCL: Patents & Royalties: UCL Business. Lucchini: Alexion: Membership on an entity's Board of Directors or advisory committees. Pule: Autolus Ltd: Employment, Equity Ownership, Research Funding; UCL: Patents & Royalties: UCL Business.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 219-219 ◽  
Author(s):  
Rebecca Gardner ◽  
Olivia Finney ◽  
Hannah Smithers ◽  
Kasey J Leger ◽  
Colleen E. Annesley ◽  
...  

Abstract Introduction: Multiply recurrent pre-B-cell ALL, and particularly relapse following allo-HSCT, has dismal outcomes due in large part to ineffectual therapies. The primary objectives of the Phase 1 portion of the PLAT-02 study (NCT02028455) were to determine the feasibility of manufacturing products of defined composition and transgene expression, the safety of the cryopreserved T cell product infusion, and to describe the full toxicity profile, including development of clinically significant GVHD in the post-allo-HSCT cohort. Methods: Subjects on the PLAT-02 study undergo apheresis, with their CD4 and CD8 T cell subsets prepared immunomagnetically. Following anti-CD3xCD28 bead stimulation, T cell lines are transduced with a SIN lentiviral vector that directs the co-expression of the FMC63scFv:IgG4hinge:CD28tm:4-1BB:ζ CAR and the selection/tracking/suicide construct EGFRt. Transduced cells are propagated using recombinant human cytokine cocktails to numbers suitable for clinical use over 10-22 days, during which time they are subjected to EGFRt immunomagnetic positive selection. Shortly following lymphodepleting chemotherapy, cryopreserved CD4/EGFRt+ and CD8/EGFRt+ T cell products are thawed and infused at the bedside such that patients receive a 1:1 ratio of EGFRt+ CD4 and CD8 T cells at the protocol-prescribed dose level. Results: 45 subjects have been enrolled and 43 have been treated from dose level 1 (5 x 105 CAR-T cells/kg) through 4 (10 x 106 CAR-T cells/kg). Therapeutic T cell products were released on all 45 enrolled subjects, with 1 subject requiring a second apheresis. Two subjects died of disease prior to their infusion. All 43 infused subjects received lymphodepletion chemotherapy prior to T cell infusion (cyclophosphamide, n=27; fludarabine/cyclophosphamide; n=14 cyclophosphamide/etoposide n=1; fludarabine n=1,). 91% (39/43) of subjects received infusions at the desired 1:1 CD4:CD8 ratio and their infusions were well tolerated with only 1 related AE >grade 2. 93% (40/43) of subjects had a documented MRD-negative CR within 21 days following CAR-T cell therapy. The 12 month event-free survival (EFS) is 50.8% (95% CI 36.6, 69.9) and 12 month OS is 69.5% (95% CI 55.8, 86.5). All responding subjects exhibited in vivo expansion of CAR-T cells. The % of CAR T cell expansion over time is not impacted by dose level or lymphodepletion but is impacted by disease burden (p=0.004) and total CD19 antigen burden (p=0.001) at the time of lymphodepletion. The median duration of functional CAR-T cell persistence as measured by ongoing B-cell aplasia (BCA) is impacted by the total CD19 antigen burden in the bone marrow at time of lymphodepletion (>15% vs <15%), with a median of 6.4 months vs 1.7 months (p=0.005), respectively. Flu/cy may also affect the duration of BCA with a median of 6.4 months vs 2.1 months for alternative lymphodepletion (p=0.15). Of the 18 relapses, 7 were CD19 negative. Loss of functional persistence of CAR T cells is associated with CD19+ relapse with a HR of 34 (95%CI 2.1, 552; p=0.013). Any grade CRS was seen in 93% (40/43) of infused subjects with severe CRS in 23% (10/43). Any grade neurotoxicity was seen in 49% (21/43) with a rate of severe neurotoxicity of 21% (10/43). Severity of CRS was only related to dose level (p=0.032), with no significant difference based on disease burden, total CD19 antigen burden or lymphodepletion regimen. Severity of neurotoxicity was only related to the occurrence of severe CRS (p=0.016). There were no toxic deaths. The recommended phase 2 dose is 1 X 106 CAR-T cells/kg with flu/cy conditioning. Conclusions: Infusions of defined composition CD4:CD8 CD19 CAR/EGFRt+ T cells/kg produce high rates of MRD-negative CR in pediatric and young adult B-cell ALL patients. Based on intent to treat analyses without a proliferation screen, we have found it is feasible to generate CAR products from each of the enrolled subjects. Despite the high rates of MRD-negative CR, the durability of remission is highly influenced by the functional persistence of CAR-T cells. Strategies to enhance persistence are currently being investigated, including episodic antigen stimulation through subject derived tAPC engineered to express truncated CD19. Disclosures Gardner: Amgen: Honoraria. Li:Juno Therapeutics: Employment, Equity Ownership. Jensen:Juno Therapeutics, Inc: Consultancy, Equity Ownership, Membership on an entity's Board of Directors or advisory committees, Research Funding.


Cancers ◽  
2021 ◽  
Vol 13 (4) ◽  
pp. 743
Author(s):  
Aleksei Titov ◽  
Ekaterina Zmievskaya ◽  
Irina Ganeeva ◽  
Aygul Valiullina ◽  
Alexey Petukhov ◽  
...  

Adoptive cell immunotherapy (ACT) is a vibrant field of cancer treatment that began progressive development in the 1980s. One of the most prominent and promising examples is chimeric antigen receptor (CAR) T-cell immunotherapy for the treatment of B-cell hematologic malignancies. Despite success in the treatment of B-cell lymphomas and leukemia, CAR T-cell therapy remains mostly ineffective for solid tumors. This is due to several reasons, such as the heterogeneity of the cellular composition in solid tumors, the need for directed migration and penetration of CAR T-cells against the pressure gradient in the tumor stroma, and the immunosuppressive microenvironment. To substantially improve the clinical efficacy of ACT against solid tumors, researchers might need to look closer into recent developments in the other branches of adoptive immunotherapy, both traditional and innovative. In this review, we describe the variety of adoptive cell therapies beyond CAR T-cell technology, i.e., exploitation of alternative cell sources with a high therapeutic potential against solid tumors (e.g., CAR M-cells) or aiming to be universal allogeneic (e.g., CAR NK-cells, γδ T-cells), tumor-infiltrating lymphocytes (TILs), and transgenic T-cell receptor (TCR) T-cell immunotherapies. In addition, we discuss the strategies for selection and validation of neoantigens to achieve efficiency and safety. We provide an overview of non-conventional TCRs and CARs, and address the problem of mispairing between the cognate and transgenic TCRs. Finally, we summarize existing and emerging approaches for manufacturing of the therapeutic cell products in traditional, semi-automated and fully automated Point-of-Care (PoC) systems.


Sign in / Sign up

Export Citation Format

Share Document