A phase I trial using CD19 CAR-T expressing PD-1/CD28 chimeric switch-receptor for refractory or relapsed B-cell lymphoma.

2019 ◽  
Vol 37 (15_suppl) ◽  
pp. 7557-7557 ◽  
Author(s):  
Hui Liu ◽  
Wen Lei ◽  
Chaoting Zhang ◽  
Chunmei Yang ◽  
Juying Wei ◽  
...  

7557 Background: The anti-CD19 chimeric antigen receptor (CAR)-T cell therapy has established efficacy in the treatment of refractory or relapsed (R/R) B-cell lymphoma. However, the existence of different immunosuppressive pathways such PD-1/PD-L1 pathway can restrict the full potential of CAR-T therapy. A recent study demonstrates that a novel PD-1/CD28 chimeric switch-receptor, comprising the extracellular domain of PD-1 and the transmembrane and cytoplasmic signaling domains of CD28, can be used a modality for augmenting antitumor activity of mesothelin- and prostate specific cancer antigen-specific CAR-T cells in either pleural mesothelioma or prostate cancer xenograft model. Here, we describe the first human trial of CD19-PD-1/CD28-CAR-T cells (a CD19-specific CAR-T expressing PD-1/CD28 chimeric switch-receptor) to evaluate safety and efficacy. Methods: This phase I, single-arm, open-label, multicenter trial enrolled patients (18-75 years) with R/R large B-cell lymphoma. PD-L1 expression was confirmed by immunohistochemistry using an anti-PD-L1 mAb (SP142), and confirmed by the review committee with a central evaluation. Seventeen patients received conditioning chemotherapy (cyclophosphamide [500 mg/m2] and fludarabine [30 mg/m2] on days -5, -4 and -3) and followed by CD19-PD-1/CD28-CART cells infusion at doses ranging 0.5 to 4 × 106 CAR+ T cells/kg on day 0. Response was assessed by F-FDG PET/CT at month 3 after CAR-T infusion, according to the International Working Group Response Criteria for Malignant Lymphoma, and duration of response was evaluated by ultrasonic and computed tomography every three months from month 3 to month 12. Results: At data cutoff, 17 patients had received CD19-PD-1/CD28-CART cells. Overall PD-L1 positivity was 15/17 (88.2%) and the mean percentage of PD-L1 positivity was 19.7% (range, 5-50%). Fourteen patients (82.35%) developed cytokine release syndrome (grade ≤ 2) and four (23.53%) developed neurotoxicity (grade = 1), which was reversible in all. The overall response rate was 58.8% (10/17) and complete remission rate was 41.2% (7/17). At a median follow-up 5 months, median overall survival for all patients was not reached. Conclusions: This trial showed the feasibility, controllable toxicities, and effective activity of CD19-PD-1/CD28-CART cells for treating patients with R/R B-cell lymphoma, providing the first proof-of-principle of the potential therapeutic value of targeting the PD-1/PD-L1 pathway in lymphoma in the clinical setting. Clinical trial information: NCT03258047.

BMJ Open ◽  
2020 ◽  
Vol 10 (2) ◽  
pp. e034629 ◽  
Author(s):  
Philip George ◽  
Nathaniel Dasyam ◽  
Giulia Giunti ◽  
Brigitta Mester ◽  
Evelyn Bauer ◽  
...  

IntroductionAutologous T-cells transduced to express a chimeric antigen receptor (CAR) directed against CD19 elicit high response rates in relapsed or refractory (r/r) B-cell non-Hodgkin lymphoma (B-NHL). However, r/r B-NHL remissions are durable in fewer than half of recipients of second-generation CAR T-cells. Third-generation (3G) CARs employ two costimulatory domains, resulting in improved CAR T-cell efficacy in vitro and in animal models in vivo. This investigator-initiated, phase I dose escalation trial, termed ENABLE, will investigate the safety and preliminary efficacy of WZTL-002, comprising autologous T-cells expressing a 3G anti-CD19 CAR incorporating the intracellular signalling domains of CD28 and Toll-like receptor 2 (TLR2) for the treatment of r/r B-NHL.Methods and analysisEligible participants will be adults with r/r B-NHL including diffuse large B-cell lymphoma and its variants, follicular lymphoma, transformed follicular lymphoma and mantle cell lymphoma. Participants must have satisfactory organ function, and lack other curative options. Autologous T-cells will be obtained by leukapheresis. Following WZTL-002 manufacture and product release, participants will receive lymphodepleting chemotherapy comprising intravenous fludarabine and cyclophosphamide. A single dose of WZTL-002 will be administered intravenously 2 days later. Targeted assessments for cytokine release syndrome and immune cell effector-associated neurotoxicity syndrome, graded by the American Society Transplantation and Cellular Therapy criteria, will be made. A modified 3+3 dose escalation scheme is planned starting at 5×104 CAR T-cells/kg with a maximum dose of 1×106 CAR T-cells/kg. The primary outcome of this trial is safety of WZTL-002. Secondary outcomes include feasibility of WZTL-002 manufacture and preliminary measures of efficacy.Ethics and disseminationEthical approval for the study was granted by the New Zealand Health and Disability Ethics Committee (reference 19/STH/69) on 23 June 2019 for Protocol V.1.2. Trial results will be reported in a peer-reviewed journal, and results presented at scientific conferences or meetings.Trial registration numberNCT04049513


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2800-2800
Author(s):  
Michael Olson ◽  
Tim Luetkens ◽  
Fiorella Iglesias ◽  
Sabarinath Radhakrishnan ◽  
Jennie Y. Law ◽  
...  

Abstract B cell lymphoma is the most common hematologic malignancy in the United States. Although treatment options have greatly improved in the past several decades, outcomes for patients with relapsed B cell lymphoma remain poor. Chimeric antigen receptor (CAR) T cells have recently entered the clinic with promise to address the gap in effective therapies for patients relapsed B cell lymphoma. However, antigen loss and poor CAR T cell persistence has been shown to drive resistance to the widely approved CD19-targeted CAR in some patients, demonstrating the need for additional therapies. Here, we demonstrate CD229-targeted CAR T cell therapy as a promising option for the treatment of relapsed B cell lymphoma, addressing an important group of patients with typically poor outcomes. CD229 is an immune-modulating receptor expressed on the surface of B cells that we recently found to be highly expressed in the plasma cell neoplasm multiple myeloma (Radhakrishnan et al. 2020). We utilized semi-quantitative PCR and flow cytometry to assess whether CD229 is also expressed on malignant B cells earlier in development as found in B cell lymphoma. Expression analysis revealed the presence of CD229 in a panel of 11 B cell lymphoma cell lines and 45 primary B cell lymphoma samples comprising several subsets of disease including aggressive B cell lymphomas such as diffuse large B cell lymphoma (DLBCL), mantle cell lymphoma (MCL) and Burkitt lymphoma as well as indolent subtypes of B cell lymphoma including chronic lymphoblastic leukemia (CLL) and follicular lymphoma. Of note, CD229 was found to be overexpressed on primary B cell lymphoma cells when compared to autologous normal B cells. Given the high levels of CD229 expression throughout all B cell lymphoma subtypes analyzed, we generated CD229 CAR T cells in order to determine whether CAR T cell therapy is an effective way to target CD229 expressing B cell lymphoma cells. CD229 CAR T cells exhibited robust cytotoxicity when cocultured with B cell lymphoma cell lines and primary samples characterized by significant production of TH1 cytokines IL-2, TNF and IFNγ and rapid loss of B cell lymphoma cell viability when compared to control CAR T cells lacking an antigen binding scFv domain (∆scFv CAR T cells). In vivo analysis revealed effective tumor control in NSG mice carrying B cell lymphoma cell lines JeKo-1 (MCL) and DB (DLBCL) when treated with CD229 CAR T cells versus ∆scFv CAR T cells. Finally, we sought to determine the efficacy of CD229 CAR T cells in the context of CD19 CAR T cell therapy relapse. Here, a 71-year-old patient with CLL had an initial response when treated with CD19 CAR T cells but quickly relapsed only 2 months after treatment. Malignant cells from the CLL patient retained CD229 expression as identified by flow cytometry and an ex vivo coculture with CD229 CAR T cells revealed robust killing of CLL cells by CD229 CAR T cells. Transfer of antigen from target cell to CAR T cell by trogocytosis was recently suggested to drive relapse following CAR T cell therapy by decreasing antigen on tumor cells and promoting CAR T cell fratricide (Hamieh et al. 2019). We cocultured CD19 and CD229 CAR T cells with primary CLL cells and assessed CD19 and CD229 expression as well as CAR T cell viability by flow cytometry. In contrast with CD19 CAR T cells, CD229 CARs did not strip their target antigen from the surface of CLL cells. The transfer of CD19 from CLL cells to CD19 CAR T cells resulted in poor CAR T cell viability while CD229 CAR T cell viability remained high following coculture. In summary, we demonstrate that CD229 is a promising therapeutic target in B cell lymphoma due to its high levels of expression throughout many subtypes of disease. CD229 CAR T cells effectively kill B cell lymphoma cells in vitro and control growth of aggressive B cell lymphomas in vivo. Finally, CD229 CAR T cells are effective against primary CLL cells from patients that have relapsed from CD19 CAR T cell therapy and do no exhibit antigen loss by trogocytosis. Taken together, these data suggest that CD229 CAR T cell therapy may be a promising option to address the poor outcomes for patients with relapsed B cell lymphoma. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 34-34
Author(s):  
Yongxian Hu ◽  
Yanlei Zhang ◽  
Houli Zhao ◽  
Yiyun Wang ◽  
Arnon Nagler ◽  
...  

Introduction Chimeric antigen receptor (CAR)-T-cell therapy has revolutionized the treatment of relapsed/refractory (R/R) B-cell hematological malignancies, primarily acute lymphoblastic leukemia (ALL), and B-cell non-Hodgkin lymphoma (NHL). CD19 CAR-T cells have been extensively studied and have been shown to yield complete remission (CR) rates of about 90% in R/R ALL, but substantially lower (50%) rates in R/R NHL. Moreover, persistence is usually limited, and antigen escape-mediated relapse is a major limitation. Dual CAR-T cells targeting both CD19 and CD22 may address these limitations. Patients and methods We developed a bispecific CAR-T cells that could concomitantly recognize CD19- and CD22-expressing targets by incorporating both CD19 and CD22 single-chain variables in a single CAR construct (Figure 1A). We designed a prospective study to assess the safety and efficacy profiles of the dual CAR-T therapy in patients with R/R aggressive B-cell lymphoma. Results The preclinical cytotoxicity evaluation of the CD19/CD22 dual-targeted CAR-T cells was performed in comparison with mono-specific CD19-BB-002 and CD22-BB-002 CAR-T cells in HeLa cells that were engineered to express CD19, CD22, or both antigens. The dual-antigen specific CAR-T cells performed equally well when compared with the mono-specific CAR-T cells when there was only a single antigen present on the target cells; better performance was observed when both antigens were present on target cells (Figure 1B). In addition, the dual-antigen specific CAR-T cells induced equal amounts of interleukin (IL)-3, granulocyte-macrophage colony-stimulating factor (GM-CSF), and interferon (IFN)-γ, when compared with the two mono-specific CAR-T cells (Figure 1C). Furthermore, the CD19 CAR-T cells induced more IL-2 and tumor necrosis factor (TNF)-α than the CD22 CAR-T cells and dual-antigen CAR-T cells. However, in the presence of both CD19 and CD22 antigens, the dual-specific CAR-T cell tended to produce more granzyme B, which may explain the higher degree of cytotoxicity when compared with the two mono-specific CAR-T cells (Figure 1D). Twenty-four patients were screened. Of the 16 eligible patients 14 (87.5%) achieved objective response (RR), with 10 (62.5%) achieving complete response (CR). The 2-year overall survival (OS) and progression-free survival (PFS) rates were 77.3% and 40.2%, respectively (Figure 2A). Achieving CR (HR: 0.017, 95% CI: 0.000-0.935; P=0.046) and number of prior lines of chemotherapy (n=2) (HR:135.784, 95% CI: 1.069-17248.110, P=0.047) were found as independent prognostic factors associated with favorable PFS. The 2-year OS and PFS of the CR patients were higher than those of the non-CR patients (100% versus 41.7%, P=0.015; 66.7% versus 0%, P < 0.001), respectively (Figure 2B). The 2-year PFS in patients received 2 prior lines of chemotherapy was higher as compared to those that received more than 2 lines of chemotherapy (68.6% versus 16.7%, P=0.049) whereas the OS in the 2 groups did not differ significantly (83.3% and 71.1%, P=0.613) (Figure 2C). Severe grade 3 cytokine release syndrome (CRS) was observed in only one patient, while 4 had grade one and 11 had grade 2, respectively. No patient developed neurotoxicity. Conclusions Immunotherapy with a novel CD19/CD22 dual targeted CAR-T cells yields a potent and durable anti-lymphoma response with no neurotoxicity or severe CRS. Bispecific CD19/CD22 CAR-T cells represent a safe and potent anti-lymphoma cellular based targeted immunotherapy. Figure 1 Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2054-2054 ◽  
Author(s):  
Yang Cao ◽  
Na Wang ◽  
Gaoxiang Wang ◽  
Yi Xiao ◽  
Liang Huang ◽  
...  

Abstract Salvage chemotherapy followed by autologous hematopoietic stem cell transplantation (HSCT) is the standard treatment for the patients with chemotherapy sensitive relapsed or refractory B-NHL. However, more than half of patients will ultimately relapse. To improve long-term remission rate, we administered adoptive T-cell immunotherapy after HSCT. Based on the findings that CD19/CD22 dual antigen targeting have been demonstrated to be a promising approach for overcoming antigen escape relapse, we conducted an open-label, single-center and single-arm pilot study of sequential infusion of anti-CD22 and anti-CD19 CAR-T cells following autologous HSCT. We aimed to evaluate its safety and efficacy in patients with relapsed, refractory and high-risk B cell lymphoma. This trial is registered with ChiCTR, number ChiCTR-OPN-16009847. Between December 2016 and March 2018, 20 patients were enrolled in this clinical trial, with a median age of 33 years (range, 24-61 years). Of these 20 treated patients, 2 had PMBCL, 4 had tFL,14 had DLBCL, 70.0% (14/20) had received at least three previous therapies. 50.0% patients (8/16) harbor a TP53 mutation. At baseline assessment (prior to conditioning), 3 participants had a CR, 8 had a PR, 8 had a PD, 1 had a SD. All patients received bis-chloroethyl nitrosourea, etoposide, Ara-C, and melphalan, CD22 and CD19 CAR T cells were infused 2 to 7 days after stem cell infusion. The median anti-CD22 and anti-CD19 CAR-T cells dose per kilogram of body weight were 4.0× 106 (range, 1.0-10.0×106) and 4.2× 106 (range, 1.8-10.0×106) respectively. Normal HSCT-associated neutrophil engraftment within 20 days in all 20 patients was observed. 11/20 (50.5%) patients experienced cytokine release syndrome and no patient had severe CRS (grade 3-4). Neurologic events occurred in 2 (10.0%) patients. At 3 months, there was a 90.0% remission rate (95% CI ,68.3-98.8) with 85.0 % complete remissions (CRs) and 5.0% partial remissions (PRs) accessed by PET-CT. The duration of responses currently ranges from 3.7 months to19.1 months. All patients (17/20) who achieved CRs are ongoing, and the current median duration of all CRs is 13.2 months. Our results indicated that patients with relapsed, refractory and high-risk B cell lymphoma who received sequential infusion of anti-CD22 and anti-CD19 CAR-T cell therapy following HSCT had high rates of durable remission, with safety profile. Anti-CD22 and Anti-CD19 CAR-T cell therapy following HSCT may become a useful treatment approach for relapsed , refractory B cell lymphoma and high-risk B cell lymphoma. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Vol 22 (16) ◽  
pp. 8706
Author(s):  
Kyung-Eun Noh ◽  
Jun-Ho Lee ◽  
So-Yeon Choi ◽  
Nam-Chul Jung ◽  
Ji-Hee Nam ◽  
...  

Chimeric antigen receptor (CAR)-T cells are effective in the treatment of hematologic malignancies but have shown limited efficacy against solid tumors. Here, we demonstrated an approach to inhibit recurrence of B cell lymphoma by co-expressing both a human anti-CD19-specific single-chain variable fragment (scFv) CAR (CD19 CAR) and a TGF-β/IL-7 chimeric switch receptor (tTRII-I7R) in T cells (CD19 CAR-tTRII-I7R-T cells). The tTRII-I7R was designed to convert immunosuppressive TGF-β signaling into immune-activating IL-7 signaling. The effect of TGF-β on CD19 CAR-tTRII-I7R-T cells was assessed by western blotting. Target-specific killing by CD19 CAR-tTRII-I7R-T cells was evaluated by Eu-TDA assay. Daudi tumor-bearing NSG (NOD/SCID/IL2Rγ-/-) mice were treated with CD19 CAR-tTRII-I7R-T cells to analyze the in vivo anti-tumor effect. In vitro, CD19 CAR-tTRII-I7R-T cells had a lower level of phosphorylated SMAD2 and a higher level of target-specific cytotoxicity than controls in the presence of rhTGF-β1. In the animal model, the overall survival and recurrence-free survival of mice that received CD19 CAR-tTRII-I7R-T cells were significantly longer than in control mice. These findings strongly suggest that CD19 CAR-tTRII-I7R-T cell therapy provides a new strategy for long-lasting, TGF-β-resistant anti-tumor effects against B cell lymphoma, which may lead ultimately to increased clinical efficacy.


2020 ◽  
Vol 55 (12) ◽  
pp. 2347-2349
Author(s):  
Eugenio Galli ◽  
Vincent Allain ◽  
Roberta Di Blasi ◽  
Sophie Bernard ◽  
Laetitia Vercellino ◽  
...  

Cancers ◽  
2020 ◽  
Vol 12 (9) ◽  
pp. 2612
Author(s):  
Lei Tao ◽  
Muhammad Asad Farooq ◽  
Yaoxin Gao ◽  
Li Zhang ◽  
Congyi Niu ◽  
...  

B cell aplasia caused by “on-target off-tumor” toxicity is one of the clinical side effects during CD19-targeted chimeric antigen receptor (CAR) T (CD19-CAR-T) cells treatment for B cell malignancies. Persistent B cell aplasia was observed in all patients with sustained remission, which increased the patients’ risk of infection. Some patients even died due to infection. To overcome this challenge, the concept of incorporating an inhibitory CAR (iCAR) into CAR-T cells was introduced to constrain the T cells response once an “on-target off-tumor” event occurred. In this study, we engineered a novel KIR/PD-1-based inhibitory CAR (iKP CAR) by fusing the extracellular domain of killer cell immunoglobulin-like receptors (KIR) 2DL2 (KIR2DL2) and the intracellular domain of PD-1. We also confirmed that iKP CAR could inhibit the CD19 CAR activation signal via the PD-1 domain and CD19-CAR-T cells bearing an iKP CAR (iKP-19-CAR-T) exerted robust cytotoxicity in vitro and antitumor activity in the xenograft model of CD19+HLA-C1− Burkitt’s lymphoma parallel to CD19-CAR-T cells, whilst sparing CD19+HLA-C1+ healthy human B cells both in vitro and in the xenograft model. Meanwhile, iKP-19-CAR-T cells exhibited more naïve, less exhausted phenotypes and preserved a higher proportion of central memory T cells (TCM). Our data demonstrates that the KIR/PD-1-based inhibitory CAR can be a promising strategy for preventing B cell aplasia induced by CD19-CAR-T cell therapy.


Sign in / Sign up

Export Citation Format

Share Document