scholarly journals Engineering Chimeric Antigen Receptors

Acta Naturae ◽  
2017 ◽  
Vol 9 (1) ◽  
pp. 6-14 ◽  
Author(s):  
S. V. Kulemzin ◽  
V. V. Kuznetsova ◽  
M. Mamonkin ◽  
A. V. Taranin ◽  
А. A. Gorchakov

Chimeric antigen receptors (CARs) are recombinant protein molecules that redirect cytotoxic lymphocytes toward malignant and other target cells. The high feasibility of manufacturing CAR-modified lymphocytes for the therapy of cancer has spurred the development and optimization of new CAR T cells directed against a broad range of target antigens. In this review, we describe the main structural and functional elements constituting a CAR, discuss the roles of these elements in modulating the anti-tumor activity of CAR T cells, and highlight alternative approaches to CAR engineering.

Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 951-951 ◽  
Author(s):  
Michael Hudecek ◽  
Anne Silva ◽  
Paula L. Kosasih ◽  
Yvonne Y. Chen ◽  
Cameron J. Turtle ◽  
...  

Abstract Abstract 951 Adoptive immunotherapy with T cells engineered by gene transfer to express CD19-specific chimeric antigen receptors (CARs) has the potential to induce remissions in patients with advanced B cell malignancies. CARs are synthetic receptors with an extracellular antigen-binding domain (scFv), a spacer domain that provides separation of the scFv from the cell membrane and an intracellular signaling module, most commonly the CD3ζ chain and one or more costimulatory domains such as CD28 or 4-1BB. Several clinical trials with CD19-CAR T cells in small cohorts of patients with B cell tumors have been reported with variable results. Although most studies have used the CD19-specific FMC63 scFv as the tumor-targeting moiety, the extracellular, transmembrane and intracellular CAR domains used in each trial have been distinct, and an emerging paradigm is that including costimulation in the design of the CAR is key to achieving anti-tumor activity in vivo. In this study, we analyzed the influence of extracellular spacer domain length on the in vitro and in vivo function of CD19-CARs. We constructed a panel of four CD19-CARs comprised of the FMC63 scFv and either a long spacer derived from the IgG4-Fc Hinge-CH2-CH3 domain (229 AA) or a short Hinge domain only spacer (12 AA). Each CAR contained a signaling module of CD3ζ with CD28 (short/CD28; long/CD28) or 4-1BB (short/4-1BB; long/4-1BB). We transduced CD8+ CD45RO+ CD62L+central memory T cells of normal donors with each of the CARs, enriched transduced T cells to >90% purity by immunomagnetic selection using a tEGFR marker encoded in the CAR vector, and expanded CAR transduced T cells using a uniform culture protocol. We compared the in vitro function of T cell lines expressing each of the CD19-CARs and confirmed specific cytolytic activity against CD19+ target cells including K562/CD19, and Raji and JeKo-1 lymphoma cells. Quantitative cytokine analyses showed higher levels of IFN-γ, TNF-α, IL-2 production in T cells expressing CD19-CARs with CD28 costimulatory domain compared to the corresponding constructs with 4-1BB, consistent with prior work. T cells expressing each of the CD19-CARs proliferated in vitro after stimulation with K562/CD19 and Raji tumor cells by CFSE dye dilution, with the strongest proliferation observed in T cells expressing the CD19-CAR ‘long/CD28’, consistent with the highest levels of IL-2 production by T cells expressing this construct. We then analyzed the in vivo anti-tumor efficacy of each CD19-CAR in immunodeficient NOD/SCID/g−/− (NSG) mice engrafted with firefly luciferase transduced Raji cells. Tumor was inoculated on day 0, and once tumor was established (day 7), the mice received a single dose of 2.5×106̂ T cells expressing each CD19-CAR, a tEGFR control vector, or were left untreated. Surprisingly, only T cells expressing CD19-CARs with a short spacer domain (short/CD28 and short/4-1BB) eradicated the Raji tumors and led to long-term tumor-free survival of all mice. T cells expressing CD19-CARs with a long spacer domain (long/CD28 and long/4-1BB) did not confer a significant anti-tumor effect and all mice expired from systemic lymphoma at a similar time as control and untreated mice. The anti-tumor efficacy in vivo of T cells modified with long spacer CD19-CARs could not be improved by increasing CAR T cell dose 4 fold, or by including additional costimulatory domains into the CD19-CAR (long/CD28:4-1BB). Serial analyses in peripheral blood, bone marrow and spleen showed dramatically lower numbers of transferred T cells in mice treated with long spacer CD19-CARs compared to mice treated with short spacer CD19-CARs or control T cells. Further analysis revealed that despite strong activation in vivo as assessed by upregulation of CD69 and CD25, CD19-CARs with long extracellular spacer domain induced a high rate of activation induced T cell death in vivo. Collectively, these results demonstrate that the extracellular spacer domain that lacks intrinsic signaling function is critical in the design of effective CD19-CARs, and illustrates that tailoring spacer length is likely to be essential for designing effective CARs specific for other tumor antigens. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Mighmig Simonian Gharghani ◽  
Miganoosh Simonian ◽  
Faezeh Bakhtiari ◽  
Mozhan Haji Ghaffari ◽  
Ghazaleh Fazli ◽  
...  

One of the main reasons that researchers pay enormous attention to immunotherapy is that, despite significant advances in conventional therapy approaches, breast cancer remains the leading cause of death from malignant tumors among women. Genetically modifying T cells with chimeric antigen receptors (CAR) is one of the novel methods that has exhibited encouraging activity with relative safety, further urging investigators to develop several CAR T cells to target overexpressed antigens in breast tumors. This article is aimed not only to present such CAR T cells and discuss their remarkable results but also indicates their shortcomings with the hope of achieving possible strategies for improving therapeutic response.


2021 ◽  
Author(s):  
Erica R. Vander Mause ◽  
Jillian M. Baker ◽  
Sabarinath V. Radhakrishnan ◽  
Patricia Davis ◽  
Jens Panse ◽  
...  

ABSTRACTT cells expressing chimeric antigen receptors have shown remarkable therapeutic activity against different types of cancer. However, their wider use has been hampered by the potential for life-threatening toxicities due to the unintended targeting of healthy cells expressing low levels of the targeted antigen. We have now developed an affinity-tuning approach for the generation of minimally modified, low-affinity antibody variants derived from existing high-affinity antibodies. Using this approach, we engineered low affinity variants of the fully human CD229-specific antibody 2D3. Parental 2D3 originally efficiently targeted multiple myeloma cells but also healthy T cells expressing low levels of CD229. We demonstrate that CAR T cells based on a low affinity variant of 2D3 maintain the parental antibody’s anti-tumor activity, but lack its targeting of healthy T cells. In addition, variant CD229 CAR T cells show reduced trogocytosis potentially augmenting CAR T cell persistence. The fast off-rate CAR produced using our affinity tuning approach eliminates a key liability of CD229 CAR T cells and paves the way for the effective and safe treatment of patients with multiple myeloma.One sentence summaryAffinity tuning approach yields low affinity CD229 CAR binding domain maintaining the parental clone’s anti-tumor activity while eliminating killing of healthy T cells, increasing CAR T cell expansion, and decreasing trogocytosis.


Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 3262-3262
Author(s):  
Daniel W. Lee ◽  
James N Kochenderfer ◽  
Christoph Rader ◽  
Rimas J Orentas ◽  
Crystal L Mackall

Abstract Abstract 3262 Relapsed acute lymphoblastic leukemia (ALL) remains a difficult challenge for both pediatric and adult patients. Chimeric antigen receptors (CARs) are genetically engineered molecules expressed in transduced T lymphocytes. CARs express both a target binding motif and TCRzeta signals needed for T cell activation, with or without other costimulatory domains. CARs that target CD19 (expressed on most ALL blasts) are being studied in several clinical trials in adults and are planned for pediatric relapsed/refractory ALL. To date, CARs have been expanded ex vivo using antigen-independent techniques. This project sought to develop antigen specific expansion of cells transduced with a CD19-specific CAR by employing artificial APCs (aAPCs) and to explore whether the method of expansion impacted functionality of CD19-CAR T cells. aAPCs used in these studies express the high affinity Fc receptor (CD64) and the costimulatory molecule CD137L (aAPC-41BBL). We created an Fc-CD19 fusion protein that when loaded onto the aAPCs, engages the chimeric antigen receptor and induces antigen specific activation. Antigen-specific vs. non-specific approaches to activation and expansion of CAR expressing T cells were compared using three different expansion protocols (EPs). CD19-CAR transduced T cells were expanded with A) irradiated aAPC-41BBL loaded with anti-OKT3, B) irradiated aAPC-41BBL loaded with rFc-CD19 or C) irradiated allogeneic PBMC feeder cells with anti-OKT3. Briefly, OKT3 and IL-2 activated T cells were transduced with CD19CAR, control CAR (Her2-specific), or non-transduced (Mock). All cultures were maintained in IL-2. Functionality was assessed in a 4-hour 51Cr release assay against 4 distinct CD19+ ALL cell lines, K562 cells, and K562 stably transfected with CD19 (K562-CD19) or NGFR (K562-NGFR). Results demonstrate similar significant levels of CD19-specific cytotoxicity in the 4h assay at E:T ratios as low as 2.5:1, regardless of the expansion protocol used (20-40% lysis of all CD19+ targets when expanded by OKT3/aAPC, 40–60% lysis with rFc-CD19/aAPC, and 35–45% lysis with allogeneic feeders and OKT3). To evaluate cytotoxicity in long term culture, CD19-CAR T cells expanded using the three protocols were co-cultured with ALL cell targets for 4 days, then flow cytometry was performed to enumerate surviving ALL cells as determined by CD22+ staining. NALM6 and K562-CD19 cells were entirely eliminated even at an E:T ratio of 2.5:1 by CD19-CAR T cells, regardless of expansion protocol, and not by any of the 3 Mock EPs. Therefore, these results demonstrate that the cytolytic potential of CAR transduced T cells is similar, regardless of whether expansion occurs via CD3 signaling or via the chimeric receptor itself. Interestingly, we did observe substantial NK mediated killing in these assays, which correlated with CD56+ cell content and was eliminated by cold target inhibition using K562 cells. Studies are underway to determine whether differences in NK killing varies with expansion protocol. In summary, Fc-bearing artificial antigen presenting cells combined with CAR specific Fc fusion proteins provide a potential off-the-shelf reagent for antigen specific expansion of T cells with chimeric antigen receptors. This could overcome variable transduction efficiencies and allow administration of a more homogenous population of CAR specific T cells. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 23-23 ◽  
Author(s):  
Surabhi Srinivasan ◽  
Nguyen Tan ◽  
Hsin-Yuan Cheng ◽  
Yi Zhang ◽  
Silvia Tacheva-Grigorova ◽  
...  

CD70, a member of the TNF superfamily, is a type II transmembrane glycoprotein that interacts with its receptor (CD27) to promote survival of primed T cells and leads to formation of effector and memory T cells. Expression of CD70 in normal tissues is restricted to activated T and B lymphocytes and mature dendritic cells. CD70 is also widely expressed in various malignancies, including renal cell carcinoma (RCC) and acute myeloid leukemia (AML). The restricted expression pattern of CD70 in normal tissues makes it an attractive target for cancer therapeutics. Adoptive transfer of T cells expressing chimeric antigen receptors (CARs) is an exciting new therapeutic modality showing great promise in hematologic malignancies. Approval of two CD19-targeting autologous CAR Ts, Kymriah® and Yescarta®, has been followed with promising results from BCMA autologous CAR T clinical trials, showing that activity can extend to other targets. We have previously described the functional screening of a library of anti-CD70 scFv-based CARs and the identification of lead CD70 allogeneic CAR T cells (AlloCAR TTM) with robust activity against RCC cell lines both in vitro and in vivo. Here, we evaluate the anti-tumor activity and safety of a lead CD70 AlloCAR T (ALLO-316) for the treatment of AML. CD70 expression was evaluated and detected on three AML cell lines and in six primary AML patient samples, with 5/6 patient samples showing expression on 24%-99% of cells. CD70 expression will be profiled in a broader subset of AML patients and preliminary data will be presented. Despite the expression of CD70 on activated T cells it was possible to generate CD70 AlloCAR T cells. No CD70 expression was observed on CAR T cells after generation, suggesting either cells are succumbing to fratricide or are being "masked" by the CAR. CD70 was also not detected on Jurkat cells expressing CARs and this data, in combination with results showing CAR expression is protective when overexpressed in RCC cells support the phenomenon of "masking". Cellectis' TALEN® gene-editing was used to inactivate the TRAC and CD52 loci with the intent to minimize the risk of graft-versus-host disease and to confer resistance to ALLO-647, an anti-CD52 antibody that can be used as part of the conditioning regimen to deplete host alloreactive immune cells potentially leading to increased persistence and efficacy of the infused allogeneic cells. ALLO-316 cells were highly effective at lysing CD70-expressing target cells and eliminated greater than 99% of cells at the high effector to target (E:T) ratio and were unable to lyse AML cells in which CD70 was knocked out. Moreover, ALLO-316 cells were able to kill primary AML blasts with CD70 expression ex vivo. An orthotopic in vivo model utilizing the AML cell line MV4-11 was developed and anti-tumor activity was observed. In addition to evaluating efficacy against AML cell lines and tumors we also explored potential toxicity liabilities related to ALLO-316 treatment. Previous studies have reported that certain AML tumor antigens can also be expressed on normal hematopoietic progenitors and such expression could potentially lead to toxicity with targeted therapeutics. No detectable CD70 was observed by flow cytometry on purified CD34+ cells from 14 healthy donors. Taken together, our results support clinical development of CD70 AlloCAR T therapy for the treatment of AML. Disclosures Srinivasan: Allogene Therapeutics: Current Employment. Tan:Allogene Therapeutics: Current Employment. Cheng:Allogene Therapeutics, Inc.: Current Employment, Current equity holder in publicly-traded company. Zhang:Allogene Therapeutics: Current Employment, Current equity holder in publicly-traded company. Tacheva-Grigorova:Allogene Therapeutics: Current Employment, Current equity holder in publicly-traded company. Van Blarcom:Allogene Therapeutics, Inc.: Current Employment, Current equity holder in publicly-traded company. Sommer:Allogene Therapeutics, Inc.: Current Employment, Current equity holder in publicly-traded company. Nguyen:Allogene Therapeutics, Inc.: Current Employment, Current equity holder in publicly-traded company. Sasu:Allogene Therapeutics, Inc.: Current Employment, Current equity holder in publicly-traded company. Panowski:Allogene Therapeutics, Inc.: Current Employment, Current equity holder in publicly-traded company.


2020 ◽  
Vol 21 (22) ◽  
pp. 8620
Author(s):  
Alain E. Andrea ◽  
Andrada Chiron ◽  
Stéphanie Bessoles ◽  
Salima Hacein-Bey-Abina

Immunoadoptive therapy with genetically modified T lymphocytes expressing chimeric antigen receptors (CARs) has revolutionized the treatment of patients with hematologic cancers. Although clinical outcomes in B-cell malignancies are impressive, researchers are seeking to enhance the activity, persistence, and also safety of CAR-T cell therapy—notably with a view to mitigating potentially serious or even life-threatening adverse events like on-target/off-tumor toxicity and (in particular) cytokine release syndrome. A variety of safety strategies have been developed by replacing or adding various components (such as OFF- and ON-switch CARs) or by combining multi-antigen-targeting OR-, AND- and NOT-gate CAR-T cells. This research has laid the foundations for a whole new generation of therapeutic CAR-T cells. Here, we review the most promising CAR-T cell safety strategies and the corresponding preclinical and clinical studies.


2017 ◽  
Vol 19 (suppl_4) ◽  
pp. iv29-iv29
Author(s):  
Daniel Landi ◽  
Kristen Fousek ◽  
Malini Mukherjee ◽  
Ankita Shree ◽  
Heba Samaha ◽  
...  

Blood ◽  
2013 ◽  
Vol 122 (18) ◽  
pp. 3138-3148 ◽  
Author(s):  
Armen Mardiros ◽  
Cedric Dos Santos ◽  
Tinisha McDonald ◽  
Christine E. Brown ◽  
Xiuli Wang ◽  
...  

Key Points CD123 CAR T cells specifically target CD123+ AML cells. AML patient-derived T cells can be genetically modified to lyse autologous tumor cells.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3921-3921 ◽  
Author(s):  
Cesar Sommer ◽  
Hsin-Yuan Cheng ◽  
Yik Andy Yeung ◽  
Duy Nguyen ◽  
Janette Sutton ◽  
...  

Autologous chimeric antigen receptor (CAR) T cells have achieved unprecedented clinical responses in patients with B-cell leukemias, lymphomas and multiple myeloma, raising interest in using CAR T cell therapies in AML. These therapies are produced using a patient's own T cells, an approach that has inherent challenges, including requiring significant time for production, complex supply chain logistics, separate GMP manufacturing for each patient, and variability in performance of patient-derived cells. Given the rapid pace of disease progression combined with limitations associated with the autologous approach and treatment-induced lymphopenia, many patients with AML may not receive treatment. Allogeneic CAR T (AlloCAR T) cell therapies, which utilize cells from healthy donors, may provide greater convenience with readily available off-the-shelf CAR T cells on-demand, reliable product consistency, and accessibility at greater scale for more patients. To create an allogeneic product, the TRAC and CD52 genes are inactivated in CAR T cells using Transcription Activator-Like Effector Nuclease (TALEN®) technology. These genetic modifications are intended to minimize the risk of graft-versus-host disease and to confer resistance to ALLO-647, an anti-CD52 antibody that can be used as part of the conditioning regimen to deplete host alloreactive immune cells potentially leading to increased persistence and efficacy of the infused allogeneic cells. We have previously described the functional screening of a library of anti-FLT3 single-chain variable fragments (scFvs) and the identification of a lead FLT3 CAR with optimal activity against AML cells and featuring an off-switch activated by rituximab. Here we characterize ALLO-819, an allogeneic FLT3 CAR T cell product, for its antitumor efficacy and expansion in orthotopic models of human AML, cytotoxicity in the presence of soluble FLT3 (sFLT3), performance compared with previously described anti-FLT3 CARs and potential for off-target binding of the scFv to normal human tissues. To produce ALLO-819, T cells derived from healthy donors were activated and transduced with a lentiviral construct for expression of the lead anti-FLT3 CAR followed by efficient knockout of TRAC and CD52. ALLO-819 manufactured from multiple donors was insensitive to ALLO-647 (100 µg/mL) in in vitro assays, suggesting that it would avoid elimination by the lymphodepletion regimen. In orthotopic models of AML (MV4-11 and EOL-1), ALLO-819 exhibited dose-dependent expansion and cytotoxic activity, with peak CAR T cell levels corresponding to maximal antitumor efficacy. Intriguingly, ALLO-819 showed earlier and more robust peak expansion in mice engrafted with MV4-11 target cells, which express lower levels of the antigen relative to EOL-1 cells (n=2 donors). To further assess the potency of ALLO-819, multiple anti-FLT3 scFvs that had been described in previous reports were cloned into lentiviral constructs that were used to generate CAR T cells following the standard protocol. In these comparative studies, the ALLO-819 CAR displayed high transduction efficiency and superior performance across different donors. Furthermore, the effector function of ALLO-819 was equivalent to that observed in FLT3 CAR T cells with normal expression of TCR and CD52, indicating no effects of TALEN® treatment on CAR T cell activity. Plasma levels of sFLT3 are frequently increased in patients with AML and correlate with tumor burden, raising the possibility that sFLT3 may act as a decoy for FLT3 CAR T cells. To rule out an inhibitory effect of sFLT3 on ALLO-819, effector and target cells were cultured overnight in the presence of increasing concentrations of recombinant sFLT3. We found that ALLO-819 retained its killing properties even in the presence of supraphysiological concentrations of sFLT3 (1 µg/mL). To investigate the potential for off-target binding of the ALLO-819 CAR to human tissues, tissue cross-reactivity studies were conducted using a recombinant protein consisting of the extracellular domain of the CAR fused to human IgG Fc. Consistent with the limited expression pattern of FLT3 and indicative of the high specificity of the lead scFv, no appreciable membrane staining was detected in any of the 36 normal tissues tested (n=3 donors). Taken together, our results support clinical development of ALLO-819 as a novel and effective CAR T cell therapy for the treatment of AML. Disclosures Sommer: Allogene Therapeutics, Inc.: Employment, Equity Ownership. Cheng:Allogene Therapeutics, Inc.: Employment, Equity Ownership. Yeung:Pfizer Inc.: Employment, Equity Ownership. Nguyen:Allogene Therapeutics, Inc.: Employment, Equity Ownership. Sutton:Allogene Therapeutics, Inc.: Employment, Equity Ownership. Melton:Allogene Therapeutics, Inc.: Employment, Equity Ownership. Valton:Cellectis, Inc.: Employment, Equity Ownership. Poulsen:Allogene Therapeutics, Inc.: Employment, Equity Ownership. Djuretic:Pfizer, Inc.: Employment, Equity Ownership. Van Blarcom:Allogene Therapeutics, Inc.: Employment, Equity Ownership. Chaparro-Riggers:Pfizer, Inc.: Employment, Equity Ownership. Sasu:Allogene Therapeutics, Inc.: Employment, Equity Ownership.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 3931-3931
Author(s):  
Martina Fontaine ◽  
Benjamin Demoulin ◽  
Simon Bornschein ◽  
Susanna Raitano ◽  
Steve Lenger ◽  
...  

Background The Natural Killer Group 2D (NKG2D) receptor is a NK cell activating receptor that binds to eight different ligands (NKG2DL) commonly over-expressed in cancer, including MICA and MICB. The product candidate CYAD-01 are chimeric antigen receptor (CAR) T-cells encoding the full length human NKG2D fused to the intracellular domain of CD3ζ. Data from preclinical models have shown that CYAD-01 cells specifically target solid and hematological tumors. Encouraging preliminary results from the Phase I clinical trial THINK, assessing CYAD-01 safety, showed initial signals of objective clinical responses in patients with r/r AML and MDS. The clinical development of CAR T-cells has been limited by several challenges including achieving sufficient numbers of cells for clinical application. We have previously shown that NKG2D ligands are transiently expressed on activated T cells and that robust cell yields are generated through the addition of a blocking antibody and a PI3K inhibitor during cell manufacture. Here, we investigated the ability of an optimized short hairpin RNA (shRNA) technology to modulate NKG2DL expression on CYAD-01 cells and to determine if there is an increase in the anti-tumor activity of NKG2D-based CAR T-cells (termed CYAD-02). Methods Molecular and cellular analyses identified MICA and MICB as the key NKG2DL expressed on activated T-cells and highly likely to participate in driving fratricide. In silico analysis and in vitro screening allowed the identification of a single shRNA targeting the conserved regions of MICA and MICB, thus downregulating both MICA and MICB expression. The selected shRNA was incorporated in the NKG2D-based CAR vector, creating the next-generation NKG2D-based CAR T-cell candidate, CYAD-02. In addition, truncated versions of the NKG2D receptor were generated to explore the mechanisms of action of NKG2D receptor activity in vivo. The in vivo persistence and anti-tumor activity of CYAD-02 cells was evaluated in an aggressive preclinical model of AML. Results Injection of CAR T-cells bearing truncated forms of the NKG2D-CAR in immunosuppressed mice resulted in similar persistence to the control T-cells. In contrast, CYAD-01 cells had reduced persistence, suggesting that the recognition of the NKG2DL by the NKG2D receptor could contribute to this effect. Analysis of cell phenotype upon CAR T-cell activation showed that MICA and MICB were transiently expressed on T-cells during manufacturing. These results collectively suggested that downregulating MICA and MICB expression in CYAD-01 cells could be a mean to increase CAR T-cell persistence in vivo. Candidate shRNA were screened for efficient targeting of both MICA and MICB at the mRNA and protein level. T-cells transduced with a single vector encoding for the NKG2D-based CAR and the selected shRNA targeting MICA and MICB (CYAD-02) demonstrated 3-fold increased expansion during in vitro culture in the absence of the blocking antibody used to increase cell yield during manufacture. When injected into immunosuppressed mice, CYAD-02 cells generated with the Optimab process showed 10-fold higher engraftment one week after injection and potent anti-tumor activity resulting in 2.6-fold increase of mouse survival in an aggressive AML model. Conclusions By using a single vector encoding the NKG2D-based CAR next to a shRNA targeting MICA and MICB and combined with improved cell culture methods, CYAD-02, the next-generation of NKG2D-based CAR T-cells, demonstrated enhanced in vivo persistence and anti-tumor activity. Following FDA acceptance of the IND application, a Phase 1 dose-escalation trial evaluating the safety and clinical activity of CYAD-02 for the treatment of r/r AML and MDS is scheduled to start in early 2020. Disclosures Fontaine: Celyad: Employment. Demoulin:Celyad: Employment. Bornschein:Celyad: Employment. Raitano:Celyad: Employment. Machado:Horizon Discovery: Employment. Moore:Avvinity Therapeutics: Employment, Other: Relationship at the time the work was performed; Horizon Discovery: Employment, Equity Ownership, Other: Relationship at the time the work was performed; Centauri Therapeutics: Consultancy, Other: Current relationship. Sotiropoulou:Celyad: Employment. Gilham:Celyad: Employment.


Sign in / Sign up

Export Citation Format

Share Document