Autologous Peripheral Blood T Lymphocytes Transduced with An Anti LewisY Chimeric Receptor Gene Persist In Patients with Lewisy Positive Acute Myeloid Leukaemia and Show Changes In Functional Polarization After Adoptive Transfer,

Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 4180-4180
Author(s):  
Paul Neeson ◽  
Amit S Khot ◽  
Stefan Peinert ◽  
Tsin Y Tai ◽  
Karen Chen ◽  
...  

Abstract Abstract 4180 High risk acute myeloid leukaemia (AML) and myelodysplastic syndrome (MDS) may be amenable to immunotherapy with adoptive transfer of chimeric antigen receptor (CAR) T cells. However, persistence and generation of central memory (CM) are essential for ongoing clinical responses (Morgan et al Science 2006). Our prior work showed that the carbohydrate antigen LewisY (LeY) is expressed in 46% of AML patients (n=33) to varying degrees, making it a suitable and novel target for CAR therapy. In our phase I study, we have assessed the trafficking, persistence and functional capacity of CAR-T cells transduced with an anti-LeY chimeric receptor gene in high-risk patients with LeY positive AML or MDS. METHOD: The CAR comprised extracellular humanized scFv recognizing the LeY Ag, linked to an extracellular CD8 hinge region, a transmembrane and cytoplasmic CD28 and CD3 zeta signalling domain. The humanized anti- LeY scFv-CD28-ζ receptor vector was produced as described previously (Westwood J et al PNAS 2005). T cells, harvested from the patient, were transduced with LeY scFv-CD28-ζ pSAMEN retroviral vector. Bulk transduced T cells (LeY-T) were re-infused into the patient after lymphodepleting fludarabine chemotherapy. AML immunophenotyping and cytogenetics were used to monitor minimal residual disease (MRD). PB and BM samples were collected prior to and post CAR-T adoptive transfer. An optimized PCR assay (sensitivity 1:1e5) for the presence of the LeY transgene (TG) was performed on genomic DNA extracted from each sample. To measure both CAR-T cell functional polarization and persistence of CAR expression, PB or BM cells were co-cultured with a cell line expressing LeY (OVCAR-3), in the presence or absence of MHC class I or II blocking antibodies. Culture supernatant was collected at 24 hours and assessed by Luminex assay for Th1 (IFN-g, IL-2), Th2 (IL-4, IL-10), pro-inflammatory (IL-6, IL-17 and TNF) cytokines and TGF-b. RESULTS: Five AML patients have been enrolled to date. Four patients have had sufficient CAR-T cells generated to provide doses of 0.5 × 109 −1.3 × 109T cells (transduced T cells: 8 – 30% of total T-cells]) with a viability of 96.1– 98.5%. 2 patients (01 and 04) had progressive disease 1 and 2 months after infusion. 2 patients (02 and 05) remain in morphologic remission with stable cytogenetic MRD, 3 and 14 months post infusion. Patients 01, 02 and 04 all had LeY TG in PB and BM samples at all time points measured, up to day 49 in patient 01 (Figure 1) and up to 10 months in patient 02. In patient 04 LeY TG was also detected in a skin biopsy (day +6) of leukemic skin infiltrate. Co-culture supernatant cytokine analysis showed LeY-T cells from patient 01, 02 and 04 secreted high levels of IFN-g (805.5 ± 198.9 pg/ml) and low levels of IL-2 (3.14 ± 0.65 pg/ml) prior to adoptive transfer. In contrast, post adoptive transfer, co-culture with OVCAR-3 cells, and MHC class I and II blocking antibody, induced these PB LeY-T cells to secrete IL-4 (24.13 ± 12.47pg/ml) and IL-10 (9.35 ± 0.44 pg/ml); no IFN-g or IL-2 was detected. This Th2 polarization of CAR-T cells was shown in PB samples taken at days 5 to 28 (patient 01), day 28 to 8 months (patient 02) and day 1 to day 28 (patient 04) post-adoptive transfer (Figure 2). In addition, when Th1/Th2 cytokines were measured in patient 02 PB and BM plasma, Th1 cytokines were detected immediately post-adoptive transfer (IFN-g=39 pg/ml, IL-2=8.6 pg/ml at peak); however these decreased to basal levels by day 3 (IFN-g=1.8 pg/ml) and day 2 (IL-2=2.6 pg/ml). The same trend in serum cytokines was observed for patient 01. Therefore, our studies reveal that, after adoptive transfer, LeY CAR-T cells persist in vivo, maintain their expression of the CAR, but show rapid polarization from a Th1 to a Th2 phenotype. Disclosures: No relevant conflicts of interest to declare.

2018 ◽  
Vol 11 (1) ◽  
Author(s):  
Jinghua Wang ◽  
Siyu Chen ◽  
Wei Xiao ◽  
Wende Li ◽  
Liang Wang ◽  
...  

Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 2205-2205 ◽  
Author(s):  
Elisa De Togni ◽  
Miriam Y Kim ◽  
Matt L Cooper ◽  
Julie Ritchey ◽  
Julie O'Neal ◽  
...  

Abstract Chimeric antigen receptor (CAR) T cells are a novel therapeutic approach which have shown good clinical outcomes in patients receiving CD19 CAR T cells for B cell acute lymphoblastic leukemia. CAR T cells are made to express a CAR that recognizes a specific surface antigen on a cell upon which they can then exert cytotoxic effects. We aim to extend the success of this therapy to acute myeloid leukemia (AML), a disease with generally poor clinical outcomes. However, due to the genetic heterogeneity characteristic of AML and the limited number of distinctive tumor markers, it has been difficult to find effective targets for CAR T cells on AML. C-type lectin like molecule-1 (CLL-1), also known as CD371, is a transmembrane glycoprotein that is expressed on about 90% of AML patient samples. CLL-1 may function as an inhibitory signaling receptor, as it contains an intracellular immunoreceptor tyrosine based inhibitory motif (ITIM). CLL-1 is primarily expressed on myeloid lineage cells in the bone marrow and in peripheral blood. While CLL-1 has been shown to be expressed on some granulocytes in the spleen, it is not reported to be expressed in non-hematopoietic tissues or on hematopoietic stem cells, which make CLL-1 a potential therapeutic target for AML. We generated two types of CLL-1 CARs, termed A and B, by using two different single chain variable fragments (scFvs) recognizing CLL-1. We used second generation CARs containing the scFvs, CD8 hinge and transmembrane domain, 4-1BB co-stimulatory domain, and CD3 zeta signaling domains. Using a lentiviral vector, we transferred the CAR gene into healthy donor human T cells and detected CAR expression by flow cytometry. We then tested the specific cytotoxic effects of CLL-1 CART-A and B on a CLL-1-expressing AML cell line, U937, by conducting a 4-hour chromium release assay. We found that both CAR T cells exhibited a dose-dependent killing of U937 (CLL-1 positive), while the untransduced (UTD) T cells had no cytotoxic effect (Figure 1A). We also found that U937 induces degranulation of CLL-1 CAR T cells as measured by CD107a expression by flow cytometry, while Ramos, a CLL-1 negative cell line, does not (Figure 1B). We then proceeded to investigate the in vivo efficacy of the CAR T cells. We injected NOD/SCID/IL2RG-null (NSG) mice with 1 x 106 THP-1 cells, a CLL-1 positive cell line. We confirmed engraftment by bioluminescent imaging (BLI) after 7 days and then injected 4 x 106 UTD, CLL-1 CART-A or CLL-1 CART-B. Surprisingly, only one of the CAR constructs, CLL-1 CART-A, showed significant activity in vivo, although both CARs had shown comparable activity in vitro. CLL-1 CART-A treated mice had delayed tumor progression and significantly increased length of survival (85 days vs. 63 days, p = 0.0021) compared to mice injected with UTD (Figure 1C and D). While CLL-1 CART-B treated mice also exhibited slower tumor growth and a trend towards better survival (72 days vs. 63 days, p=0.0547) this was not statistically significant. Post-mortem analysis showed that human T cells that continued to express CAR were present in the tumor, bone marrow and spleen of mice treated with CLL-1 CART-A only, while the UTD and CLL-1 CART-B treated mice showed tumor in all organs and no T cells. In summary, we show that CLL-1 CAR T cells can selectively eliminate CLL-1 positive target cells in vitro and in vivo, albeit with different degrees of efficacy modulated by the scFv. Studies are ongoing to investigate the mechanism behind the differential activity of these CAR constructs and to increase the long-term antitumor efficacy. Our results demonstrate that targeting CLL-1 using CAR T cell therapy holds promise for the treatment of AML. Disclosures Cooper: WUGEN: Consultancy, Equity Ownership.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 2555-2555 ◽  
Author(s):  
Roman Galetto ◽  
Céline Lebuhotel ◽  
Agnès Gouble ◽  
Nuria Mencia-Trinchant ◽  
Cruz M Nicole ◽  
...  

Abstract The remissions achieved using autologous T-cells expressing chimeric antigen receptors (CARs) in patients with advanced B cell leukemia and lymphomas have encouraged the use of CAR technology to treat different types of cancers by targeting distinct tumor-specific antigens. Since the current autologous approach utilizes CAR T-cells manufactured on a "per patient" basis, we propose an alternative approach based on the use of a standardized platform for manufacturing T-cells from third-party healthy donors to generate allogeneic "off-the-shelf" CAR T-cell-based frozen products. In the present work we have adapted this allogeneic platform to the production of T-cells targeting CD123, the transmembrane alpha chain of the interleukin-3 receptor, which is expressed on tumor cells from the majority of patients with Acute Myeloid Leukemia (AML). Multiple antigen recognition domains were screened in the context of different CAR architectures to identify candidates displaying activity against cells expressing variable levels of the CD123 antigen. The three lead candidates were tested in an orthotopic human AML cell line xenograft mouse model. From the three candidates that displayed comparable activity in vitro, we found two candidates capable of eradicating tumor cells in vivo with high efficiency. Subsequently, Transcription Activator-Like Effector Nuclease (TALEN) gene editing technology was used to inactivate the TCRα constant (TRAC) gene, eliminating the potential for engineered T-cells to mediate Graft versus Host Disease (GvHD). Editing of the TRAC gene can be achieved at high frequencies, and allows efficient amplification of TCR-deficient T-cells that no longer mediate alloreactivity in a xeno-GvHD mouse model. In addition, we show that TCR-deficient T-cells display equivalent in vitro and in vivo activity to non-edited T-cells expressing the same CAR. We have performed an initial evaluation of the expression of CD123 in AML patients and found an average cell surface expression of CD123 was of 67% in leukemic blasts (95% CI 48-82), 71% in CD34+CD38+ cells (95% CI 56-86), and 64% in CD34+CD38- (95% CI 41-87). Importantly, we have found that CD123 surface expression persists in CD34+CD38-CD90- cells after therapy in at least 20% of patients in remission (n=25), thus emphasizing the relevance of the target. Currently, the sensitivity of primary AML cells to CAR T-cells is being tested. Finally, we will also present our large scale manufacturing process of allogeneic CD123 specific T-cells from healthy donors, showing the feasibility for this off-the-shelf T-cell product that could be available for administration to a large number of AML patients. Disclosures Galetto: Cellectis SA: Employment. Lebuhotel:Cellectis SA: Employment. Gouble:Cellectis SA: Employment. Smith:Cellectis: Employment, Patents & Royalties.


2015 ◽  
Vol 33 (15_suppl) ◽  
pp. 3041-3041
Author(s):  
Roman Galetto ◽  
Celine Lebuhotel ◽  
Patricia Francon ◽  
Agnes Gouble ◽  
Julianne Smith

Blood ◽  
2015 ◽  
Vol 125 (22) ◽  
pp. 3466-3476 ◽  
Author(s):  
Rachel C. Lynn ◽  
Mathilde Poussin ◽  
Anna Kalota ◽  
Yang Feng ◽  
Philip S. Low ◽  
...  

Key PointsHuman FRβ-specific CAR T cells target AML in vitro and in vivo without toxicity against healthy bone marrow HSCs. Combination with ATRA-mediated receptor upregulation may augment FRβ-directed CAR therapy of AML.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 4058-4058 ◽  
Author(s):  
Degang Song ◽  
Michael H. Swartz ◽  
Steve G. Biesecker ◽  
Fernando Borda ◽  
Rutul R. Shah ◽  
...  

Abstract Relapsed acute myeloid leukemia (AML) is an aggressive disease with very poor outcomes. Redirection of T-cell specificity via chimeric antigen receptor (CAR) has shown promising anti-tumor activity in clinical trials, particularly for B cell linage malignancies. CD33 is a transmembrane protein expressed on normal and malignant myeloid-derived cells as well (as on subsets of activated T cells and NK cells). Since this protein is commonly expressed on AML cells, we sought to evaluate the efficacy of targeting AML with CD33-specific CAR-T cells. We generated a lentiviral construct to co-express CD33-specific CAR and a kill switch based on a tag derived from the epidermal growth factor receptor. The latter allows for the conditional elimination of CAR-T cells in vivo. Following transduction of primary T cells, we confirmed CAR and kill switch co-expression by flow cytometry and western blot analyses. Elimination of genetically modified T cells was demonstrated using the clinically-available antibody, cetuximab. CD33 CAR-T cells demonstrated specific cytotoxicity to CD33+ target cell lines. CD33 CAR-T cells were also activated to produce IFNg, TNF, and IL-2 cytokines in response to CD33+ target cells. Furthermore, adoptive transfer of CD33 CAR-T in immunocompromised (NSG) mice bearing established CD33+(CD19neg) AML (MOLM-13) tumor resulted in reduction of tumor burden and improvement of overall survival, compared to control mice receiving CD19 CAR-T cells or no immunotherapy (Figure). Sampling of blood demonstrated the persistence of the CD33 CAR-T cells with no detection of AML (MOLM-13) tumor cells. These pre-clinical data demonstrate the effectiveness of CD33 CAR-T cells in targeting CD33+ AML tumor cells and provide a rationale for future clinical evaluation in AML patients with unmet medical need. Disclosures Song: Intrexon Corporation: Employment, Equity Ownership. Swartz:Intrexon Corporation: Employment, Equity Ownership. Biesecker:Intrexon Corporation: Employment, Equity Ownership. Borda:Intrexon Corporation: Employment. Shah:Intrexon Corporation: Employment, Equity Ownership. Wierda:Genentech: Research Funding; Gilead: Research Funding; Abbvie: Research Funding; Novartis: Research Funding; Acerta: Research Funding. Cooper:MD Anderson Cancer Center: Employment; Intrexon: Equity Ownership; Sangamo BioSciences: Patents & Royalties; Targazyme,Inc.,: Equity Ownership; City of Hope: Patents & Royalties; ZIOPHARM Oncology: Employment, Equity Ownership, Patents & Royalties; Miltenyi Biotec: Honoraria; Immatics: Equity Ownership. Chan:Intrexon Corporation: Employment, Equity Ownership.


Children ◽  
2020 ◽  
Vol 7 (2) ◽  
pp. 14
Author(s):  
Rebecca Epperly ◽  
Stephen Gottschalk ◽  
Mireya Paulina Velasquez

Outcomes for pediatric patients with acute myeloid leukemia (AML) remain poor, highlighting the need for improved targeted therapies. Building on the success of CD19-directed immune therapy for acute lymphocytic leukemia (ALL), efforts are ongoing to develop similar strategies for AML. Identifying target antigens for AML is challenging because of the high expression overlap in hematopoietic cells and normal tissues. Despite this, CD123 and CD33 antigen targeted therapies, among others, have emerged as promising candidates. In this review we focus on AML-specific T cell engaging bispecific antibodies and chimeric antigen receptor (CAR) T cells. We review antigens being explored for T cell-based immunotherapy in AML, describe the landscape of clinical trials upcoming for bispecific antibodies and CAR T cells, and highlight strategies to overcome additional challenges facing translation of T cell-based immunotherapy for AML.


Sign in / Sign up

Export Citation Format

Share Document