scholarly journals Author Correction: CAR-T cells targeting a nucleophosmin neoepitope exhibit potent specific activity in mouse models of acute myeloid leukaemia

Author(s):  
Guozhu Xie ◽  
Nikola A. Ivica ◽  
Bin Jia ◽  
Yingzhong Li ◽  
Han Dong ◽  
...  
2021 ◽  
Vol 10 (5) ◽  
Author(s):  
Aiko Hasegawa ◽  
Shoji Saito ◽  
Shogo Narimatsu ◽  
Shigeru Nakano ◽  
Mika Nagai ◽  
...  

Blood ◽  
2020 ◽  
Vol 136 (Supplement 1) ◽  
pp. 10-11
Author(s):  
Warren Hazelton ◽  
Sara Ghorashian ◽  
Martin Pule

Background: Chimeric Antigen Receptor (CAR) T-cell therapy represents a paradigm shift in therapy of advanced haematological malignancies in particular Acute Lymphoblastic Leukaemia. We have engineered multi-antigen specific CAR T-cells targeting three acute myeloid leukaemia (AML) associated antigens through incorporation of nanobody binding domains. Target antigens were selected for their broad expression on both on AML blasts as well as a high expression on Leukaemic Stem Cells (LSC). LSCs are a primitive self-renewing population that can contribute to relapse and may be phenotypically distinct from bulk AML blasts within the same patient. Aims: Our multi-antigenic approach thus aims to overcome antigen negative escape, overcome intra-tumour heterogeneity, and clear both LSCs and bulk disease by designing a tandem CAR capable of sensing CD33, CD123 and CLL1 on target cells. Methods: We initially screened libraries of nanobody binders for each antigen in a second generation CAR format. Once lead binders for each antigen were identified, Tandem CAR (TanCAR) structures were designed taking into consideration data on antigen structure from structural prediction servers and published crystal structures, nanobody domain mapping and antigen expression profiles. A TanCAR was then rationally designed comprising of three nanobodies joined by G4S linkers, fused to a hinge spacer, CD8 transmembrane domains, and 41BBz signalling domains. TanCAR T-cell function was compared to that of CAR T-cells targeting single antigens through in vitro co-culture assays with engineered target cells expressing single antigens (n=3). Antigen-specific cytotoxicity and proliferation were measured by flow cytometry assays; IL-2 and IFNy production was assayed by ELISA. Results: The ability of nanobody binding domains to bind individual antigens within the TanCAR format were confirmed using soluble antigen formats with a murine Fc tags via flow cytometry (Fig 1). In co-culture assays against engineered cell lines expressing single antigens (E:T=1:8, 24hrs) the TanCAR demonstrated antigen specific cytolytic activity (median cytotoxicity = 63%, 59%, and 47% against SupT1-CD33, SupT1-CD123, and SupT1-CLL1 respectively) approximately equal to or greater than single antigen targeting nanobody CAR T-cells (Fig 2a). In further co-culture experiments TanCAR T-cells produced high levels of IL-2 and IFNy when co cultured with cell lines expressing CD33 or CD123 only (n=3, median IL-2 = 9237pg/ml and 8190pg/ml, median IFNy 3924pg/ml and 4842pg/ml respectively) compared to SupT1 NT target cells, whereas low level cytokine production was observed with cell lines that express CLL-1 only (Fig 2b). Proliferation data matched this trend with greater levels of antigen specific proliferation observed when TanCAR T-cells were co-cultured with cells expressing CD33 and CD123 (7day co-culture, E:T=1:10, median fold expansion = 22, 8, and 5 fold for TanCAR co-cultured with cell lines expressing CD33, CD123, and CLL1 respectively), compared to antigen negative SupT1 target cells (Fig 2c). Summary/Conclusion: Collectively, our data demonstrate that TanCAR T-cells operate as an OR gate mediating potent reactivity against target cell lines expressing single AML antigens in vitro. We now aim to test for optimal TanCAR formats and potential additive or synergistic responses where TanCAR T cells are co-cultured with AML cell lines in-vivo (Molm14, PDX). Disclosures Ghorashian: Novartis: Honoraria; UCLB: Patents & Royalties; Amgen: Honoraria. Pule:Mana Therapeutics: Other: entitled to share of revenue from patents filed by UCL; UCLB: Patents & Royalties; Autolus: Current Employment, Other: owns stock in and receives royalties, Patents & Royalties.


Cancers ◽  
2021 ◽  
Vol 13 (12) ◽  
pp. 2941
Author(s):  
Luciana R. C. Barros ◽  
Emanuelle A. Paixão ◽  
Andrea M. P. Valli ◽  
Gustavo T. Naozuka ◽  
Artur C. Fassoni ◽  
...  

Immunotherapy has gained great momentum with chimeric antigen receptor T cell (CAR-T) therapy, in which patient’s T lymphocytes are genetically manipulated to recognize tumor-specific antigens, increasing tumor elimination efficiency. In recent years, CAR-T cell immunotherapy for hematological malignancies achieved a great response rate in patients and is a very promising therapy for several other malignancies. Each new CAR design requires a preclinical proof-of-concept experiment using immunodeficient mouse models. The absence of a functional immune system in these mice makes them simple and suitable for use as mathematical models. In this work, we develop a three-population mathematical model to describe tumor response to CAR-T cell immunotherapy in immunodeficient mouse models, encompassing interactions between a non-solid tumor and CAR-T cells (effector and long-term memory). We account for several phenomena, such as tumor-induced immunosuppression, memory pool formation, and conversion of memory into effector CAR-T cells in the presence of new tumor cells. Individual donor and tumor specificities are considered uncertainties in the model parameters. Our model is able to reproduce several CAR-T cell immunotherapy scenarios, with different CAR receptors and tumor targets reported in the literature. We found that therapy effectiveness mostly depends on specific parameters such as the differentiation of effector to memory CAR-T cells, CAR-T cytotoxic capacity, tumor growth rate, and tumor-induced immunosuppression. In summary, our model can contribute to reducing and optimizing the number of in vivo experiments with in silico tests to select specific scenarios that could be tested in experimental research. Such an in silico laboratory is an easy-to-run open-source simulator, built on a Shiny R-based platform called CARTmath. It contains the results of this manuscript as examples and documentation. The developed model together with the CARTmath platform have potential use in assessing different CAR-T cell immunotherapy protocols and its associated efficacy, becoming an accessory for in silico trials.


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.


2021 ◽  
Vol 13 (591) ◽  
pp. eabd8836
Author(s):  
Axel Hyrenius-Wittsten ◽  
Yang Su ◽  
Minhee Park ◽  
Julie M. Garcia ◽  
Josef Alavi ◽  
...  

The first clinically approved engineered chimeric antigen receptor (CAR) T cell therapies are remarkably effective in a subset of hematological malignancies with few therapeutic options. Although these clinical successes have been exciting, CAR T cells have hit roadblocks in solid tumors that include the lack of highly tumor-specific antigens to target, opening up the possibility of life-threatening “on-target/off-tumor” toxicities, and problems with T cell entry into solid tumor and persistent activity in suppressive tumor microenvironments. Here, we improve the specificity and persistent antitumor activity of therapeutic T cells with synthetic Notch (synNotch) CAR circuits. We identify alkaline phosphatase placental-like 2 (ALPPL2) as a tumor-specific antigen expressed in a spectrum of solid tumors, including mesothelioma and ovarian cancer. ALPPL2 can act as a sole target for CAR therapy or be combined with tumor-associated antigens such as melanoma cell adhesion molecule (MCAM), mesothelin, or human epidermal growth factor receptor 2 (HER2) in synNotch CAR combinatorial antigen circuits. SynNotch CAR T cells display superior control of tumor burden when compared to T cells constitutively expressing a CAR targeting the same antigens in mouse models of human mesothelioma and ovarian cancer. This was achieved by preventing CAR-mediated tonic signaling through synNotch-controlled expression, allowing T cells to maintain a long-lived memory and non-exhausted phenotype. Collectively, we establish ALPPL2 as a clinically viable cell therapy target for multiple solid tumors and demonstrate the multifaceted therapeutic benefits of synNotch CAR T cells.


Sign in / Sign up

Export Citation Format

Share Document