In Vitro Pre-Clinical Validation of Suicide Gene Modified Anti-CD33 Redirected Chimeric Antigen Receptor T-Cells for Acute Myeloid Leukemia

Kentaro Minagawa, Muhammad O Jamil, Mustafa Al-Obaidi, Larisa Pereboeva, Donna Salzman, Harry P Erba, Lawrence S Lamb, Ravi Bhatia, Shin Mineishi, Antonio Di Stasi, Kentaro Minagawa, Muhammad O Jamil, Mustafa Al-Obaidi, Larisa Pereboeva, Donna Salzman, Harry P Erba, Lawrence S Lamb, Ravi Bhatia, Shin Mineishi, Antonio Di Stasi

Abstract

Background: Approximately fifty percent of patients with acute myeloid leukemia can be cured with current therapeutic strategies which include, standard dose chemotherapy for patients at standard risk of relapse as assessed by cytogenetic and molecular analysis, or high-dose chemotherapy with allogeneic hematopoietic stem cell transplant for high-risk patients. Despite allogeneic hematopoietic stem cell transplant about 25% of patients still succumb to disease relapse, therefore, novel strategies are needed to improve the outcome of patients with acute myeloid leukemia.

Methods and findings: We developed an immunotherapeutic strategy targeting the CD33 myeloid antigen, expressed in ~ 85-90% of patients with acute myeloid leukemia, using chimeric antigen receptor redirected T-cells. Considering that administration of CAR T-cells has been associated with cytokine release syndrome and other potential off-tumor effects in patients, safety measures were here investigated and reported. We genetically modified human activated T-cells from healthy donors or patients with acute myeloid leukemia with retroviral supernatant encoding the inducible Caspase9 suicide gene, a ΔCD19 selectable marker, and a humanized third generation chimeric antigen receptor recognizing human CD33. ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells had a 75±3.8% (average ± standard error of the mean) chimeric antigen receptor expression, were able to specifically lyse CD33+ targets in vitro, including freshly isolated leukemic blasts from patients, produce significant amount of tumor-necrosis-factor-alpha and interferon-gamma, express the CD107a degranulation marker, and proliferate upon antigen specific stimulation. Challenging ΔCD19 selected inducible Caspase9-CAR.CD33 T-cells with programmed-death-ligand-1 enriched leukemia blasts resulted in significant killing like observed for the programmed-death-ligand-1 negative leukemic blasts fraction. Since the administration of 10 nanomolar of a non-therapeutic dimerizer to activate the suicide gene resulted in the elimination of only 76.4±2.0% gene modified cells in vitro, we found that co-administration of the dimerizer with either the BCL-2 inhibitor ABT-199, the pan-BCL inhibitor ABT-737, or mafosfamide, resulted in an additive effect up to complete cell elimination.

Conclusions: This strategy could be investigated for the safety of CAR T-cell applications, and targeting CD33 could be used as a 'bridge" therapy for patients coming to allogeneic hematopoietic stem cell transplant, as anti-leukemia activity from infusing CAR.CD33 T-cells has been demonstrated in an ongoing clinical trial. Albeit never performed in the clinical setting, our future plan is to investigate the utility of iC9-CAR.CD33 T-cells as part of the conditioning therapy for an allogeneic hematopoietic stem cell transplant for acute myeloid leukemia, together with other myelosuppressive agents, whilst the activation of the inducible Caspase9 suicide gene would grant elimination of the infused gene modified T-cells prior to stem cell infusion to reduce the risk of engraftment failure as the CD33 is also expressed on a proportion of the donor stem cell graft.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Generation and genetic modification of…
Fig 1. Generation and genetic modification of human activated T-cells.
(a) Diagram of CAR.CD33, and iC9-ΔCD19-CAR.CD33 plasmid constructs used for the genetic modification of human activated T-cells (ATCs). (b) Schematic representation of the protocol for ATCs generation and transduction. (c) Representative experiment displaying the CAR expression on the surface of ATCs generated from a healthy donor. (d) CAR expression on non-transduced (NT), CAR.CD33, or iC9-ΔCD19-CAR.CD33 transduced ATCs assessed on day 11 after transduction, and on ΔCD19 selected (sel.) iC9-CAR.CD33 assessed on day 28 after transduction. (e) Lymphocyte subset marker’s expression on NT, CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs assessed by flow cytometry on day 11 after transduction. (f) Memory marker’s expression on NT, CAR.CD33, and ΔCD19 sel. iC9-CAR.CD33 ATCs (assessed on day 11, 11, or 28 after transduction, respectively); (mean± standard error of the mean (SEM); N: 4–7 experiments from 5 healthy donors for all the experiments). LTR: long terminal repeat; Ψ: packaging signal; scFv: single chain variable fragment; ζ: Zeta chain of the T-cell receptor; PBMC: peripheral blood mononuclear cells; CM: central memory; EM: effector memory; TEMRA: terminal differentiated effector memory.
Fig 2. CAR.CD33, and ΔCD19 sel. iC9-CAR.CD33…
Fig 2. CAR.CD33, and ΔCD19 sel. iC9-CAR.CD33 ATCs displayed effectors function after stimulation with the CD33+ acute myeloid leukemia cell line, MV4-11.
(a) Overnight co-culture experiments using non transduced (NT), CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 activated T-cells (ATCs) against the MV 4-11eGFP+ CD33+ cell line, followed by evaluation of enhanced green fluorescent protein (eGFP)/CD33+ cell killing by flow cytometry, as compared with co-culture employing NT ATCs as effectors; (mean±SEM of 3 experiments using ATCs from 3 healthy donors) (left). Annexin V and 7-AAD expression from one representative co-culture experiment (right). (b) Luciferase-based cytotoxicity assay using NT, CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs after overnight incubation with the CD33+ MV4-11 cell line engineered to express the eGFP-firefly-luciferase (eGFP-ffLuc) construct. Effector:target (E:T) ratios ranged from 10:1 to 0.6:1; (N: 3–7 experiments with ATCs generated from 5 healthy donors). (c) Evaluation of CD107a degranulation marker expression on CAR.CD33 or ΔCD19 sel. iC9-CAR.CD33 ATCs (as compared with NT ATCs) after 4 hours of stimulation with MV4-11 CD33+ targets (E:T = 4:1); (N: mean±SEM of 4–6 experiments with ATCs from 4 healthy donors). (d) CFSE-based proliferation assay stimulating NT, CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs with irradiated (40 Gy) MV4-11 CD33+ cell line (gray), or no target cells (blank), and cultured for additional 5 days (E:T = 1:1). Data from one representative experiment is shown. (e) Cytokines production on supernatant collected after overnight incubation of NT, CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs with the MV4-11 CD33+ cell line (E:T = 4:1); (mean±SEM of 3–9 independent experiments from 3–4 different healthy donors). SEM: standard error of the mean; NS: not statistically significant.
Fig 3. CAR.CD33, and ΔCD19 sel. iC9-CAR.CD33…
Fig 3. CAR.CD33, and ΔCD19 sel. iC9-CAR.CD33 ATCs exhibit anti-tumor activity against samples from patients with active AML.
(a) CD33 expression on freshly isolated samples from patients with active acute myeloid leukemia (AML) evaluated by flow cytometry after gating on side scatterlow/CD45dim cells; (N:9 peripheral blood; N:5 bone marrow samples from different patients). (b) Overnight co-culture assay of non transduced (NT), CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs, generated from 3 healthy donors, with samples collected from patients with active AML labeled with the PKH-26 dye (effector:target (E:T) = 4:1), (patients#C, P, Q, R, and S). Results from a representative co-culture experiment (patient#C) gated on viable residual cells displaying the expression of (i) PKH vs.CD33, or (ii) PKH vs.CD3, or (iii) CD3 vs. CD33, (left). Summary results from 3–9 independent experiments displaying average (±SEM) percentage of cell killing is shown, (right). (c) Correlation (coefficient R2) between the reduction of PKH+CD33+ cells and CD33 expression level of the ΔCD19 sel. iC9-CAR.CD33 ATCs co-cultured with patient samples as represented in Fig 2B; (N: 9 independent experiments using ATCs from 3 healthy donors). (d) Evaluation of CD107a expression on CAR.CD33 or ΔCD19 sel. iC9-CAR.CD33 ATCs (as compared with NT ATCs) assessed 4 hours after stimulating them with samples collected from patients with AML (effector:target (E:T) = 4:1); (N:8–12 experiments using ATCs from 4 healthy donors). (e) Cytokines production (picograms per mL: pg/mL) on supernatant collected after overnight incubation of NT, CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs generated from healthy donor with samples isolated from 8 patients with AML (#C, G, L, N, O, P, Q, R, S, and U) (E:T = 4:1); (mean±SEM from 5–18 experiments using ATCs generated from 4 different healthy donors). ATCs: activated T-cells; Pt(s): patient(s); lymph: lymphocyte; SEM: standard error of the mean; NS: not statistically significant.
Fig 4. ΔCD19 sel. iC9-CAR.CD33 ATCs generated…
Fig 4. ΔCD19 sel. iC9-CAR.CD33 ATCs generated from a patient with AML.
(a) Peripheral blood mononuclear cells isolated from a patient with acute myeloid leukemia (AML) (patient#3) were used to generate activated T-cells (ATCs). Non transduced (NT), CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs were stained with the appropriate monoclonal antibody, and analyzed by flow cytometry after gating on the CD3+ population to analyze CD8 and CAR expression, (left) or CD45RA and CD62L expression, (right). (b) Dot plots from a representative co-culture experiment employing NT, CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs from patient#3 challenged with the MV4-11 CD33+ AML cell line genetically modified to express the enhanced green fluorescent protein (eGFP) marker (left), or with an autologous CD33+ AML sample (right); the co-cultures were performed in the presence of medium (top panels) or in the presence of autologous patient’s plasma (bottom panels).
Fig 5. Co-culture of CAR T-cells with…
Fig 5. Co-culture of CAR T-cells with PD-L1+ AML blasts.
(a) Non Transduced (NT), CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 activated T-cells (ATCs) generated from a healthy donor were co-cultured overnight with PD-L1+(top panels) or PD-L1neg(bottom panels) blasts collected from a patient with acute myeloid leukemia (AML), (patient#T). AML blasts were sorted by flow cytometry for PD-L1 expression, and labeled with the PKH-26 dye prior to co-culture. After overnight incubation 20,000 events were acquired by flow cytometry after gating on the PKH+ population and the reduction of CD33+ AML cells, as compared with the retained CD3+ cells is shown in the dot-plots. (b) ATCs co-cultured as per 5a were stained with a PD-1 monoclonal antibody, and PD-1 expression on the PKHneg/CD3+ population was analyzed by flow cytometry.
Fig 6. ΔCD19 sel. iC9-CAR.CD33 ATCs demonstrate…
Fig 6. ΔCD19 sel. iC9-CAR.CD33 ATCs demonstrate limited toxicity against normal hematopoietic stem cells.
(a) Human CD34+-selected hematopoietic stem cells (CD34+ HSC) from healthy volunteer donors were co-cultured overnight with or without non transduced (NT) or ΔCD19 sel. iC9-CAR.CD33 activated T-cells (ATCs), generated from healthy donors at an effector:target (E:T) ratio of 4:1. Afterwards, CD34+ HSC were isolated with hematopoietic lineage depletion kit, and enumerated by flow cytometry; (mean±SEM, N:4, from duplicate experiments using ATCs from 2 different healthy donors). (b) 500 CD34+lyneage- HSC, from each respective condition outlined above, were plated on cytokine conditioned semisolid medium, (MethoCult H4434). After 14 days, hematopoietic colony formation was scored using standard morphologic criteria; (mean±SEM, N:4, from duplicate experiments using ATCs from 2 different healthy donors). CFU: Colony-forming unit: BFU-E: burst-forming unit-erythroid: CFU-M/G/GM: CFU-macrophage/granulocyte/granulocyte, macrophage; CFU-GEMM: CFU-granulocyte, erythroid, macrophage, megakaryocyte; SEM: standard error of the mean; NS: not statistically significant.
Fig 7. Pharmacologic elimination of ΔCD19 sel.…
Fig 7. Pharmacologic elimination of ΔCD19 sel. iC9-CAR.CD33 ATCs.
(a) NT, CAR.CD33, or ΔCD19 sel. iC9-CAR.CD33 ATCs generated from healthy donors were treated overnight with ABT-199 or ABT-737 [both at 2 or 10μM], or mafosfamide [0.5 or 2 μg/mL] with or without the CID [10 nM], and thereafter washed by centrifugation. On day 1, cells were harvested, stained with Annexin V/7-AAD and analyzed by flow cytometry. Ten to twenty thousand total events were acquired for each sample (the same number of events was acquired within each experiment). Average percentage (±SEM) of cell killing is represented in the histogram graph. Conditions without application of the CID are shown as shaded bars, while conditions involving the application of the CID are shown as black bars; (N:3–15 experiments using ATCs from 3–8 different healthy donors). (b) Cells treated overnight with ABT-199 or ABT-737 [both at 2 μM], or mafosfamide [0.5 μg/mL] (with or without CID [10 nM]) were harvested, washed by centrifugation and cultured for additional 4–7 days in the presence of culture medium supplemented with rhIL2 50 I.U./mL. Outline of the experimental plan, (left), and histogram data from a representative experiment displaying residual viable cells is shown, (right). UnTx: untreated; SEM: standard error of the mean; CID: chemical inducer of dimerization to activate the iC9 suicide gene; rhIL2: recombinant human interleukin-2; ABT-199: BCL-2 inhibitor; ABT-737: Pan-BCL inhibitor; MAF: Mafosfamide; NS: not statistically significant.

References

    1. Thomas S, Stauss HJ, Morris EC. Molecular immunology lessons from therapeutic T-cell receptor gene transfer. Immunology. 2010;129(2):170–7. 10.1111/j.1365-2567.2009.03227.x
    1. Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proceedings of the National Academy of Sciences of the United States of America. 1993;90(2):720–4.
    1. Garrido F, Ruiz-Cabello F, Cabrera T, Perez-Villar JJ, Lopez-Botet M, Duggan-Keen M, et al. Implications for immunosurveillance of altered HLA class I phenotypes in human tumours. Immunology today. 1997;18(2):89–95.
    1. Minagawa K, Zhou X, Mineishi S, Di Stasi A. Seatbelts in CAR therapy: How Safe Are CARS? Pharmaceuticals (Basel, Switzerland). 2015;8(2):230–49.
    1. Gill S, Tasian SK, Ruella M, Shestova O, Li Y, Porter DL, et al. Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood. 2014;123(15):2343–54. 10.1182/blood-2013-09-529537
    1. Marin V, Pizzitola I, Agostoni V, Attianese GM, Finney H, Lawson A, et al. Cytokine-induced killer cells for cell therapy of acute myeloid leukemia: improvement of their immune activity by expression of CD33-specific chimeric receptors. Haematologica. 2010;95(12):2144–52. 10.3324/haematol.2010.026310
    1. Tettamanti S, Marin V, Pizzitola I, Magnani CF, Giordano Attianese GM, Cribioli E, et al. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor. British journal of haematology. 2013;161(3):389–401. 10.1111/bjh.12282
    1. Dutour A, Marin V, Pizzitola I, Valsesia-Wittmann S, Lee D, Yvon E, et al. In Vitro and In Vivo Antitumor Effect of Anti-CD33 Chimeric Receptor-Expressing EBV-CTL against CD33 Acute Myeloid Leukemia. Advances in hematology. 2012;2012:683065 10.1155/2012/683065
    1. Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, Lassailly F, Tettamanti S, Spinelli O, et al. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia. 2014;28(8):1596–605. 10.1038/leu.2014.62
    1. Casucci M, Nicolis di Robilant B, Falcone L, Camisa B, Norelli M, Genovese P, et al. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood. 2013;122(20):3461–72. 10.1182/blood-2013-04-493361
    1. Mardiros A, Dos Santos C, McDonald T, Brown CE, Wang X, Budde LE, et al. T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia. Blood. 2013;122(18):3138–48. 10.1182/blood-2012-12-474056
    1. Kenderian SS, Ruella M, Shestova O, Klichinsky M, Aikawa V, Morrissette JJ, et al. CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia. Leukemia. 2015;29(8):1637–47. 10.1038/leu.2015.52
    1. Ritchie DS, Neeson PJ, Khot A, Peinert S, Tai T, Tainton K, et al. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Molecular therapy: the journal of the American Society of Gene Therapy. 2013;21(11):2122–9.
    1. Wang QS, Wang Y, Lv HY, Han QW, Fan H, Guo B, et al. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Molecular therapy: the journal of the American Society of Gene Therapy. 2015;23(1):184–91.
    1. McMillan SJ, Crocker PR. CD33-related sialic-acid-binding immunoglobulin-like lectins in health and disease. Carbohydrate research. 2008;343(12):2050–6. 10.1016/j.carres.2008.01.009
    1. Pearce DJ, Taussig D, Zibara K, Smith LL, Ridler CM, Preudhomme C, et al. AML engraftment in the NOD/SCID assay reflects the outcome of AML: implications for our understanding of the heterogeneity of AML. Blood. 2006;107(3):1166–73. 10.1182/blood-2005-06-2325
    1. Nakahata T, Okumura N. Cell surface antigen expression in human erythroid progenitors: erythroid and megakaryocytic markers. Leukemia & lymphoma. 1994;13(5–6):401–9.
    1. Buckley SA, Walter RB. Update on antigen-specific immunotherapy of acute myeloid leukemia. Current hematologic malignancy reports. 2015;10(2):65–75. 10.1007/s11899-015-0250-9
    1. Stasi R. Gemtuzumab ozogamicin: an anti-CD33 immunoconjugate for the treatment of acute myeloid leukaemia. Expert opinion on biological therapy. 2008;8(4):527–40. 10.1517/14712598.8.4.527
    1. Pagano L, Fianchi L, Caira M, Rutella S, Leone G. The role of Gemtuzumab Ozogamicin in the treatment of acute myeloid leukemia patients. Oncogene. 2007;26(25):3679–90. 10.1038/sj.onc.1210364
    1. Di Stasi A, Tey SK, Dotti G, Fujita Y, Kennedy-Nasser A, Martinez C, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. The New England journal of medicine. 2011;365(18):1673–83. 10.1056/NEJMoa1106152
    1. Zhou X, Di Stasi A, Tey SK, Krance RA, Martinez C, Leung KS, et al. Long-term outcome after haploidentical stem cell transplant and infusion of T cells expressing the inducible caspase 9 safety transgene. Blood. 2014;123(25):3895–905. 10.1182/blood-2014-01-551671
    1. Zhou X, Dotti G, Krance RA, Martinez CA, Naik S, Kamble RT, et al. Inducible caspase-9 suicide gene controls adverse effects from alloreplete T cells after haploidentical stem cell transplantation. Blood. 2015;125(26):4103–13. 10.1182/blood-2015-02-628354
    1. Gargett T, Brown MP. The inducible caspase-9 suicide gene system as a "safety switch" to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Frontiers in pharmacology. 2014;5:235 10.3389/fphar.2014.00235
    1. Barese CN, Felizardo TC, Sellers SE, Keyvanfar K, Di Stasi A, Metzger ME, et al. Regulated apoptosis of genetically modified hematopoietic stem and progenitor cells via an inducible caspase-9 suicide gene in rhesus macaques. Stem cells (Dayton, Ohio). 2015;33(1):91–100.
    1. Zhou X, Naik S, Dakhova O, Dotti G, Heslop HE, Brenner MK. Serial Activation of the Inducible Caspase 9 Safety Switch After Human Stem Cell Transplantation. Molecular therapy: the journal of the American Society of Gene Therapy. 2016;24(4):823–31.
    1. Pan R, Hogdal LJ, Benito JM, Bucci D, Han L, Borthakur G, et al. Selective BCL-2 inhibition by ABT-199 causes on-target cell death in acute myeloid leukemia. Cancer discovery. 2014;4(3):362–75. 10.1158/-13-0609
    1. de Jonge ME, Huitema AD, Rodenhuis S, Beijnen JH. Clinical pharmacokinetics of cyclophosphamide. Clinical pharmacokinetics. 2005;44(11):1135–64. 10.2165/00003088-200544110-00003
    1. Baev DV, Krawczyk J, M OD, Szegezdi E. The BH3-mimetic ABT-737 effectively kills acute myeloid leukemia initiating cells. Leukemia research reports. 2014;3(2):79–82. 10.1016/j.lrr.2014.06.001
    1. Straathof KC, Pule MA, Yotnda P, Dotti G, Vanin EF, Brenner MK, et al. An inducible caspase 9 safety switch for T-cell therapy. Blood. 2005;105(11):4247–54. 10.1182/blood-2004-11-4564
    1. Iuliucci JD, Oliver SD, Morley S, Ward C, Ward J, Dalgarno D, et al. Intravenous safety and pharmacokinetics of a novel dimerizer drug, AP1903, in healthy volunteers. Journal of clinical pharmacology. 2001;41(8):870–9.
    1. Cheng S, Zheng J, Zhu J, Xie C, Zhang X, Han X, et al. PD-L1 gene polymorphism and high level of plasma soluble PD-L1 protein may be associated with non-small cell lung cancer. The International journal of biological markers. 2015;30(4):e364–8. 10.5301/jbm.5000170
    1. Wang L, Wang H, Chen H, Wang WD, Chen XQ, Geng QR, et al. Serum levels of soluble programmed death ligand 1 predict treatment response and progression free survival in multiple myeloma. Oncotarget. 2015;6(38):41228–36. 10.18632/oncotarget.5682
    1. Finkelmeier F, Canli O, Tal A, Pleli T, Trojan J, Schmidt M, et al. High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. European journal of cancer (Oxford, England: 1990). 2016;59:152–9.
    1. Zhou Q, Munger ME, Veenstra RG, Weigel BJ, Hirashima M, Munn DH, et al. Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood. 2011;117(17):4501–10. 10.1182/blood-2010-10-310425
    1. Krupka C, Kufer P, Kischel R, Zugmaier G, Lichtenegger FS, Kohnke T, et al. Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell-induced immune escape mechanism. Leukemia. 2016;30(2):484–91. 10.1038/leu.2015.214
    1. Bhatia S. Caring for the long-term survivor after allogeneic stem cell transplantation. Hematology / the Education Program of the American Society of Hematology American Society of Hematology Education Program. 2014;2014(1):495–503.
    1. Jan M, Snyder TM, Corces-Zimmerman MR, Vyas P, Weissman IL, Quake SR, et al. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Science translational medicine. 2012;4(149):149ra18.
    1. Ding L, Ley TJ, Larson DE, Miller CA, Koboldt DC, Welch JS, et al. Clonal evolution in relapsed acute myeloid leukaemia revealed by whole-genome sequencing. Nature. 2012;481(7382):506–10. 10.1038/nature10738
    1. Maus MV, Haas AR, Beatty GL, Albelda SM, Levine BL, Liu X, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer immunology research. 2013;1:26–31.
    1. Chang EC, Liu H, West JA, Zhou X, Dakhova O, Wheeler DA, et al. Clonal Dynamics In Vivo of Virus Integration Sites of T Cells Expressing a Safety Switch. Molecular therapy: the journal of the American Society of Gene Therapy. 2016;24(4):736–45.
    1. Roberts AW, Davids MS, Pagel JM, Kahl BS, Puvvada SD, Gerecitano JF, et al. Targeting BCL2 with Venetoclax in Relapsed Chronic Lymphocytic Leukemia. The New England journal of medicine. 2016;374(4):311–22. 10.1056/NEJMoa1513257
    1. Karlsson H, Lindqvist AC, Fransson M, Paul-Wetterberg G, Nilsson B, Essand M, et al. Combining CAR T cells and the Bcl-2 family apoptosis inhibitor ABT-737 for treating B-cell malignancy. Cancer gene therapy. 2013;20(7):386–93. 10.1038/cgt.2013.35
    1. Srinivasula SM, Hegde R, Saleh A, Datta P, Shiozaki E, Chai J, et al. A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. Nature. 2001;410(6824):112–6. 10.1038/35065125
    1. Hu Y, Benedict MA, Wu D, Inohara N, Nunez G. Bcl-XL interacts with Apaf-1 and inhibits Apaf-1-dependent caspase-9 activation. Proceedings of the National Academy of Sciences of the United States of America. 1998;95(8):4386–91.
    1. Wilkie S, Picco G, Foster J, Davies DM, Julien S, Cooper L, et al. Retargeting of human T cells to tumor-associated MUC1: the evolution of a chimeric antigen receptor. Journal of immunology (Baltimore, Md: 1950). 2008;180(7):4901–9.
    1. Savoldo B, Rooney CM, Di Stasi A, Abken H, Hombach A, Foster AE, et al. Epstein Barr virus specific cytotoxic T lymphocytes expressing the anti-CD30zeta artificial chimeric T-cell receptor for immunotherapy of Hodgkin disease. Blood. 2007;110(7):2620–30. 10.1182/blood-2006-11-059139
    1. Di Stasi A, De Angelis B, Rooney CM, Zhang L, Mahendravada A, Foster AE, et al. T lymphocytes coexpressing CCR4 and a chimeric antigen receptor targeting CD30 have improved homing and antitumor activity in a Hodgkin tumor model. Blood. 2009;113(25):6392–402. 10.1182/blood-2009-03-209650

Source: PubMed

3
S'abonner