Systematic preclinical evaluation of CD33-directed chimeric antigen receptor T cell immunotherapy for acute myeloid leukemia defines optimized construct design

Haiying Qin, Lila Yang, John A Chukinas, Nirali Shah, Samiksha Tarun, Marie Pouzolles, Christopher D Chien, Lisa M Niswander, Anthony R Welch, Naomi Taylor, Sarah K Tasian, Terry J Fry, Haiying Qin, Lila Yang, John A Chukinas, Nirali Shah, Samiksha Tarun, Marie Pouzolles, Christopher D Chien, Lisa M Niswander, Anthony R Welch, Naomi Taylor, Sarah K Tasian, Terry J Fry

Abstract

Background: Successful development of chimeric antigen receptor (CAR) T cell immunotherapy for children and adults with relapsed/refractory acute myeloid leukemia (AML) is highly desired given their poor clinical prognosis and frequent inability to achieve cure with conventional chemotherapy. Initial experiences with CD19 CAR T cell immunotherapy for patients with B-cell malignancies highlighted the critical impact of intracellular costimulatory domain selection (CD28 vs 4-1BB (CD137)) on CAR T cell expansion and in vivo persistence that may impact clinical outcomes. However, the impact of costimulatory domains on the efficacy of myeloid antigen-directed CAR T cell immunotherapy remains unknown.

Methods: In this preclinical study, we developed six CAR constructs targeting CD33, a highly expressed and validated AML target, comprised of one of three single-chain variable fragments with CD3ζ and either CD28 or 4-1BB costimulatory domains. We systematically compared the preclinical in vitro and in vivo efficacy of T cells lentivirally transduced with CD33 CAR constructs (CD33CARTs) against human AML.

Results: We observed potent in vitro cytokine production and cytotoxicity of CD33CARTs incubated with human CD33+ AML cell lines, as well as robust in vivo antileukemia activity in cell line and childhood AML patient-derived xenograft (PDX) models. Gemtuzumab-based CD33CARTs were unexpectedly toxic in vivo in animal models despite observed in vitro anti-leukemia activity. CD28-based CD33CARTs consistently induced more robust inhibition of leukemia proliferation in AML cell line and PDX models than did 4-1BB-based CD33CARTs. A 'best-in-class' lintuzumab-CD28/CD3ζ CAR construct was thus selected for clinical translation.

Conclusions: CD33 is a critical antigen for potential immunotherapeutic targeting in patients with AML. Based on this rigorous preclinical evaluation, our validated clinical grade lintuzumab-CD28/CD3ζ CD33CART immunotherapy is now under evaluation in a first-in-child/first-in-human phase 1 clinical trial for children and adolescents/young adults with relapsed/refractory AML.

Trial registration number: clinicaltrials.gov; NCT03971799.

Keywords: adoptive; chimeric antigen; hematologic neoplasms; immunotherapy; pediatrics; receptors; translational medical research.

Conflict of interest statement

Competing interests: HQ and TJF have filed United States patent number WO2019178382A1 ‘Anti-CD33 Chimeric Antigen Receptors and Their Uses.’ SKT is a scientific advisory board member for Aleta Biotherapeutics and Kura Oncology and has received research funding from Incyte Corporation and Gilead Sciences for unrelated studies. TJF has received research funding from Lentigen and Elevate Bio for unrelated studies and is a current employee of Sana Biotechnology. The remaining authors declare no relevant conflicts of interest.

© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
CD33 chimeric antigen receptor construct (CD33CART) design and validation. (A) Schema of second-generation CD33-redirectedCAR constructs. Single-chain variable fragments (ScFvs) were derived from commercially available CD33 antibodies gemtuzumab ozogamicin (hP67.6; green), lintuzumab (SGN-33; light blue), and M195 (dark blue) and paired with a CD8 hinge or CD28 transmembrane (28tm) domain linker, 4-1BB or CD28 costimulatory endodomain, and a CD3ζ endodomain (designated BBz and 28z, respectively). CD123 CAR T cells with a 32 716 ScFv were used as a positive control for some studies. (B) Flow cytometry analysis of protein L expression demonstrating transduction efficiency of T cells transduced with gemtuzumab-based (gem), lintuzumab-based (lin), and M195-based CD33 CAR constructs or CD123 CAR constructs. (C) Flow cytometry quantification of CD33 site density (molecules/cell) on AML cell lines (MOLM14, MV4;11, THP-1, U937) and childhood acute myeloid leukemia (AML) patient-derived xenograft (PDX) models (JMML117, AML290) used for in vitro and/or in vivo testing of CD33CARTs. CD123 site density is shown as a comparator. The B-ALL cell line NALM-6 was used as a CD33-negative control.
Figure 2
Figure 2
Robust in vitro anti-leukemia activity of CD33 chimeric antigen receptor constructs (CD33CARTs). (A) CD33CART or CD123CART (positive acute myeloid leukemia (AML) killing control) cells were co-incubated with target-positive AML cells in vitro in a 1:1 ratio as indicated for 18 hours, and IL-2 and IFN-γ levels were quantified by ELISA in the culture supernatant. NALM-6 was again used as a negative control cell line. Experiments were performed in triplicate (technical replicates) using two normal T cell donors (biologic replicates). Data are displayed as mean±SD. (B) Cell-Titer Glo viability assays of human AML cell lines co-incubated with CD33CARTs or CD123CARTs in 1:1 effector: T cell show marked inhibition of cell growth over time with all tested CAR T cell products using two normal T cell donors. The B-ALL cell line NALM-6 was used as a CD33-negative control. T cell transduction efficiency ranged from 41% to 94% (mean 75%) depending on the CAR construct used. 28z=CD28/CD3ζ endodomains, BBz=4–1BB/CD3ζ endodomains. Cytokine and viability assays were performed in technical triplicates with biologic duplicates (T cell donor number 1 and number 2). Mean data are displayed for each condition with SD also shown for the viability assays. Data were analyzed by two-way ANOVA with multiple comparisons analysis of column means. All six CD33CARTs induced statistically significant killing of CD33 +AML cell lines at day 4 versus no T cell or GFP-transduced ((-)TC) negative controls (p<0.0001 for all comparisons), but were not statistically different from one another.
Figure 3
Figure 3
CD33 chimeric antigen receptor construct (CD33CART) inhibits acute myeloid leukemia (AML) proliferation in vivo. (A) Luciferase-transduced MOLM14 cells (1×106) were injected intravenously via tail vein into NSG mice on day 0. Once engraftment was documented by bioluminescent imaging (BLI) on day 7, cohorts of three mice were randomized to intravenous treatment with saline, GFP-transduced T cells ((-)TC), or one of the CD33CARTs as designated (5×106 total cells/mouse; red arrow). Mice were followed by weekly BLI and euthanized when a predetermined maximal radiance level of 1×106 photons/s/cm2/sr was detected, indicative of leukemia progression. All three gemtuzumab-based (gem), lintuzumab-based (lin), and M195-based CD33CARTs demonstrated some anti-leukemia activity in vivo with complete inhibition of MOLM14 proliferation observed only in the 4-1BB/CD3ζ (BBz) construct-containing T cells versus partial activity with CD28/CD3ζ(28z) versions. (B) Graphic representation of summary BLI radiance data was performed in Prism. 28z, CD28/CD3ζ endodomains, BBz, 4–1BB/CD3ζ endodomains.
Figure 4
Figure 4
CD33 chimeric antigen receptor construct (CD33CART) inhibits leukemia proliferation in vivo in childhood acute myeloid leukemia (AML) patient-derived xenograft models. Busulfan-conditioned NSGS mice engrafted with primary AML cells (tertiary PDX models (A) JMML117 and (B) AML290) were randomized (n=5 mice/cohort) to intravenous treatment with saline, irrelevant target GFP-transduced T cells ((-)TC), or one of the CD33CARTs as designated (5×106 total cells/mouse). CD33CART transduction efficiency ranged from 19.9% to 88.3% for these experiments with information detailed in online supplemental table 1. Mice were followed by weekly flow cytometric quantification of human CD33+/CD45 +AML and CD3+ CAR T cells in peripheral blood. Animals were euthanized at 2 or 4 weeks after treatment depending on rate of leukemia progression in control animals or >15% wt loss. AML cells and T cells were quantified in murine bone marrow and/or spleens as above. Superior leukemia clearance was again seen with the lintuzumab-based CD33CART with a CD28/CD3ζendodomain (lin-28z). *P<0.05, **p<0.01, ***p<0.001 by ANOVA with Dunnett post-test for multiple comparisons of treatment group versus saline control (black bars). Absence of symbol indicates lack of statistical significance.
Figure 5
Figure 5
In vivo exhaustion potential of lintuzumab-based CD33 chimeric antigen receptor constructs (CD33CARTs) in an acute myeloid leukemia (AML) xenograft model. (A) Luciferase-expressing MOLM14 xenograft mice were created as in figure 3. Cohort of five mice were treated with one 4×106 dose of GFP-transduced T cells ((-)TC), lintuzumab-CD28/CD3ζ (lin-28z), or lintuzumab-4-1BB/CD3z (lin-BBz) CD33CART intravenous on day 4 (red arrow), then followed by bioluminescent imaging. CD33CART transduction efficiency ranged from 19.9% to 88.3% for these experiments with information detailed in online supplemental table 1. (B) Representative dot plots show flow cytometric detection of percentages of human CD3+ CD33 CART cells in harvested end-study murine spleens at day 26 following T cell treatment (upper panels). T cell quantification (lower panels) detected greater numbers in lin-BBz-treated versus lin-28z-treated animals. (C) Representative dot plots show flow cytometric analysis of CD4/CD8 T cell profiles (upper panels) with ratio calculation (lower panel) in end-study murine spleens. (D) Similar percentages of cell surface CD33 CAR positivity (bound to recombinant CD33-Fc fusion protein) were detected in CD4+ T cells, but not CD8+ T cells, when comparing lin-28z and lin-BBz CD33CARTs. (E) Naïve (N), effector memory (EM), and effector memory re-expressing CD45RA (TEMRA) human T cells in murine spleens at the day 26 end-study timepoint were further characterized by CCCR7+ CD45RA+, CCR7-CD45RA-, and CCR7-CD45RA+ flow cytometric profiling, respectively. Representative plots for CD4+ and CD8+CD33CARTs are shown (upper panels) and quantification of the different populations within the CD4+ (left) and CD8+ (right) subsets are shown. (F) Exhaustion phenotypes of CD33CARTs were evaluated as a function of PD1 and PD1/LAG3 expression profiles and representative dot plots are presented with %cells designated numerically for each gated quadrant (top). Quantification of PD1+ and PD1+/LAG3 +CD4+ and CD8+ T cells in murine spleens at the day 26 end-study timepoint are shown for lin-28z versus lin-4BBz CD33CART treatments. Paired data were analyzed by Student’s t-test with *p<0.05, **p<0.01, ***p<0.001, and ****p<0.0001.
Figure 6
Figure 6
Minimal bystander toxicity of clinical CD33 chimeric antigen receptor construct (CD33CART) product. (A) 1×105 GFP-transduced T cells ((-)TC) or clinical-grade lintuzumab-CD28/CD3ζ (lin-28z) cells were co-incubated in a 1:1 ratio with induced pluripotent stem cell lines of the designated normal tissue types for 18 hours. IFN-γ levels were quantified by ELISA in the culture supernatant as in figure 2. MOLM14 cells were used as a CD33 +positive control. No appreciable cytokine production was detected in any of the normal tissue conditions.
Figure 7
Figure 7
Preclinical validation of anti-leukemia activity of clinical-grade CD33 chimeric antigen receptor construct (CD33CART). The lintuzumab-based CD33CART with a CD28/CD3ζendodomain (lin-28z) was selected for further development and evaluation in a phase 1 clinical trial. (A) Transduction efficiency of clinical grade lin-28z CD33CART was 30.3% for this experiment. (B) Luciferase-transduced MOLM14 cells (1×106) were injected intravenous into NSG mice on day 0. Animals were assessed by bioluminescent imaging (BLI) and then randomized to treatment with GFP-transduced T cells ((-)TC) or clinical-grade lin-28z CD33CART cells (5×106 total cells/mouse) on day 3 (red arrow). Mice were followed by weekly BLI and euthanized when a predetermined maximal radiance level of 1×1010 photons/s/cm2/sr was detected Complete inhibition of leukemia proliferation was again observed with CD33CART.

References

    1. Tarlock K, Meshinchi S. Pediatric acute myeloid leukemia: biology and therapeutic implications of genomic variants. Pediatr Clin North Am 2015;62:75–93. 10.1016/j.pcl.2014.09.007
    1. Zwaan CM, Kolb EA, Reinhardt D, et al. . Collaborative efforts driving progress in pediatric acute myeloid leukemia. J Clin Oncol 2015;33:2949–62. 10.1200/JCO.2015.62.8289
    1. Rasche M, Zimmermann M, Borschel L, et al. . Successes and challenges in the treatment of pediatric acute myeloid leukemia: a retrospective analysis of the AML-BFM trials from 1987 to 2012. Leukemia 2018;32:2167–77. 10.1038/s41375-018-0071-7
    1. Lee DW, Kochenderfer JN, Stetler-Stevenson M, et al. . T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 2015;385:517–28. 10.1016/S0140-6736(14)61403-3
    1. Gardner RA, Finney O, Annesley C, et al. . Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood 2017;129:3322–31. 10.1182/blood-2017-02-769208
    1. Maude SL, Laetsch TW, Buechner J, et al. . Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med 2018;378:439–48. 10.1056/NEJMoa1709866
    1. Park JH, Rivière I, Gonen M, et al. . Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 2018;378:449–59. 10.1056/NEJMoa1709919
    1. Ghorashian S, Kramer AM, Onuoha S, et al. . Enhanced CAR T cell expansion and prolonged persistence in pediatric patients with ALL treated with a low-affinity CD19 CAR. Nat Med 2019;25:1408–14. 10.1038/s41591-019-0549-5
    1. Lamble AJ, Tasian SK. Opportunities for immunotherapy in childhood acute myeloid leukemia. Blood Adv 2019;3:3750–8. 10.1182/bloodadvances.2019000357
    1. Ehninger A, Kramer M, Röllig C, et al. . Distribution and levels of cell surface expression of CD33 and CD123 in acute myeloid leukemia. Blood Cancer J 2014;4:e218. 10.1038/bcj.2014.39
    1. Pollard JA, Alonzo TA, Loken M, et al. . Correlation of CD33 expression level with disease characteristics and response to gemtuzumab ozogamicin containing chemotherapy in childhood AML. Blood 2012;119:3705–11. 10.1182/blood-2011-12-398370
    1. Pollard JA, Loken M, Gerbing RB, et al. . CD33 expression and its association with Gemtuzumab Ozogamicin response: results from the randomized phase III children's Oncology Group trial AAML0531. J Clin Oncol 2016;34:747–55. 10.1200/JCO.2015.62.6846
    1. Hills RK, Castaigne S, Appelbaum FR, et al. . Addition of gemtuzumab ozogamicin to induction chemotherapy in adult patients with acute myeloid leukaemia: a meta-analysis of individual patient data from randomised controlled trials. Lancet Oncol 2014;15:986–96. 10.1016/S1470-2045(14)70281-5
    1. Gamis AS, Alonzo TA, Meshinchi S, et al. . Gemtuzumab ozogamicin in children and adolescents with de novo acute myeloid leukemia improves event-free survival by reducing relapse risk: results from the randomized phase III Children’s Oncology Group trial AAML0531. J Clin Oncol 2014;32:3021–32. 10.1200/JCO.2014.55.3628
    1. Appelbaum FR, Bernstein ID. Gemtuzumab ozogamicin for acute myeloid leukemia. Blood 2017;130:2373–6. 10.1182/blood-2017-09-797712
    1. Norsworthy KJ, Ko C-W, Lee JE, et al. . Fda approval summary: Mylotarg for treatment of patients with relapsed or refractory CD33-Positive acute myeloid leukemia. Oncologist 2018;23:1103–8. 10.1634/theoncologist.2017-0604
    1. Raza A, Jurcic JG, Roboz GJ, et al. . Complete remissions observed in acute myeloid leukemia following prolonged exposure to lintuzumab: a phase 1 trial. Leuk Lymphoma 2009;50:1336–44. 10.1080/10428190903050013
    1. Feldman EJ, Brandwein J, Stone R, et al. . Phase III randomized multicenter study of a humanized anti-CD33 monoclonal antibody, lintuzumab, in combination with chemotherapy, versus chemotherapy alone in patients with refractory or first-relapsed acute myeloid leukemia. J Clin Oncol 2005;23:4110–6. 10.1200/JCO.2005.09.133
    1. Ravandi F, Walter RB, Subklewe M, et al. . Updated results from phase I dose-escalation study of AMG 330, a bispecific T-cell engager molecule, in patients with relapsed/refractory acute myeloid leukemia (R/R AML). J Clin Oncol 2020;38:supplement:7508. abstract 7508. 10.1200/JCO.2020.38.15_suppl.7508
    1. Subklewe M, Stein A, Walter RB, et al. . Preliminary results from a phase 1 first-in-human study of AMG 673, a novel half-life extended (HLE) Anti-CD33/CD3 BiTE® (bispecific T-cell Engager) in patients with relapsed/refractory (R/R) acute myeloid leukemia (AML). Blood 2019;134:833. 10.1182/blood-2019-127977
    1. Tambaro FP, Singh H, Jones E, et al. . Autologous CD33-CAR-T cells for treatment of relapsed/refractory acute myelogenous leukemia. Leukemia 2021. doi:10.1038/s41375-021-01232-2. [Epub ahead of print: 08 Apr 2021].
    1. Guedan S, Calderon H, Posey AD, et al. . Engineering and design of chimeric antigen receptors. Mol Ther Methods Clin Dev 2019;12:145–56. 10.1016/j.omtm.2018.12.009
    1. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 2020;17:147–67. 10.1038/s41571-019-0297-y
    1. Long AH, Haso WM, Shern JF, et al. . 4-1BB costimulation ameliorates T cell exhaustion induced by tonic signaling of chimeric antigen receptors. Nat Med 2015;21:581–90. 10.1038/nm.3838
    1. Li S, Zhang J, Wang M, et al. . Treatment of acute lymphoblastic leukaemia with the second generation of CD19 CAR-T containing either CD28 or 4-1BB. Br J Haematol 2018;181:360–71. 10.1111/bjh.15195
    1. Mueller KT, Maude SL, Porter DL, et al. . Cellular kinetics of CTL019 in relapsed/refractory B-cell acute lymphoblastic leukemia and chronic lymphocytic leukemia. Blood 2017;130:2317–25. 10.1182/blood-2017-06-786129
    1. Finney OC, Brakke HM, Rawlings-Rhea S, et al. . CD19 CAR T cell product and disease attributes predict leukemia remission durability. J Clin Invest 2019;129:2123–32. 10.1172/JCI125423
    1. Fry TJ, Shah NN, Orentas RJ, et al. . CD22-targeted CAR T cells induce remission in B-ALL that is naive or resistant to CD19-targeted CAR immunotherapy. Nat Med 2018;24:20–8. 10.1038/nm.4441
    1. Shah NN, Highfill SL, Shalabi H, et al. . CD4/Cd8 T-cell selection affects chimeric antigen receptor (CAR) T-cell potency and toxicity: updated results from a phase I anti-CD22 CAR T-cell trial. J Clin Oncol 2020;38:1938–50. 10.1200/JCO.19.03279
    1. Tasian SK. Acute myeloid leukemia chimeric antigen receptor T-cell immunotherapy: how far up the road have we traveled? Ther Adv Hematol 2018;9:135–48. 10.1177/2040620718774268
    1. Niktoreh N, Lerius B, Zimmermann M, et al. . Gemtuzumab ozogamicin in children with relapsed or refractory acute myeloid leukemia: a report by Berlin-Frankfurt-Münster Study Group. Haematologica 2019;104:120–7. 10.3324/haematol.2018.191841
    1. Corbacioglu S, Jabbour EJ, Mohty M. Risk factors for development of and progression of hepatic veno-occlusive Disease/Sinusoidal obstruction syndrome. Biol Blood Marrow Transplant 2019;25:1271–80. 10.1016/j.bbmt.2019.02.018
    1. Cowan AJ, Laszlo GS, Estey EH, et al. . Antibody-based therapy of acute myeloid leukemia with gemtuzumab ozogamicin. Front Biosci 2013;18:1311–34. 10.2741/4181
    1. Co MS, Avdalovic NM, Caron PC, et al. . Chimeric and humanized antibodies with specificity for the CD33 antigen. J Immunol 1992;148:1149–54.
    1. Qin H, Cho M, Haso W, et al. . Eradication of B-ALL using chimeric antigen receptor-expressing T cells targeting the TSLPR oncoprotein. Blood 2015;126:629–39. 10.1182/blood-2014-11-612903
    1. Qin H, Ramakrishna S, Nguyen S, et al. . Preclinical development of bivalent chimeric antigen receptors targeting both CD19 and CD22. Mol Ther Oncolytics 2018;11:127–37. 10.1016/j.omto.2018.10.006
    1. Forman S, Mardiros A. Cd123-specific chimeric antigen receptor redirected t cells and methods of their use. US Patent. 2014.
    1. Mardiros A, Dos Santos C, McDonald T, 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;122:3138–48. 10.1182/blood-2012-12-474056
    1. Gill S, Tasian SK, Ruella M, et al. . Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 2014;123:2343–54. 10.1182/blood-2013-09-529537
    1. Tasian SK, Kenderian SS, Shen F, et al. . Optimized depletion of chimeric antigen receptor T cells in murine xenograft models of human acute myeloid leukemia. Blood 2017;129:2395–407. 10.1182/blood-2016-08-736041
    1. Qin H, Edwards JP, Zaritskaya L, et al. . Chimeric antigen receptors incorporating D domains targeting CD123 direct potent mono- and bi-specific antitumor activity of T cells. Mol Ther 2019;27:1262–74. 10.1016/j.ymthe.2019.04.010
    1. Zheng Z, Chinnasamy N, Morgan RA. Protein L: a novel reagent for the detection of chimeric antigen receptor (CAR) expression by flow cytometry. J Transl Med 2012;10:29. 10.1186/1479-5876-10-29
    1. Jacoby E, Nguyen SM, Fountaine TJ, et al. . CD19 CAR immune pressure induces B-precursor acute lymphoblastic leukaemia lineage switch exposing inherent leukaemic plasticity. Nat Commun 2016;7:12320. 10.1038/ncomms12320
    1. Ramakrishna S, Highfill SL, Walsh Z, et al. . Modulation of target antigen density improves CAR T-cell functionality and persistence. Clin Cancer Res 2019;25:5329–41. 10.1158/1078-0432.CCR-18-3784
    1. Walter RB, Gooley TA, van der Velden VHJ, et al. . CD33 expression and P-glycoprotein-mediated drug efflux inversely correlate and predict clinical outcome in patients with acute myeloid leukemia treated with gemtuzumab ozogamicin monotherapy. Blood 2007;109:4168–70. 10.1182/blood-2006-09-047399
    1. Lamba JK, Pounds S, Cao X, et al. . Coding polymorphisms in CD33 and response to gemtuzumab ozogamicin in pediatric patients with AML: a pilot study. Leukemia 2009;23:402–4. 10.1038/leu.2008.185
    1. Lamba JK, Chauhan L, Shin M, et al. . CD33 splicing polymorphism determines Gemtuzumab Ozogamicin response in de novo acute myeloid leukemia: report from randomized phase III children's Oncology Group trial AAML0531. J Clin Oncol 2017;35:2674–82. 10.1200/JCO.2016.71.2513
    1. Gale RE, Popa T, Wright M, et al. . No evidence that CD33 splicing SNP impacts the response to GO in younger adults with AML treated on UK MRC/NCRI trials. Blood 2018;131:468–71. 10.1182/blood-2017-08-802157
    1. Schneider D, Xiong Y, Hu P, et al. . A unique human immunoglobulin heavy chain variable domain-only CD33 CAR for the treatment of acute myeloid leukemia. Front Oncol 2018;8:539. 10.3389/fonc.2018.00539
    1. Ruella M, Barrett DM, Kenderian SS, et al. . Dual CD19 and CD123 targeting prevents antigen-loss relapses after CD19-directed immunotherapies. J Clin Invest 2016;126:3814–26. 10.1172/JCI87366
    1. van der Stegen SJC, Hamieh M, Sadelain M. The pharmacology of second-generation chimeric antigen receptors. Nat Rev Drug Discov 2015;14:499–509. 10.1038/nrd4597
    1. Reading JL, Gálvez-Cancino F, Swanton C, et al. . The function and dysfunction of memory CD8+ T cells in tumor immunity. Immunol Rev 2018;283:194–212. 10.1111/imr.12657
    1. Kenderian SS, Ruella M, Shestova O, et al. . CD33-specific chimeric antigen receptor T cells exhibit potent preclinical activity against human acute myeloid leukemia. Leukemia 2015;29:1637–47. 10.1038/leu.2015.52
    1. Kim MY, Yu K-R, Kenderian SS, et al. . Genetic inactivation of CD33 in hematopoietic stem cells to enable CAR T cell immunotherapy for acute myeloid leukemia. Cell 2018;173:e19:1439–53. 10.1016/j.cell.2018.05.013
    1. Wang Q-shun, Wang Y, Lv H-yan, et al. . Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther 2015;23:184–91. 10.1038/mt.2014.164
    1. Li S, Tao Z, Xu Y, et al. . CD33-Specific chimeric antigen receptor T cells with different co-stimulators showed potent anti-leukemia efficacy and different phenotype. Hum Gene Ther 2018;29:626–39. 10.1089/hum.2017.241

Source: PubMed

3
Prenumerera