Abstract
The majority of patients presenting with acute myeloid leukemia (AML) initially respond to chemotherapy but post-remission therapy is required to consolidate this response and achieve long-term disease-free survival. The most effective form of post-remission therapy relies on T-cell immunotherapy in the form of allogeneic hematopoietic cell transplantation (HCT). However, patients with active disease cannot usually expect to be cured with HCT. This inherent dichotomy implies that traditional T cell-based immunotherapy in the form of allogeneic HCT stops being efficacious somewhere between the measurable residual disease (MRD) and the morphologically obvious range. This is in part because the full power of T cells must be restrained in order to avoid lethal graft-versus-host disease (GVHD) and partly because only a sub-population of donor T cells are expected to be able to recognize AML cells via their T cell receptor. Chimeric antigen receptor (CAR) T cell therapy, most advanced in the treatment of patients with B-cell malignancies, may circumvent some of these limitations. However, major challenges remain to be overcome before CAR T cell therapy can be safely applied to AML.
Keywords: acute myeloid leukemia, AML, chimeric antigen receptor, CAR, graft- versus-host disease, GVHD, hematopoietic cell transplantation, HCT, measurable residual disease, MRD, T-cell immunotherapy
Introduction
Immunotherapy has revolutionized the treatment of a variety of advanced malignancies. Complete remissions have been reported in over 90% of patients with relapsed B-cell acute lymphoblastic leukemia (B-ALL) who receive anti-CD19 chimeric antigen receptor redirected T cells (CTL019 or CART-19) [1–3]. How to translate the success of CART cell therapy to other malignancies with unmet medical need such as acute myeloid leukemia (AML) remains an important question in the field.
CART cells recognize their target antigen via an interaction between the chimeric immunoreceptor and a cell surface ligand. The success of CART-19 is predicated on two factors: (1) massive expansion and persistence of the infused T cells, and (2) tolerability of CD19+ (B cell) aplasia. The most common side effect of CART-19 is depletion of endogenous normal B cells, yet protracted B-cell aplasia is well tolerated by patients [4,5]. Thus, a critical requirement of CART cell therapy is that the target tissue be expendable. AML is a malignancy of the hematopoietic stem/progenitor cells (HSPC) and shares cell surface antigens with normal HSPC and with normal myeloid progeny such as neutrophils and monocytes [6,7]. Hence, there is no truly AML-specific surface molecule. Several groups have demonstrated in mouse xenografts that anti-CD33 or anti-CD123 CAR T cells (CART-33 or CART-123) can eradicate AML but also lead to profound myeloablation [8–11]. Thus, although the efficacy of anti-AML CAR T cells appears equivalent to that of anti-ALL CAR T cells, hematopoietic toxicity is likely to be unacceptable. Here, I will review the absolute requirements for successful CAR T-cell therapy of AML (potency, target specificity, cell surface antigen expression, and persistence), describe what progress has been made in the field, and outline what challenges remain.
Potency
CAR T-cell-based therapeutics are likely more potent than equivalent monoclonal antibodies with which they share a targeting domain (single chain variable fragment). In fact, it is likely that CAR T cells are more potent than the equivalent bi-specific T-cell engagers as well [1,8,12–14]. Thus, clinical outcomes and toxicities observed on therapeutic trials of monoclonal antibodies or antibody-drug conjugates (ADC) cannot be extrapolated to CAR T cells. In AML there is extensive clinical experience with the anti-CD33 ADC gemtuzumab ozogamicin (GO) and experience is accruing with “naked” as well as conjugated CD123-specific compounds. Overall, responses to these agents as monotherapy are very limited [15–17]15–17 and toxicity is not prohibitive. In contrast, single administration of anti-CD33 or anti-CD123 CAR T cells leads to eradication of AML in xenograft mouse models along with irreversible marrow aplasia, related to expression of these antigens on normal marrow progenitors [9]. Thus, it would seem that potency against malignant myeloid cells correlates with toxicity against normal myeloid cells.
Target specificity to hematopoietic tissue
Hematopoietic toxicity is manageable with good supportive care, particularly if transient. However, transgenic T cells can traffic to non-hematopoietic organs and have been found throughout the body at autopsy of patients dying from on-target specificity against non-hematopoietic tissues [18–20]. Thus, it is critical that putative targets of anti-AML CAR T cells be restricted to hematopoietic tissues. In this context, Table 1 lists some of the cell surface targets in AML that have been evaluated or are under evaluation for CAR or antibody-based therapeutics along with their potential for off-target toxicity.
Table 1.
Target | Reference | Comments |
---|---|---|
CD123 | Mardiros 2013[10] Gill 2014 [8] Pizzitola 2014 [11] |
Hematopoietic toxicity and possibly endothelial toxicity |
CD33 | Dutour 2012 [27] Pizzitola 2014 [11] Kenderian 2014 [9] |
Hematopoietic toxicity and concern for hepatic toxicity |
CD44v6 | Casucci 2013 [28] | Concern for skin toxicity |
FLT3 | None | Neurologic tissue expression and hematopoietic toxicity |
CD34 | None | Endothelial expression and hematopoietic toxicity |
Others: Lewis Y antigen, CD38, CD96, CD99, IL1RAP, NKG2D ligands
Cell surface antigen target
Since CAR T cells rely on antibody-like recognition, only cell surface antigens are suitable for targeting. While the advantage is non MHC-restricted recognition and the lack of requirement for antigen presentation, the disadvantage is that most tumor-specific antigens are intracellular and thus not accessible to CAR T cells. One potential way to target intracellular antigens is using novel constructs that are based on antibodies recognizing peptide/MHC complexes [21]. While this approach paves the way to targeting leukemia-associated antigens (LAA) such as WT1 or PR3 and even leukemia-specific mutations (if the relevant peptides are presented on MHC, which is not a given), it is significantly limited by the same issues that bedevil T cell receptor (TCR)-based therapeutics, namely HLA restriction (each antibody will only recognize peptide in the context of a specific HLA molecule) and HLA dependency (downregulation of HLA molecules is a classic tumor escape mechanism), as well as the sheer heterogeneity of AML-associated mutations [22]. Nonetheless, LAA-specific adoptive T cell immunotherapy has been performed and its feasibility is established [23].
Persistence
Results from CART-19 studies in the setting of ALL and CLL indicate that persistence of the infused T cells correlates with prolonged responses. As expected, malignant and normal B cells are generally undetectable if CART cells are still present and conversely early loss of CART cells is a harbinger of relapse [1,5]. While B-cell aplasia and attendant hypogammaglobulinemia are easily tolerated, prolonged absence of myelopoiesis (particularly neutropenia) are likely to be poorly tolerated. There is therefore an inherent issue in the treatment of AML with CART cells: prolonged persistence is required for disease eradication yet is not clinically feasible. The main approach to mitigating this problem is depletion of CART cells followed by a “rescue” alloHCT thus combining an initial anti-myeloid effect with resumption of hematopoiesis from a donor source. This can be done by infusing CART cells where the CAR protein is translated from electroporated mRNA (thus not permanently expressed), or by engineering the CAR with a depletion marker such as EGFR (target of the monoclonal antibody cetuximab) or a suicide gene (such as inducible caspase 9) [9,10,24].
Clinical results to date
Few patients have been treated to date. Ritchie et al treated 4 patients with AML using autologous, retrovirally transduced second-generation T cells re-directed to the tumor antigen Lewis Y. The patients received up to 1.3x109 total cells with a transduction efficiency ranging from 14%–38%. Using radiolabelled T cells they demonstrated trafficking to the bone marrow. In a patient with leukemia cutis they demonstrated CAR T-cell infiltration of sites of disease. Of 3 patients treated in cytogenetic measurable residual disease 1 had stable disease and progressed at 49 days, 1 had stable disease and progressed at 23 months after infusion, and 1 had a transient cytogenetic remission and progressed at 5 months. The 1 patient who was treated in morphologic active disease (70% marrow blasts) experienced fever and rigors, a transient flare in the skin, and transient reduction in blast count [25]. Wang et al reported 1 patient treated with approximately 4x108 anti-CD33 CART cells who experienced cytokine release syndrome, moderate hepatotoxicity, and transient reduction in marrow blasts [26]. The same group reported in abstract form a single case treated with anti-CD123 CART cells, showing likely cytokine release syndrome but there was no clear anti-leukemic effect.
Current clinical trials
Unlike the profusion of clinical trials for B-cell malignancies, many fewer groups are attempting CART cell therapy for AML. CD123 is being targeted using a lentiviral approach by the City of Hope (NCT02159495) and using a transient mRNA-based approach by the University of Pennsylvania (NCT02623582). The Dana-Farber Cancer Institute has an NKG2D-based CAR trial for myeloid malignancies and multiple myeloma (NCT02203825). The Beijing group has an open anti-CD33 CAR trial (NCT01864902). The relative dearth of clinical trials stands in stark contrast to the relative incidence of AML and ALL (at least in adults) and highlights the challenges faced by investigators in the field, as highlighted in this brief review.
Summary
At present, there is no compelling case for an AML-specific cell surface antigen that can be safely used in order to unleash the power of CART cells. The apparent tolerability of targeting CD33 and CD123 using antibody-based approaches cannot be extrapolated to the CART cell arena due to the higher activity of the latter technology. Furthermore, infused CART cells must persist long-term to ensure eradication of the last leukemic cell and in order to ensure immunosurveillance against relapse. However, long-term persistence of myeloid-directed CART cells is likely incompatible with normal myelopoiesis, likely rendering the patient aplastic. Current clinical trials rely on transient CART activity or on the ability to deplete CART cells using clinically available monoclonal antibodies, with the plan to rescue hematopoiesis with an allogeneic HCT. Results from these trials and accumulating data from the CART-19 studies will inform future progress on CART cells and other potent immunotherapies for AML.
Footnotes
Conflict of interest:
Saar Gill receives research funding from Novartis and has intellectual property with the University of Pennsylvania and Novartis related to chimeric antigen receptor T cell therapy.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371:1507–1517. doi: 10.1056/NEJMoa1407222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med. 2014;6:224ra25. doi: 10.1126/scitranslmed.3008226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, 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;385:517–528. doi: 10.1016/S0140-6736(14)61403-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, et al. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368:1509–1518. doi: 10.1056/NEJMoa1215134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med. 2015;7:303ra139. doi: 10.1126/scitranslmed.aac5415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Konopleva MY, Jordan CT. Leukemia stem cells and microenvironment: biology and therapeutic targeting. J Clin Oncol. 2011;29:591–599. doi: 10.1200/JCO.2010.31.0904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Levine JH, Simonds EF, Bendall SC, Davis KL, Amir e, Tadmor MD, et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell. 2015;162:184–197. doi: 10.1016/j.cell.2015.05.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.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;123:2343–2354. doi: 10.1182/blood-2013-09-529537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.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;29:1637–1647. doi: 10.1038/leu.2015.52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Mardiros A, Dos SC, 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;122:3138–3148. doi: 10.1182/blood-2012-12-474056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pizzitola I, njos-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;28:1596–1605. doi: 10.1038/leu.2014.62. [DOI] [PubMed] [Google Scholar]
- 12.Jin L, Lee EM, Ramshaw HS, Busfield SJ, Peoppl AG, Wilkinson L, et al. Monoclonal antibody-mediated targeting of CD123, IL-3 receptor alpha chain, eliminates human acute myeloid leukemic stem cells. Cell Stem Cell. 2009;5:31–42. doi: 10.1016/j.stem.2009.04.018. [DOI] [PubMed] [Google Scholar]
- 13.Du X, Ho M, Pastan I. New immunotoxins targeting CD123, a stem cell antigen on acute myeloid leukemia cells. J Immunother. 2007;30:607–613. doi: 10.1097/CJI.0b013e318053ed8e. [DOI] [PubMed] [Google Scholar]
- 14.Topp MS, Gokbuget N, Stein AS, Zugmaier G, O’Brien S, Bargou RC, et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2015;16:57–66. doi: 10.1016/S1470-2045(14)71170-2. [DOI] [PubMed] [Google Scholar]
- 15.Loke J, Khan JN, Wilson JS, Craddock C, Wheatley K. Mylotarg has potent anti-leukaemic effect: a systematic review and meta-analysis of anti-CD33 antibody treatment in acute myeloid leukaemia. Ann Hematol. 2015;94:361–373. doi: 10.1007/s00277-014-2218-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.He SZ, Busfield S, Ritchie DS, Hertzberg MS, Durrant S, Lewis ID, et al. A Phase 1 study of the safety, pharmacokinetics and anti-leukemic activity of the anti-CD123 monoclonal antibody CSL360 in relapsed, refractory or high-risk acute myeloid leukemia. Leuk Lymphoma. 2015;56:1406–1415. doi: 10.3109/10428194.2014.956316. [DOI] [PubMed] [Google Scholar]
- 17.Frankel A, Liu JS, Rizzieri D, Hogge D. Phase I clinical study of diphtheria toxin-interleukin 3 fusion protein in patients with acute myeloid leukemia and myelodysplasia. Leuk Lymphoma. 2008;49:543–553. doi: 10.1080/10428190701799035. [DOI] [PubMed] [Google Scholar]
- 18.Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case report of a serious adverse event following the administration of T cells transduced with a chimeric antigen receptor recognizing ERBB2. Mol Ther. 2010;18:843–851. doi: 10.1038/mt.2010.24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Cameron BJ, Gerry AB, Dukes J, Harper JV, Kannan V, Bianchi FC, et al. Identification of a Titin-derived HLA-A1-presented peptide as a cross-reactive target for engineered MAGE A3-directed T cells. Sci Transl Med. 2013;5:197ra103. doi: 10.1126/scitranslmed.3006034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Linette GP, Stadtmauer EA, Maus MV, Rapoport AP, Levine BL, Emery L, et al. Cardiovascular toxicity and titin cross-reactivity of affinity-enhanced T cells in myeloma and melanoma. Blood. 2013;122:863–871. doi: 10.1182/blood-2013-03-490565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Dao T, Yan S, Veomett N, Pankov D, Zhou L, Korontsvit T, et al. Targeting the intracellular WT1 oncogene product with a therapeutic human antibody. Sci Transl Med. 2013;5:176ra33. doi: 10.1126/scitranslmed.3005661. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND, et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016;374:2209–2221. doi: 10.1056/NEJMoa1516192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Chapuis AG, Ragnarsson GB, Nguyen HN, Chaney CN, Pufnock JS, Schmitt TM, et al. Transferred WT1-reactive CD8+ T cells can mediate antileukemic activity and persist in post-transplant patients. Sci Transl Med. 2013;5:174ra27. doi: 10.1126/scitranslmed.3004916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.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. N Engl J Med. 2011;365:1673–1683. doi: 10.1056/NEJMoa1106152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.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. Mol Ther. 2013;21:2122–2129. doi: 10.1038/mt.2013.154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.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. Mol Ther. 2015;23:184–191. doi: 10.1038/mt.2014.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.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. Adv Hematol. 2012;2012:683065. doi: 10.1155/2012/683065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Casucci M, Nicolis di RB, 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;122:3461–3472. doi: 10.1182/blood-2013-04-493361. [DOI] [PubMed] [Google Scholar]