Abstract
Chemotherapy resistance and relapse remain significant sources of mortality for children and adults with acute myeloid leukemia (AML). Further intensification of conventional cytotoxic chemotherapy is likely not feasible due to the severity of acute and long-term side effects upon normal tissues commonly induced by these drugs. Successful development and implementation of new precision medicine treatment approaches for patients with AML, which may improve leukemia remission and diminish toxicity, is thus a major priority. Tumor antigen-redirected chimeric antigen receptor (CAR) T-cell immunotherapies have induced remarkable responses in patients with relapsed or chemorefractory B-lymphoblastic leukemia, and similar strategies are now under early clinical study in adults with relapsed/refractory AML. However, potential on target/off tumor toxicity of AML CAR T-cell immunotherapies, notably aplasia of normal myeloid cells, may limit broader implementation of such approaches. Careful selection of optimal target antigens, consideration of toxicity mitigation strategies, and development of methodologies to circumvent potential CAR T-cell resistance are essential for successful implementation of cellular immunotherapies for patients with high-risk AML.
Keywords: acute myeloid leukemia, chimeric antigen receptor, clinical trial, cytokine release syndrome, immunotherapy, T cell
Introduction
Acute myeloid leukemia (AML) is the most common leukemia occurring in adults and the second most common leukemia of childhood. Intensive multiagent chemotherapy regimens for patients with AML have changed little during the past four decades and are associated with considerable acute and chronic toxicities. Despite consolidation with hematopoietic stem cell transplantation (HSCT) for those with high-risk AML, relapse-free and overall survival for many patients remains poor.1–5 New therapeutic approaches are compulsory to improve outcomes.
Targeted antibody-based or cellular immunotherapies are particularly attractive for patients with demonstrated chemotherapy-resistant disease, who often have few remaining curative treatment options. Studies have demonstrated improved outcomes for subsets of patients with high-risk AML treated with chemotherapy and the CD33-targeted antibody–drug conjugate (ADC) gemtuzumab ozogamicin,6–8 which led to approval of gemtuzumab by the United States Food and Drug Administration (FDA) and the European Medicines Agency in adults and children with de novo or relapsed AML. Early phase clinical trials of other antibody-based therapeutics, including new CD33 and CD123 [interleukin (IL)-3Rα] targeting ADCs and bispecific T-cell engager (BiTE)/dual affinity retargeting (DART) antibodies are currently underway in adults with relapsed/refractory AML.9–14
Particular progress has recently been made with adoptive cellular therapies using autologous or allogeneic T cells engineered with synthetic chimeric antigen receptors (CARs) redirected against tumor antigens with remarkable early-phase clinical trial results in patients with B-lymphoblastic leukemia (B-ALL) treated with CD19 or CD22 CAR T cells.15–22 The logistics and mechanics of CAR T-cell engineering for patients with acute leukemias and potential safety modifications have been delineated in numerous recent reviews.23–28 In contrast to earlier T-cell receptor (TCR)-directed T cells, genetically engineered CAR T cells (usually autologous and permanently modified via retroviral or lentiviral transduction) bind to cell surface antigens without the need for traditional matching of major histocompatibility complex (MHC) antigens to prevent alloimmunization. Upon binding of the synthetic CAR to its target antigen, intracellular signaling via costimulatory domains induces T-cell activation and marked expansion, often resulting in rapid and complete cancer cell cytotoxicity. However, ‘on target/on tumor’ sequelae of CAR T-cell activation and proliferation can result in life-threatening toxicities, including neurologic dysfunction, cytokine release syndrome (CRS), and macrophage activation syndrome.29–33 Concomitant ‘on target/off tumor’ effects of CAR T cells caused by indiscriminate cellular binding to the same antigens on nonmalignant normal cells can also be quite detrimental to the host and have been described in detail elsewhere.24,34–36
In patients with AML, hematologic toxicity with potential CAR T-cell-induced myeloablation is a legitimate particular concern given the lack of currently known AML-only surface proteins and expression of targeted candidate antigens on normal myeloid precursor cells (Figure 1). As such, rescue of CAR T-cell-treated patients with HSCT to restore normal myelopoiesis may be required. High potency CAR T-cell exhaustion37 and immune escape with target antigen loss or immunophenotype switching38–40 are also emerging as major mechanisms of resistance to CAR T-cell and antibody-based immunotherapies, a reproducible lesson now well learned from treated patients.19,41 This review discusses the current bench-to-bedside landscape of antigen-redirected CAR T-cell immunotherapies for patients with AML, continued challenges in the field, and emerging strategies that may optimize therapeutic efficacy while reducing potential toxicity.
Figure 1.
Balancing efficacy and toxicity of chimeric antigen receptor (CAR) T-cell immunotherapy for acute myeloid leukemia (AML). CNS, central nervous system.
Particulars of AML CAR T-cell development: picking target antigens, potency, persistence, and potential problems
Prolonged B-cell aplasia is an expected (and perhaps desired) bystander toxicity in patients treated with CD19 CAR T cells given the concomitant presence of CD19 on normal B cells.16,17,42 Patients with B-ALL treated with these targeted immunotherapies are now monitored closely for continued B-cell loss as a biomarker of CD19 CAR T-cell persistence. Surprisingly, few untoward effects of this toxicity have been observed to date, as CD19 expression is restricted to B cells, and patients with continued B-cell depletion and resultant hypogammaglobulinemia can be safely supported with monthly intravenous immunoglobulin infusions to minimize infectious complications.43 Similar B-cell aplasia has now been observed in patients with relapsed/refractory B-ALL treated with the CD22 ADC inotuzumab ozogamicin or CD22 CAR T cells given similar levels of CD22 expression on normal B lymphocytes.19,44,45
Ideal AML surface antigen characteristics for successful immunotherapeutic targeting include restriction to malignant myeloblasts without expression on normal hematopoietic stem cells or on vital normal nonhematopoietic tissues. Preferably, the antigens should be critical to leukemogenesis initiation or maintenance or expressed on leukemia-initiating cells to maximize AML eradication versus expressed only on more mature, often clonally heterogeneous bulk leukemia cell populations. Given the lack of AML cell-only antigens identified to date, most preclinical and clinical immunotherapy efforts have instead attempted to identify a therapeutic window of targeting myeloid antigens overexpressed on AML blasts that are present at lower or minimal levels on normal tissues. It is thus possible, even probable, that permanently modified AML CAR T cells under current study will eradicate normal antigen-expressing myeloid progenitor cells or induce significant myeloablation. As many AML-associated surface proteins of interest for CAR T-cell targeting are not restricted to the hematopoietic compartment (e.g. CD33 on hepatic Kupffer cells, CD44 on keratinocytes, CD123 on endothelial cells, CD135 on neural and testis tissues), risk of appreciable nonhematologic side effects must also be carefully considered. Optimal AML immunotherapy approaches should thus aim when possible to target surface antigens occurring only in the hematopoietic compartment or those where ‘bystander’ toxicity to antigen-expressing normal tissues will be minimal or clinically tolerable.
To address the issue of leukemia cell antigen specificity, a recent transcriptomic and surfaceome proteomic analysis profiled human AML cell lines and primary patient samples and reported unique pairings of antigens detected on AML blasts, but not co-occurring on normal hematopoietic cells. The investigators suitably concluded that dual antigen-cotargeted CAR T cells will likely induce less toxicity upon normal hematopoietic and non-hematopoietic tissues in which only single antigen expression occurs.46 Another recent preclinical study demonstrated greater in vivo anti-leukemia activity and prolonged animal survival in AML cell line xenograft models treated with compound CD33xCD123-targeting CAR T cells versus single antigen-redirected T cells.47 These combinatorial therapeutic strategies to increase efficacy and potentially reduce toxicity are analogous to that of bispecific CD19xCD22 CAR T cells48 under current clinical evaluation in adults and children with relapsed B-cell ALL [ClinicalTrials.gov identifier: NCT03241940].
While antecedent experience with and demonstrated safety of monoclonal antibody-based immunotherapeutics provides some reassurance that targeting of the same antigens with CAR T cells will be similarly well tolerated, it is imperative to remember that these ‘living drugs’ are likely far more potent and longer lasting than their antibody counterparts, thereby increasing potential for both increased anti-AML efficacy and prolonged toxicity. This paradox must be carefully balanced: CAR T-cell persistence for some duration is unquestionably necessary to eradicate leukemia, yet potential ensuing myeloablation caused by such therapies poses great hematologic and infectious risk to the host without additional intervention. As such, investigators focused on AML CAR T-cell development have appropriately attempted to maximize efficacy and minimize toxicity via subsequent T-cell termination, or rescue with allogeneic HSCT to restore hematopoiesis.36,49–51
Preclinical studies of AML CAR T-cell immunotherapy
In the past decade, numerous preclinical studies have reported promising in vitro and in vivo activity of CAR T cells targeting various myeloid antigens in human AML cells and patient-derived xenograft (PDX) models, including Lewis-Y,52 CD33,53–59 CD123,35,47,50,51,54,60–64 CD44v6,49 the folate receptor β,65 CD38,66,67 and the FLT3 receptor (CD135),68–70 and CLL-1 (CLEC12A).71–73 All of these targeted CAR T cells have demonstrated potent in vitro cytotoxicity and cytokine production when incubated with antigen-positive AML cell lines or primary cells (Table 1). In many of these studies, a single dose of permanently modified CAR T cells also completely eradicated in vivo human AML cell proliferation in engrafted immunocompromised mice.
Table 1.
Candidate AML antigens for CAR T-cell immunotherapy and data from preclinical studies.
| Target antigen | Protein type | Present on AML blasts? | Present on LICs? | Present on HSCs? | Other normal tissue expression | In vitro studies: cytokine production | In vivo studies | Potential toxicity | Key references in AML |
|---|---|---|---|---|---|---|---|---|---|
| CD7 | Immunoglobulin superfamily protein | Some | Possibly | No or transient | T cells | Yes, specific data n/a | Anti-AML and anti-LIC activity in PDX models, minimal depletion of MPs | Lymphodepletion | Gomes Silva et al.74,75 |
| CD25 (IL-2Rα) | Type I transmembrane protein | Some | Possibly | No | T and B cells, myeloid precursors, thymocytes, oligodendrocytes | n/a | n/a | Lymphodepletion, myeloablation, neural toxicity | n/a |
| CD32 (FcγRIII) | Immunoglobulin superfamily glycoprotein | Some | Possibly | No | Monocytes, macrophages, granulocytes, platelets and B cells | n/a | n/a | Lymphodepletion, myeloablation, thrombocytopenia | n/a |
| CD33 (sieglec-3) | Transmembrane receptor / adhesion molecule | Yes | Yes | Yes | Kupffer cells | GM-CSF, IFN-γ, IL-2, MIP1α, MIP1β | Activity in AML PDX models, depletion of normal hematopoietic cells | Hematopoietic depletion, sinusoidal obstruction syndrome | Dutour et al.,50 Pizzitola et al.,54 Kenderian et al.,55 O’Hear et al.,56 Zheng et al.59 |
| CD38 (cyclic ADP ribose hydrolase) | Glycoprotein | Yes | Rare | No | B, T, and NK cells | IFN-γ, IL-2, TNF-α | Activity in MM PDX models | Lymphodepletion | Drent et al.66 |
| CD44v6 | Glycoprotein | Yes | Yes | No | Keratinocytes | IFN-γ, IL-2 | Activity in MM and AML PDX models, monocytopenia | Skin toxicity, monocytopenia | Casucci et al.49 |
| CD47 | Immunoglobulin superfamily glycoprotein | Yes | Yes | Low | Keratinocytes, immune cells | n/a | n/a | Immunosuppression, skin toxicity | n/a |
| CD56 (neural cell adhesion molecule) | Glycoprotein | Yes | Possibly | No | Dendritic, NK, and T cells, neural and neuroendocrine tissues | n/a | n/a | Immunosuppression, neural toxicity | n/a |
| CD90 (Thy1) | Immunoglobulin superfamily glycoprotein | Some | Possibly | Yes | Endothelial cells, nervous tissue, thymocytes | n/a | n/a | Endothelial toxicity, neurotoxicity | n/a |
| CD96 | Type I transmembrane protein | Yes | Yes | Minimal | T and NK cells, epithelial cells | n/a | n/a | Lymphodepletion, epithelial toxicity | n/a |
| CD117 (c-KIT) | Type III receptor tyrosine kinase | Yes | Yes | Yes | Lymphoid and myeloid progenitors, mast cells, melanocytes, interstitial cells of Cajal | n/a | n/a | Myeloablation | n/a |
| CD123 (IL3Rα) | Type I cytokine receptor | Yes | Yes | Yes | Myeloid progenitors, dendritic and endothelial cells, basophils | GM-CSF, IFN-γ, IL-2, MIP1α, MIP1β | Activity in AML PDX models, depletion of normal hematopoietic cells | Hematopoietic depletion, cardiac endothelial damage | Mardiros et al.,50 Tettamanti et al.,60 Gill et al.,61 Pizzitola et al.,54 Arcangeli et al.,67 Tasian et al.51 |
| CD135 (FLT3R) | Type III cytokine receptor | Yes | Yes | Yes | Neurons, testis | IFN-γ, IL-2 | Activity in AML PDX models, depletion of normal hematopoietic cells | Hematopoietic depletion, neurotoxicity, GU toxicity | Chien et al.,68 Sauter et al.,76 Chen et al.,69 Jetani et al.70 |
| CD174 (Lewis-Y) | Glycosphingolipid (fucosyltransferase) | Yes | Likely | Yes | Intestinal epithelial cells | IFN-γ, IL-2 | Activity in AML and MM PDX models | GI toxicity | Peinert et al.52 |
| CLL-1 (CLEC12A) | Type II membrane glycoprotein | Yes | Yes | No | Myeloid, lung, GI epithelial cells | GM-CSF, IFN-γ, IL-2, IL-13, MIP1α, TNF-α | Activity in AML PDX models, no effects upon HSCs | Myeloablation | Laborda et al.,71 Wang et al.73 |
| Folate receptor-b | Folate-binding protein receptor | Yes | Yes | No | Myeloid cells | IFN-γ | Activity in AML cell line xenograft models, no effects upon HSCs | Myeloablation | Lynn et al.65 |
| IL1RAP | Accessory protein | Yes | Yes | Yes | Liver, GI tissues, GU tissues | n/a | n/a | Hepatic, GI, and GU toxicity | n/a |
| MUC1 | Glycoprotein | Yes | Yes | Minimal | Lymphatic tissues, lung epithelium, GI epithelium, GU tissues, placenta, skin | IFN-γ, IL-2, TNF-α | Activity in T-cell leukemia and solid tumor xenograft models | Multiorgan epithelial toxicity | Posey et al.77 |
| NKG2D / NKG2DL | C-type lectin-like receptor protein | Yes | Possibly | No | Treg cells, endothelial cells, myeloid-derived suppressor cells | IFN-γ, TNF-α | Activity in MM and solid tumor xenograft models | Lymphodepletion, myeloablation, inflammatory responses | Barber et al.78–80 |
| TIM-3 (HAVCR2) | Immunoglobulin superfamily protein | Yes | Yes | No | T cells, lung tissue | n/a | n/a | Lymphodepletion, pulmonary toxicity | n/a |
| WT1 | Zinc finger DNA-binding protein | Yes | Yes | Minimal | Kidney fallopian tube, endometrium and testis | IFN-γ, IL-2, IL-12 | Activity in AML and ovarian carcinoma xenograft models | Renal/GU toxicity | Rafiq et al.81 |
Some HSC antigen expression data obtained from Yoriko Saito et al.82
AML, acute myeloid leukemia; GI, gastrointestinal; GM-CSF, granulocyte macrophage colony stimulating factor; GU, genitourinary; HSC, hematopoietic stem cell; LIC, leukemia-initiating cell; IFN-γ, interferon γ; IL, interleukin; MIP1, macrophage inhibitory protein 1 (α or β); MM, multiple myeloma; MP, myeloid progenitor cells; n/a, not available; NK, natural killer; PDX, patient-derived xenograft model; TNF-α, tumor necrosis factor α.
However, several groups have also reported potent in vivo hematologic toxicity due to T-cell persistence in AML PDX models,49,50,61,76 highlighting potential necessity of T-cell termination strategies to decrease toxicity to normal tissues. Such approaches include incorporation of suicide genes in the CAR construct for subsequent chemical- or antibody-mediated T-cell termination, CAR T-cell depletion with monoclonal antibodies directed against T-cell surface proteins, and mRNA CAR-electroporated T cells that last for only a few days in vivo.36 Each of these strategies has particular advantages and disadvantages. The suicide switches herpes simplex virus thymidine kinase (HSV-tk) and inducible caspase 9 (iC9) can induce rapid, but potentially incomplete, CAR T-cell death following administration of the antiviral medication ganciclovir or chemically inert inducer of dimerization (CID) prodrugs, respectively. The HSV-tk strategy was initially developed to control T-cell alloreactivity in graft-versus-host disease following HSCT83 and has been integrated into some preclinical CAR T-cell studies,49 but has proven to be fairly immunogenic in patients. Conversely, the iC9 approach with administration of the CID rimiducid (AP1903) is minimally immunogenic and does not depend upon cell division to induce apoptosis, which may be more clinically desirable.66,84,85 Several groups have also incorporated coexpression of synthetic surface proteins [e.g. truncated epidermal growth factor receptor (tEGFR), CD20] in CAR T cells and demonstrated potent T-cell depletion following administration of specific monoclonal antibodies, such as anti-EGFR cetuximab or anti-CD20 rituximab. Administration of the anti-CD52 antibody alemtuzumab has also been used to eliminate CAR T cells via targeting of endogenously occurring cell surface CD52.47,51 Use of ‘biodegradable’ RNA CAR T cells is another attractive approach given their self-limited in vivo lifespan and thus lower risk of inducing sustained off tumor toxicity.51,55,86 However, RNA CAR T cells also have limited expansion potential that is likely required for sustained leukemia cure, and administration of multiple cell infusions is likely necessary to achieve initial remission. Based on promising preclinical data, the tEGFR/cetuximab, CD20/rituximab, alemtuzumab, and iC9/AP1903 approaches have been incorporated into clinical CAR T-cell products under current investigation in adults with relapsed/refractory acute leukemias,18,20,87,88 although actual depletion of CAR T cells in patients with leukemia via these methods has not been reported.
Phase I clinical trial testing of AML CAR T-cell immunotherapy
Despite the recent explosion of preclinical studies demonstrating exciting cytotoxic potential of various AML antigen-targeting CAR T cells, translation to the clinic has been much slower and highlights inherent challenges and risks in developing these treatment strategies for patients with AML. To date, a small number of adults have been treated with AML CAR T-cell immunotherapies via early phase clinical trials (Table 2).
Table 2.
Current phase I clinical trials of CAR T-cell immunotherapy for patients with relapsed/refractory AML.
| AML target antigen | ClinicalTrials.gov identifier | Vector | Costimulatory domains | T-cell source | Age eligibility | Site of conduction | Trial results |
|---|---|---|---|---|---|---|---|
| CD33 | NCT03126864 | Lentivirus | Not listed | Autologous | 1–80 years | MDACC, USA | Not yet available |
| NCT01864902 | Not listed | 4–1BB/CD3ζ, CD3ζ only | Autologous | 5–90 years | PLA Hospital, Beijing, China | One patient treated: PR, then PD (Wang et al.89) | |
| NCT02799680 | Not listed | Not listed | Allogeneic | ⩾50 years | AHCAMMS, Beijing, China | Not yet available | |
| CD38 | NCT03222674 | Not listed | Not listed | Not listed | 6 months–65 years | Geno-Immune Medical Institute, Shenzhen, China | Not yet available |
| CD56 | NCT03222674 | Not listed | Not listed | Not listed | 6 months–65 years | Geno-Immune Medical Institute, Shenzhen, China | Not yet available |
| CD117 | NCT03222674 | Not listed | Not listed | Not listed | 6 months–65 years | Geno-Immune Medical Institute, Shenzhen, China | Not yet available |
| CD123 | NCT02159495 | Lentivirus | CD28/CD3ζ/tEGFR | Autologous or allogeneic | ⩾18 years | City of Hope, USA | 5 patients treated: 5 PD, tolerable CRS (Budde et al.87) |
| NCT02623582 | Electroporated RNA | TCR/4-1BB | Autologous | ⩾18 years | University of Pennsylvania, USA | 6 patients treated: 3 CR, 2 PR, 1 PD (Cummins et al.90) | |
| NCT03114670 | Not listed | 4-1BB/CD3ζ/tEGFR | Allogeneic (donor-derived) | ⩾18 years | AHCAMMS, Beijing, China | Not yet available | |
| NCT03190278 | Lentivirus | Not listed | Gene-edited allogeneic | 18–75 years | Cornell and MDACC, USA | ||
| Lewis-Y | NCT01716364 | Retrovirus | CD28/CD3ζ | Autologou s |
⩾18 years | Peter MacCallum Cancer Centre, Australia | 4 patients treated: 4 PR/SD, then PD (Ritchie et al.91) |
| Muc-1 | NCT03222674 | Not listed | Not listed | Not listed | 6 months–65 years | Geno-Immune Medical Institute, Shenzhen, China | Not yet available |
| NKG2DL | NCT02203825 | Not listed | Not listed | Not listed | ⩾18 years | DFCI, USA | 9 patients treated, no DLTs identified (Nikiforow et al.92) |
| NCT03018405 | Not listed | Not listed | Not listed | ⩾18 years | Belgium and USA | Not yet available (Verma et al.93, Lonez et al.94) |
Data were assimilated from publicly available information on www.clinicaltrials.gov.
AHCAMMS, Affiliated Hospital of the Chinese Academy of Military Medical Sciences; AML, acute myeloid leukemia; CAR, chimeric antigen receptor; CR, complete response; CRS, cytokine release syndrome; DFCI, Dana-Farber Cancer Institute; DLT, dose-limiting toxicity; MDACC, MD Anderson Cancer Center; PD, progressive disease; PLA, People’s Liberation Army General Hospital; PR, partial response; TCR, T-cell receptor; tEGFR, truncated epidermal growth factor receptor.
A phase I study conducted at the Peter MacCallum Cancer Centre in Melbourne [ClinicalTrials.gov identifier: NCT01716364] treated four adult patients with AML with a single dose of radioactively labeled second-generation CAR T cells redirected against Lewis-Y (LeY), a carbohydrate antigen overexpressed on many cancer types, including AML.91 Three patients had minimal residual disease (MRD)-level marrow involvement at study entry, while the fourth patient had active AML with 70% marrow involvement at the time of T-cell infusion with up to 1 × 109 cells at 14–38% transduction efficiency. Single photon emission computed tomography (SPECT) imaging of infused patients and quantitative polymerase chain reaction (PCR) monitoring of peripheral blood transgene levels demonstrated initial pulmonary trafficking of T cells with subsequent localization to spleen and bone marrow. While transient responses (range 1–23 months) with stable disease or leukemia reduction were observed in the four patients, all eventually relapsed. Importantly, this initial trial provided first clues about cytokine release syndrome (CRS) as a potential biomarker of CAR T-cell-induced responses, as the patient with high-level marrow involvement treated with anti-LeY cells experienced pyrexia, rigors, and skin rash exacerbation with biopsy-proven lymphocytic infiltrates at approximately 1 week post T-cell infusion. These studies also demonstrated PCR-detectable persistence of CAR T cells in vivo lasting several months.91
A current phase I trial in Beijing [ClinicalTrials.gov identifier: NCT01864902] is assessing the safety of second-generation CD33-redirected CAR T cells in patients with relapsed/refractory AML. The investigators reported their first treated patient, a 41-year-old man with multiply relapsed AML, who received an infusion of 1 × 109 CD33 CAR T cells (38% transduction efficiency) and experienced a partial response with marked reduction in marrow blasts prior to leukemia progression at 9 weeks from T-cell infusion. This patient also experienced clinically significant CRS with fever and elevated inflammatory cytokine levels. Importantly, the patient did not appear to have clinical or laboratory signs of sinusoidal obstruction syndrome/veno-occlusive disease, a potential on target/off tumor toxicity given CD33 expression on hepatic Kupffer cells.89 This trial remains open for patient accrual.
Several additional phase I trials of CAR T cells for patients with AML are ongoing across the globe (Table 2). The MD Anderson Cancer Center (MDACC) is conducting a study of CD33-redirected CAR T cells and will allow pediatric patient participation if a safe and tolerable dose of T cells is first defined in adults with relapsed/refractory AML [ClinicalTrials.gov identifier: NCT03126864]. A separate Pediatric Bone Marrow Transplantation Consortium-sponsored CD33 CAR T-cell trial specifically for children, adolescents, and young adults with relapsed/refractory AML is also in development at the National Cancer Institute and Children’s Hospital of Philadelphia and anticipated to open in 2018.
Various centers in the United States [City of Hope Comprehensive Cancer Center, University of Pennsylvania (Penn) Abramson Cancer Center, Weill Cornell Medicine, MDACC] and in Beijing are conducting phase I trials of RNA-modified or lentivirally transduced CD123 CAR T cells. In the Penn trial [ClinicalTrials.gov identifier: NCT02623582], five adults with relapsed/refractory AML were treated with lymphodepleting chemotherapy followed by one to three doses of short-persisting RNA-electroporated CD123 CAR-modified autologous T cells administered every 48–72 h in a pilot trial to study manufacturing feasibility, safety, and potential myelotoxicity.90 In this study, less than 60% of planned T-cell doses were able to be manufactured successfully, and all treated patients had leukemia progression within a month of cell infusion. T-cell persistence (as measured by flow cytometry and PCR) was expectedly undetectable given the very short half life of these biodegradable mRNA-transfected cells. However, all patients experienced fever or CRS necessitating tocilizumab administration for toxicity management. In addition, no obvious hematologic, neurologic, or vascular toxicity was observed given CD123 expression in normal tissues.90 Given the lack of anti-leukemia efficacy, but also minimal on target/off tumor toxicity, this trial was terminated early with plans for development of a subsequent lentiviral CD123 CAR T-cell trial. The City of Hope group also recently reported early results from its phase I CD123 CAR T-cell trial [ClinicalTrials.gov identifier: NCT02159495].87 Six adults with relapsed AML after HSCT were treated with 50 × 106 (dose level 1, DL1) or 100 × 106 (DL2) lentivirally transduced CAR+ CD28/CD3ζ autologous T cells following lymphodepleting chemotherapy. Retreatment with a second T-cell infusion was permitted for eligible patients. One of two DL1 patients had transient morphologic remission and then responded to a second cell dose upon AML reprogression, ultimately achieving an MRD-level disease response. Two of four DL2 patients had complete responses (CRs) following CD123 CAR T-cell treatment and were able to undergo a second HSCT. The other two patients treated at DL2 had partial responses. Grade 2 or less CRS was observed in most patients, and no dose-limiting toxicity, including myeloablation, has been observed to date. A patient with a refractory blastic plasmacytoid dendritic cell neoplasm (BPDCN) treated with these CD123 CAR T cells also experienced a CR.87 This trial is currently enrolling.
Additional phase I trials of CAR T cells for adults with relapsed/refractory AML include those targeting the transmembrane NKG2D C-type lectin-like receptor or its cell surface ligand (NKG2DL) at Dana-Farber Cancer Institute [ClinicalTrials.gov identifier: NCT02203825] or at several US and European sites [ClinicalTrials.gov identifier: NCT03018405].92–94 A phase I trial in Shenzhen, Guandong, China is testing the safety of CD38, CD56, CD117, or Muc-1 CAR T cells in children and adults with relapsed/refractory AML [ClinicalTrials.gov identifier: NCT03222674]. No data are yet publically available from these trials (Table 2).
Conclusion
The remarkable cure rates achieved in patients with multiply relapsed B-ALL treated with antigen-redirected CAR T cells has had transformative scientific and clinical impact and recently led to expedited FDA approval of two CD19 CAR T- cell products, tisagenlecleucel and axicabtagene ciloleucel. While similar successes are certainly equally desired for patients with AML treated with immunotherapy, issues of myeloid antigen selection for CAR targeting and the delicate balance of on target/on tumor and on target/off tumor toxicity remain significant unsolved challenges. Moderate to longer-term persistence of AML CAR T cells likely required for achievement of deep molecular remissions is potentially incompatible with maintenance of normal hematopoiesis. The sequela of CAR T-cell-induced myeloablation may be rescued with allogeneic HSCT, but steps must be taken to ensure that engineered T cells do not attack incoming donor cells and induce graft rejection.
Strategies to optimize AML CAR T-cell activity and reduce potential resistance are critically important for successful actualization of these new immunotherapies. In addition to the aforementioned bispecific antigen-targeting approaches to maximize AML specificity, use of genome-editing technologies, such as clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR associated protein 9 (Cas9) and transcription activator-like effector nucleases (TALEN), or checkpoint inhibitors may further fine-tune AML CAR T-cell precision, potency, and persistence.28,95–97 Combining CAR T cells with relevant kinase inhibitors may also decrease potential immunotherapeutic resistance leading to leukemia relapse. This concept has been recently successfully demonstrated in preclinical studies of CD19 CAR T cells and ibrutinib in ALL, FLT3 CAR T cells with the FLT3 inhibitor crenolanib in AML, and CD33 or CD19 CAR T cells with the PI3K inhibitor LY294002 in AML and ALL, respectively.59,70,98 Finally, successful engineering and treatment of patients with allogeneic donor-derived99 or universal CAR T cells 62,88 without appreciable graft-versus-host or host-versus-graft reactivity may minimize typical delays often occurring between autologous T-cell pheresis to manufacture to infusion, as well as more readily increase therapy availability to a larger patient population in need.
As nascent bench-to-bedside translation of cellular immunotherapies for patients with relapsed/refractory AML is beginning to occur, biologic questions persist that may only be answerable with further clinical experience and careful monitoring of patients: (1) What are optimal AML-associated surface antigens (or antigen pairings) for targeting with CAR T cells and other immunotherapies? (2) Is there a therapeutic window for AML CAR T cells to maximize leukemia remission and minimize hematologic and non-hematologic toxicity? (3) Will hematopoietic stem cell transplantation be necessary to consolidate potential CAR T-cell-induced remissions or to rescue patients from on target/off tumor myeloablation? (4) Should AML CAR T cells be terminated via suicide switches or antibody depletion, particularly prior to HSCT, minimize allogeneic graft rejection? (5) Can HSCT donor-derived or ‘off the shelf’ allogeneic T cells successfully induce deep leukemia remission with minimal or tolerable graft-versus-host and host-versus-graft effects? (6) Will combining AML CAR T cells with checkpoint inhibitors or kinase inhibitors increase anti-leukemia efficacy or decrease immunotherapeutic resistance? (7) Can gene-editing strategies such as CRISPR/Cas9 and TALEN be used for further optimization of AML CAR T-cell specificity, persistence, toxicity, and availability to patients?
Acknowledgments
SKT wrote the manuscript.
Footnotes
Funding: This work was supported by a National Cancer Institute 1K08CA184418 career development award, the Rally Foundation for Childhood Cancer Research, the Gabrielle’s Angel Foundation for Cancer Research, and a St Baldrick’s Foundation/Stand Up to Cancer Pediatric Dream Team Translational Research Grant (SU2C-AACR-DT2727). Stand Up To Cancer is a program of the Entertainment Industry Foundation administered by the American Association for Cancer Research.
Conflict of interest statement: SKT has no relevant financial conflicts of interest.
ORCID iD: Sarah K Tasian
https://orcid.org/0000-0003-1327-1662
References
- 1. Döhner H, Estey EH, Amadori S, et al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European leukemianet. Blood 2010; 115: 453–474. [DOI] [PubMed] [Google Scholar]
- 2. Rowe JM, Tallman MS. How I treat acute myeloid leukemia. Blood 2010; 116: 3147–3156. [DOI] [PubMed] [Google Scholar]
- 3. Pui CH, Carroll WL, Meshinchi S, et al. Biology, risk stratification, and therapy of pediatric acute leukemias: an update. J Clin Oncol 2011; 29: 551–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Creutzig U, van den Heuvel-Eibrink MM, Gibson B, et al. Diagnosis and management of acute myeloid leukemia in children and adolescents: recommendations from an international expert panel. Blood 2012; 120: 3187–3205. [DOI] [PubMed] [Google Scholar]
- 5. Rubnitz JE. How I treat pediatric acute myeloid leukemia. Blood 2012; 119: 5980–5988. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Larson RA, Sievers EL, Stadtmauer EA, et al. Final report of the efficacy and safety of gemtuzumab ozogamicin (Mylotarg) in patients with CD33-positive acute myeloid leukemia in first recurrence. Cancer 2005; 104: 1442–1452. [DOI] [PubMed] [Google Scholar]
- 7. 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: 3021–3032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. 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: 986–996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Kung Sutherland MS, Walter RB, Jeffrey SC, et al. SGN-CD33A: a novel CD33-targeting antibody-drug conjugate using a pyrrolobenzodiazepine dimer is active in models of drug-resistant AML. Blood 2013; 122: 1455–1463. [DOI] [PubMed] [Google Scholar]
- 10. Geiger TL, Rubnitz JE. New approaches for the immunotherapy of acute myeloid leukemia. Discov Med 2015; 19: 275–284. [PMC free article] [PubMed] [Google Scholar]
- 11. Al-Hussaini M, Rettig MP, Ritchey JK, et al. Targeting CD123 in acute myeloid leukemia using a T-cell-directed dual-affinity retargeting platform. Blood 2016; 127: 122–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Lichtenegger FS, Krupka C, Haubner S, et al. Recent developments in immunotherapy of acute myeloid leukemia. J Hematol Oncol 2017; 10: 142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Uy GL, Godwin J, Rettig MP, et al. Preliminary results of a phase 1 study of flotetuzumab, a CD123 X CD3 bispecific dart protein, in patients with relapsed/refractory acute myeloid leukemia and myelodysplastic syndrome. Blood 2017; 130: 637. [Google Scholar]
- 14. Li F, Sutherland MK, Yu C, et al. Characterization of SGN-CD123A, a potent CD123-directed antibody-drug conjugate for acute myeloid leukemia. Mol Cancer Ther 2018; 17: 554–564. [DOI] [PubMed] [Google Scholar]
- 15. Brentjens RJ, Davila ML, Riviere I, et al. CD19-targeted T cells rapidly induce molecular remissions in adults with chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 2013; 5: 177ra138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. 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: 517–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014; 371: 1507–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Turtle CJ, Hanafi LA, Berger C, et al. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients. J Clin Invest 2016; 126: 2123–2138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. 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: 20–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. 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: 3322–3331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. 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: 439–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Park JH, Riviere I, Gonen M, et al. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 2018; 378: 449–459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Fry TJ, Mackall CL. T-cell adoptive immunotherapy for acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program 2013; 2013: 348–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Tasian SK, Gardner RA. CD19-redirected chimeric antigen receptor-modified T cells: a promising immunotherapy for children and adults with b-cell acute lymphoblastic leukemia (ALL). Ther Adv Hematol 2015; 6: 228–241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Maus MV, June CH. Making better chimeric antigen receptors for adoptive T-cell therapy. Clin Cancer Res 2016; 22: 1875–1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Jain MD, Davila ML. Concise review: emerging principles from the clinical application of chimeric antigen receptor T cell therapies for B cell malignancies. Stem Cells 2018; 36: 36–44. [DOI] [PubMed] [Google Scholar]
- 27. Majzner RG, Heitzeneder S, Mackall CL. Harnessing the immunotherapy revolution for the treatment of childhood cancers. Cancer Cell 2017; 31: 476–485. [DOI] [PubMed] [Google Scholar]
- 28. Perales MA, Kebriaei P, Kean LS, et al. Building a safer and faster CAR: seatbelts, airbags, and CRISPR. Biol Blood Marrow Transplant 2018; 24: 27–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Bonifant CL, Jackson HJ, Brentjens RJ, et al. Toxicity and management in CAR T-cell therapy. Mol Ther Oncolytics 2016; 3: 16011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition and management. Blood 2016; 127: 3321–3330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Frey NV, Porter DL. Cytokine release syndrome with novel therapeutics for acute lymphoblastic leukemia. Hematology Am Soc Hematol Educ Program 2016; 2016: 567–572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Teachey DT, Lacey SF, Shaw PA, et al. Identification of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-cell therapy for acute lymphoblastic leukemia. Cancer Discov 2016; 6: 664–679. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy – assessment and management of toxicities. Nat Rev Clin Oncol 2018; 15: 47–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Morgan RA, Yang JC, Kitano M, et al. 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] [PMC free article] [PubMed] [Google Scholar]
- 35. Bonifant CL, Szoor A, Torres D, et al. CD123-engager T cells as a novel immunotherapeutic for acute myeloid leukemia. Mol Ther 2016; 24: 1615–1626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Gross G, Eshhar Z. Therapeutic potential of T cell chimeric antigen receptors (CARS) in cancer treatment: counteracting off-tumor toxicities for safe CAR T cell therapy. Annu Rev Pharmacol Toxicol 2016; 56: 59–83. [DOI] [PubMed] [Google Scholar]
- 37. 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: 581–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART-19 immunotherapy. Cancer Discov 2015; 5: 1282–1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Gardner R, Wu D, Cherian S, et al. Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood 2016; 127: 2406–2410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Ruella M, Kenderian SS, Shestova O, et al. The addition of the BTK inhibitor ibrutinib to anti-CD19 chimeric antigen receptor T cells (CART19) improves responses against mantle cell lymphoma. Clin Cancer Res 2016; 22: 2684–2696. [DOI] [PubMed] [Google Scholar]
- 42. Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2014; 6: 224ra225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Doan A, Pulsipher MA. Hypogammaglobulinemia due to CAR T-cell therapy. Pediatr Blood Cancer 2018; 65. DOI: 10.1002/pbc.26914 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Kantarjian HM, Deangelo DJ, Stelljes M, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med 2016; 375: 740–753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Bhojwani D, Sposto R, Shah N, et al. Inotuzumab ozogamicin in pediatric patients with relapsed/refractory acute lymphoblastic leukemia (R/R ALL). J Clin Oncol 2017; 35: 10512–10512. [Google Scholar]
- 46. Perna F, Berman SH, Soni RK, et al. Integrating proteomics and transcriptomics for systematic combinatorial chimeric antigen receptor therapy of AML. Cancer Cell 2017; 32: 506–519.e505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Petrov JC, Wada M, Pinz KG, et al. Compound CAR T-cells as a double-pronged approach for treating acute myeloid leukemia. Leukemia 2018. DOI: 10.1038/s41375-018-0075-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Qin H, Nguyen SM, Ramakrishna S, et al. Novel CD19/CD22 bicistronic chimeric antigen receptors outperform single or bivalent cars in eradicating CD19+CD22+, CD19–, and CD22– Pre-B leukemia. Blood 2017; 130: 810. [Google Scholar]
- 49. Casucci M, Nicolis di, Robilant B, Falcone L, et al. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood 2013; 122: 3461–3472. [DOI] [PubMed] [Google Scholar]
- 50. 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: 3138–3148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. 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: 2395–2407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Peinert S, Prince HM, Guru PM, et al. Gene-modified T cells as immunotherapy for multiple myeloma and acute myeloid leukemia expressing the Lewis Y antigen. Gene Ther 2010; 17: 678–686. [DOI] [PubMed] [Google Scholar]
- 53. Dutour A, Marin V, Pizzitola I, 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] [PMC free article] [PubMed] [Google Scholar]
- 54. Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, et al. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia 2014; 28: 1596–1605. [DOI] [PubMed] [Google Scholar]
- 55. 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: 1637–1647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. O’hear C, Heiber JF, Schubert I, et al. Anti-CD33 chimeric antigen receptor targeting of acute myeloid leukemia. Haematologica 2015; 100: 336–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Cartellieri M, Feldmann A, Koristka S, et al. Switching car T cells on and off: a novel modular platform for retargeting of T cells to AML blasts. Blood Cancer J 2016; 6: e458. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Li S, Tao Z, Xu Y, et al. CD33 specific chimeric antigen receptor T cells with different costimulators showed potent anti-leukemia efficacy and different phenotype. Hum Gene Ther 2018; 29(5): 626–639. DOI: 10.1089/hum.2017.241. [DOI] [PubMed] [Google Scholar]
- 59. Zheng W, O’Hear CE, Alli R, et al. PI3K Orchestration of the in vivo persistence of chimeric antigen receptor-modified T cells. Leukemia 2018. DOI: 10.1038/s41375-017-0008-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tettamanti S, Marin V, Pizzitola I, et al. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor. Br J Haematol 2013; 161: 389–401. [DOI] [PubMed] [Google Scholar]
- 61. 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: 2343–2354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Guzman ML, Sugita M, Zong H, et al. Allogeneic TCRα/β deficient CAR T-cells targeting CD123 prolong overall survival of AML patient-derived xenografts. Blood 2016; 128: 765–765. [Google Scholar]
- 63. Zhou L, Liu X, Wang X, et al. CD123 redirected multiple virus-specific T cells for acute myeloid leukemia. Leuk Res 2016; 41: 76–84. [DOI] [PubMed] [Google Scholar]
- 64. Arcangeli S, Rotiroti MC, Bardelli M, et al. Balance of anti-CD123 chimeric antigen receptor binding affinity and density for the targeting of acute myeloid leukemia. Mol Ther 2017; 25: 1933–1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Lynn RC, Poussin M, Kalota A, et al. Targeting of folate receptor beta on acute myeloid leukemia blasts with chimeric antigen receptor-expressing T cells. Blood 2015; 125: 3466–3476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Drent E, Groen RW, Noort WA, et al. Pre-clinical evaluation of CD38 chimeric antigen receptor engineered T cells for the treatment of multiple myeloma. Haematologica 2016; 101: 616–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Yoshida T, Mihara K, Takei Y, et al. All-trans retinoic acid enhances cytotoxic effect of T cells with an anti-CD38 chimeric antigen receptor in acute myeloid leukemia. Clin Transl Immunology 2016; 5: e116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Chien CD, Sauter CT, Ishii K, et al. Preclinical development of FLT3-redirected chimeric antigen receptor T cell immunotherapy for acute myeloid leukemia. Blood 2016; 128: 1072. [Google Scholar]
- 69. Chen L, Mao H, Zhang J, et al. Targeting FLT3 by chimeric antigen receptor T cells for the treatment of acute myeloid leukemia. Leukemia 2017; 31: 1830–1834. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Jetani H, Garcia-Cadenas I, Nerreter T, et al. CAR T-cells targeting FLT3 have potent activity against FLT3(–)ITD(+) AML and act synergistically with the FLT3-inhibitor crenolanib. Leukemia 2018. DOI: 10.1038/s41375-018-0009-0 [DOI] [PubMed] [Google Scholar]
- 71. Laborda E, Mazagova M, Shao S, et al. Development of a chimeric antigen receptor targeting C-type lectin-like molecule-1 for human acute myeloid leukemia. Int J Mol Sci 2017; 18 pii: E2259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Tashiro H, Sauer T, Shum T, et al. Treatment of acute myeloid leukemia with T cells expressing chimeric antigen receptors directed to C-type lectin-like molecule 1. Mol Ther 2017; 25: 2202–2213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Wang J, Chen S, Xiao W, et al. Car-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J Hematol Oncol 2018; 11: 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Silva Gomes, et al. 2016 http://www.bloodjournal.org/content/128/22/4555
- 75. Silva Gomes, et al. 2017 http://www.bloodjournal.org/content/130/3/285 [Google Scholar]
- 76. Sauter CT, Chien CD, Shen F, et al. Evaluating on-target toxicity of hematopoietic-targeting Cars demonstrates target-nonspecific suppression of marrow progenitors. Blood 2016; 128: 3357. [Google Scholar]
- 77. Posey, et al. Engineered CAR T Cells Targeting the Cancer-Associated Tn-Glycoform of the Membrane Mucin MUC1 Control Adenocarcinoma. 2016; 44(6):1444–54. DOI: 10.1016/j.immuni.2016.05.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Barber, et al. Chimeric NKG2D receptor-bearing T cells as immunotherapy for ovarian cancer. 2007; 15;67(10):5003–8. [DOI] [PubMed] [Google Scholar]
- 79. Barber, et al. Chimeric NKG2D receptor-expressing T cells as an immunotherapy for multiple myeloma. 2008; 36(10):1318–28. DOI: 10.1016/j.exphem.2008.04.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Barber, et al. NKG2D receptor regulates human effector T-cell cytokine production. 2011; 117(24): 6571–81. DOI: 10.1182/blood-2011-01-329417 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Rafiq, et al. Optimized T-cell receptor-mimic chimeric antigen receptor T cells directed toward the intracellular Wilms Tumor 1 antigen. 2017, https://www.ncbi.nlm.nih.gov/m/pubmed/27924074/ [DOI] [PMC free article] [PubMed]
- 82. Saito, et al. Identification of therapeutic targets for quiescent, chemotherapy-resistant human leukemia stem cells. 2010; 3;2(17):17ra9. DOI: 10.1126/scitranslmed.3000349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Bonini C, Ferrari G, Verzeletti S, et al. HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia. Science 1997; 276: 1719–1724. [DOI] [PubMed] [Google Scholar]
- 84. Di Stasi A, Tey SK, Dotti G, et al. Inducible apoptosis as a safety switch for adoptive cell therapy. N Engl J Med 2011; 365: 1673–1683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Minagawa K, Jamil MO, Al-Obaidi M, et al. In vitro pre-clinical validation of suicide gene modified anti-CD33 redirected chimeric antigen receptor T-cells for acute myeloid leukemia. PLoS One 2016; 11: e0166891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Barrett DM, Liu X, Jiang S, et al. Regimen-specific effects of RNA-modified chimeric antigen receptor T cells in mice with advanced leukemia. Hum Gene Ther 2013; 24: 717–727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Budde L, Song JY, Kim Y, et al. Remissions of acute myeloid leukemia and blastic plasmacytoid dendritic cell neoplasm following treatment with CD123-specific CAR T cells: a first-in-human clinical trial. Blood 2017; 130: abstract 811. [Google Scholar]
- 88. Qasim W, Zhan H, Samarasinghe S, et al. Molecular remission of infant B-ALL after infusion of universal TALEN gene-edited CAR T cells. Sci Transl Med 2017; 9. [DOI] [PubMed] [Google Scholar]
- 89. Wang QS, Wang Y, Lv HY, 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] [PMC free article] [PubMed] [Google Scholar]
- 90. Cummins KD, Frey N, Nelson AM, et al. Treating Relapsed / Refractory (RR) AML with biodegradable Anti-CD123 CAR modified T cells. Blood 2017; 130: abstract 1359. [Google Scholar]
- 91. Ritchie DS, Neeson PJ, Khot A, 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] [PMC free article] [PubMed] [Google Scholar]
- 92. Nikiforow S, Murad J, Daley H, et al. A first-in-human phase I trial of NKG2D chimeric antigen receptor-T cells in AML/MDS and multiple myeloma. J Clin Oncol 2016; 34: TPS3102-TPS3102. [Google Scholar]
- 93. Verma B, Aftimos PG, Awada A, et al. A NKG2D-based CAR-T therapy in a multinational phase I dose escalation and expansion study targeting multiple solid and hematologic tumor types. J Clin Oncol 2017; 35: TPS3093–TPS3093. [Google Scholar]
- 94. Lonez C, Verma B, Hendlisz A, et al. Study protocol for THINK: a multinational open-label phase I study to assess the safety and clinical activity of multiple administrations of NKR-2 in patients with different metastatic tumour types. BMJ Open 2017; 7: e017075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Poirot L, Philip B, Schiffer-Mannioui C, et al. Multiplex genome-edited T-cell manufacturing platform for ‘off-the-shelf’ adoptive T-cell immunotherapies. Cancer Res 2015; 75: 3853–3864. [DOI] [PubMed] [Google Scholar]
- 96. Georgiadis C, Qasim W. Emerging applications of gene edited T cells for the treatment of leukemia. Expert Rev Hematol 2017; 10: 753–755. [DOI] [PubMed] [Google Scholar]
- 97. Singh N, Shi J, June CH, et al. Genome-editing technologies in adoptive T cell immunotherapy for cancer. Curr Hematol Malig Rep 2017; 12: 522–529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Ruella M, Maus MV. Catch me if you can: leukemia escape after CD19-directed T cell immunotherapies. Comput Struct Biotechnol J 2016; 14: 357–362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Yang Y, Jacoby E, Fry TJ. Challenges and opportunities of allogeneic donor-derived CAR T cells. Curr Opin Hematol 2015; 22: 509–515. [DOI] [PMC free article] [PubMed] [Google Scholar]

