Abstract
Recurrent and/or refractory (R/R) pediatric acute myeloid leukemia (AML) remains a recalcitrant disease with poor outcomes. Cell therapy with genetically modified immune effector cells holds the promise to improve outcomes for R/R AML since it relies on cytotoxic mechanisms that are distinct from chemotherapeutic agents. While T cells expressing chimeric antigen receptors (CAR T cells) showed significant anti-AML activity in preclinical models, early phase clinical studies have demonstrated limited activity, irrespective of the targeted AML antigen. Lack of efficacy is most likely multifactorial, including: (i) a limited array of AML-specific targets and target antigen heterogeneity; (ii) the aggressive nature of R/R AML and heavy pretreatment of patients; (iii) T-cell product manufacturing, and (iv) limited expansion and persistence of the CAR T cells, which is in part driven by the immunosuppressive AML microenvironment. Here we review the results of early phase clinical studies with AML-specific CAR T cells, and avenues investigators are exploring to improve their effector function.
Introduction
The overall survival of pediatric patients with newly diagnosed acute myeloid leukemia. (AML) has improved over the last two decades with rates reported to be close to 70%.1-4 However, the event-free survival of these patients has plateaued at about 55% despite the use of intensive therapies.1-3 Furthermore, the outcome of children with relapsed and/or refractory (R/R) disease remains poor. Recent data from the Berlin Frankfurt Münster (BFM) group and Children’s Oncology Group (COG) showed that the overall survival rates of pediatric patients with first relapse of AML were 42% and 35%, respectively.4 For patients with second relapse of AML, the overall survival at 5 years remains dismal (15 ± 4%) based on BFM and COG data.5 Similar outcomes were reported in a recent analysis by a Nordic-Dutch-Belgian-Spain-Hong-Kong-Israel-Portugal (NOPHO-DB-SHIP) consortium.6
This high rate of treatment failure has been attributed in part to the existence of chemotherapy-resistant leukemic stem cells, a minor fraction of leukemic cells that are capable of maintaining and re-initiating the disease.7, 8 While allogeneic hematopoietic stem cell transplant (HSCT) at present offers the highest probability of long-term sustained remission,3 its success relies on several factors with remission status prior to HSCT being one of the most important prognostic factors.9 However, it is challenging to re-induce remission in these patients with salvage chemotherapy due to toxicities and limited efficacy of standard or investigational therapies. Many patients are therefore unable to proceed to allogeneic HSCT.
Adoptive transfer of chimeric antigen receptor (CAR) T cells has emerged as a promising treatment option for pediatric patients with CD19-positive B-cell acute lymphoblastic leukemia.10,11 CD19-CAR T cells have shown potent anti-leukemia activity in the R/R setting, leading to their approval by the Food and Drug Administration in 2017.10-14 In contrast, the clinical experience with AML-specific CAR T cells is limited with information on several studies only reported in abstract form. Overall, the clinical activity of AML-specific CAR T cells, irrespective of the targeted antigen, has been underwhelming and has shown variable complete response (CR) rates.15-33 Importantly, early phase clinical and preclinical studies have highlighted several roadblocks (Figure 1), including: (i) a limited array of AML-specific targets and target antigen heterogeneity; (ii) the aggressive nature of R/R AML and heavy pretreatment of patients; (iii) T-cell product manufacturing; and (iv) limited expansion and persistence of the CAR T cells, which is in part driven by the immunosuppressive AML microenvironment.18,34-39
In this review we summarize published results of early phase clinical studies with AML-specific CAR T cells from centers around the globe, including the USA, Europe, Australia and China, presenting pediatric as well as adult studies. In the second part of the review, we then outline potential ways to overcome current roadblocks to effective CAR T-cell therapies for AML. This review is not meant to be comprehensive, and we refer the reader to other excellent reviews on CAR immune cell therapies for AML that have been published recently.36,40-42 Likewise, due to space limitations, we will not review combinatorial approaches in which CAR T cells are combined with other treatment modalities.
Immunotherapy targets in acute myeloid leukemia
An ideal antigen for CAR T-cell therapy is a cell surface antigen that is highly expressed in all malignant cells but not expressed in healthy tissues. As for many other malignancies, such an ideal target remains elusive for AML. To date, CD33, CD123, and CLL1 (CLEC12A) have been the most frequently targeted AML antigens.43-49 While these are overexpressed on the cell surface of AML blasts, they are also expressed on normal hematopoietic cells, raising concerns about myelotoxicity, with CD33 and CD123 being pan-myeloid antigens that are also expressed on hematopoietic stem cells and CLL1 being expressed on mature myeloid cells, including mature granulocytes. Nevertheless, the safety of targeting CD33 and CD123 has been shown in the pediatric clinical setting with the use of gemtuzumab, a CD33-specific monoclonal antibody drug conjugate,50,51 leading to its approval by the Food and Drug Administration, and flotetuzumab, a bispecific CD123xCD3 antibody.52 Early phase clinical trials of CAR T cells targeting CD123, CD33 and CLL1, as described in the next section, have used either safety switches or adopted a bridge-to-transplant strategy to overcome this issue. Targets that are expressed on the cell surface of AML blasts and not on normal myeloid cells are also being explored, including NKG2D, CD70, and GRP78.29,53-55 However, these are also associated with potential on-target/off-cancer toxicity, including being upregulated in normal cells under conditions of cellular stress (NKG2D, GRP78) or in subsets of activated immune cells (CD70). Other AML targets that have been or are being actively explored include CD38, CD44v6, CD117 (c-KIT) CD276 (B7-H3), CD327 (Siglec-6), FLT3, FRβ, GRP78, LILRB3, LILRB4, and TIM3.56-65 While these promise to have limited myelotoxicity, this aspect needs to be carefully evaluated in preclinical studies. For example, CD38-CAR T cells had potent anti-AML activity and prolonged survival in xenograft models, but limited hematopoietic stem cell toxicity was observed, consistent with the pattern of CD38 expression.56 Finally, a recent single-cell transcriptomic study of AML blasts discovered and validated CD86 and CSF-1R as AML-CAR T-cell therapy targets.35
Clinical experience with chimeric antigen receptor T cells for acute myeloid leukemia
In the following section we summarize results of clinical studies (Table 1) which have been either published or presented in abstract form. Data from the USA, UK, Europe, and China are included.
Lewis-Y-chimeric antigen receptor T cells
Lewis-Y (LeY) is an aberrantly glycosylated carbohydrate antigen that is overexpressed on AML blasts but has limited expression on normal tissues. While to our knowledge, LeY is no longer being pursued as an AML target, LeY-CAR T cells were the first CAR T-cell product that was evaluated in a phase I clinical study for AML (NCT03851146).15 Four adult patients with AML received LeY.CD28.ζ-CAR T cells. No significant toxicities were reported, but only transient responses or stable disease were observed. However, this study provided valuable information since CAR T cells were labeled with indium enabling tracking by single-photon emission computed tomography, which demonstrated CAR T-cell trafficking to bone marrow and at extramedullary disease sites, highlighting the potential benefit of using CAR T-cell therapy for AML.
CD123-chimeric antigen receptor T cells
Autologous CD123-chimeric antigen receptor T cells
Investigators evaluated T cells that expressed a CD123. CD28.ζ-CAR and a truncated EGFR safety switch which were infused after lymphodepletion induced by fludarabine/cyclophosphamide (NCT02159495). From this study, the outcome of six patients with AML, who were infused with CD123-CAR T cells at either dose level 1 (DL1: 5x107 CAR T cells) or dose level 2 (DL2: 2x108 CAR T cells), have been reported.16 Two patients achieved a CR and proceeded to allogeneic HSCT. Blood counts recovered in all patients following initial pancytopenia. Four patients showed either grade 1 or 2 cytokine release syndrome (CRS) but no dose-limiting toxicities were observed.
T cells that express CD123.CD28.ζ-CAR and a CD20 safety switch are actively being evaluated in a phase I clinical trial (NCT04318678) and preliminary results have been presented.17 Following lymphodepletion with fludarabine/ cyclophosphamide, two patients were infused at DL1 (3x105 CAR T cells/kg), and four patients at DL2 (3x105 CAR T cells/ kg), with one patient being infused off protocol. Isolated fevers occurred after infusion but resolved within 24 hours, and no dose-limiting toxicities were observed. At DL2, one patient with extramedullary disease achieved a CR that lasted for 2 months; she then received a second infusion and again achieved a short-lived CR. One additional patient was infused at DL2 off protocol and achieved a morphological CR but with detectable low-level minimal residual disease (MRD).
To overcome the potential concern regarding prolonged myelosuppression and vascular toxicity, investigators have evaluated T cells that were transfected with messenger RNA encoding a CD123.41BB.ζ-CAR, which results in transient CAR expression in adoptively transferred T cells (NCT02623582). Five patients received one to three serial infusions of CD123-CAR T cells following lymphodepleting chemotherapy. All five patients had grade ≥2 CRS, but the study was terminated as no patient had a clinical response. However, the lack of significant vascular toxicity prompted the development of a follow-up study utilizing a lentiviral vector to generate CD123-CAR T cells (NCT03766126).18,19 Another unique approach to overcome the risk of myelosuppression has been the use of switchable CAR. The universal CAR platform (UniCAR) is a two-component, second-generation CD28.ζ-based CAR T-cell platform in which an adapter molecule, targeting module (TM), confers specificity against the cancer antigen of choice.66 The targeting module has a short half-life of less than 30 minutes, enabling a rapid switch-off. Ten patients have been treated on a phase I trial with UniCAR-T cells directed at the CD123 antigen (NCT04230265).20,21 Following lymphodepletion, patients received continuous intravenous infusion of a CD123 targeting module (TM123; 0.5 to 1 mg/day) from days 0 to 24. CD123-directed UniCAR-T-cell therapy was safe and tolerable with limited toxicities. Encouraging anti-AML activity was observed, including one CR, two CR with incomplete hematologic recovery (CRi), and four partial responses (PR).
Allogeneic CD123-chimeric antigen receptor T cells
To overcome the concern about T-cell fitness and in an effort to enhance persistence, allogeneic CD123-CAR T cells have been evaluated, including an off-the-shelf, ‘universal’ CD123-CAR T-cell product, which is generated by transduction with a lentiviral vector encoding a CD123.41BB.ζ-CAR and gene edited with transcription activator-like effector nuclease to disrupt the T-cell receptor α constant gene locus to minimize risk of graft-versus-host disease and the CD52 locus to enable the use of alemtuzumab (anti-CD52) as part of the lymphodepletion regimen (NCT04106076).22 Sixteen patients received off-the-shelf CD123-CAR T-cell infusions with half undergoing lymphodepletion with fludarabine/cyclophosphamide and the other half receiving fludarabine/cyclophosphamide plus alemtuzumab. Four patients experienced dose-limiting toxicities including grade 4 CRS (n=2), grade 5 CRS (n=1), and grade 3 immune effector cell-associated neurotoxicity syndrome (ICANS) (n=1) irrespective of the lymphodepletion used. Low-grade (<3) CRS occurred in 12 patients. Adding alemtuzumab to fludarabine/cyclophosphamide was associated with improved lymphodepletion and significantly higher CAR T-cell expansion and persistence. Four patients had a response, including two cases of stable disease, one morphological leukemia-free state, and one durable MRD-negative CR. In addition to this study, a case report of a single patient given donor-derived CD123-CAR T cells as part of a reduced intensity conditioning regimen prior to HSCT has been published.67
CD33-chimeric antigen receptor T cells
Investigators evaluated the feasibility and safety of autologous T cells modified to express a CD33.41BB.ζ-CAR and truncated human epidermal growth factor receptor (HER1t) (NCT03126864).23 Ten adults with R/R AML were enrolled; T cells were collected by apheresis from eight, although only four T-cell products were successfully manufactured, and three patients were infused at the lowest dose level of 0.3x106 cells/kg. There were no dose-limiting toxicities; two patients had CRS (grades 2 and 3) and one had ICANS (grade 2). Although CD33-CAR T cells were detected in blood samples from patients following infusion, with associated symptoms and increased cytokine levels, no anti-leukemic responses were seen. One case report on the use of CD33.41BB.ζ-CAR T cells for R/R AML described transient clinical activity but also pancytopenia.24 Finally, there is an ongoing multicenter study, being conducted through the Pediatric Transplantation and Cellular Therapy Consortium, evaluating the safety and efficacy of autologous CD33.CD28.ζ-CAR T cells in pediatric patients with R/R AML (NCT03971799). Interim results from this study were presented at the American Society of Hematology annual meeting in 2023. Nineteen of the 24 patients enrolled were infused with CD33 CAR T cells. CRS was early in onset and was reported in 13 (68%) patients, being high grade (≥3) in four (21%) of these patients. Complete remission was seen at DL4, with two patients achieving MRD-negative CR. Dose escalation is currently underway in the phase II portion of the study.68
CLL1-chimeric antigen receptor T cells
CLL1.41BB.ζ-CAR T-cell products have been evaluated in a phase I clinical trial in eight pediatric patients with AML.25 The children were given 0.35-1x106 CAR T cells/kg as a single dose after fludarabine/cyclophosphamide lymphodepleting chemotherapy. All developed grade 1-2 CRS and pancytopenia following the infusion. Five patients achieved a morphological leukemia-free state, being MRD-negative in four cases and MRD-positive in one case, one patient had an MRD-positive CRi, one patient had a PR, and one patient has stable disease. In a second report, four pediatric patients received T cells expressing CLL1.CD28.CD27.ζ-CAR and an inducible caspase 9 suicide gene after lymphodepleting chemotherapy. Infusions were well tolerated with only low-grade CRS (≤2) and/or ICANS (≤2). Three patients achieved an MRD-negative CR.26 The investigators subsequently compared autologous T cells expressing CLL1.CD28. CD27.ζ-CAR and the inducible caspase 9 suicide gene (n=4) or CLL1.41BB.ζ-CAR (n=3) in seven children with R/R AML following lymphodepleting chemotherapy.27 The CAR T-cell infusions were well tolerated and only grade ≤2 CRS and one grade 2 ICANS were observed. Five patients achieved CR (3 negative and 2 positive for MRD) with a 1-year overall survival rate of 57.1%. Due to the small number of patients, no significant differences were noted between the two CAR T-cell products.
Finally, CLL1.41BB.ζ-CAR T cells have been evaluated in ten adults with R/R AML.28 Patients received fludarabine/cyclophosphamide lymphodepleting chemotherapy followed by an infusion of a single dose of 1-2 x106 CAR T cells/kg. All patients developed CRS, including grade 2 (n=4) and grade 3 (n=3) cases, and one patient developed T cell-related encephalopathy syndrome. Severe pancytopenia occurred in all patients, and two patients died of severe infection due to chronic agranulocytosis. The CR/CRi rate was 70% and, at a median follow-up time of 173 days, six patients were alive.
NKG2D ligand-chimeric antigen receptor T cells
NKG2D ligand (NKG2D-L)-positive tumor cells can be targeted with human leukocyte antigen (HLA) killer immunoglobulin-like receptor mismatched natural killer (NK) cells.69 Another approach relies on expressing a NKG2D-L-CAR which consists of NKG2D directly linked to the CD3ζ chain.70 NKG2D-L-CAR T cells were infused without prior lymphodepletion to alleviate concerns that chemotherapy might induce NKG2D-L expression on healthy tissues.29 Seven patients with NKG2D-L-positive R/R AML received NKG2D-L-CAR T cells in four escalating dose levels (1x106 to 3x107 total viable T cells). No dose-limiting toxicities were reported, but no objective responses were noted either. It is likely that the low cell dose without antecedent chemotherapy played a role in limited expansion and persistence of CAR T cells, resulting in limited anti-AML activity.
Building on these initial results and preclinical data supporting the use of multiple NKG2D-L-CAR T-cell infusions, a follow-up study was conducted (NCT03018405).30,31 This multicenter study enrolled 25 patients of whom 16 were infused at three dose levels, DL1: 3x108, DL2: 1x109, and DL3: 3x109 T cells per infusion. Patients received three infusions at 2-week intervals followed by potential consolidation treatment consisting of three additional infusions. Of the infused patients, one had myelodysplastic syndrome, three had multiple myeloma, and 12 had R/R AML. Five patients across all dose levels had grade 3 or 4 CRS, and one dose-limiting toxicity was observed in a patient on DL3. Three of 12 evaluable patients with R/R AML (n=11) or myelodysplastic syndrome (n=1) had CR/CRi. Among responders, two patients with R/R AML proceeded to allogeneic HSCT with durable ongoing remissions (at 5 and 61 months).
Multi-antigen-specific chimeric antigen receptor T cells
In addition to monospecific CAR T-cell products, multi-antigen-specific CAR T cells are starting to be evaluated in early phase clinical studies.32,33 Nine patients received escalating doses (1-3x106/kg) of CAR T cells expressing CLL-1- and CD33-CAR as single or split doses following lymphodepletion with fludarabine/cyclophosphamide. The T-cell products were autologous in eight cases and from an HLA-matched sibling donor in one case. Toxicities included grade 4 pancytopenia (n=9), grade 1-3 CRS (n=8), and grade 1-3 neurotoxicity (n=4). Seven of the nine patients achieved a CR/CRi (MRD negative by flow cytometry), while two had no response (the AML blasts of one of the non-responders expressed only CD33). Six of the seven patients who achieved MRD-negative CR proceeded to a subsequent HSCT.
The prospect of generating effective chimeric antigen receptor T-cell products for acute myeloid leukemia
Based on the available clinical experience with CAR T cells for AML, there is a long road ahead before these cells can be applied in the routine clinical setting akin to CD19-CAR T cells. The timeline until CAR T-cell therapy for AML achieves similar effectiveness as CD19-CAR T cells for B-cell acute lymphoblastic leukemia remains undefined. The current status of the field of AML-CAR immune cell therapy is reminiscent of the early days of HSCT in the 1960s, when outcomes were poor and many experts believed that this approach would never become part of our treatment armamentarium for patients with hematologic malignancies.71 Current preclinical and clinical studies have identified formidable roadblocks that center around: (i) which antigen(s) to target and heterogenous antigen expression (the antigen dilemma); (ii) limited T-cell effector function through intrinsic and extrinsic mechanisms; and (iii) the ideal starting cell source for generating AML-specific T-cell products. In the following we summarize current efforts to overcome these roadblocks,, which are also illustrated in Figure 1.
Roadblock #1: the antigen dilemma
A major obstacle to AML-specific CAR T-cell therapies is the identification of target antigens that are expressed on leukemic blasts but are absent or expressed at low levels on normal myeloid cells and/or other healthy tissues.34,35,72,73 Moreover, AML blasts and leukemic stem cells are highly heterogeneous cell populations and more than one antigen may need to be targeted to increase antitumor activity.7 While target antigen discovery is ongoing, we believe that targeting a single antigen is highly unlikely to achieve outstanding cure rates, i.e., we will not suddenly discover a CD19-equivalent pan-AML target that is consistently expressed with an acceptable on-target/off-cancer toxicity profile. We will likely have to develop CAR T-cell products directed at certain AML subsets. The majority of CAR-targeting approaches have relied on targeting cell surface molecules. However several studies have highlighted that CAR with an antigen recognition domain which recognize a peptide in the context of an HLA molecule can target intracellular proteins in cancer cells, including AML.74-76 While this approach increases the repertoire of targetable antigens it is HLA-restricted and therefore raises feasibility concerns for small populations unless we revolutionize cell production to enable the generation of patient-specific genetically modified T-cell products in a cost-effective and timely manner.
In addition to antigen discovery, investigators have focused on developing dual targeting strategies for AML.34,72,77,78 This approach has the potential not only to mitigate the risk of antigen loss variants, but also specificity since CAR immune cells will only be fully activated in the presence of both antigens. For example, a multiomic approach systematically analyzed potential AML target antigen combinations,34 and based on these data an AML-specific T-cell product was developed that expresses an ADGRE2-CAR and a CLEC12A-chimeric co-stimulatory receptor (CCR).78 Other CAR/CCR combinations that are being explored include CD123-CAR/CD33-CCR and CLEC12A-CAR/TIM3-CCR.79,80
Other studies have suggested that targeting either CD33 and TIM3 or CLL1 and TIM3 has the potential to overcome heterogenous antigen expression of AML blasts.72 Finally, dual targeting of CD123 and GRP78 is being explored as an approach to prevent immune escape.81 In this regard, utilizing NK cells as immune cells to express CAR may be beneficial due to their inherent ability to target NK-G2D-L-positive tumor cells (see the section on NKG2D-L CAR T cells). Other efforts are focused on limiting on-target/ off-cancer toxicities, including myelotoxicity, by deleting AML target antigens, including CD33, from hematopoietic stem cells or by using base editing to delete a specific epitope of an AML target in the hematopoietic stem cells (e.g. CD45).82,83 Finally, investigators have focused on fine-tuning antigen recognition and logical gating approaches to drive the specificity of CAR T cells.84-88
Roadblock #2: limited effector function through T-cell intrinsic and extrinsic mechanisms
Intrinsic and extrinsic mechanisms contribute to the limited effector function of CAR T cells for cancers including AML. The immunosuppressive AML microenvironment presents a key extrinsic mechanism, which has been recently reviewed, and we defer the interested reader to these publications.37-39 Clearly, the microenvironment needs to be studied in pediatric as well as adult AML since these are distinct disease entities.89 Several strategies have been evaluated to enhance the effector function of CAR T cells in preclinical models focusing on genetic engineering.90,91 While many of these have not been directly evaluated in AML models, they could potentially also enhance the anti-AML activity of CAR T cells. Conceptually, additional genetic modifications of CAR T cells can be divided into two approaches. One approach is focused on identifying and deleting negative regulators of immune cell function, and the other approach relies on transgenic expression of molecules to enhance their function. The former strategy includes targeting molecules that play a role in (i) limiting T cell activation,92,93 and (ii) regulating T-cell fate through epigenetic mechanisms.94-98 The latter approach includes expressing (i) transcription factors,99 (ii) secretory or membrane-bound cytokines,100-104 (iii) chimeric cytokine receptors that are either constitutively active or ligand-activated,105-107 (iv) chimeric switch receptors that convert a T-cell inhibitory signal into an activating signal,108,109 or (v) co-stimulatory receptors.110,111 Specific examples for each approach are listed in Table 2. Some of these approaches are actively being explored for CAR immune cell therapies for solid tumors, and it is unclear which approach will have the desired outcome of achieving long-term CR. It is likely that several strategies will have to be combined to generate effective AML-specific CAR T cells, for example improving T-cell activation and manipulating epigenetic pathways. In addition, other factors need to be considered. For example, pro-inflammatory cytokines have been reported to promote leukemogenesis, which would make the transgenic expression of cytokines in AML-specific immune cells less desirable.112
Roadblock #3: acute myeloid leukemia-specific immune cell production
The optimal CAR design, CAR immune cell production, as well as the donor source of AML-specific CAR immune cells remain unknown. The original CAR design celebrates its 30th anniversary this year,113,114 and is still being used by the majority of synthetic T-cell receptors that are currently being evaluated in clinical studies, including those on AML. It has a cytoplasmic signaling domain that consists of a signaling domain derived from one or two co-stimulatory molecules and an activation domain derived from the CD3ζ chain.115 Several preclinical studies have highlighted that adding a signaling domain from cytokine receptors or replacing CD3ζ with ZAP70 improves CAR functionality.85,116 Likewise, adding a domain to enhance recruitment of intracellular signaling molecules has been shown to improve the effector function of CAR T and NK cells.117 Novel synthetic T-cell receptors have also been developed, including T-cell antigen couplers (TAC),118 T-cell receptor fusion constructs (TRuC),119 HLA-independent T-cell receptors (HIT),120 and synthetic T-cell receptor and antigen receptors (STAR),121 which endow T cells with enhanced effector function in comparison to CAR T cells. Finally, modifying T cells with a bispecific T-cell engager is another approach to generate T cells that recognize AML blasts.122 While it will be impossible to compare all these different approaches in clinical studies, revisiting which genetic engineering approach is optimal to generate tumor-specific immune cells for AML in preclinical models seems advisable, given their limited clinical activity thus far.
Currently, most investigators use the same CAR T-cell-product manufacturing approach irrespective of the targeted disease. At least for autologous products it is likely that cytokine requirements might differ to generate CAR T cells with optimal antitumor activity, since for example T cells from patients with AML or acute lymphoblastic leukemia have been exposed to very different classes of chemotherapeutic agents, and the leukemia microenvironments differ. To our knowledge, there are no published reports of studies investigating the effects of different cytokine cocktails or other culture media supplements on the anti-AML activity of autologous CAR T cells. Integrative bulk- and single-cell sequencing approaches have been used to characterize CD19-CAR T-cell products in detail and track infused T cells at a clonal level.123,124 We are hopeful that insights gained from these types of studies conducted on autologous AML-specific CAR T-cell products will direct the development of a manufacturing process to optimize their effector function in the future.
What is the ideal donor source of immune cells to develop AML-specific immune cell therapies? This of course depends on the type of immune cells. For example, while autologous CAR T cells can be generated from AML patients, this might be less than ideal since the effector function of autologous T cells is compromised by previous exposure to chemotherapeutic agents.125,126 Likewise, it presents a logistical challenge since patients must stop their AML-targeted therapies for 1 to 2 weeks (or even longer) before T cells can be collected for manufacturing.
These challenges could be overcome by using allogeneic donors. These include the previous HSCT donor for patients who relapse after a transplant or off-the-shelf CAR T-cell products from healthy donors. While generating CAR T-cell products from the previous HSCT donor requires no additional genetic modifications, off-the-shelf CAR T-cell products require additional modifications to prevent graft-versus-host disease and/or allorejection.127,128 In addition, allogeneic NK cells, including cytokine-induced memory NK cells and CAR NK cells, are actively being explored as a source of immune effector cells for the immunotherapy of AML.76,129-131
Conclusions
We believe that developing safe and effective CAR T-cell therapy for AML is not a pipe dream. However, there are formidable challenges that have to be addressed, including targeting multiple antigens to prevent immune escape and on-target/off-cancer toxicities, and to overcome the immunosuppressive AML microenvironment. Likewise, T-cell production as well as evaluating these therapies initially in heavily pretreated patients poses significant challenges. Clearly a concerted team effort will be required in which basic/translational science and clinical investigators from multiple centers work together to achieve this goal. We hope that this review will stimulate discussions on how to reach the summit of effective cell therapies for AML.
Footnotes
- Received November 7, 2023
- Accepted February 28, 2024
Correspondence
Disclosures
SG and MPV are co-inventors of products with patents or patent applications in the fields of T-cell and gene therapy for cancer. SG is a member of the Scientific Advisory Board of Be Biopharma and CARGO, and the Data and Safety Monitoring Board (DSMB) of Immatics and has received honoraria from TESSA Therapeutics within the last year.
Contributions
SN, MPV, and SG co-wrote the manuscript.
Funding
MPV is supported by a St. Baldrick’s scholar grant. The research efforts of SN, MPV, and SG focused on hematologic malignancies are supported by grants from the V Foundation, the Leukemia Lymphoma Society, Cookies for Kids’ Cancer, Assisi Foundation of Memphis and American Lebanese Syrian Associated Charities (ALSAC).
References
- Hoffman AE, Schoonmade LJ, Kaspers GJ. Pediatric relapsed acute myeloid leukemia: a systematic review. Expert Rev Anticancer Ther. 2021; 21(1):45-52. https://doi.org/10.1080/14737140.2021.1841640PubMedGoogle Scholar
- Miller KD, Fidler-Benaoudia M, Keegan TH. Cancer statistics for adolescents and young adults, 2020. CA Cancer J Clin. 2020; 70(6):443-459. https://doi.org/10.3322/caac.21637PubMedGoogle Scholar
- Rasche M, Zimmermann M, Borschel L. 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(10):2167-2177. https://doi.org/10.1038/s41375-018-0071-7PubMedPubMed CentralGoogle Scholar
- Rasche M, Zimmermann M, Steidel E. Survival following relapse in children with acute myeloid leukemia: a report from AML-BFM and COG. Cancers (Basel). 2021; 13(10):2336. https://doi.org/10.3390/cancers13102336PubMedPubMed CentralGoogle Scholar
- Rasche M, Steidel E, Zimmermann M. Second relapse of pediatric patients with acute myeloid leukemia: a report on current treatment strategies and outcome of the AML-BFM study group. Cancers (Basel). 2021; 13(4):789. https://doi.org/10.3390/cancers13040789PubMedPubMed CentralGoogle Scholar
- White T, Kaspers G, Abrahamsson J. Clinical outcomes of second relapsed and refractory first relapsed paediatric AML: a retrospective study within the NOPHO-DB SHIP consortium. Br J Haematol. 2022; 197(6):755-765. https://doi.org/10.1111/bjh.18039PubMedPubMed CentralGoogle Scholar
- Thomas D, Majeti R. Biology and relevance of human acute myeloid leukemia stem cells. Blood. 2017; 129(12):1577-1585. https://doi.org/10.1182/blood-2016-10-696054PubMedPubMed CentralGoogle Scholar
- Ishikawa F, Yoshida S, Saito Y. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol. 2007; 25(11):1315-1321. https://doi.org/10.1038/nbt1350PubMedGoogle Scholar
- Jentzsch M, Grimm J, Bill M. Prognostic relevance of remission and measurable residual disease status in AML patients prior to reduced intensity or non-myeloablative allogeneic stem cell transplantation. Blood Cancer J. 2021; 11(4):80. https://doi.org/10.1038/s41408-021-00471-xPubMedPubMed CentralGoogle Scholar
- Maude SL, Laetsch TW, Buechner J. Tisagenlecleucel in children and young adults with B-cell lymphoblastic leukemia. N Engl J Med. 2018; 378(5):439-448. https://doi.org/10.1056/NEJMoa1709866PubMedPubMed CentralGoogle Scholar
- Maude SL, Frey N, Shaw PA. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014; 371(16):1507-1517. https://doi.org/10.1056/NEJMoa1407222PubMedPubMed CentralGoogle Scholar
- Fabrizio VA, Phillips CL, Lane A. Tisagenlecleucel outcomes in relapsed/refractory extramedullary ALL: a Pediatric Real World CAR Consortium report. Blood Adv. 2022; 6(2):600-610. https://doi.org/10.1182/bloodadvances.2021005564PubMedPubMed CentralGoogle Scholar
- Ghorashian S, Jacoby E, De Moerloose B. Tisagenlecleucel therapy for relapsed or refractory B-cell acute lymphoblastic leukaemia in infants and children younger than 3 years of age at screening: an international, multicentre, retrospective cohort study. Lancet Haematol. 2022; 9(10):e766-e775. https://doi.org/10.1016/S2352-3026(22)00225-3PubMedGoogle Scholar
- Moskop A, Pommert L, Baggott C. Real-world use of tisagenlecleucel in infant acute lymphoblastic leukemia. Blood Adv. 2022; 6(14):4251-4255. https://doi.org/10.1182/bloodadvances.2021006393PubMedPubMed CentralGoogle Scholar
- Ritchie DS, Neeson PJ, Khot A. Persistence and efficacy of second generation CAR T cell against the LeY antigen in acute myeloid leukemia. Mol Ther. 2013; 21(11):2122-2129. https://doi.org/10.1038/mt.2013.154PubMedPubMed CentralGoogle Scholar
- Budde L, Song JY, Kim Y. Remissions of acute myeloid leukemia and blastic plasmacytoid dendritic cell neoplasm following treatment with CD123-specific CAR T cells: a first-inhuman clinical trial. Blood. 2017; 130(Supplement 1):811. https://doi.org/10.1182/blood.V130.Suppl_1.811.811Google Scholar
- Naik S, Madden RM, Lipsitt A. Safety and anti-leukemic activity of CD123-CAR T cells in pediatric patients with AML: preliminary results from a phase 1 trial. Blood. 2022; 140(Supplement 1):4584-4585. https://doi.org/10.1182/blood-2022-170201Google Scholar
- Mardiana S, Gill S. CAR T cells for acute myeloid leukemia: state of the art and future directions. Front Oncol. 2020; 10:697. https://doi.org/10.3389/fonc.2020.00697PubMedPubMed CentralGoogle Scholar
- Cummins KD, Gill S. Will CAR T cell therapy have a role in AML? Promises and pitfalls. Semin Hematol. 2019; 56(2):155-163. https://doi.org/10.1053/j.seminhematol.2018.08.008PubMedGoogle Scholar
- Wermke M, Kraus S, Ehninger A. Proof of concept for a rapidly switchable universal CAR-T platform with UniCAR-T-CD123 in relapsed/refractory AML. Blood. 2021; 137(22):3145-3148. https://doi.org/10.1182/blood.2020009759PubMedPubMed CentralGoogle Scholar
- Ehninger G, Kraus S, Sala E. Phase 1 dose escalation study of the rapidly switchable universal CAR-T therapy Unicar-T-CD123 in relapsed/refractory AML. Blood. 2022; 140(Suppl 1):2367-2368. https://doi.org/10.1182/blood-2022-168877Google Scholar
- Sallman DA, DeAngelo DJ, Pemmaraju N. Ameli-01: a phase I trial of UCART123v1.2, an anti-CD123 allogeneic CAR-T cell product, in adult patients with relapsed or refractory (R/R) CD123+ acute myeloid leukemia (AML). Blood. 2022; 140(Suppl 1):2371-2373. https://doi.org/10.1182/blood-2022-169928Google Scholar
- Tambaro FP, Singh H, Jones E. Autologous CD33-CAR-T cells for treatment of relapsed/refractory acute myelogenous leukemia. Leukemia. 2021; 35(11):3282-3286. https://doi.org/10.1038/s41375-021-01232-2PubMedPubMed CentralGoogle Scholar
- Wang QS, Wang Y, Lv HY. Treatment of CD33-directed chimeric antigen receptor-modified T cells in one patient with relapsed and refractory acute myeloid leukemia. Mol Ther. 2015; 23(1):184-191. https://doi.org/10.1038/mt.2014.164PubMedPubMed CentralGoogle Scholar
- Zhang H, Bu C, Peng Z. Characteristics of anti-CLL1 based CAR-T therapy for children with relapsed or refractory acute myeloid leukemia: the multi-center efficacy and safety interim analysis. Leukemia. 2022; 36(11):2596-2604. https://doi.org/10.1038/s41375-022-01703-0PubMedGoogle Scholar
- Zhang H, Wang P, Li Z. Anti-CLL1 chimeric antigen receptor T-cell therapy in children with relapsed/refractory acute myeloid leukemia. Clin Cancer Res. 2021; 27(13):3549-3555. https://doi.org/10.1158/1078-0432.CCR-20-4543PubMedGoogle Scholar
- Pei K, Xu H, Wang P. Anti-CLL1-based CAR T-cells with 4-1-BB or CD28/CD27 stimulatory domains in treating childhood refractory/relapsed acute myeloid leukemia. Cancer Med. 2023; 12(8):9655-9661. https://doi.org/10.1002/cam4.5916PubMedPubMed CentralGoogle Scholar
- Jin X, Zhang M, Sun R. First-in-human phase I study of CLL-1 CAR-T cells in adults with relapsed/refractory acute myeloid leukemia. J Hematol Oncol. 2022; 15(1):88. https://doi.org/10.1186/s13045-022-01308-1PubMedPubMed CentralGoogle Scholar
- Baumeister SH, Murad J, Werner L. Phase I trial of autologous CAR T cells targeting NKG2D ligands in patients with AML/MDS and multiple myeloma. Cancer Immunol Res. 2019; 7(1):100-112. https://doi.org/10.1158/2326-6066.CIR-18-0307PubMedPubMed CentralGoogle Scholar
- Sallman DA, Kerre T, Havelange V. CYAD-01, an autologous NKG2D-based CAR T-cell therapy, in relapsed or refractory acute myeloid leukaemia and myelodysplastic syndromes or multiple myeloma (THINK): haematological cohorts of the dose escalation segment of a phase 1 trial. Lancet Haematol. 2023; 10(3):e191-e202. https://doi.org/10.1016/S2352-3026(22)00378-7PubMedGoogle Scholar
- Sallman DA, Brayer J, Sagatys EM. NKG2D-based chimeric antigen receptor therapy induced remission in a relapsed/ refractory acute myeloid leukemia patient. Haematologica. 2018; 103(9):e424-e426. https://doi.org/10.3324/haematol.2017.186742PubMedPubMed CentralGoogle Scholar
- Liu F, Hongyu Z, Sun L. First-in-human CLL1-CD33 compound CAR (CCAR) T cell therapy in relapsed and refractory acute myeloid leukemia. EHA Library. 2020; 294969:S149. Google Scholar
- Liu F, Cao Y, Pinz K. First-in-human CLL1-CD33 compound CAR T cell therapy induces complete remission in patients with refractory acute myeloid leukemia: update on phase 1 clinical trial. Blood. 2018; 132:901. https://doi.org/10.1182/blood-2018-99-110579Google Scholar
- Perna F, Berman SH, Soni RK. Integrating proteomics and transcriptomics for systematic combinatorial chimeric antigen receptor therapy of AML. Cancer Cell. 2017; 32(4):506-519. https://doi.org/10.1016/j.ccell.2017.09.004PubMedPubMed CentralGoogle Scholar
- Gottschlich A, Thomas M, Grunmeier R. Single-cell transcriptomic atlas-guided development of CAR-T cells for the treatment of acute myeloid leukemia. Nat Biotechnol. 2023; 41(11):1618-1632. https://doi.org/10.1038/s41587-023-01684-0PubMedPubMed CentralGoogle Scholar
- Daver N, Alotaibi AS, Bucklein V, Subklewe M. T-cell-based immunotherapy of acute myeloid leukemia: current concepts and future developments. Leukemia. 2021; 35(7):1843-1863. https://doi.org/10.1038/s41375-021-01253-xPubMedPubMed CentralGoogle Scholar
- Epperly R, Gottschalk S, Velasquez MP. A bump in the road: how the hostile AML microenvironment affects CAR T cell therapy. Front Oncol. 2020; 10:262. https://doi.org/10.3389/fonc.2020.00262PubMedPubMed CentralGoogle Scholar
- Tettamanti S, Pievani A, Biondi A, Dotti G, Serafini M. Catch me if you can: how AML and its niche escape immunotherapy. Leukemia. 2022; 36(1):13-22. https://doi.org/10.1038/s41375-021-01350-xPubMedPubMed CentralGoogle Scholar
- Vadakekolathu J, Rutella S. Escape from T-cell targeting immunotherapies in acute myeloid leukemia. Blood. 2023. https://doi.org/10.1182/blood.2023019961PubMedGoogle Scholar
- Atilla E, Benabdellah K. The black hole: CAR T cell therapy in AML. Cancers (Basel). 2023; 15(10):2713. https://doi.org/10.3390/cancers15102713PubMedPubMed CentralGoogle Scholar
- Vishwasrao P, Li G, Boucher JC, Smith DL, Hui SK. Emerging CAR T cell strategies for the treatment of AML. Cancers (Basel). 2022; 14(5):1241. https://doi.org/10.3390/cancers14051241PubMedPubMed CentralGoogle Scholar
- Gurney M, O’Dwyer M. Realizing innate potential: CAR-NK cell therapies for acute myeloid leukemia. Cancers (Basel). 2021; 13(7):1568. https://doi.org/10.3390/cancers13071568PubMedPubMed CentralGoogle Scholar
- O’Hear C, Heiber JF, Schubert I, Fey G, Geiger TL. Anti-CD33 chimeric antigen receptor targeting of acute myeloid leukemia. Haematologica. 2015; 100(3):336-344. https://doi.org/10.3324/haematol.2014.112748PubMedPubMed CentralGoogle Scholar
- Tettamanti S, Marin V, Pizzitola I. Targeting of acute myeloid leukaemia by cytokine-induced killer cells redirected with a novel CD123-specific chimeric antigen receptor. Br J Haematol. 2013; 161(3):389-401. https://doi.org/10.1111/bjh.12282PubMedGoogle Scholar
- Riberdy JM, Zhou S, Zheng F. The art and science of selecting a CD123-specific chimeric antigen receptor for clinical testing. Mol Ther Methods Clin Dev. 2020; 18:571-581. https://doi.org/10.1016/j.omtm.2020.06.024PubMedPubMed CentralGoogle Scholar
- Gill S, Tasian SK, Ruella M. Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood. 2014; 123(15):2343-2354. https://doi.org/10.1182/blood-2013-09-529537PubMedPubMed CentralGoogle Scholar
- Mardiros A, Dos SC, McDonald T. T cells expressing CD123-specific chimeric antigen receptors exhibit specific cytolytic effector functions and antitumor effects against human acute myeloid leukemia. Blood. 2013; 122(18):3138-3148. https://doi.org/10.1182/blood-2012-12-474056PubMedPubMed CentralGoogle Scholar
- Tashiro H, Sauer T, Shum T. Treatment of acute myeloid leukemia with T cells expressing chimeric antigen receptors directed to C-type lectin-like molecule 1. Mol Ther. 2017; 25(9):2202-2213. https://doi.org/10.1016/j.ymthe.2017.05.024PubMedPubMed CentralGoogle Scholar
- Wang J, Chen S, Xiao W. CAR-T cells targeting CLL-1 as an approach to treat acute myeloid leukemia. J Hematol Oncol. 2018; 11(1):7. https://doi.org/10.1186/s13045-017-0553-5PubMedPubMed CentralGoogle Scholar
- Niktoreh N, Lerius B, Zimmermann M. Gemtuzumab ozogamicin in children with relapsed or refractory acute myeloid leukemia: a report by Berlin-Frankfurt-Münster study group. Haematologica. 2019; 104(1):120-127. https://doi.org/10.3324/haematol.2018.191841PubMedPubMed CentralGoogle Scholar
- Pollard JA, Guest E, Alonzo TA. Gemtuzumab ozogamicin improves event-free survival and reduces relapse in pediatric KMT2A-rearranged AML: results from the phase III Children’s Oncology Group trial AAML0531. J Clin Oncol. 2021; 39(28):3149-3160. https://doi.org/10.1200/JCO.20.03048PubMedPubMed CentralGoogle Scholar
- Lamble AJ, Liu X, Minard C. Safety and activity of flotetuzumab in pediatric and young adult patients with relapsed/refractory acute myeloid leukemia: results from the COG PEPN1812 phase 1 trial. Blood. 2022; 40(Suppl 1):6209-6210. https://doi.org/10.1182/blood-2022-158029Google Scholar
- Sauer T, Parikh K, Sharma S. CD70-specific CAR T-cells have potent activity against acute myeloid leukemia (AML) without HSC toxicity. Blood. 2021; 138(4):318-330. https://doi.org/10.1182/blood.2020008221PubMedPubMed CentralGoogle Scholar
- Leick MB, Silva H, Scarfo I. Non-cleavable hinge enhances avidity and expansion of CAR-T cells for acute myeloid leukemia. Cancer Cell. 2022; 40(5):494-508. https://doi.org/10.1016/j.ccell.2022.04.001PubMedPubMed CentralGoogle Scholar
- Hebbar N, Epperly R, Vaidya A. CAR T cells redirected to cell surface GRP78 display robust anti-acute myeloid leukemia activity and do not target hematopoietic progenitor cells. Nat Commun. 2022; 13(1):587. https://doi.org/10.1038/s41467-022-28243-6PubMedPubMed CentralGoogle Scholar
- Glisovic-Aplenc T, Diorio C, Chukinas JA. CD38 as a pan-hematologic target for chimeric antigen receptor T cells. Blood Adv. 2023; 7(16):4418-4430. https://doi.org/10.1182/bloodadvances.2022007059PubMedPubMed CentralGoogle Scholar
- Casucci M, Nicolis di Robilant B, Falcone L. CD44v6-targeted T cells mediate potent antitumor effects against acute myeloid leukemia and multiple myeloma. Blood. 2013; 122(20):3461-3472. https://doi.org/10.1182/blood-2013-04-493361PubMedGoogle Scholar
- Myburgh R, Kiefer JD, Russkamp NF. Anti-human CD117 CAR T-cells efficiently eliminate healthy and malignant CD117-expressing hematopoietic cells. Leukemia. 2020; 34(10):2688-2703. https://doi.org/10.1038/s41375-020-0818-9PubMedGoogle Scholar
- Lichtman EI, Du H, Shou P. Preclinical evaluation of B7-H3-specific chimeric antigen receptor T cells for the treatment of acute myeloid leukemia. Clin Cancer Res. 2021; 27(11):3141-3153. https://doi.org/10.1158/1078-0432.CCR-20-2540PubMedPubMed CentralGoogle Scholar
- Jetani H, Navarro-Bailon A, Maucher M. Siglec-6 is a novel target for CAR T-cell therapy in acute myeloid leukemia. Blood. 2021; 138(19):1830-1842. https://doi.org/10.1182/blood.2020009192PubMedPubMed CentralGoogle Scholar
- Niswander LM, Graff ZT, Chien CD. Potent preclinical activity of FLT3-directed chimeric antigen receptor T-cell immunotherapy against FLT3-mutant acute myeloid leukemia and KMT2A-rearranged acute lymphoblastic leukemia. Haematologica. 2023; 108(2):457-471. https://doi.org/10.3324/haematol.2022.281456PubMedPubMed CentralGoogle Scholar
- Lynn RC, Poussin M, Kalota A. Targeting of folate receptor beta on acute myeloid leukemia blasts with chimeric antigen receptor-expressing T cells. Blood. 2015; 125(22):3466-3476. https://doi.org/10.1182/blood-2014-11-612721PubMedPubMed CentralGoogle Scholar
- Mai S, Hodges A, Chen HM. LILRB3 modulates acute myeloid leukemia progression and acts as an effective target for CAR T-cell therapy. Cancer Res. 2023; 83(24):4047-4062. https://doi.org/10.1158/0008-5472.CAN-22-2483PubMedGoogle Scholar
- John S, Chen H, Deng M. A novel anti-LILRB4 CAR-T cell for the treatment of monocytic AML. Mol Ther. 2018; 26(10):2487-2495. https://doi.org/10.1016/j.ymthe.2018.08.001PubMedPubMed CentralGoogle Scholar
- Lee WS, Ye Z, Cheung AMS. Effective killing of acute myeloid leukemia by TIM-3 targeted chimeric antigen receptor T Cells. Mol Cancer Ther. 2021; 20(9):1702-1712. https://doi.org/10.1158/1535-7163.MCT-20-0155PubMedGoogle Scholar
- Loff S, Dietrich J, Meyer JE. Rapidly switchable universal CAR-T cells for treatment of CD123-positive leukemia. Mol Ther Oncolytics. 2020; 17:408-420. https://doi.org/10.1016/j.omto.2020.04.009PubMedPubMed CentralGoogle Scholar
- Yao S, Jianlin C, Yarong L. Donor-derived CD123-targeted CAR T cell serves as a RIC regimen for haploidentical transplantation in a patient with FUS-ERG+ AML. Front Oncol. 2019; 9:1358. https://doi.org/10.3389/fonc.2019.01358PubMedPubMed CentralGoogle Scholar
- Shah NN, Tasian SK, Kohler ME. CD33 CAR T-cells (CD33CART) for children and young adults with relapsed/ refractory AML: dose-escalation results from a phase I/II multicenter trial. Blood. 2023; 142(Suppl 1):771. https://doi.org/10.1182/blood-2023-179667Google Scholar
- Davis CT, Rizzieri D. Immunotherapeutic applications of NK cells. Pharmaceuticals (Basel). 2015; 8(2):250-256. https://doi.org/10.3390/ph8020250PubMedPubMed CentralGoogle Scholar
- Zhang T, Lemoi BA, Sentman CL. Chimeric NK-receptor-bearing T cells mediate antitumor immunotherapy. Blood. 2005; 106(5):1544-1551. https://doi.org/10.1182/blood-2004-11-4365PubMedPubMed CentralGoogle Scholar
- Little MT, Storb R. History of haematopoietic stem-cell transplantation. Nat Rev Cancer. 2002; 2(3):231-238. https://doi.org/10.1038/nrc748PubMedGoogle Scholar
- Haubner S, Perna F, Kohnke T. Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML. Leukemia. 2019; 33(1):64-74. https://doi.org/10.1038/s41375-018-0180-3PubMedPubMed CentralGoogle Scholar
- Willier S, Rothamel P, Hastreiter M. CLEC12A and CD33 coexpression as a preferential target for pediatric AML combinatorial immunotherapy. Blood. 2021; 137(8):1037-1049. https://doi.org/10.1182/blood.2020006921PubMedGoogle Scholar
- Rafiq S, Purdon TJ, Daniyan AF. Optimized T-cell receptormimic chimeric antigen receptor T cells directed toward the intracellular Wilms tumor 1 antigen. Leukemia. 2017; 31(8):1788-1797. https://doi.org/10.1038/leu.2016.373PubMedPubMed CentralGoogle Scholar
- Kirkey DC, Loeb AM, Castro S. Therapeutic targeting of PRAME with mTCRCAR T cells in acute myeloid leukemia. Blood Adv. 2023; 7(7):1178-1189. https://doi.org/10.1182/bloodadvances.2022008304PubMedPubMed CentralGoogle Scholar
- Dong H, Ham JD, Hu G. Memory-like NK cells armed with a neoepitope-specific CAR exhibit potent activity against NPM1 mutated acute myeloid leukemia. Proc Natl Acad Sci U S A. 2022; 119(25):e2122379119. https://doi.org/10.1073/pnas.2122379119PubMedPubMed CentralGoogle Scholar
- Nixdorf D, Sponheimer M, Berghammer D. Adapter CAR T cells to counteract T-cell exhaustion and enable flexible targeting in AML. Leukemia. 2023; 37(6):1298-1310. https://doi.org/10.1038/s41375-023-01905-0PubMedPubMed CentralGoogle Scholar
- Haubner S, Mansilla-Soto J, Nataraj S. Cooperative CAR targeting to selectively eliminate AML and minimize escape. Cancer Cell. 2023; 41(11):1871-1891. https://doi.org/10.1016/j.ccell.2023.09.010PubMedPubMed CentralGoogle Scholar
- Perriello VM, Rotiroti MC, Pisani I. IL-3-zetakine combined with a CD33 costimulatory receptor as a dual CAR approach for safer and selective targeting of AML. Blood Adv. 2023; 7(12):2855-2871. https://doi.org/10.1182/bloodadvances.2022008762PubMedPubMed CentralGoogle Scholar
- Van Der Schans JJ, Vishwasrao P, Van Gils N. Dual split-signaling TIM3+CLEC12a targeting CAR T-cells with optimized signaling as a safe potential therapy for acute myeloid leukemia. Blood. 2023; 142(Suppl 1):883. https://doi.org/10.1182/blood-2023-180799Google Scholar
- Zoine JT, Immadisetty K, Ibanez-Vega J. Peptide-scFv antigen recognition domains effectively confer CAR T cell multiantigen specificity. Cell Rep Med. 2024; 5(2):101422. https://doi.org/10.1016/j.xcrm.2024.101422PubMedPubMed CentralGoogle Scholar
- Borot F, Wang H, Ma Y. Gene-edited stem cells enable CD33-directed immune therapy for myeloid malignancies. Proc Natl Acad Sci U S A. 2019; 116(24):11978-11987. https://doi.org/10.1073/pnas.1819992116PubMedPubMed CentralGoogle Scholar
- Wellhausen N, O’Connell RP, Lesch S. Epitope base editing CD45 in hematopoietic cells enables universal blood cancer immune therapy. Sci Transl Med. 2023; 15(714):eadi1145. https://doi.org/10.1126/scitranslmed.adi1145PubMedPubMed CentralGoogle Scholar
- Hamieh M, Mansilla-Soto J, Riviere I, Sadelain M. Programming CAR T cell tumor recognition: tuned antigen sensing and logic gating. Cancer Discov. 2023; 13(4):829-843. https://doi.org/10.1158/2159-8290.CD-23-0101PubMedPubMed CentralGoogle Scholar
- Tousley AM, Rotiroti MC, Labanieh L. Co-opting signalling molecules enables logic-gated control of CAR T cells. Nature. 2023; 615(7952):507-516. https://doi.org/10.1038/s41586-023-05778-2PubMedPubMed CentralGoogle Scholar
- Choe JH, Watchmaker PB, Simic MS. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 2021; 13(591):eabe7378. https://doi.org/10.1126/scitranslmed.abe7378PubMedPubMed CentralGoogle Scholar
- Hyrenius-Wittsten A, Su Y, Park M. SynNotch CAR circuits enhance solid tumor recognition and promote persistent antitumor activity in mouse models. Sci Transl Med. 2021; 13(591):eabd8836. https://doi.org/10.1126/scitranslmed.abd8836PubMedPubMed CentralGoogle Scholar
- Richards RM, Zhao F, Freitas KA. NOT-gated CD93 CAR T cells effectively target AML with minimized endothelial cross-reactivity. Blood Cancer Discov. 2021; 2(6):648-665. https://doi.org/10.1158/2643-3230.BCD-20-0208PubMedPubMed CentralGoogle Scholar
- Bolouri H, Farrar JE, Triche T Jr. The molecular landscape of pediatric acute myeloid leukemia reveals recurrent structural alterations and age-specific mutational interactions. Nat Med. 2018; 24(1):103-112. https://doi.org/10.1038/nm.4439PubMedPubMed CentralGoogle Scholar
- Wagner J, Wickman E, DeRenzo C, Gottschalk S. CAR T cell therapy for solid tumors: bright future or dark reality?. Mol Ther. 2020; 28(11):2320-2339. https://doi.org/10.1016/j.ymthe.2020.09.015PubMedPubMed CentralGoogle Scholar
- Labanieh L, Mackall CL. CAR immune cells: design principles, resistance and the next generation. Nature. 2023; 614(7949):635-648. https://doi.org/10.1038/s41586-023-05707-3PubMedGoogle Scholar
- Carnevale J, Shifrut E, Kale N. RASA2 ablation in T cells boosts antigen sensitivity and long-term function. Nature. 2022; 609(7925):174-182. https://doi.org/10.1038/s41586-022-05126-wPubMedPubMed CentralGoogle Scholar
- Wei J, Long L, Zheng W. Targeting REGNASE-1 programs long-lived effector T cells for cancer therapy. Nature. 2019; 576(7787):471-476. https://doi.org/10.1038/s41586-019-1821-zPubMedPubMed CentralGoogle Scholar
- Prinzing B, Zebley CC, Petersen CT. Deleting DNMT3A in CAR T cells prevents exhaustion and enhances antitumor activity. Sci Transl Med. 2021; 13(620)https://doi.org/10.1126/scitranslmed.abh0272PubMedPubMed CentralGoogle Scholar
- Jain N, Zhao Z, Feucht J. TET2 guards against unchecked BATF3-induced CAR T cell expansion. Nature. 2023; 615(7951):315-322. https://doi.org/10.1038/s41586-022-05692-zPubMedPubMed CentralGoogle Scholar
- Talleur AC, Qudeimat A, Metais JY. Preferential expansion of CD8+ CD19-CAR T cells postinfusion and the role of disease burden on outcome in pediatric B-ALL. Blood Adv. 2022; 6(21):5737-5749. https://doi.org/10.1182/bloodadvances.2021006293PubMedPubMed CentralGoogle Scholar
- Weber EW, Parker KR, Sotillo E. Transient rest restores functionality in exhausted CAR-T cells through epigenetic remodeling. Science. 2021; 372(6537):eaba1786. https://doi.org/10.1126/science.aba1786PubMedPubMed CentralGoogle Scholar
- Fraietta JA, Nobles CL, Sammons MA. Disruption of TET2 promotes the therapeutic efficacy of CD19-targeted T cells. Nature. 2018; 558(7709):307-312. https://doi.org/10.1038/s41586-018-0178-zPubMedPubMed CentralGoogle Scholar
- Lynn RC, Weber EW, Sotillo E. c-Jun overexpression in CAR T cells induces exhaustion resistance. Nature. 2019; 576(7786):293-300. https://doi.org/10.1038/s41586-019-1805-zPubMedPubMed CentralGoogle Scholar
- Krenciute G, Prinzing BL, Yi Z. Transgenic expression of IL15 improves antiglioma activity of IL13Ralpha2-CAR T cells but results in antigen loss variants. Cancer Immunol Res. 2017; 5(7):571-581. https://doi.org/10.1158/2326-6066.CIR-16-0376PubMedPubMed CentralGoogle Scholar
- Avanzi MP, Yeku O, Li X. Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both directly and through activation of the endogenous immune system. Cell Rep. 2018; 23(7):2130-2141. https://doi.org/10.1016/j.celrep.2018.04.051PubMedPubMed CentralGoogle Scholar
- Pegram HJ, Lee JC, Hayman EG. Tumor-targeted T cells modified to secrete IL-12 eradicate systemic tumors without need for prior conditioning. Blood. 2012; 119(18):4133-4141. https://doi.org/10.1182/blood-2011-12-400044PubMedPubMed CentralGoogle Scholar
- Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert Opin Biol Ther. 2015; 15(8):1145-1154. https://doi.org/10.1517/14712598.2015.1046430PubMedGoogle Scholar
- Hurton LV, Singh H, Najjar AM. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells. Proc Natl Acad Sci U S A. 2016; 113(48):E7788-E7797. https://doi.org/10.1073/pnas.1610544113PubMedPubMed CentralGoogle Scholar
- Sockolosky JT, Trotta E, Parisi G. Selective targeting of engineered T cells using orthogonal IL-2 cytokine-receptor complexes. Science. 2018; 359(6379):1037-1042. https://doi.org/10.1126/science.aar3246PubMedPubMed CentralGoogle Scholar
- Bell M, Gottschalk S. Engineered cytokine signaling to improve CAR T cell effector function. Front Immunol. 2021; 12:684642. https://doi.org/10.3389/fimmu.2021.684642PubMedPubMed CentralGoogle Scholar
- Shum T, Omer B, Tashiro H. Constitutive signaling from an engineered IL7 receptor promotes durable tumor elimination by tumor-redirected T cells. Cancer Discov. 2017; 7(11):1238-1247. https://doi.org/10.1158/2159-8290.CD-17-0538PubMedPubMed CentralGoogle Scholar
- Leen AM, Sukumaran S, Watanabe N. Reversal of tumor immune inhibition using a chimeric cytokine receptor. Mol Ther. 2014; 22(6):1211-1220. https://doi.org/10.1038/mt.2014.47PubMedPubMed CentralGoogle Scholar
- Wilkie S, Burbridge SE, Chiapero-Stanke L. Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4. J Biol Chem. 2010; 285(33):25538-25544. https://doi.org/10.1074/jbc.M110.127951PubMedPubMed CentralGoogle Scholar
- Omer B, Cardenas MG, Pfeiffer T. A costimulatory CAR improves TCR-based cancer immunotherapy. Cancer Imunol Res. 2022; 10(4):512-524. https://doi.org/10.1158/2326-6066.CIR-21-0307PubMedPubMed CentralGoogle Scholar
- Katsarou A, Sjostrand M, Naik J. Combining a CAR and a chimeric costimulatory receptor enhances T cell sensitivity to low antigen density and promotes persistence. Sci Transl Med. 2021; 13(623):eabh1962. https://doi.org/10.1126/scitranslmed.abh1962PubMedPubMed CentralGoogle Scholar
- Binder S, Luciano M, Horejs-Hoeck J. The cytokine network in acute myeloid leukemia (AML): a focus on pro- and anti-inflammatory mediators. Cytokine Growth Factor Rev. 2018; 43:8-15. https://doi.org/10.1016/j.cytogfr.2018.08.004PubMedGoogle Scholar
- Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxic lymphocytes through chimeric single chains consisting of antibody-binding domains and the gamma or zeta subunits of the immunoglobulin and T-cell receptors. Proc Natl Acad Sci U S A. 1993; 90(2):720-724. https://doi.org/10.1073/pnas.90.2.720PubMedPubMed CentralGoogle Scholar
- Brocker T, Peter A, Traunecker A, Karjalainen K. New simplified molecular design for functional T cell receptor. Eur J Immunol. 1993; 23(7):1435-1439. https://doi.org/10.1002/eji.1830230705PubMedGoogle Scholar
- June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018; 379(1):64-73. https://doi.org/10.1056/NEJMra1706169PubMedPubMed CentralGoogle Scholar
- Kagoya Y, Tanaka S, Guo T. A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat Med. 2018; 24(3):352-359. https://doi.org/10.1038/nm.4478PubMedPubMed CentralGoogle Scholar
- Chockley PJ, Ibanez-Vega J, Krenciute G, Talbot LJ, Gottschalk S. Synapse-tuned CARs enhance immune cell anti-tumor activity. Nat Biotechnol. 2023; 41(10):1434-1445. https://doi.org/10.1038/s41587-022-01650-2PubMedPubMed CentralGoogle Scholar
- Helsen CW, Hammill JA, Lau VWC. The chimeric TAC receptor co-opts the T cell receptor yielding robust anti-tumor activity without toxicity. Nat Commun. 2018; 9(1):3049. https://doi.org/10.1038/s41467-018-05395-yPubMedPubMed CentralGoogle Scholar
- Baeuerle PA, Ding J, Patel E. Synthetic TRuC receptors engaging the complete T cell receptor for potent anti-tumor response. Nat Commun. 2019; 10(1):2087. https://doi.org/10.1038/s41467-019-10097-0PubMedPubMed CentralGoogle Scholar
- Mansilla-Soto J, Eyquem J, Haubner S. HLA-independent T cell receptors for targeting tumors with low antigen density. Nat Med. 2022; 28(2):345-352. https://doi.org/10.1038/s41591-021-01621-1PubMedPubMed CentralGoogle Scholar
- Liu Y, Liu G, Wang J. Chimeric STAR receptors using TCR machinery mediate robust responses against solid tumors. Sci Transl Med. 2021; 13(586):eabb5191. https://doi.org/10.1126/scitranslmed.abb5191PubMedGoogle Scholar
- Bonifant CL, Szoor A, Torres D. CD123-engager T cells as a novel immunotherapeutic for acute myeloid leukemia. Mol Ther. 2016; 24(9):1615-1626. https://doi.org/10.1038/mt.2016.116PubMedPubMed CentralGoogle Scholar
- Wilson TL, Kim H, Chou CH. Common trajectories of highly effective CD19-specific CAR T cells identified by endogenous T-cell receptor lineages. Cancer Discov. 2022; 12(9):2098-2119. https://doi.org/10.1158/2159-8290.CD-21-1508PubMedPubMed CentralGoogle Scholar
- Chen GM, Chen C, Das RK. Integrative bulk and single-cell profiling of premanufacture T-cell populations reveals factors mediating long-term persistence of CAR T-cell therapy. Cancer Discov. 2021; 11(9):2186-2199. https://doi.org/10.1158/2159-8290.CD-20-1677PubMedPubMed CentralGoogle Scholar
- Das RK, Vernau L, Grupp SA, Barrett DM. Naive T-cell deficits at diagnosis and after chemotherapy impair cell therapy potential in pediatric cancers. Cancer Discov. 2019; 9(4):492-499. https://doi.org/10.1158/2159-8290.CD-18-1314PubMedPubMed CentralGoogle Scholar
- Das RK, O’Connor RS, Grupp SA, Barrett DM. Lingering effects of chemotherapy on mature T cells impair proliferation. Blood Adv. 2020; 4(19):4653-4664. https://doi.org/10.1182/bloodadvances.2020001797PubMedPubMed CentralGoogle Scholar
- Caldwell KJ, Gottschalk S, Talleur AC. Allogeneic CAR cell therapy-more than a pipe dream. Front Immunol. 2020; 11:618427. https://doi.org/10.3389/fimmu.2020.618427PubMedPubMed CentralGoogle Scholar
- Aparicio C, Acebal C, Gonzalez-Vallinas M. Current approaches to develop “off-the-shelf” chimeric antigen receptor (CAR)-T cells for cancer treatment: a systematic review. Exp Hematol Oncol. 2023; 12(1):73. https://doi.org/10.1186/s40164-023-00435-wPubMedPubMed CentralGoogle Scholar
- Bednarski JJ, Zimmerman C, Berrien-Elliott MM. Donor memory-like NK cells persist and induce remissions in pediatric patients with relapsed AML after transplant. Blood. 2022; 139(11):1670-1683. https://doi.org/10.1182/blood.2021013972PubMedPubMed CentralGoogle Scholar
- Berrien-Elliott MM, Foltz JA, Russler-Germain DA. Hematopoietic cell transplantation donor-derived memory-like NK cells functionally persist after transfer into patients with leukemia. Sci Transl Med. 2022; 14(633):eabm1375. https://doi.org/10.1126/scitranslmed.abm1375PubMedPubMed CentralGoogle Scholar
- Daher M, Melo Garcia L, Li Y, Rezvani K. CAR-NK cells: the next wave of cellular therapy for cancer. Clin Transl Immunology. 2021; 10(4):e1274. https://doi.org/10.1002/cti2.1274PubMedPubMed CentralGoogle Scholar
Figures & Tables
Article Information
This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License.