Abstract
Acute myeloid leukaemia (AML) is a fatal and refractory haematologic cancer that primarily affects adults. It interferes with bone marrow cell proliferation. Patients have a 5 years survival rate of less than 30% despite the availability of several treatments, including chemotherapy, allogeneic haematopoietic stem cell transplantation (Allo‐HSCT), and receptor antagonist drugs. Allo‐HSCT is the mainstay of acute myeloid leukaemia treatment. Although it does work, there are severe side effects, such as graft‐versus‐host disease (GVHD). In recent years, chimeric antigen receptor (CAR)‐T cell therapies have made significant progress in the treatment of cancer. These engineered T cells can locate and recognize tumour cells in vivo and release a large number of effectors through immune action to effectively kill tumour cells. CAR‐T cells are among the most effective cancer treatments because of this property. CAR‐T cells have demonstrated positive therapeutic results in the treatment of acute myeloid leukaemia, according to numerous clinical investigations. This review highlights recent progress in new targets for AML immunotherapy, and the limitations, and difficulties of CAR‐T therapy for AML.
Keywords: acute myeloid leukaemia, chimeric antigen receptor T cells, scFv, target antigen
1. INTRODUCTION
Acute myelocytic leukaemia (AML) is a clonal, malignant, proliferative condition that affects the myeloid primitive cells of the haematopoietic system. Acute myeloid leukaemia is a clonal expansion in bone marrow and is highly malignant. Haematopoietic progenitor cells that have experienced malignant transformation at different phases of the differentiation and development of normal myeloid cells can turn into diverse illnesses. 1 Allogeneic haematopoietic stem cell transplantation (Allo‐HSCT) is now the only option for patients with high‐risk or recurrent AML, however, it requires a matched donor. 2 The therapeutic efficiency of Allo‐HSCT is driven by the graft‐versus‐leukaemia phenomenon, which occurs when donor T cells recognize invading pathogens on the host's CAR‐T cells in the haematopoietic tissue of bone marrow malignancies and subsequently mediate tumour elimination. This GvL phenomenon illustrates how bone marrow responds to T‐lymphocytes. Unfortunately, graft‐versus‐host disease (GVHD), which has a high morbidity and mortality rate, can be caused by donor immune effector cells attacking healthy organs. Numerous immunotherapies, such as checkpoint inhibitors, antibody‐drug conjugates, T‐cell splicing agents, and cellular treatments, have been launched in clinical trials as our knowledge of the leukaemia microenvironment and immune milieu has grown. In select subgroups of AML patients with immune‐enriched characteristics, checkpoint inhibitors have had some success. 3 Early findings with fletuzumab (CD3‐CD123 bispecific antibody) and Magrolimab (anti‐CD47 monoclonal antibody) are anticipated following the approval of a modest split dosage of Gemtuzumab ozogamici (gituzumab), paving the way for AML immune‐based treatments. 4 , 5 In an innovative form of immunotherapy known as chimeric antigen receptor (CAR) T cells, naturally existing T cells are genetically altered to produce specific CARs that allow them to recognize and destroy tumour cells. After many years of preclinical and clinical study, CAR‐T cells are currently acknowledged as the best therapeutic approach. CAR‐T cell treatment has been effective in treating B‐cell lymphoma and acute lymphoblastic leukaemia (ALL). 6 , 7 , 8 , 9 , 10 , 11 , 12 , 13 , 14 , 15 , 16 Additionally, CAR‐T cell treatment for AML is presently being investigated. Unfortunately, the lack of suitable cancer antigens prevented the development of CAR‐T cells in AML. To minimize the side effects of CAR‐T cell treatment, the optimal antigen should be produced primarily on malignant tumour cells. Finding the best target for this illness can be challenging because the bulk of the antigens produced by AML cells are also present in healthy haematopoietic cells. 17 The development of various CAR‐T cell therapy targets for AML, as well as the drawbacks and difficulties of CAR‐T for AML, will be the main topics of this review.
2. STRUCTURE OF CAR
2.1. Evolution of CAR design
CAR is an artificial receptor molecule created by genetic engineering technology that confers specificity to immune effector cells against a target antigenic epitope, thereby enhancing T‐lymphocyte recognition of antigenic signals and activation. 18 The CAR structure consists of an extracellular binding region that is usually the single‐chain fragment variable (scFv) region, a hinge, a transmembrane domain, and an intracellular signalling domain. 19 , 20 A typical intracellular signalling region contains a CD3 activation domain and a co‐stimulatory proliferative signalling domain. T cells can behave as tumour‐killing cells. 21 Cytotoxic T cells (CTLs) are key effector cells of the anti‐tumor immune response, specifically recognizing tumour cells when the body is stimulated by antigens through the interaction of the T cell receptor (TCR) with the major histocompatibility complex. The CAR has undergone an evolution spanning five successive generations thus far. Zelig Eshhar developed the first‐generation CAR in 1989 by splicing the heavy and light chain variable sections of the monoclonal antibody into T cells together with a fixed percentage of the TCR. 22 The first generation of CARs only contained one intracellular region, primarily CD3, which caused T cells to become activated but had a limited ability to wake up dormant T cells and to maintain a sustained T cell response or cytokine release. 23 Second‐generation CARs include not only the CD3ζ‐activated region but also a single co‐stimulatory molecule, CD28 or 4‐BB, and in the third‐generation CARs, a second co‐stimulatory molecule, OX‐40, are added, 24 and these additional stimulatory molecules enhance T‐cell activation, persistence and proliferation. With the addition of CAR‐inducible genes that encode diverse cytokines and mediators to promote CAR activation, fourth‐generation CARs are built based on second‐generation CARs, and T cells transduced with fourth‐generation CARs can be employed for universal cytokine‐mediated killing. 25 Significantly improved therapeutic efficacy and decreased systemic toxicity have been reported in preclinical studies. Fifth‐generation CAR‐T cells, also known as next‐generation CAR‐T cells, are currently being studied. The next‐generation CAR‐T cells include one more functional element in CAR‐T structure compared with previous versions. A truncated intracellular domain of cytokine receptors with a binding site for transcription factors STAT3/5 was integrated into the CAR‐T intracellular structure (Figure 1). The addition of extra functional domains helped the CAR‐T cells to better achieve their goals. 26
FIGURE 1.
CAR structure and its evolution.
2.1.1. Single‐chain fragment variable (scFv)
The antibody heavy chain variable region and its light chain variable region were joined together to form the scFv, which serves as the CAR's antigen‐binding element, which can specifically recognize antigens on the surface of the target cell. 27 Because of the high specificity of this kind of antigen–antibody interaction, scFv is pivotal for the CAR‐T cells to execute their precise attack on cancer cells. 28
2.1.2. Extracellular spacer region
Also known as the ‘hinge region’, it provides flexibility and is typically made up of immunoglobulin‐based components like CH2, CH3 and CH2CH3. 29 Hinges derived from native T cell molecules, such as CD8 and CD28 hinge regions, were approved for clinical CAR‐T cell therapies.
2.1.3. Transmembrane structural domains
The transmembrane domain linked the extracellular hinge to the intracellular signalling domain. 30 The majority of transmembrane structures are derived from CD3ζ, CD4, CD8 or CD28 molecules. 31 Signals will be transduced via this domain from extracellular to intercellular, which can significantly affect the killing efficiency of CAR‐T cells.
2.1.4. Intracellular signalling domains
The cytoplasmic structure of CAR‐T was composed of signalling domains necessary for the activation of T cells. Co‐stimulation domains were added to the cytoplasmic structure in the later‐developed CAR‐T version. CD3 structural domain is the primary signalling region within the cell, while other co‐stimulatory molecules like CD28, 4‐1BB, or OX40 may also be present. 32 Nevertheless, they do not function as effectively as CD3ζ does. Other immunoreceptor tyrosine‐activated motifs (ITAMs), such as the IgE‐structural domain of the Fc receptor, have also been suggested as intracellular activating structural domains. 33
3. CAR‐T CELL THERAPY
T cells are separated from the patient's peripheral blood mononuclear cells (PBMC). These gathered T cells are stimulated using magnetic beads or exogenous cytokines. Then, using mRNA electroporation, gene editing techniques, liposomes, lentiviruses or retroviruses, CAR‐expressed genes are transferred into these proliferated T cells to drive the cells to detect the tumour. These CAR‐T cells are multiplied and then put back into the patient while they are being watched 34 (Figure 2).
FIGURE 2.
CAR‐T cell therapy flow.
4. HISTORICAL PROGRESS OF CAR‐T CELLS
From 1989, when Cross G and three other scientists first introduced the concept of ‘CAR’ in human history, 23 to 2022, when the FDA approved BCMA CAR‐T cells for the treatment of multiple myeloma, 35 it has been more than 30 years since the development of CAR‐T immune cell therapy (Figure 3). CAR‐T immune cell therapy has helped some patients with haematologic cancers achieve remissions of up to 10 years, 36 and future breakthroughs in the field of CAR‐T cells are sure to follow, with major trends in the field involving targets, disease treatment modalities and more.
FIGURE 3.
The history of CAR‐T cell's progress and milestones. CLL, chronic lymphocytic leukaemia; ALL, acute lymphoblastic leukaemia; DLBCL, diffuse large B‐cell lymphoma; MCL, mantle cell lymphoma; MM, multiple myeloma; LBCL, diffuse large B‐cell lymphoma.
5. DIFFERENT TARGETS OF CAR‐T FOR AML
In recent years, many studies have also screened and identified new target antigens for AML and designed various CAR‐T cells against these target antigens to evaluate their preclinical and clinical effects (Table 1). The latest advances in various targets are now discussed (Figure 4).
TABLE 1.
Ongoing clinical trials of CAR‐T cells for acute myeloid leukaemia.
NCT number | Study title | Study status | Phases | Enrollment | Targets |
---|---|---|---|---|---|
NCT05445765 | Anti‐CD33 CAR‐T Cells for the Treatment of Relapsed/Refractory CD33+ Acute Myeloid Leukaemia 131 | Not Yet Recruiting | 1 | 10 | CD33 |
NCT05248685 | Optimized Dual CD33/CLL1 CAR‐T Cells in Subjects with Refractory or Relapsed Acute Myeloid Leukaemia 132 | Recruiting | 1 | 20 | CD33, CLL1 |
NCT03896854 | CAR‐T Cell in CD19 Positive Relapsed or Refractory Acute Myeloid Leukaemia (AML) 133 | Recruiting | 1,2 | 15 | CD19 |
NCT05239689 | Clinical Study of CD38 CAR‐T Cells in the Treatment of Hematological Malignancies 134 | Recruiting | Early 1 | 36 | CD38 |
NCT01716364 | Safety Study of Anti Lewis Y Chimeric Antigen Receptor in Myeloma, Acute Myeloid Leukaemia or Myelodysplastic Syndrome 135 | Unknown | 1 | 6 | Lewis Y |
NCT05252572 | Clinical Study of CLL1 CAR‐T cells in the Treatment of Hematological Malignancies 136 | Recruiting | Early 1 | 36 | CLL1 |
NCT05473221 | Evaluate the Safety and Efficacy of CD33 CAR‐T in Patients With R/R AML 137 | Not Yet Recruiting | 1 | 20 | CD33 |
NCT03126864 | Study of Adoptive Cellular Therapy Using Autologous T Cells Transduced with Lentivirus to Express a CD33 Specific Chimeric Antigen Receptor in Patients with Relapsed or Refractory CD33‐Positive Acute Myeloid Leukaemia 138 | Terminated | 1 | 11 | CD33 |
NCT04803929 | Clinical Study of Anti‐ILT3 CAR‐T Therapy for R/R AML(M4/M5) 139 | Recruiting | Early 1 | 25 | ILT3 |
NCT03927261 | PRGN‐3006 Adoptive Cellular Therapy for CD33‐Positive Relapsed or Refractory AML, MRD Positive AML or Higher Risk MDS 140 | Recruiting | 1 | 88 | CD33 |
NCT04318678 | CD123‐Directed Autologous T‐Cell Therapy for Acute Myelogenous Leukaemia (CATCHAML) 141 | Recruiting | 1 | 32 | CD123 |
NCT05488132 | Administration of Anti‐siglec‐6 CAR‐T Cell Therapy in Relapsed and Refractory Acute Myeloid Leukaemia (rr/AML) 142 | Recruiting | 1,2 | 20 | Siglec‐6 |
NCT04662294 | CD 70 CAR‐T for Patients with CD70 Positive Malignant Hematologic Diseases 143 | Recruiting | Early 1 | 108 | CD70 |
NCT04272125 | Safety and Efficacy of CD123‐Targeted CAR‐T Therapy for Relapsed/Refractory Acute Myeloid Leukaemia 144 | Recruiting | 1,2 | 40 | CD123 |
NCT05949125 | Dose‐escalating Trial with Allo‐RevCAR01‐T Cells in Combination with CD123 Target Module (R‐TM123) for Participants with Selected Hematologic Malignancies Positive for CD123 145 | Not Yet Recruiting | 1 | 37 | CD123 |
NCT02203825 | Safety Study of Chimeric Antigen Receptor Modified T‐cells Targeting NKG2D‐Ligands 146 | Completed | 1 | 12 | NKG2D |
NCT04835519 | Phase I/II Study of Enhanced CD33 CAR‐T Cells in Subjects with Relapsed or Refractory Acute Myeloid Leukaemia 147 | Recruiting | 1,2 | 25 | CD33 |
NCT05454241 | CD7 CAR‐T for Patients With r/r CD7+ Hematologic Malignancies 148 | Recruiting | 1 | 22 | CD7 |
NCT05945849 | CD33KO‐HSPC Infusion Followed by CART‐33 Infusion(s) for Refractory/Relapsed AML 149 | Not Yet Recruiting | 1 | 16 | CD33 |
NCT05105152 | PLAT‐08: A Study Of SC‐DARIC33 CAR‐T Cells in Paediatric and Young Adults with Relapsed or Refractory CD33+ AML 150 | Recruiting | 1 | 18 | CD33 |
NCT05467254 | Evaluate the Safety and Efficacy of CLL1 + CD33 CAR‐T in Patients With R/R AML 151 | Not Yet Recruiting | 1 | 20 | CD33 |
NCT04257175 | CAR‐T CD19 for Acute Myelogenous Leukaemia With t 8:21 and CD19 Expression 152 | Recruiting | 2,3 | 10 | CD19 |
NCT05943314 | Clinical Study on Safety and Efficacy of Anti‐CLL1 /+CD33 CAR‐T Cells in the Treatment of Acute Myeloid Leukaemia 153 | Not Yet Recruiting | NA | 5 | CLL1 + CD33 |
NCT04599543 | IL3 CAR‐T Cell Therapy for Patients with CD123 Positive Relapsed and/or Refractory Acute Myeloid Leukaemia 154 | Not Yet Recruiting | Early 1 | 36 | CD123 |
NCT05023707 | Anti‐FLT3 CAR‐T Cell Therapy in FLT3 Positive Relapsed/Refractory Acute Myeloid Leukaemia 155 | Recruiting | 1,2 | 5 | FLT3 |
NCT04351022 | CD38‐targeted Chimeric Antigen Receptor T Cell (CAR‐T) in Relapsed or Refractory Acute Myeloid Leukaemia 156 | Recruiting | 1,2 | 20 | CD38 |
NCT05672147 | CD33 CAR‐T Cell Therapy for the Treatment of Recurrent or Refractory Acute Myeloid Leukaemia 157 | Recruiting | 1 | 27 | CD33 |
NCT04219163 | Chimeric Antigen Receptor T‐cells for The Treatment of AML Expressing CLL‐1 Antigen 158 | Recruiting | 1 | 18 | CLL1 |
NCT02623582 | CD123 Redirected Autologous T Cells for AML 159 | Terminated | Early 1 | 7 | CD123 |
NCT05266950 | Safety and Efficacy Study of CI‐135 CAR‐T Cells in Subjects with Relapsed or Refractory Acute Myeloid Leukaemia 160 | Recruiting | 1 | 7 | CI‐135 |
NCT03672851 | Study Evaluating Safety and Efficacy of CAR‐T Cells Targeting CD123 in Patients with Acute Leukaemia 160 | Terminated | 1 | 2 | CD123 |
NCT04762485 | Humanized CD7 CAR‐T Cell Therapy for r/r CD7+ Acute Leukaemia 160 | Recruiting | 1,2 | 20 | CD7 |
NCT05377827 | Dose‐Escalation and Dose‐Expansion Study to Evaluate the Safety and Tolerability of Anti‐CD7 Allogeneic CAR‐T Cells (WU‐CART‐007) in Patients with CD7+ Hematologic Malignancies 160 | Not Yet Recruiting | 1 | 48 | CD7 |
NCT03795779 | CLL1‐CD33 cCAR in Patients with Relapsed and/or Refractory, High Risk Hematologic Malignancies 160 | Unknown | Early 1 | 20 | CLL1, CD33 |
NCT05654779 | CLL‐1/CD33 Targeted LCAR‐AMDR Cells in Patients with Relapsed or Refractory Acute Myeloid Leukaemia 160 | Recruiting | 1 | 34 | CLL1/CD33 |
NCT04265963 | CD123‐Targeted CAR‐T Cell Therapy for Relapsed/Refractory Acute Myeloid Leukaemia 160 | Recruiting | 1,2 | 45 | CD123 |
NCT05016063 | Dual CD33‐CLL1‐CAR‐T Cells in the Treatment of Relapsed/Refractory Acute Myeloid Leukaemia 160 | Not Yet Recruiting | Early 1 | 32 | CD33 |
NCT03556982 | CAR‐T‐123 for Relapsed/ Refractory Acute Myelocytic Leukaemia(AML) 160 | Unknown | 1,2 | 10 | CD123 |
NCT03585517 | Safety and Efficacy Evaluation of IM23 CAR‐T Cells (IM23CAR‐T) 160 | Completed | 1 | 10 | IM23 |
NCT04658004 | NKG2D CAR‐T Cell Therapy for Patients with Relapsed and/or Refractory Acute Myeloid Leukaemia 160 | Not Yet Recruiting | Early 1 | 36 | NKG2D |
NCT05442580 | CAR‐T‐38 in Adult AML and MM Patients 160 | Recruiting | 1 | 36 | CD38 |
NCT03766126 | Lentivirally Redirected CD123 Autologous T Cells in AML 160 | Active, not recruiting | 1 | 12 | CD123 |
NCT03796390 | Study Evaluating Safety and Efficacy of CAR‐T Cells Targeting CD123 in Patients with Acute Myelocytic Leukaemia 160 | Unknown | 1 | 15 | CD123 |
NCT04156256 | CD123‐CD33 cCAR in Patients with Relapsed and/or Refractory, High Risk Hematologic Malignancies 160 | Unknown | Early 1 | 20 | CD123‐CD33 |
NCT04678336 | CD123 Redirected T Cells for AML in Paediatric Subjects 160 | Active, not recruiting | 1 | 12 | CD123 |
NCT05445011 | Anti‐FLT3 CAR‐T Cell (TAA05 Cell Injection) in the Treatment of Relapsed / Refractory Acute Myeloid Leukaemia 160 | Recruiting | 1 | 12 | FLT3 |
NCT04014881 | Safety and Efficacy of Anti‐CD123 CAR‐T Therapy in Patients with Refractory/ Relapsed CD123+ Acute Myeloid Leukaemia 160 | Unknown | 1 | 50 | CD123 |
NCT02799680 | Allogeneic CAR‐T‐33 for Relapsed/Refractory CD33+ AML 161 | Unknown | 1 | 12 | CD33 |
NCT04033302 | Multi‐CAR‐T Cell Therapy Targeting CD7‐positive Malignancies 161 | Recruiting | 1,2 | 30 | CD7 |
NCT05432401 | TAA05 Injection in the Treatment of Adult Patients with FLT3‐positive Relapsed/Refractory Acute Myeloid Leukaemia 161 | Recruiting | Early 1 | 18 | FLT3 |
NCT05463640 | Evaluate the Safety and Efficacy of ADGRE2 CAR‐T in Patients With R/R AML 161 | Not Yet Recruiting | 1 | 20 | ADGRE2 |
NCT03904069 | Study Evaluating the Safety, Tolerability, and Efficacy of FLT3 CAR‐T AMG 553 in FLT3‐positive Relapsed/Refractory AML 161 | Withdrawn | 1 | 0 | FLT3 |
NCT03631576 | CD123/CLL1 CAR‐T Cells for R/R AML (STPHI_0001) 161 | Unknown | 2,3 | 20 | CD123/CLL1 |
NCT05467202 | Evaluate the Safety and Efficacy of CLL1 CAR‐T in Patients With R/R AML 161 | Not Yet Recruiting | 1 | 20 | CLL1 |
NCT01864902 | Treatment of Relapsed and/or Chemotherapy Refractory CD33 Positive Acute Myeloid Leukaemia by CAR‐T‐33 161 | Unknown | 1,2 | 10 | CD33 |
NCT04884984 | Anti‐CLL1 CAR‐T Cell Therapy in CLL1 Positive Relapsed/Refractory Acute Myeloid Leukaemia (AML) 161 | Recruiting | 1,2 | 20 | CLL1 |
NCT03114670 | Donor‐derived Anti‐CD123 CAR‐T Cells for Recurred AML After Allo‐HSCT 161 | Unknown | 1 | 20 | CD123 |
NCT03971799 | Study of Anti‐CD33 Chimeric Antigen Receptor‐Expressing T Cells (CD33 CAR‐T) in Children and Young Adults with Relapsed/Refractory Acute Myeloid Leukaemia 161 | Recruiting | 1,2 | 37 | CD33 |
Note: CAR‐T cell therapy for AML on Clinicaltrials.gov as of 7 August 2023.
FIGURE 4.
Diagram of different target antigens of AML.
5.1. CD33
CD33, also known as SIGLEC3, is a single‐pass type I membrane protein that belongs to the immunoglobulin superfamily and SIGLEC (sialic acid binding Ig‐like lectin) family. 37 Studies have demonstrated that CD33, a myeloid differentiation antigen, is highly expressed in acute myeloid leukaemia progenitor cells, 38 but it also can be detected in normal haematopoietic stem cells. 39 Thus, it was used as a target for antibody‐based cancer therapy for years. Gemtuzumab ozogamicin (GO), the first antibody‐drug conjugate (ADC) for AML treatment, was approved by the FDA in 2017. 40 When used with other traditional drugs, the long‐term survival rate can reach over 70%. 39 The potential for long‐lasting marrow clearance by CD33 CAR‐T cells has led to deep concerns, and numerous methods are being investigated to reduce the side effects. Through blood cell restoration, bispecific CAR‐T or genetic modification of CD33 CAR‐T vector, CD33 CAR‐T clinical trials are currently undergoing. 37 , 41 , 42
5.2. CD123
CD123, the interleukin 3 (IL‐3) receptor alpha chain, is structurally a type I membrane protein with three extracellular areas, a transmembrane region and an intracellular portion. 43 CD123 was considered a potential target, highly expressed on leukaemia stem cells (LSCs) and acute myeloid leukaemia (AML) primordial cells like other AML markers. 44 CAR‐engineered T cells that target CD123 can cause myeloids to be cleared from immunodeficient mouse models of human AML reference. In vivo, co‐stimulated 4‐1BB CAR‐T cells were able to eliminate transplanted primary human AML cells, but a xenograft model revealed impaired haematopoiesis. 45 Research has revealed that 70% of AML cells express CD33 and CD123, which supports the case for targeting both of these molecules simultaneously in AML treatment. 46 Bispecific CARs with CD33 and CD123 single‐stranded variable fragments (ScFv) have superior proliferative capability and less HSC toxicity both in vitro and in vivo. 47 Many clinical studies including numerous antigens have been started based on preclinical data. 48 , 49 , 50 Yet, a significant issue with the development of AML CAR‐T treatments is the probable loss of proper bone marrow and haematopoietic precursor cells. 51 , 52 These antigens were selected because studies have consistently demonstrated that they express more strongly on AML cells than on healthy haematopoietic stem cells (HSCs), indicating that they have little effect on healthy haematopoiesis.
5.3. CLL‐1
CLL‐1, also known as CLEC12A, is a type II transmembrane glycoprotein that belongs to clade V of the C‐type lectin‐like receptor family. 53 According to research, the glycoprotein C‐type lectin‐like molecule‐1 (CLL‐1) expressed in myeloid cells, AML blasts and leukaemia stem cells, while hard to detect in haematopoietic stem cells. 54 AML cell lines and transgenic mice models have been used to test the efficiency of many CLL‐1 CAR‐T cells. Due to the powerful in vitro effects and eradication of leukaemic cells in mice models without significant myelotoxicity, CLL‐1 is theoretically a promising target for AML. Notably, CLL‐1 was also expressed in some early haematopoietic cells and normal monocytes, indicating the potential off‐target risk. 55 Both CLL‐1+ progenitor cells and mature granulocytes were effectively eliminated, albeit to varied degrees, after co‐culturing normal autologous HSCs with CLL‐1 CAR‐T cells from healthy normal donors. 56 Targeting CLL‐1 has significant drawbacks due to the antigen's differential expression in mature myeloid cells. The caspase‐9 suicide gene system was thus developed to control CLL‐1 CAR‐T cells and reduce the damage to normal myeloid cells. 57
5.4. CD70
CD70, a type 2 transmembrane glycoprotein from the tumour necrosis factor (TNF) family, is a compromising target for CAR‐T cell therapy in AML because it is present on both leukaemic blasts and leukaemia stem cells in AML patients. But different from CD33 or CD123, CD70 barely can be detected in normal HSCs. 58 In tumour cell lines and animal xenograft models, it was discovered that CD27z‐CARs, which comprise CD70 ligand and CD3z chain fusion, displayed superior proliferative and anticancer activity than CD70 scFv CARs while retaining normal HSCs. 59 The Marcela Maus laboratory evaluated the anti‐tumor activity of CD70 CAR‐T cells through the NSG Molm13 AML model and found that in co‐culture with AML, CD70‐CAR‐T cell activity was impaired by the soluble CD27 form was attenuated. 60 In response to the soluble ligand limitation, a series of new hinge CAR variants (truncated, deletion, flexible and ‘CD8 hinge & TM’) were evaluated, and the CD8 hinge and TM variants were found to have enhanced anti‐tumour activity and better expansion capacity in vitro and in vivo compared to natural 41BB‐based CAR‐T cells. A clinical trial using CD70 CAR‐T cells is currently being conducted for the therapy of individuals with CD70‐positive haematologic malignancies due to minimal myelotoxicity (NCT04662294).
5.5. CD38
CD38 is a type II transmembrane glycoprotein, one of the target antigens of AML, which is expressed in AML progenitor cells but not in normal human haematopoietic stem cells. 61 It is expressed in a variety of immune cells, including B‐lymphocytes, T‐lymphocytes, monocytes and NK cells, and influences tumour‐killing activity by regulating the release of cytokines. 62 The number and intensity of CD38 antigens, which are expressed by over 80% of AML cells, should be increased to achieve better results when designing CD38‐CAR‐T cells against these cells. Accordingly, CD38 expression in AML cells can be induced by all‐trans retinoic acid (ATRA), a treatment agent for acute promyelocytic leukaemia (APL). In preclinical research, all‐trans retinoic acid (ATRA) and CD38 CAR‐T cells were coupled to treat AML cell lines, and increased CD38 expression strengthened the cytotoxic effect on AML cell lines. 63 , 64 If ATRA is not available, the cytotoxic effect of CD38 CAR‐T cells is limited. 64 In another study, CD38 CAR‐T‐cell therapy was administered to six patients with AML who relapsed after Allo‐HSCT, 65 and the study showed that CR was achieved in four cases, grades 2–3 CRS were seen in 88% of cases, and grades III and IV haematologic toxicity was seen in all three patients.
5.6. Le Y
Lewis Y (Le Y) is a carbohydrate tumour‐associated antigen that promotes tumour survival, invasion and metastasis. 66 Le Y is abundantly expressed in haematologic tumours, including acute myeloid leukaemia, and is minimally expressed in healthy tissues, making Le Y a potentially suitable target for the treatment of AML. 67 In a preclinical study, investigators tested the effectiveness of anti‐Le Y CAR‐T cells against AML and MM (multiple myeloma) cells. 67 The Peter MacCallum Cancer Center is conducting an important clinical study evaluating the potential of CAR‐T therapy in patients with AML, with a phase I clinical trial demonstrating the feasibility and safety of targeting Lewis Y and CAR‐T cells in skin and bone marrow in proven leukaemia for up to 10 months. 68 It has been reported that Le Y CAR‐T cells specifically target Le Y+ cells by producing interferon‐γ. Neeson et al. designed anti‐Lewis Y CAR‐T cells using autologous T cells and found that high levels of interferon‐γ were produced in the killing of Lewis Y‐positive AML cells. 69
5.7. CD7
Transmembrane glycoprotein CD7, which is produced by T cells, NK cells and cord blood myeloid progenitor cells, has a synergistic stimulatory effect on the contacts between B and T cells during lymphatic development. AML cells express CD7, while healthy myeloid cells do not, and CD7 is not harmful to normal cells. 70 Consequently, it might be a suitable target for the elimination of malignant cells. since T cells also express CD7, removal of the CD7 gene in T cells is critical for successful treatment. In two experiments, Gomes‐Silva et al. used a transgenic model of AML and CD7 CAR‐T cells to target CD7+ tumour cells. 71 The CD7 gene was deleted from primary activated T cells using a CRISPR/Cas9 technique before producing CAR‐T cells. Then, by employing scFv generated from anti‐CD7 antibodies, second‐generation CD28‐ CD3‐CD7 CAR ‐T cells were developed with de novo CD7 knockout. Surprisingly, the study's findings revealed that their CAR‐T cells had a strong killing impact on AML, significantly eliminating both primary AML cells and leukaemic colony‐forming cells while causing no harm to healthy cells or erythrocytes. Therefore, CD7 CAR‐T cells may be a potential successful therapy for refractory or relapsed AML.
5.8. FLT‐3
FMS‐like tyrosine kinase‐3 (Flt‐3) is a glycosylated protein of the class III receptor tyrosine kinase family, 72 which is mainly expressed on haematopoietic stem cells and myeloid cells and participates in normal haematopoiesis by controlling cell survival, proliferation and differentiation. 73 Flt3 expression is increased on primitive cells in 70% of AML patients, and 30% of AML patients are associated with mutations in Flt3. 74 Midostaurin played a favourable role in suppressing AML and improving survival when combined with chemotherapeutic agents. 75 In a preclinical study, investigators evaluated the killing effect of second‐generation Flt3‐41BB CAR‐T cells in a xenograft mouse model and found that the mice prolonged their lives after input of Flt3‐41BB CAR‐T cells. 76 In another preclinical study, second‐generation CAR‐T cells with Flt3 ligands prolonged the survival of Flt3 mutant mice, showing limited myelotoxicity. 64 , 77
5.9. B7‐H3
B7‐H3, also known as CD273, is a type I transmembrane protein expressed in a variety of cancer types including AML mother cells. 78 As a T‐lymphocyte stimulating protein, B7‐H3 reduces T‐cell cytotoxicity and promotes immune evasion within cancer cells. 79 Lichtman et al. evaluated the ability of B7‐H3‐specific CAR‐T cells to combat leukaemia in an in vitro xenograft model, which was found to be cytotoxic and show significant interferon‐γ and IL‐2 release in AML cell lines at an E: T ratio of 1:5. 80 Yang et al. designed tandem anti‐B7‐H3 and anti‐CD70 CAR‐T cells, and both in vitro and in vivo experiments showed significant antigenic toxicity but little damage to haematopoietic cells. 81 More research is needed on B7‐H3 as a potential CAR‐T cell target for the treatment of AML.
5.10. TIM‐3
T‐cell immunoglobulin mucin‐3 (Tim‐3) is a transmembrane protein belonging to the TIM family that is expressed on almost all forms of AML primitive cells but not on HSCs, making it a promising target. 82 The main cause of leukaemia relapse after chemotherapy is resistance to LSCs because they cannot target the elimination of LSCs. In one study, researchers created a phage library from which anti‐human TIM antibodies were isolated and evaluated the killing ability of second‐generation TIM3 CAR‐T cells against leukaemia cell lines and primary AML cells in vitro and found that they had good anti‐leukaemia activity. 83 In another study, Xin He et al. evaluated the therapeutic efficacy of TIM‐3 CAR‐T cells in vivo through a xenograft model and found that the levels of cytotoxic agents, such as interferon‐gamma, granzyme B, perforin, and interleukin 5, were markedly increased, and that the TIM‐3 CAR‐T cells were effective in killing LSCs. 84 Effective CAR‐T therapies are being developed one after another, and some studies have shown that CD13 and TIM3 bispecific CAR‐T cells have a better killing effect on AML, and also reduce the cytotoxicity of stem cells and peripheral myeloid cells. 84
5.11. CD44v6
CD44v6 is a splice variant of the hyaluronan CD44 receptor and belongs to class I membrane glycoproteins. It is highly expressed in other haematologic malignancies, including acute myeloid leukaemia, and is not expressed on healthy progenitor cells, and haematopoietic stem cells 85 , 86 ; therefore, CD44v6 is expected to be an effective target antigen for AML therapy. Casucci et al. evaluated the killing ability of second‐generation CD44v6 CAR‐T cells targeting CD44+ AML cells using scFv with humanized anti‐CD44v6 single‐chain antibody. The results showed that CD44v6 CAR ‐T cells produced anti‐tumour cytokines and effectively cleared CD44+ tumour cells. 86 Notably, CD44v6 CAR‐T cells produced haematological toxicity, so‐called extra‐tumour toxicity, to monocytes during the selective removal of tumour cells. To avoid this side effect, non‐immune inducible Caspase9 (IC9) and thymidine kinase suicide genes were used to efficiently eliminate CAR‐T cells on CD44v6 CAR‐T cells. 87 DNA methylation has been associated with CD44v6 expression in AML cells and T‐cell exhaustion and function, and in AML therapy, the hypomethylating agents (HAMs) decitabine (Dec) and azacitidine (Aza) have been widely used. 88 , 89 A recent study has shown that combining Dec or Aza with CD44v6 CAR‐T cells improves the function of CD44v6 CAR‐T cells and enhances anti‐tumour capacity against AML. 90
5.12. CD93
CD93, also known as C1QR1, is a cell surface C‐type lectin transmembrane receptor protein that is differentially expressed in approximately 55% of AML patients, but not in HSC or other precursor myeloid cells. 91 CD93 plays an important role in host defence and intercellular adhesion. Recently, Richards et al. developed a new humanized CD93‐specific binding protein and evaluated the role of CD93 CAR‐T cells in leukaemia, and in vivo experiments showed that CD93 CAR‐T cells specifically cleared AML but not HSPC. 92 Notably, human endothelial cells express CD93, so CD93 CAR‐T cells exert a targeted, non‐tumorigenic toxic effect on vascular endothelial cells. To limit the cytotoxic effects of CAR‐T cell recognition and killing, Richards et al. recently developed a non‐gated anti‐CD93 CAR‐T cell. 92
5.13. CD99
CD99 is a transmembrane glycoprotein and is strongly expressed in the majority of AML. CD99 is a marker expressed on the surface of leukaemic stem cells but not normal haematopoietic stem cells, thus can be a promising therapeutic target for AML. 16 , 93 Previous studies show that expression of CD99 can separate LSCs from functionally normal HSCs in AML and have higher CD99 levels in relapse AML blasts. 94 CD99 single‐chain variable fragment antibody was proved to induce apoptotic cell death in AML cell lines and extended the survival time of AML xenograft mouse model. 95 Currently, purine nucleotide analogs clofarabine and cladribine that can inhibit CD99 dimerization were approved by the FDA to treat Ewing's sarcoma. 96 , 97 CD99 drugs for AML are still under development.
6. EXPRESSION OF AML TARGETS
Annotation of antigen expression can predict potential targeted tumour effects. We performed expression analysis of the above AML targets based on the Gene Expression Profiling Interactive Analysis (GEPIA) database in the hope of helping researchers select appropriate antigens. The results showed that the expression of CD33, CD123, CLL‐1, CD38, CD7, FLT‐3, B7‐H3, TIM‐3, CD44V6, CD93 and CD99 was significantly higher than that of normal tissues in tumour tissues, and the expression of CD70 and Le Y in tumour tissues was not significantly different from that of normal tissues (Figure 5). Ideal targets should ensure that CAR‐T cells specifically kill tumour cells while not harming or rarely harming normal cells, which is difficult to achieve in practice. Analysing the expression of various potential targets can provide researchers with some ideas, such as using two single‐targeted CAR‐T cells in combination, dual‐targeted CAR‐T cells, triple‐targeted CAR‐T cells and tandem CAR‐T cells, to overcome off‐target effects, antigen escape, and other problems.
FIGURE 5.
Expression analysis of AML targets in the GEPIA database. The filter conditions are p‐value <0.05 and the absolute value of the difference (| log2 (Fold Change) |) > 1.
7. LIMITATIONS AND CHALLENGES OF CAR‐T FOR AML
Despite the tremendous development of CAR‐T cells in the treatment of haematologic malignancies, however, CAR‐T cells still face many problems in the treatment of AML, such as target antigen loss, off‐target effects, cytokine release syndrome (CRS) and neurotoxicity, CAR‐T persistence, preclinical models and CAR‐T production. We summarize the current dilemmas facing CAR‐T cell therapy for AML (Figure 6).
FIGURE 6.
Limitations and challenges of CAR‐T cell therapy for acute myeloid leukaemia.
7.1. Antigen loss
The toxic effect of CAR‐T cells on tumours is dependent on the recognition and binding of CAR antigens, and loss or down‐regulation of target antigens results in AML progenitor cells evading immune surveillance, thus limiting the killing effect of CAR‐T cells. 98 Despite the CAR‐T cell therapy's fast advancement, treatment resistance has grown to be a significant barrier to its use in clinical settings. The cause of most resistance in CAR‐T cell therapy is target antigen loss. The CAR study CTL109 by Novartis (NCT01626495) demonstrated an amazing 93% efficiency. 99 Yet, the likelihood of a complete response after 1 year has dropped to about 55%, indicating that drug resistance developed in about 50% of patients during this period. This implies that any CAR‐T cell therapy may not be effective enough if it just targets one antigen.
7.2. On–Off‐target effects
On–off‐target effects include normal cell on‐target and tumour cell off‐target. Normal cell on target means that normal cells expressing target antigens are inevitably killed by CAR‐T, whereas tumour cell off‐target means that the density of target antigens expressed by tumour cells is reduced or lost, resulting in the inability of CAR‐T to recognize and attack tumour cells. For example, CD19‐CAR‐T cells recognize and kill B‐cell lymphoma and leukaemia cells while also killing normal CD19‐positive B‐cells in the patient. Due to the target antigens' high levels of expression on normal HSC, off‐target consequences such as haematological toxicity and protracted myeloid elimination are some of the main drawbacks of CAR‐T cells in the therapy of AML. 100 B‐cell malignancies' CAR‐T cells specifically target CD19 and their depletion can be managed without endangering life. Acute myeloid leukaemia lacks a disposable antigen, unlike B‐cell malignancies, and targeting myeloid antigens causes long‐term myelotoxicity. Prolonged myelotoxicity raises the risk of sepsis as well as invasive fungal infections, which is clinically undesirable.
7.3. CRS and neurotoxicity
Systemic immune inflammation in which patients rapidly produce and secrete inflammatory cytokines after injection of CAR‐T cells is called CRS. CRS is one of the major side effects of CAR‐T cell therapy because when CAR‐T cells exert their effector functions, large amounts of cytokines, such as IL‐6, IFN‐γ, IL‐1β, GM‐CSF and TNF‐α production, cause a systemic inflammatory response, leading to signs such as hypoxia, fever, myalgia, hypotension and neurologic dysfunction, which threaten patients' lives. 101 , 102 The main mechanism by which CRS occurs is the disruption of the balance between pro‐ and anti‐inflammatory in the body, where monocytes are activated and secrete cytokines such as IL‐6 and IL‐1 in response to the interaction of CAR‐T cells and tumour cells, followed by peripheral molecules that initiate the activation of endothelial cells and initiate pro‐inflammatory pathways, thus disrupting the balance between pro‐ and anti‐inflammatory. 103 Neurotoxicity generally occurs a few days after the onset of CRS, when excessive cytokines diffuse in the brain, and patients may experience adverse conditions, such as mild headaches, hallucinations, confusion, psychosis and seizures. 104 , 105
7.4. CAR‐T persistence
Clinical trials have confirmed that reduced CAR‐T cell persistence is one of the major reasons for CAR‐T cell therapy failure. CAR‐T cells do not sustain their tumour‐killing effect in patients due to immunorecognition of CAR‐derived exogenous peptides and subsequent disruption of immune‐mediated modification of T‐lymphocytes. 106 , 107 CAR‐T cells in patients must be used for a longer period to destroy AML cells. According to data from the original CD19 CAR in the juvenile B‐cell ALL research, the ideal length of CAR‐T cells to prolong remission is up to 8–10 months, which seems unlikely in AML patients given the danger of infection and its possibly fatal consequences.
7.5. Preclinical models
Fundamental in vitro processes including proliferation, antigen‐targeted lysis, cytokine production and allergen stimulation tests are examples of quantitative processes that may not be able to properly predict in vivo performance. Utilizing preclinical animal models of adaptive T cell transfer for transplantation, the CAR‐T cell design was validated. The immunological microenvironments in mice and human lymphoid organs are so different from one another, however, that these mouse models do not always adequately describe human T cell function, despite their initial effectiveness in removing tumour cells. 108
7.6. CAR‐T production
Unlike other immunotherapeutic methods, CAR‐T cell therapy is highly personalized, which requires the isolation of T cells from the patient and their modification and transformation in vitro by genetic engineering before being infused back into the patient's body. CAR‐T cells cannot be mass‐produced or generalized, and the constraints in the whole process of CAR‐T production and treatment include cost, raw materials, equipment, environment, etc. CAR‐T cells can be costly due to the complex preparation process. Culture media and reagents used to better enable cell expansion are not yet standardized, and raw materials and reagents may be difficult to obtain if manufactured according to GMP specifications. A variety of advanced instruments and techniques are required to genetically modify T cells. In addition, a highly sterile and controlled environment and continuous monitoring are essential for patients undergoing CAR‐T cell therapy. These constraints are undoubtedly challenging for CAR‐T cell therapy for haematologic tumours. 109 , 110
8. OPTIMIZATION STRATEGIES RELATED TO CAR‐T FOR AML
To address the many problems faced in CAR‐T cell therapy for AML, several studies have been undertaken to improve the efficacy of CAR‐T cells. Such as CAR structure optimization, multi‐targeted CAR‐T cells, universal CAR‐T, combination therapy and immunosuppression of tumour microenvironment. We summarize current optimization strategies related to enhancing CAR‐T efficacy (Figure 7).
FIGURE 7.
Schematic diagram of optimization strategy for CAR‐T cell therapy for AML.
8.1. CAR structure optimization
Two important features of successful CAR‐T cell therapy are durability and efficacy, and factors such as weak CARS structure, lack of co‐stimulatory molecules, and insufficient expansion or activation in vitro and in vivo can affect the durability and efficacy of CAR‐T. CD28 or 4‐1BB stimulation of structural domains can enhance the durability of CAR‐T cells and their function. 111 , 112 The third‐generation CAR showed a strong anti‐tumour response compared to the first‐ and second‐generation CARs. 113 CD27 and CD28 are the two most important co‐stimulatory receptors for T‐cell differentiation, with CD28 co‐stimulatory signals mainly triggering cell proliferation and CD27 co‐stimulatory signals promoting cell survival and differentiation. CAR‐T cells using CD27 co‐stimulators are impressive in terms of safety. Another beneficial co‐stimulatory molecule, TLR2, has also been used to enhance the anti‐tumour activity of CAR‐T cells. 114 Notably, CAR‐T cells with MyD88/CD40 structural domains showed potential killing effects on tumour cells. 115 For the survival and effectiveness of CAR‐T cells to become better, powerful co‐stimulatory molecules arming CAR‐T cells or modifying optimized CAR‐T cells are necessary.
8.2. Multi‐targeted CAR‐T cells
CAR‐T cells targeting only one antigen cannot be used in all AML patients because tumour‐associated antigens (TAAs) on the surface of tumour cells are different in different pathologies, and the TAAs targeted by CAR‐T cells are also present in normal tissue cells, resulting in the risk of the inadvertent killing of their normal cells during the treatment process. In addition, the escape, reduction or loss of tumour antigens can directly reduce the effectiveness of CAR‐T cells. To overcome the problems of off‐target effect and antigen escape, a multi‐targeted CAR‐T cell strategy has been introduced into clinical trials. 33 , 116 Multi‐targeted CAR‐T cell applications include dual‐targeted CAR‐T cells, triple‐targeted CAR‐T cells, a combination of two single‐targeted CAR‐T cells, and tandem CAR‐T cells. Dual‐targeted and triple‐targeted CAR‐T cells refer to the design of two or three separate CARs on a single T‐lymphocyte, each targeting a different antigen, thus providing dual or triple targeting. The combination of two CAR‐T cells refers to two single‐targeted CAR‐T cells, each CAR targeting a separate antigen to improve tumour recognition. Tandem CAR‐T cells are two different scFv regions within one CAR, each targeting a different antigen. The researchers used Boolean logic operations to elucidate how these various types of multi‐targeted CAR‐T cells function, including ‘AND’, ‘OR’ and ‘NOT’. CAR‐T cells with ‘AND’ are activated only if both target antigens are recognized, CAR‐T cells with ‘OR’ need either of the two target antigens to be recognized to be activated, and CAR‐T cells with ‘NOT’ avoid killing normal cells by stopping when a specific antigen is detected. 117 Therefore, the Boolean logic operations ‘AND’, ‘OR’ and ‘NOT’ in logic gates can improve the recognition and killing ability of CAR‐T cells on tumour cells and can also effectively avoid off‐target effects.
8.3. Universal CAR‐T cells
Based on the second‐generation CAR‐T, Cartellieri et al. designed a flexible CAR platform, the universal CAR‐T cell, utilizing CD28 as a co‐stimulatory structural domain. 118 The universal CAR‐T cell has a switch structure consisting of two components that can be utilized to regulate the function of the CAR‐T cell. Notably, no extra‐tumour toxicity or xenograft‐versus‐host response was demonstrated in experiments using the universal CAR dual‐targeting CD33 and CD123 to lysate AML primary cells and AML cell lines. Graft‐versus‐host response (GVHR) or host‐versus‐graft response (HVGR) affects CAR‐T therapeutic efficacy, and the development of universal CAR‐T cell therapies necessarily addresses the immune rejection induced by allogeneic cell therapy. A commonly adopted means is to knock out the TCR of CAR‐T through gene editing technology to reduce its recognition and attack on patients and reduce the risk of GvHD. Knockdown of HLA‐like molecules responsible for antigen presentation reduces the immunogenicity of allogeneic‐derived CAR‐T cells and enhances their killing and expansion activities. 119 , 120
8.4. Combination therapy
Haematopoietic stem cell transplantation (HSCT), oncolytic viruses, checkpoint inhibitors and chemotherapeutic agents in combination with CAR‐T cells have shown great potential as several new therapeutic strategies in the treatment of human haematological malignancies.CD33 CAR‐T cells in combination with Allo‐HSCT have shown less haematopoietic toxicity in patients with CD33+ AML.CD33 CAR‐T cells eliminated CD33+ primitive cells without effect on CD33‐haematopoietic stem cells (HSPC). 121 , 122 The use of chemokine‐encoding gene vectors of lysosomal viruses allows for the recruitment of more CAR‐T cells, prolonging the duration of CAR‐T cells and directly attacking malignant cells, thereby increasing the durability and effectiveness of CAR‐T cells. 123 , 124 PD‐1, or programmed death receptor 1, is the most common immune checkpoint identified for T cells. PD‐1 regulates the immune system by down‐regulating the immune system's response to human cells and by suppressing the inflammatory activity of T cells. PD‐1 inhibitors have been the hottest method of tumour immunotherapy in recent years. By blocking the PD‐1/PD‐L1 signalling pathway with PD‐1 inhibitors, CAR‐T cells can target PD‐L1‐expressing tumour cells and play the role of tumour killing. In addition, the combination of corticosteroids and tocilizumab with CAR‐T cell therapy can reduce side effects in patients. 125 Both drugs rapidly reverse CRS symptoms in most patients, but the long treatment interval between the two drugs Precise regulation of CAR‐T cell proliferation and differentiation is also needed to mitigate CRS. Siltuximab, an IL‐6 antagonist monoclonal antibody that prevents IL‐6 translocation from the blood–brain barrier (BBB), also plays an important role in managing neurotoxicity. 126
8.5. Immunosuppressive tumour microenvironment
Tumour microenvironment (TME) refers to the surrounding microenvironment in which tumour cells exist, including surrounding blood vessels, immune cells, fibroblasts, bone marrow‐derived inflammatory cells, various signalling molecules and extracellular matrix (ECM). Depletion and decreased activity of CAR‐T cells are strongly associated with immunosuppression of TME. 101 , 127 Synergistic treatment with checkpoint inhibitors and CAR‐T cells improves the function of CAR‐T cells in TME and helps to overcome the resistance of cancer cells to treatment. Extensive transcriptional and epigenetic alterations and increased expression of inhibitory receptors lead to T cell exhaustion. 128 Reducing CAR‐T cell depletion as well as enhancing the persistence of CAR‐T cells are necessary for better therapeutic outcomes. Increased expression of nuclear receptor subfamily 4A (NR4A) and thymocyte selection‐associated high mobility group box proteins 1 and 2 (TOX and TOX2) transcription factors can up‐regulate genes encoding repressor molecules to induce T cell exhaustion. It has been shown that CAR‐T cells knocking down the NR4A gene or lacking both TOX and TOX2 can reduce CAR‐T cell depletion in TME and have enhanced anticancer activity. 129 , 130
9. SUMMARY AND OUTLOOK
In recent years, a new class of experimental medicines known as CAR‐T cell therapy has emerged, showing promise in treating haematologic malignancies. Approximately 190 clinical studies are now being conducted in the field of immunotherapy. The outcomes of these investigations have been incorporated into phase I clinical trials. Functional evaluation of CAR‐T cells has advanced. With each study, a significant advancement in our comprehension of CAR‐T cell biology has been made. Despite the early clinical success of CAR‐T in the treatment of AML, research in this field is still in its early stages, and the high risk of relapse and variable levels of toxicity connected with CAR‐T therapy calls for more advancements in this technology. Examples include refining the CAR structure, studying new targets, modifying the immune milieu, creating universal CAR‐T treatment, using combination therapies, and other tactics. Our ongoing efforts are focused on improving targeted killing, combating targeted tumour antigen escape, decreasing the rate of recurrence, and minimizing harmful side effects. In the future, we think that more investigations by researchers will benefit tumour patients.
AUTHOR CONTRIBUTIONS
Chi Gao: Data curation (equal); formal analysis (equal); investigation (equal); methodology (equal); resources (equal); writing – original draft (equal). Xin Li: Data curation (supporting); formal analysis (supporting); resources (supporting); writing – original draft (supporting). Yao Xu: Formal analysis (supporting); funding acquisition (equal); project administration (supporting); resources (supporting); writing – review and editing (equal). Tongcun Zhang: Formal analysis (equal); funding acquisition (lead); project administration (equal); supervision (equal); writing – review and editing (equal). Haichuan Zhu: Project administration (equal); resources (equal); supervision (equal); validation (equal); writing – review and editing (equal). Di Yao: Conceptualization (lead); data curation (lead); formal analysis (lead); funding acquisition (equal); investigation (lead); project administration (lead); resources (lead); supervision (lead); validation (lead); writing – original draft (lead); writing – review and editing (lead).
FUNDING INFORMATION
This work was supported by a grant from the Wuhan Science and Technology Plan Project (2019030703011533), Wuhan East Lake High‐tech Zone ‘JieBangGuaShuai’ Project (2022KJB113), and Hubei Province Supporting Enterprise Technology Innovation Development Project (2021BAB12).
CONFLICT OF INTEREST STATEMENT
The authors confirm that there are no conflicts of interest.
Gao C, Li X, Xu Y, Zhang T, Zhu H, Yao D. Recent advances in CAR‐T cell therapy for acute myeloid leukaemia. J Cell Mol Med. 2024;28:e18369. doi: 10.1111/jcmm.18369
DATA AVAILABILITY STATEMENT
The datasets used and/or analysed during the current study are available from the corresponding authoron reasonable request.
REFERENCES
- 1. Shallis RM, Wang R, Davidoff A, Ma X, Zeidan AM. Epidemiology of acute myeloid leukemia: recent progress and enduring challenges. Blood Rev. 2019;36:70‐87. doi: 10.1016/j.blre.2019.04.005 [DOI] [PubMed] [Google Scholar]
- 2. Scott BL, Pasquini MC, Fei M, et al. Myeloablative versus reduced‐intensity conditioning for hematopoietic cell transplantation in acute myelogenous leukemia and myelodysplastic syndromes‐Long‐term follow‐up of the BMT CTN 0901 clinical trial. Transplant Cell Ther. 2021;27:483.e1‐483.e6. doi: 10.1016/j.jtct.2021.02.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Zeidner JF, Vincent BG, Ivanova A, et al. Phase II trial of pembrolizumab after high‐dose cytarabine in relapsed/refractory acute myeloid leukemia. Blood Cancer Discov. 2021;2:616‐629. doi: 10.1158/2643-3230.Bcd-21-0070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Daver N, Konopleva M, Maiti A, et al. Phase I/II study of Azacitidine (AZA) with Venetoclax (VEN) and Magrolimab (Magro) in patients (pts) with newly diagnosed older/unfit or high‐risk acute myeloid leukemia (AML) and relapsed/refractory (R/R) AML. Blood. 2021;138:371. doi: 10.1182/blood-2021-153638 [DOI] [Google Scholar]
- 5. Uy GL, Aldoss I, Foster MC, et al. Flotetuzumab as salvage immunotherapy for refractory acute myeloid leukemia. Blood. 2021;137:751‐762. doi: 10.1182/blood.2020007732 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Badar T, Shah NN. Chimeric antigen receptor T cell therapy for acute lymphoblastic leukemia. Curr Treat Options Oncol. 2020;21:16. doi: 10.1007/s11864-020-0706-6 [DOI] [PubMed] [Google Scholar]
- 7. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene Ciloleucel CAR T‐cell therapy in refractory large B‐cell lymphoma. N Engl J Med. 2017;377:2531‐2544. doi: 10.1056/NEJMoa1707447 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Park JH, Rivière I, Gonen M, et al. Long‐term follow‐up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med. 2018;378:449‐459. doi: 10.1056/NEJMoa1709919 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Ren A, Tong X, Xu N, Zhang T, Zhou F, Zhu H. CAR T‐cell immunotherapy treating T‐ALL: challenges and opportunities. Vaccines (Basel). 2023;11:165. doi: 10.3390/vaccines11010165 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Zhang P, Yang X, Cao Y, et al. Autologous stem cell transplantation in tandem with anti‐CD30 CAR T‐cell infusion in relapsed/refractory CD30(+) lymphoma. Exp Hematol Oncol. 2022;11:72. doi: 10.1186/s40164-022-00323-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Zhang S, Gu C, Huang L, et al. The third‐generation anti‐CD30 CAR T‐cells specifically homing to the tumor and mediating powerful antitumor activity. Sci Rep. 2022;12:10488. doi: 10.1038/s41598-022-14523-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Wei J, Xiao M, Mao Z, et al. Outcome of aggressive B‐cell lymphoma with TP53 alterations administered with CAR T‐cell cocktail alone or in combination with ASCT. Signal Transduct Target Ther. 2022;7:101. doi: 10.1038/s41392-022-00924-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Cao Y, Xiao Y, Wang N, et al. CD19/CD22 chimeric antigen receptor T cell cocktail therapy following autologous transplantation in patients with relapsed/refractory aggressive B cell lymphomas. Transplant Cell Ther. 2021;27:910.e1‐910.e911. doi: 10.1016/j.jtct.2021.08.012 [DOI] [PubMed] [Google Scholar]
- 14. Wei J, Mao Z, Wang N, et al. Long‐term outcomes of relapsed/refractory double‐hit lymphoma (r/r DHL) treated with CD19/22 CAR T‐cell cocktail therapy. Clin Transl Med. 2020;10:e176. doi: 10.1002/ctm2.176 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Wang N, Hu X, Cao W, et al. Efficacy and safety of CAR19/22 T‐cell cocktail therapy in patients with refractory/relapsed B‐cell malignancies. Blood. 2020;135:17‐27. doi: 10.1182/blood.2019000017 [DOI] [PubMed] [Google Scholar]
- 16. Shi J, Zhang Z, Cen H, et al. CAR T cells targeting CD99 as an approach to eradicate T‐cell acute lymphoblastic leukemia without normal blood cells toxicity. J Hematol Oncol. 2021;14:162. doi: 10.1186/s13045-021-01178-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Hofmann S, Schubert ML, Wang L, et al. Chimeric antigen receptor (CAR) T cell therapy in acute myeloid leukemia (AML). J Clin Med. 2019;8:200. doi: 10.3390/jcm8020200 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Cao Y, Efetov SK, He M, et al. Updated clinical perspectives and challenges of chimeric antigen receptor‐T cell therapy in colorectal cancer and invasive breast cancer. Arch Immunol Ther Exp (Warsz). 2023;71:19. doi: 10.1007/s00005-023-00684-x [DOI] [PubMed] [Google Scholar]
- 19. Mavi AK, Gaur S, Gaur G, et al. CAR T‐cell therapy: reprogramming patient's immune cell to treat cancer. Cell Signal. 2023;105:110638. doi: 10.1016/j.cellsig.2023.110638 [DOI] [PubMed] [Google Scholar]
- 20. Chen YJ, Abila B, Mostafa Kamel Y. CAR‐T: what is next? Cancers (Basel). 2023;15:663. doi: 10.3390/cancers15030663 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Brudno JN, Somerville RPT, Shi V, et al. Allogeneic T cells that express an anti‐CD19 chimeric antigen receptor induce remissions of B‐cell malignancies that Progress after allogeneic hematopoietic stem‐cell transplantation without causing graft‐versus‐host disease. J Clin Oncol. 2016;34:1112‐1121. doi: 10.1200/jco.2015.64.5929 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Cheadle EJ, Gornall H, Baldan V, Hanson V, Hawkins RE, Gilham DE. CAR T cells: driving the road from the laboratory to the clinic. Immunol Rev. 2014;257:91‐106. doi: 10.1111/imr.12126 [DOI] [PubMed] [Google Scholar]
- 23. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin‐T‐cell receptor chimeric molecules as functional receptors with antibody‐type specificity. Proc Natl Acad Sci U S A. 1989;86:10024‐10028. doi: 10.1073/pnas.86.24.10024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Au R. Immunooncology: can the right chimeric antigen receptors T‐cell design Be made to cure all types of cancers and will it Be covered? J Pharm. 2017;2017:7513687. doi: 10.1155/2017/7513687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Brown CE, Alizadeh D, Starr R, et al. Regression of glioblastoma after chimeric antigen receptor T‐cell therapy. N Engl J Med. 2016;375:2561‐2569. doi: 10.1056/NEJMoa1610497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Alnefaie A, Albogami S, Asiri Y, et al. Chimeric antigen receptor T‐cells: An overview of concepts, applications, limitations, and proposed solutions. Front Bioeng Biotechnol. 2022;10:797440. doi: 10.3389/fbioe.2022.797440 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Sadelain M, Rivière I, Brentjens R. Targeting tumours with genetically enhanced T lymphocytes. Nat Rev Cancer. 2003;3:35‐45. doi: 10.1038/nrc971 [DOI] [PubMed] [Google Scholar]
- 28. Hombach A, Wieczarkowiecz A, Marquardt T, et al. Tumor‐specific T cell activation by recombinant immunoreceptors: CD3 zeta signaling and CD28 costimulation are simultaneously required for efficient IL‐2 secretion and can be integrated into one combined CD28/CD3 zeta signaling receptor molecule. J Immunol (Baltimore, Md: 1950). 2001;167:6123‐6131. doi: 10.4049/jimmunol.167.11.6123 [DOI] [PubMed] [Google Scholar]
- 29. Marofi F, Rahman HS, al‐Obaidi ZMJ, et al. Novel CAR T therapy is a ray of hope in the treatment of seriously ill AML patients. Stem Cell Res Ther. 2021;12:465. doi: 10.1186/s13287-021-02420-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Jayaraman J, Mellody MP, Hou AJ, et al. CAR‐T design: elements and their synergistic function. EBioMedicine. 2020;58:102931. doi: 10.1016/j.ebiom.2020.102931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Huang Z, Zhang C, Xing XH. Design and construction of chimeric linker library with controllable flexibilities for precision protein engineering. Methods Enzymol. 2021;647:23‐49. doi: 10.1016/bs.mie.2020.12.004 [DOI] [PubMed] [Google Scholar]
- 32. Hudecek M, Sommermeyer D, Kosasih PL, et al. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol Res. 2015;3:125‐135. doi: 10.1158/2326-6066.Cir-14-0127 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Elahi R, Khosh E, Tahmasebi S, Esmaeilzadeh A. Immune cell hacking: challenges and clinical approaches to create smarter generations of chimeric antigen receptor T cells. Front Immunol. 2018;9:1717. doi: 10.3389/fimmu.2018.01717 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Marofi F, Tahmasebi S, Rahman HS, et al. Any closer to successful therapy of multiple myeloma? CAR‐T cell is a good reason for optimism. Stem Cell Res Ther. 2021;12:217. doi: 10.1186/s13287-021-02283-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Yang J, Zhou W, Li D, Niu T, Wang W. BCMA‐targeting chimeric antigen receptor T‐cell therapy for multiple myeloma. Cancer Lett. 2023;553:215949. doi: 10.1016/j.canlet.2022.215949 [DOI] [PubMed] [Google Scholar]
- 36. Melenhorst JJ, Chen GM, Wang M, et al. Decade‐long leukaemia remissions with persistence of CD4(+) CAR T cells. Nature. 2022;602:503‐509. doi: 10.1038/s41586-021-04390-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Tambaro FP, Singh H, Jones E, et al. Autologous CD33‐CAR‐T cells for treatment of relapsed/refractory acute myelogenous leukemia. Leukemia. 2021;35:3282‐3286. doi: 10.1038/s41375-021-01232-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Liu Y, Wang S, Schubert ML, et al. CD33‐directed immunotherapy with third‐generation chimeric antigen receptor T cells and gemtuzumab ozogamicin in intact and CD33‐edited acute myeloid leukemia and hematopoietic stem and progenitor cells. Int J Cancer. 2022;150:1141‐1155. doi: 10.1002/ijc.33865 [DOI] [PubMed] [Google Scholar]
- 39. 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: 10.1038/leu.2015.52 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Swaminathan M, Cortes JE. Update on the role of gemtuzumab‐ozogamicin in the treatment of acute myeloid leukemia. Ther Adv Hematol. 2023;14:20406207231154708. doi: 10.1177/20406207231154708 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. 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: 10.1038/mt.2014.164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Sallman DA, Elmariah H, Sweet K, et al. Phase 1/1b safety study of Prgn‐3006 Ultracar‐T in patients with relapsed or refractory CD33‐positive acute myeloid leukemia and higher risk myelodysplastic syndromes. Blood. 2021;138:825. doi: 10.1182/blood-2021-152692 [DOI] [Google Scholar]
- 43. Testa U, Pelosi E, Castelli G. CD123 as a therapeutic target in the treatment of hematological malignancies. Cancers (Basel). 2019;11:1358. doi: 10.3390/cancers11091358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Jordan CT, Upchurch D, Szilvassy SJ, et al. The interleukin‐3 receptor alpha chain is a unique marker for human acute myelogenous leukemia stem cells. Leukemia. 2000;14:1777‐1784. doi: 10.1038/sj.leu.2401903 [DOI] [PubMed] [Google Scholar]
- 45. Liu K, Zhu M, Huang Y, Wei S, Xie J, Xiao Y. CD123 and its potential clinical application in leukemias. Life Sci. 2015;122:59‐64. doi: 10.1016/j.lfs.2014.10.013 [DOI] [PubMed] [Google Scholar]
- 46. Qi H, Ratnam M. Synergistic induction of folate receptor beta by all‐trans retinoic acid and histone deacetylase inhibitors in acute myelogenous leukemia cells: mechanism and utility in enhancing selective growth inhibition by antifolates. Cancer Res. 2006;66:5875‐5882. doi: 10.1158/0008-5472.Can-05-4048 [DOI] [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;32:1317‐1326. doi: 10.1038/s41375-018-0075-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Griffin JD, Linch D, Sabbath K, Larcom P, Schlossman SF. A monoclonal antibody reactive with normal and leukemic human myeloid progenitor cells. Leuk Res. 1984;8:521‐534. doi: 10.1016/0145-2126(84)90001-8 [DOI] [PubMed] [Google Scholar]
- 49. Taussig DC, Pearce DJ, Simpson C, et al. Hematopoietic stem cells express multiple myeloid markers: implications for the origin and targeted therapy of acute myeloid leukemia. Blood. 2005;106:4086‐4092. doi: 10.1182/blood-2005-03-1072 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Kossman SE, Scheinberg DA, Jurcic JG, Jimenez J, Caron PC. A phase I trial of humanized monoclonal antibody HuM195 (anti‐CD33) with low‐dose interleukin 2 in acute myelogenous leukemia. Clin Cancer Res. 1999;5:2748‐2755. [PubMed] [Google Scholar]
- 51. Harrington KH, Gudgeon CJ, Laszlo GS, et al. The broad anti‐AML activity of the CD33/CD3 BiTE antibody construct, AMG 330, is impacted by disease stage and risk. PLoS One. 2015;10:e0135945. doi: 10.1371/journal.pone.0135945 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Laszlo GS, Gudgeon CJ, Harrington KH, Walter RB. T‐cell ligands modulate the cytolytic activity of the CD33/CD3 BiTE antibody construct, AMG 330. Blood Cancer J. 2015;5:e340. doi: 10.1038/bcj.2015.68 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Ma H, Padmanabhan IS, Parmar S, Gong Y. Targeting CLL‐1 for acute myeloid leukemia therapy. J Hematol Oncol. 2019;12:41. doi: 10.1186/s13045-019-0726-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. 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: 10.1016/j.ymthe.2017.05.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. 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: 10.1186/s13045-017-0553-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Wiersma VR, de Bruyn M, Shi C, et al. C‐type lectin‐like molecule‐1 (CLL1)‐targeted TRAIL augments the tumoricidal activity of granulocytes and potentiates therapeutic antibody‐dependent cell‐mediated cytotoxicity. MAbs. 2015;7:321‐330. doi: 10.1080/19420862.2015.1007811 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. van Rhenen A, van Dongen GAMS, Kelder A, et al. The novel AML stem cell associated antigen CLL‐1 aids in discrimination between normal and leukemic stem cells. Blood. 2007;110:2659‐2666. doi: 10.1182/blood-2007-03-083048 [DOI] [PubMed] [Google Scholar]
- 58. Laughlin MJ, Barker J, Bambach B, et al. Hematopoietic engraftment and survival in adult recipients of umbilical‐cord blood from unrelated donors. N Engl J Med. 2001;344:1815‐1822. doi: 10.1056/nejm200106143442402 [DOI] [PubMed] [Google Scholar]
- 59. Sauer T, Parikh K, Sharma S, et al. CD70‐specific CAR T cells have potent activity against acute myeloid leukemia without HSC toxicity. Blood. 2021;138:318‐330. doi: 10.1182/blood.2020008221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Riether C, Pabst T, Höpner S, et al. Targeting CD70 with cusatuzumab eliminates acute myeloid leukemia stem cells in patients treated with hypomethylating agents. Nat Med. 2020;26:1459‐1467. doi: 10.1038/s41591-020-0910-8 [DOI] [PubMed] [Google Scholar]
- 61. Glisovic‐Aplenc T, Diorio C, Chukinas JA, et al. CD38 as a pan‐hematologic target for chimeric antigen receptor T cells. Blood Adv. 2023;7:4418‐4430. doi: 10.1182/bloodadvances.2022007059 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Schorr C, Perna F. Targets for chimeric antigen receptor T‐cell therapy of acute myeloid leukemia. Front Immunol. 2022;13:1085978. doi: 10.3389/fimmu.2022.1085978 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Terstappen LW, Safford M, Unterhalt M, et al. Flow cytometric characterization of acute myeloid leukemia: IV. Comparison to the differentiation pathway of normal hematopoietic progenitor cells. Leukemia. 1992;6:993‐1000. [PubMed] [Google Scholar]
- 64. 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 Immunol. 2016;5:e116. doi: 10.1038/cti.2016.73 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Cui Q, Qian C, Xu N, et al. CD38‐directed CAR‐T cell therapy: a novel immunotherapy strategy for relapsed acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. J Hematol Oncol. 2021;14:82. doi: 10.1186/s13045-021-01092-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Zhuang H, Hu Z, Tan M, et al. Overexpression of Lewis y antigen promotes human epididymis protein 4‐mediated invasion and metastasis of ovarian cancer cells. Biochimie. 2014;105:91‐98. doi: 10.1016/j.biochi.2014.06.022 [DOI] [PubMed] [Google Scholar]
- 67. 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: 10.1038/gt.2010.21 [DOI] [PubMed] [Google Scholar]
- 68. 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: 10.1038/mt.2013.154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Neeson P, Shin A, Tainton KM, et al. Ex vivo culture of chimeric antigen receptor T cells generates functional CD8+ T cells with effector and central memory‐like phenotype. Gene Ther. 2010;17:1105‐1116. doi: 10.1038/gt.2010.59 [DOI] [PubMed] [Google Scholar]
- 70. Saxena A, Sheridan DP, Card RT, McPeek AM, Mewdell CC, Skinnider LF. Biologic and clinical significance of CD7 expression in acute myeloid leukemia. Am J Hematol. 1998;58:278‐284. [DOI] [PubMed] [Google Scholar]
- 71. Gomes‐Silva D, Srinivasan M, Sharma S, et al. CD7‐edited T cells expressing a CD7‐specific CAR for the therapy of T‐cell malignancies. Blood. 2017;130:285‐296. doi: 10.1182/blood-2017-01-761320 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Nitika WJ, Hui AM. Role of biomarkers in FLT3 AML. Cancers (Basel). 2022;14:1164. doi: 10.3390/cancers14051164 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Pedersen MG, Møller BK, Bak RO. Recent advances in the development of anti‐FLT3 CAR T‐cell therapies for treatment of AML. Biomedicine. 2022;10:2441. doi: 10.3390/biomedicines10102441 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Castaño‐Bonilla T, Alonso‐Dominguez JM, Barragán E, et al. Prognostic significance of FLT3‐ITD length in AML patients treated with intensive regimens. Sci Rep. 2021;11:20745. doi: 10.1038/s41598-021-00050-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Stone RM. What FLT3 inhibitor holds the greatest promise? Best Pract Res Clin Haematol. 2018;31:401‐404. doi: 10.1016/j.beha.2018.09.008 [DOI] [PubMed] [Google Scholar]
- 76. 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. doi: 10.1182/blood.V128.22.1072.1072 [DOI] [Google Scholar]
- 77. Wang Y, Xu Y, Li S, et al. Targeting FLT3 in acute myeloid leukemia using ligand‐based chimeric antigen receptor‐engineered T cells. J Hematol Oncol. 2018;11:60. doi: 10.1186/s13045-018-0603-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Fan S, Wang T, You F, et al. B7‐H3 chimeric antigen receptor‐modified T cell shows potential for targeted treatment of acute myeloid leukaemia. Eur J Med Res. 2023;28:129. doi: 10.1186/s40001-023-01049-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Dong P, Xiong Y, Yue J, Hanley SJB, Watari H. B7H3 As a promoter of metastasis and promising therapeutic target. Front Oncol. 2018;8:264. doi: 10.3389/fonc.2018.00264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Lichtman EI, du H, Shou P, et al. Preclinical evaluation of B7‐H3‐specific chimeric antigen receptor T cells for the treatment of acute myeloid leukemia. Clin Cancer Res. 2021;27:3141‐3153. doi: 10.1158/1078-0432.Ccr-20-2540 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Yang M, Tang X, Zhang Z, et al. Tandem CAR‐T cells targeting CD70 and B7‐H3 exhibit potent preclinical activity against multiple solid tumors. Theranostics. 2020;10:7622‐7634. doi: 10.7150/thno.43991 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Bueno C, Martínez A, Romecin PA, et al. TIM3, a human acute myeloid leukemia stem cell marker, does not enrich for leukemia‐initiating stem cells in B‐cell acute lymphoblastic leukemia. Haematologica. 2023;108:2229‐2233. doi: 10.3324/haematol.2022.282394 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Wang QJ, Yu Z, Hanada KI, et al. Preclinical evaluation of chimeric antigen receptors targeting CD70‐expressing cancers. Clin Cancer Res. 2017;23:2267‐2276. doi: 10.1158/1078-0432.Ccr-16-1421 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. He X, Feng Z, Ma J, et al. Bispecific and split CAR T cells targeting CD13 and TIM3 eradicate acute myeloid leukemia. Blood. 2020;135:713‐723. doi: 10.1182/blood.2019002779 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Tang L, Huang H, Tang Y, et al. CD44v6 chimeric antigen receptor T cell specificity towards AML with FLT3 or DNMT3A mutations. Clin Transl Med. 2022;12:e1043. doi: 10.1002/ctm2.1043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. 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: 10.1182/blood-2013-04-493361 [DOI] [PubMed] [Google Scholar]
- 87. 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: 10.1056/NEJMoa1106152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Stahl M, Menghrajani K, Derkach A, et al. Clinical and molecular predictors of response and survival following venetoclax therapy in relapsed/refractory AML. Blood Adv. 2021;5:1552‐1564. doi: 10.1182/bloodadvances.2020003734 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. DiNardo CD, Pratz K, Pullarkat V, et al. Venetoclax combined with decitabine or azacitidine in treatment‐naive, elderly patients with acute myeloid leukemia. Blood. 2019;133:7‐17. doi: 10.1182/blood-2018-08-868752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Tang, L. , Kong Y., Wang H., Zou P., Sun T., Liu Y., Zhang J., Jin N., Mao H., Zhu X., Wang J., Meng F., You Y. Demethylating therapy increases cytotoxicity of CD44v6 CAR‐T cells against acute myeloid leukemia. Front Immunol 14, 1145441, 10.3389/fimmu.2023.1145441 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Jia J, Liu B, Wang D, et al. CD93 promotes acute myeloid leukemia development and is a potential therapeutic target. Exp Cell Res. 2022;420:113361. doi: 10.1016/j.yexcr.2022.113361 [DOI] [PubMed] [Google Scholar]
- 92. Richards RM, Zhao F, Freitas KA, et al. NOT‐gated CD93 CAR T cells effectively target AML with minimized endothelial cross‐reactivity. Blood Cancer Discov. 2021;2:648‐665. doi: 10.1158/2643-3230.Bcd-20-0208 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Li K, du Y, Cai Y, et al. Single‐cell analysis reveals the chemotherapy‐induced cellular reprogramming and novel therapeutic targets in relapsed/refractory acute myeloid leukemia. Leukemia. 2023;37:308‐325. doi: 10.1038/s41375-022-01789-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Chung SS, Eng WS, Hu W, et al. CD99 is a therapeutic target on disease stem cells in myeloid malignancies. Sci Transl Med. 2017;9:2025. doi: 10.1126/scitranslmed.aaj2025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Vaikari VP, Park M, Keossayan L, MacKay JA, Alachkar H. Anti‐CD99 scFv‐ELP nanoworms for the treatment of acute myeloid leukemia. Nanomedicine. 2020;29:102236. doi: 10.1016/j.nano.2020.102236 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Sevim H, Çelik H, Düşünceli L, et al. Clofarabine induces ERK/MSK/CREB activation through inhibiting CD99 on Ewing sarcoma cells. PLoS One. 2021;16:e0253170. doi: 10.1371/journal.pone.0253170 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Celik H, Sciandra M, Flashner B, et al. Clofarabine inhibits Ewing sarcoma growth through a novel molecular mechanism involving direct binding to CD99. Oncogene. 2018;37:2181‐2196. doi: 10.1038/s41388-017-0080-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Benmebarek MR, Karches C, Cadilha B, Lesch S, Endres S, Kobold S. Killing mechanisms of chimeric antigen receptor (CAR) T cells. Int J Mol Sci. 2019;20:1283. doi: 10.3390/ijms20061283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Sheth VS, Gauthier J. Taming the beast: CRS and ICANS after CAR T‐cell therapy for ALL. Bone Marrow Transplant. 2021;56:552‐566. doi: 10.1038/s41409-020-01134-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. 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: 10.1182/blood-2013-09-529537 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Chohan KL, Siegler EL, Kenderian SS. CAR‐T cell therapy: the efficacy and toxicity balance. Curr Hematol Malig Rep. 2023;18:9‐18. doi: 10.1007/s11899-023-00687-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Schubert ML, Schmitt M, Wang L, et al. Side‐effect management of chimeric antigen receptor (CAR) T‐cell therapy. Ann Oncol. 2021;32:34‐48. doi: 10.1016/j.annonc.2020.10.478 [DOI] [PubMed] [Google Scholar]
- 103. Orning P, Lien E, Fitzgerald KA. Gasdermins and their role in immunity and inflammation. J Exp Med. 2019;216:2453‐2465. doi: 10.1084/jem.20190545 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Pan K, Farrukh H, Chittepu VCSR, Xu H, Pan CX, Zhu Z. CAR race to cancer immunotherapy: from CAR T, CAR NK to CAR macrophage therapy. J Exp Clin Cancer Res. 2022;41:119. doi: 10.1186/s13046-022-02327-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Shimabukuro‐Vornhagen A, Gödel P, Subklewe M, et al. Cytokine release syndrome. J Immunother Cancer. 2018;6:56. doi: 10.1186/s40425-018-0343-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Maus MV, Haas AR, Beatty GL, et al. T cells expressing chimeric antigen receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013;1:26‐31. doi: 10.1158/2326-6066.Cir-13-0006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Till BG, Jensen MC, Wang J, et al. Adoptive immunotherapy for indolent non‐Hodgkin lymphoma and mantle cell lymphoma using genetically modified autologous CD20‐specific T cells. Blood. 2008;112:2261‐2271. doi: 10.1182/blood-2007-12-128843 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med. 2014;371(16):1507‐1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Skorka K, Ostapinska K, Malesa A, Giannopoulos K. The application of CAR‐T cells in Haematological malignancies. Arch Immunol Ther Exp (Warsz). 2020;68:34. doi: 10.1007/s00005-020-00599-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Hettle R, Corbett M, Hinde S, et al. The assessment and appraisal of regenerative medicines and cell therapy products: an exploration of methods for review, economic evaluation and appraisal. Health Technol Assess. 2017;21:1‐204. doi: 10.3310/hta21070 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Kowolik CM, Topp MS, Gonzalez S, et al. CD28 costimulation provided through a CD19‐specific chimeric antigen receptor enhances in vivo persistence and antitumor efficacy of adoptively transferred T cells. Cancer Res. 2006;66:10995‐11004. doi: 10.1158/0008-5472.Can-06-0160 [DOI] [PubMed] [Google Scholar]
- 112. 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: 10.1038/nm.3838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor‐modified T cells in chronic lymphoid leukemia. N Engl J Med. 2011;365:725‐733. doi: 10.1056/NEJMoa1103849 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Lai Y, Weng J, Wei X, et al. Toll‐like receptor 2 costimulation potentiates the antitumor efficacy of CAR T cells. Leukemia. 2018;32:801‐808. doi: 10.1038/leu.2017.249 [DOI] [PubMed] [Google Scholar]
- 115. Mata M, Gerken C, Nguyen P, Krenciute G, Spencer DM, Gottschalk S. Inducible activation of MyD88 and CD40 in CAR T cells results in controllable and potent antitumor activity in preclinical solid tumor models. Cancer Discov. 2017;7:1306‐1319. doi: 10.1158/2159-8290.Cd-17-0263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Tahmasebi S, Elahi R, Esmaeilzadeh A. Solid tumors challenges and new insights of CAR T cell engineering. Stem Cell Rev Rep. 2019;15:619‐636. doi: 10.1007/s12015-019-09901-7 [DOI] [PubMed] [Google Scholar]
- 117. Han X, Wang Y, Wei J, Han W. Multi‐antigen‐targeted chimeric antigen receptor T cells for cancer therapy. J Hematol Oncol. 2019;12:128. doi: 10.1186/s13045-019-0813-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. 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: 10.1038/bcj.2016.61 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Naeem M, Hazafa A, Bano N, et al. Explorations of CRISPR/Cas9 for improving the long‐term efficacy of universal CAR‐T cells in tumor immunotherapy. Life Sci. 2023;316:121409. doi: 10.1016/j.lfs.2023.121409 [DOI] [PubMed] [Google Scholar]
- 120. Zhao J, Lin Q, Song Y, Liu D. Universal CARs, universal T cells, and universal CAR T cells. J Hematol Oncol. 2018;11:132. doi: 10.1186/s13045-018-0677-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Kim MY, Yu KR, Kenderian SS, et al. Genetic inactivation of CD33 in hematopoietic stem cells to enable CAR T cell immunotherapy for acute myeloid leukemia. Cell. 2018;173:1439‐1453. doi: 10.1016/j.cell.2018.05.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Cummins KD, Gill S. Will CAR T cell therapy have a role in AML? Promises and pitfalls. Semin Hematol. 2019;56:155‐163. doi: 10.1053/j.seminhematol.2018.08.008 [DOI] [PubMed] [Google Scholar]
- 123. Nishio N, Diaconu I, Liu H, et al. Armed oncolytic virus enhances immune functions of chimeric antigen receptor‐modified T cells in solid tumors. Cancer Res. 2014;74:5195‐5205. doi: 10.1158/0008-5472.Can-14-0697 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Rosewell Shaw A, Porter CE, Watanabe N, et al. Adenovirotherapy delivering cytokine and checkpoint inhibitor augments CAR T cells against metastatic head and neck cancer. Mol Ther. 2017;25:2440‐2451. doi: 10.1016/j.ymthe.2017.09.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Laborda E. Strategies to control CAR‐T cell therapy: perspective on next‐generation CARs. Cell Gene Ther Insights. 2018;4:275‐285. doi: 10.18609/cgti.2018.028 [DOI] [Google Scholar]
- 126. Gust J, Hay KA, Hanafi LA, et al. Endothelial activation and blood‐brain barrier disruption in neurotoxicity after adoptive immunotherapy with CD19 CAR‐T cells. Cancer Discov. 2017;7:1404‐1419. doi: 10.1158/2159-8290.Cd-17-0698 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Zhu X et al. RUNX3 improves CAR‐T cell phenotype and reduces cytokine release while maintaining CAR‐T function. Med Oncol. 2023;40:89. doi: 10.1007/s12032-022-01913-7 [DOI] [PubMed] [Google Scholar]
- 128. Cappell KM, Kochenderfer JN. A comparison of chimeric antigen receptors containing CD28 versus 4‐1BB costimulatory domains. Nat Rev Clin Oncol. 2021;18:715‐727. doi: 10.1038/s41571-021-00530-z [DOI] [PubMed] [Google Scholar]
- 129. Gumber D, Wang LD. Improving CAR‐T immunotherapy: overcoming the challenges of T cell exhaustion. EBioMedicine. 2022;77:103941. doi: 10.1016/j.ebiom.2022.103941 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Seo H, Chen J, González‐Avalos E, et al. TOX and TOX2 transcription factors cooperate with NR4A transcription factors to impose CD8(+) T cell exhaustion. Proc Natl Acad Sci U S A. 2019;116:12410‐12415. doi: 10.1073/pnas.1905675116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05445765.
- 132. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05248685.
- 133. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT03896854.
- 134. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05239689.
- 135. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT01716364.
- 136. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05252572.
- 137. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05473221.
- 138. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT03126864.
- 139. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04803929.
- 140. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT03927261.
- 141. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04318678.
- 142. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05488132.
- 143. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04662294.
- 144. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04272125.
- 145. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05949125.
- 146. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT02203825.
- 147. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04835519.
- 148. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05454241.
- 149. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05945849.
- 150. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05105152.
- 151. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05467254.
- 152. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04257175.
- 153. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05943314.
- 154. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04599543.
- 155. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05023707.
- 156. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04351022.
- 157. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05672147.
- 158. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT04219163.
- 159. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT02623582.
- 160. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT05266950.
- 161. ClinicalTrials.gov . Available online:https://clinicaltrials.gov/study/NCT02799680.
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The datasets used and/or analysed during the current study are available from the corresponding authoron reasonable request.