Skip to main content
Clinical and Translational Medicine logoLink to Clinical and Translational Medicine
. 2025 Nov 25;15(12):e70536. doi: 10.1002/ctm2.70536

CAR‐DC combined with CAR‐T therapy for relapsed/refractory acute myeloid leukaemia: Research progress and future perspectives

Rui Zhang 1, Jinlin Zhang 2, Hongkai Zhang 3, Mingfeng Zhao 1,
PMCID: PMC12647367  PMID: 41292193

Abstract

Acute myeloid leukaemia (AML) remains the most common type of leukaemia in adults. Despite advances in conventional therapies, high relapse rates persist, underscoring the need for novel approaches such as chimeric antigen receptor T (CAR‐T) cell therapy. C‐type lectin‐like molecule‐1 (CLL1)‐targeted CAR‐T emerges as a promising treatment for relapsed/refractory (R/R) AML. Although approximately 70% patients achieved remission, only a subset achieved minimal residual disease‐negative remission, which still has much room for improvement. The main reasons for the failure of CLL1 CAR‐T‐cell therapy include: (1) persistence of CLL1‐negative AML cells persist due to antigen escape; (2) downregulation of interleukin (IL)‐12 and other cytokines by the immunosuppressive tumour microenvironment (TME), contributing to the exhaustion of both endogenous T cells and CLL1 CAR‐T cells.

We synthesise a combination of CAR‐engineered dendritic cells (CAR‐DCs) and CLL1 CAR‐T cells to overcome current limitations. CAR‐DCs enhance antigen cross‐presentation to activate endogenous T cells against antigen‐negative clones, secrete immunostimulatory cytokines (e.g., IL‐12) to sustain CAR‐T activity, and remodel the TME. Key challenges involve optimising CAR designs (e.g., incorporating Fms‐like tyrosine kinase 3 ligand [FLT‐3L] or CD40 signalling domains), mitigating toxicity and establishing clinical administration protocols.

In this review, a focused discussion was provided on the specific challenges limiting CLL1‐targeted CAR‐T‐cell therapy in R/R AML, namely antigen escape and the TME, and a novel combination strategy of CAR‐DCs with CLL1 CAR‐T cells was proposed as a promising approach to mitigate these barriers. Here, the rationale, current research advances, and future perspectives of this synergistic strategy were critically examined.

Highlights

  • Our earlier clinical trials showed that C‐type lectin‐like molecule‐1 (CLL1)‐targeted therapy for refractory/relapse acute myeloid leukaemia (AML) was validated, which still has a considerable room for improvement.

  • We summarise the clinical trials and basic research on the dendritic cell (DC) therapy and chimeric antigen receptor‐engineered DC (CAR‐DC) therapy.

  • We explored the synergistic mechanism and prospects of CLL1 CAR‐DC cells combined with CLL1 CAR‐T cells in AML.

Keywords: acute myeloid leukaemia, CAR‐DCs, CLL1 CAR‐T


1. Chimeric antigen receptor T (CAR‐T) therapy has made some progress in the treatment of acute myeloid leukaemia (AML), among which C‐type lectin‐like molecule‐1 (CLL1) CAR‐T‐cell therapy has shown certain efficacy in the treatment of relapsed/refractory (R/R) AML. However, 30% of patients exhibit primary treatment resistance. Key factors limiting the efficacy of CAR‐T‐cell therapy include antigen escape, interleukin (IL)‐12 and tumour microenvironment (TME).

2. Dendritic cell (DC) mediates antitumour immunity by phagocytosing tumour material, processing tumour antigens and presenting peptide‐MHC complexes to activate tumour specific. CAR‐engineered DCs (CAR‐DCs) can specifically recognise tumour antigens, efficiently phagocytose CAR‐targeted tumour cells and debris, and subsequently activate endogenous tumour‐specific T‐cell responses. Activated CAR‐DC cells can secrete IL‐12 that may help counteract the TME and enhance immune cell function.

3. CAR‐DC cells interaction with CAR‐T cells significantly enhanced anti‐AML cytotoxic activity. CAR‐DC cells phagocytoses AML cells via CAR targeting and secretes IL‐12; activated endogenous T cells and CAR‐T cells work together to eliminate both CLL1‐positive and CLL1‐negative AML clones. Therefore, CAR‐DC and CAR‐T therapy have a good complementarity.

4. The selection and design of the intracellular signal transduction region of CAR is the key in DC cell therapy. The CAR‐DC cells with an FLT3‐integrated intracellular domain enabled conventional DC differentiation and enhanced cross‐presentation of tumour antigens. The CD40 signalling cascade upregulates MHC class II, co‐stimulatory molecules and cytokines.

graphic file with name CTM2-15-e70536-g002.jpg

1. CURRENT RESEARCH STATUS OF acute myeloid leukaemia TREATMENT

The conventional treatment of acute myeloid leukaemia (AML) mainly includes chemotherapy, targeted therapy and haematopoietic stem cell transplantation, but most patients face the risk of relapse. 1 , 2 Chimeric antigen receptor T (CAR‐T) therapy represents a promising novel approach for relapsed/refractory (R/R) AML, although significant challenges remain. Currently, CAR‐T‐cell therapy has achieved impressive clinical outcomes in haematologic malignancies, including acute lymphoblastic leukaemia, lymphoma and multiple myeloma, by targeting CD19, CD22 and b‐cell maturation antigen (BCMA). 3 , 4 , 5 , 6 However, CAR‐T‐cell therapy remains clinically immature for AML, with limited overall efficacy in current studies. 7 , 8 The primary reasons include the lack of specific targets, 9 , 10 the immunosuppressive tumour microenvironment (TME) 11 , 12 and antigen escape. 13 , 14 , 15 Early CAR‐T therapies for AML targeted CD33 and CD123, but their toxicity or poor efficacy has limited their clinical application. 7 , 16 , 17 Other potential targets include C‐type lectin‐like molecule‐1 (CLL1), FLT3, NKG2D, CD7, CD38, etc. 18 , 19 , 20 , 21 , 22 , 23 CLL1‐targeted CAR‐T cells have demonstrated promising efficacy in the treatment of R/R AML.

2. THERAPEUTIC EFFICACY OF CLL1 CAR‐T CELLS IN RELAPSED/REFRACTORY AML

CLL1 is a C‐type lectin‐like receptor that is highly expressed on leukaemia stem cells (LSCs) (about 45%) and leukaemia progenitor cells (77.5%‒92%). Tashiro et al. developed the first CLL1 CAR‐T cells, which selectively killing leukemic progenitor cells and leukaemia cells. 24 CLL1 CAR‐T cells have shown superior AML killing in vitro and in mice. 18 A case report described complete remission (CR) in a 10‐year‐old patient after CLL1 CAR‐T therapy. 25 Our group reported a 70% CR rate in 10 adult AML patients treated with CLL1 CAR‐T cells. 26 At present, a total of 48 patients received CLL1 CAR‐T‐cell therapy. All patients underwent efficacy evaluation between days 14 and 16 post‐infusion. CR was achieved in 34 patients (70.83%), including 13 with minimal residual disease (MRD)‐positive CR and 21 with MRD‐negative CR. The remaining 14 patients (29.17%) showed no response to the treatment. Regarding adverse events, cytokine release syndrome (CRS) and immune effector cell‐associated neurotoxicity syndrome (ICANS) occurred in 95.83% and 20.83% of patients, respectively. Severe (grade 3/4) CRS and ICANS were observed in 41.67% and 10.42% of patients. Haematologic toxicities were the most common complications, with near‐universal incidence of leukopaenia, granulocytopaenia, anaemia and thrombocytopaenia. Notably, severe and prolonged granulocytopaenia significantly increased the risk of infectious complications. Among the reported infections, 38 patients experienced bacterial infections, while viral and fungal infections were documented in nine and 10 patients, respectively. Comparative analysis further revealed that patients with severe CRS (grade 3/4) exhibited more pronounced cytokine release, particularly elevated levels of interleukin (IL)‐2, IL‐6, IL‐10, C‐reactive protein and ferritin, compared to those with mild CRS (grade 1/2). 27 Although CLL1‐targeted CAR‐T cells have shown clinical efficacy in R/R AML, approximately 30% of patients exhibit primary treatment resistance. Furthermore, only 50% of responders achieve MRD‐negative remission, underscoring the need for enhanced therapeutic strategies.

The key limitations include the following: (1) antigen escape—AML cells with low CLL1 expression are difficult for CAR‐T cells to recognise, and relapsed patients frequently present weak expression of target antigens. 28 , 29 (2) TME and IL‐12—the TME reduces the efficacy of CAR‐T cells via myeloid‐derived suppressor cells (MDSCs), regulatory T cells (Tregs) and tumour‐associated macrophages (TAMs). 30 , 31 , 32 Dysregulated cytokines, including IL‐12, further impair CAR‐T‐cell function. Notably, mKRAS‐specific NeoCARs with inducible IL‐12 secretion and T‐cell receptor (TCR) knockout (KO) exhibit strong in vivo antitumour activity and favourable safety. 33 Co‐expression of IL‐12 and interferon‐gamma (IFN)α2 with CAR enhances the proinflammatory microenvironment and mitigates T‐cell exhaustion. 34 Moreover, mbIL‐12‐engineered CAR‐T cells have demonstrated safety and efficacy in overcoming the TME. 35 IL‐12 not only increases CAR‐T cytotoxicity, but also remodels the TME by increasing proinflammatory CD4+ T‐cell infiltration, decreasing Tregs, and activating myeloid cells, with minimal systemic toxicity in glioblastoma multiforme‐targeted CAR‐T‐cell therapy. 36 Therefore, integrated strategies combining CAR‐T modification and TME remodelling are essential for improving clinical outcomes of CLL1 CAR‐T‐cell therapy.

3. RESEARCH PROGRESS AND CHALLENGES ASSOCIATED WITH DENDRITIC CELL AND CAR‐ENGINEERED DENDRITIC CELL THERAPY

Dendritic cells (DCs) regulate antitumour immunity by phagocytosing tumour material, processing tumour antigens and presenting peptide‒major histocompatibility complex (MHC) complexes to activate tumour‐specific T cells. 37 , 38 Mature DCs directly engage T cells through co‐stimulatory molecules (CD80/CD86), secrete IL‐12 and initiate endogenous tumour‐specific T‐cell priming. 39 , 40 , 41 Mature DCs trigger tumour‐specific CD8+ T‐cell immunity by migrating from tumours to lymph nodes, capturing antigens and activating naïve T cells. 42

Conventional DCs (cDCs) are classified into two major subsets: type 1 (cDC1) and type 2 (cDC2). 43 cDC1s prime CD8+ T cells in draining lymph nodes (dLNs) and activate Toll‐like receptors to induce IL‐12p70 and IFN‐α, promoting Th1‐type immunity. Their presence correlates with favourable prognosis in cancer patients. 44 cDC2s, more abundant than cDC1s, express CD11c, CSF‐1R, MHC‐II, CD11b, BDCA1 and SIRPα produce various cytokines, such as IL‐23 and IL‐10, and present antigens to CD4+ helper T cells, activating Th2 and Th17 subsets. 45 Novel DC subsets, including LAMP3+ DCs 46 , 47 and AXL+SIGLEC6+ (AS) DCs 48 have been identified. Studies have described DC precursors in bone marrow and blood, showing both shared and distinct regulatory networks, and suggesting underlying heterogeneity within pre‐DCs reflecting early lineage bias. 49 , 50

Single‐cell and bulk RNA‐seq revealed enrichment of immunosuppressive CCL22⁺DCs, termed ‘exhausted DCs’, which were reversed by the herbal formula SBJDD and its component berberine; TMEM131‐mediated tumor necrosis factor (TNF) signalling, promoted CCL22⁺DC development, inhibited by berberine. 51 Lineage tracing showed DC3s arise from monocyte‐DC progenitors via Lyz2⁺Ly6C⁺CD11c precursors, distinct from DC2 lineage, indicating DC3 as a separate lineage phenotypically related, but developmentally independent from monocytes. 52 scRNA‐seq of sepsis revealed major immune cell changes and a distinct cDC subcluster with maturation, migration and immunoregulatory signatures, consistent with mregDCs; this sepsis‐induced mregDC subset was validated and shown to activate naïve CD4⁺ T cells while promoting Tregs differentiation. 53

Functional status depends on maturation: mature DCs express high MHC and co‐stimulatory molecules (e.g., CD80/86), and promote effector T‐cell activation, whereas immature DCs often induce Tregs and immune tolerance. Tumours impair DC maturation to evade immunity. In AML, DC frequency and function are frequently impaired, contributing to disease progression and therapeutic resistance. 54 cDC1s specifically perform antigen cross‐presentation in vivo and are essential for adaptive antitumour immunity; without cDC1, tumours escape immune elimination. 55 Due to their superior T‐cell activation and cross‐presentation capacity, cDC1 is a preferred subset for CAR‐DC‐based immunotherapy.

DC/tumour fusion vaccines or tumour lysate‐loaded DCs can activate and sustain antitumour T‐cell responses and expand tumour‐specific T‐cell clones. 56 A phase II clinical trial (NCT03059485) showed DC/AML fusion vaccination without maintenance led to 73% 2‐year overall survival and 36% progression‐free survival in elderly AML patients. 57 Eps8‐DCs enhance CD19 CAR‐T function, increasing cytokines, CD107a degranulation and cytotoxicity. 58 However, DC dysfunction contributes to immune evasion and limits DC vaccine efficacy in elderly patients. 59 Clinical moDC vaccine efficacy remains variable, limited by antigen‐loading challenges, incomplete maturation and tumour heterogeneity. IL‐12 secretion directly influences endogenous DC1 function. 57 , 60 , 61 cDC vaccines face significant limitations, including the dysfunction of DCs (especially in the elderly), poor and variable efficacy due to challenges in antigen loading and incomplete maturation, and susceptibility to tumour heterogeneity. These limitations underscore the need for advanced strategies such as CAR‐DC platforms. Supporting Information

TME impairs DC function via multiple inhibitory mechanisms, including: (1) induction of DC apoptosis; 62 , 63 (2) inhibition of DC maturation and antigen presentation; 64 , 65 (3) promotion of tolerogenic DC phenotypes; 66 , 67 and (4) downregulation of DC‐recruiting chemokines to limit tumour infiltration. 68 , 69 , 70 , 71

Given the critical role of functional DCs and their vulnerability in AML, strategies to reprogram or ‘rejuvenate’ DCs have become a key research focus. The DC growth factor Fms‐like tyrosine kinase 3 ligand (Flt3L) enhances T‐cell‐mediated antitumour immunity by expanding and activating DC populations. 55 Highly activated DCs can induce CD4+ T cells to acquire cytotoxic and antitumour functions in aged mice. 58 PU.1‐, BATF3‐ and IRF8‐mediated DC reprogramming reduces exhaustion and increases memory‐ and stem‐cell‐like T‐cell infiltration. 72 KO of BCL9/BCL9L in cDC1 enhances CD8+ T‐cell activation, antigen presentation, epitope expansion and promotes antitumour activity. 73 , 74 Although promising approaches such as Flt3L expansion and transcriptional reprogramming via PU.1, BATF3 and IRF8 have been developed, DC reprogramming still faces considerable challenges. Key limitations include the translational gap between mouse models and human patients, the difficulty and safety concerns of controlling genetic or transcriptional programs in clinical settings, and the instability of the reprogrammed states in the immunosuppressive TME.

Moreover, CAR‐DCs can specifically recognise tumour antigens, efficiently phagocytose CAR‐targeted tumour cells and debris, and subsequently activate endogenous tumour‐specific T‐cell responses. Antitumour T‐cell responses can directly eliminate CAR‐targeted antigen‐positive tumours and indirectly eliminate antigen‐negative tumours (which are not directly recognised by the CAR) through cross‐presentation and epitope spreading. 69 , 75 Activated CAR‐DCs secrete immunostimulatory cytokines such as IL‐12. 35 , 69 , 76 , 77 The intracellular signalling domain of the CAR can remain continuously activated during tumour recognition, enabling DCs to sustain homeostasis and resist the suppressive microenvironment, allowing tolerant DCs to reacclimate.

The mechanism by which CAR‐DCs overcome antigen escape and target CAR antigen‐negative cells is based on their unique capacity for antigen cross‐presentation and subsequent epitope spreading. (1) Phagocytosis of antigen‐positive cells: CAR‐DCs use their CAR to specifically recognise and phagocytose CAR‐positive (e.g., CLL1⁺) AML cells. (2) Cross‐presentation and endogenous T‐cell activation: after phagocytosis, CAR‐DCs process whole‐cell contents and present a broad repertoire of tumour‐derived peptides via MHC‐I to activate endogenous, CD8⁺ T cells. (3) Elimination of antigen‐negative clones via epitope spreading: these activated endogenous T cells are polyclonal and can recognise multiple tumour antigens, allowing them to eradicate AML cell populations that escape CAR‐T therapy by losing the target antigen (e.g., CLL1‐negative cells). This immune expansion from a single antigen to additional antigens is termed epitope spreading.

The process by which CAR‐DCs target and eliminate CAR antigen‐negative cells is as follows: CAR‐mediated phagocytosis → broad antigen processing and cross‐presentation → activation of endogenous T cells against multiple tumour antigens → elimination of antigen‐negative clones via epitope spreading.

4. VALIDATION OF TARGET SPECIFICITY USING GENE KO MODELS

In vitro validation often involves the use of CRISPR‐Cas9 or RNA interference to KO the target antigen (e.g., CLL1) in AML cell lines. For instance, CLL1‐KO AML cells show significantly reduced susceptibility to CLL1‐targeted CAR‐T‐cell killing, while retaining sensitivity to CAR‐T cells targeting alternative antigens (e.g., CD33 or CD123). 18 , 24 This confirms the antigen specificity of the CAR construct. Similarly, when CAR‐DCs are co‐cultured with CLL1‐KO AML cells, their phagocytic capacity and subsequent T‐cell activation are markedly diminished, underscoring the dependence on CAR‐mediated recognition. 69 , 75

In vivo studies further validate these findings using xenograft models established with CLL1‐deficient AML cells. In such models, CLL1 CAR‐T cells exhibit reduced leukemic control compared to their activity against CLL1‐positive tumours, highlighting the role of antigen expression in therapeutic efficacy. 18 , 26 Moreover, in dual‐flank tumour models—where one tumour expresses CLL1 and the other is CLL1‐KO—adoptive transfer of CLL1 CAR‐T cells combined with CAR‐DCs results in preferential regression of the CLL1‐positive tumour, with limited impact on the KO tumour unless epitope spreading occurs via CAR‐DC‐mediated cross‐presentation. 75 , 82 These KO models not only confirm the on‐target mechanism of action but also help elucidate escape mechanisms. For example, residual disease in CLL1‐KO models often emerges from antigen‐low or antigen‐negative clones, reinforcing the need for combination strategies that address heterogeneity, such as the incorporation of CAR‐DCs to broaden antigen recognition.

Genetic KO models provide indispensable evidence for the specificity and mechanism of CAR‐DCs and CAR‐T therapies. They validate that targeting CLL1 is both necessary and sufficient for initiating antitumour responses, while also revealing limitations that inform the design of next‐generation combinatorial immunotherapies.

5. CAR‐DC COMBINED WITH CAR‐T‐CELL THERAPY

CAR‐DC therapy demonstrates antitumour efficacy in AML models and is under clinical evaluation for solid epithelial malignancies (NCT05631899 and NCT05631886). NCT05631899 is a pilot clinical trial assessing the safety, immune activity and efficacy of an EphA2‐targeting CAR‐DC vaccine loaded with a KRAS mutant peptide (KRAS‐EphA2‐CAR‐DC) plus immune checkpoint inhibitors (ICIs) in patients with locally advanced or metastatic solid tumours. Preclinical findings indicate that engineered CAR‐DCs enhance the cytotoxicity of co‐administered CAR‐T cells in solid tumour mouse models. NCT05631886 is a parallel pilot trial evaluating an EphA2‐directed CAR‐DC vaccine loaded with a TP53 mutant peptide (TP53‐EphA2‐CAR‐DCs) plus ICIs in solid tumours or R/R lymphomas.

Multiple studies indicate that intratumoural DC delivery can safely augment CAR‐T‐cell activity and alleviate the immunosuppressive TME. In vitro differentiation of DCs expressing 4‐1BB is enriched for the CD141+/ClEC9A+ DC subset. CAR‐DC and CAR‐T‐cell interactions synergistically enhance anti‐AML cytotoxicity. 78 DC‐derived cytokines, such as IL‐12 and type I IFNs, provide additional T‐cell stimulation during antigen presentation, while CAR‐T cells retain their intrinsic cytotoxic capacity independent of DCs. 79 These data indicate that intratumoural CAR‐DC delivery establishes an ‘immunological niche’. CAR‐DCs secrete IL‐12 and type I IFNs to reverse T‐cell exhaustion and remodel the TME. This synergy addresses CAR‐T‐cell limitations, including inadequate tumour infiltration and antigen loss, through DC‐mediated antigen spreading and localised immune activation.

Studies further demonstrate that 4‐1BB‐engineered autologous DCs enhance the efficacy of anti‐CD33 CAR‐T cells in AML by secreting cytokines and facilitating CAR‐T‐cell recruitment to the bone marrow niche. 80 The cooperative interaction between CAR‐DCs and CAR‐T cells may represent a powerful strategy to improve antitumour immunotherapy.

Mechanistic overview of CAR‐DC and CAR‐T‐cell collaboration (Figure 1): (1) CAR‐DCs phagocytose AML cells via CAR targeting, process antigens and cross‐present tumour‐derived peptides via MHC‐I/II to activate endogenous T cells. 40 , 41 (2) CAR‐DCs secrete cytokines such as IL‐12 to promote CAR‐T‐cell support the activation, proliferation and persistence, reducing exhaustion and supporting memory formation. 35 , 40 , 79 , 81 (3) Activated endogenous T cells and CAR‐T cells cooperatively eradicate both CLL1‐positive and CLL1‐negative AML populations through direct cytotoxicity and epitope spreading. 82 , 83 Comparative characteristics of CAR‐DCs are summarised in Table 1.

FIGURE 1.

FIGURE 1

Mechanistic overview of chimeric antigen receptor‐engineered dendritic cell (CAR‐DC) cells and chimeric antigen receptor T (CAR‐T) cell collaboration: (1) CAR‐DC cells phagocytoses acute myeloid leukaemia (AML) cells via CAR targeting, processes antigens and cross‐presents tumour‐derived peptides via major histocompatibility complex (MHC)‐I/II to activate endogenous T cells. (2) CAR‐DC secretes cytokines (e.g., interleukin [IL]‐12) to support the activation, proliferation and persistence of C‐type lectin‐like molecule‐1 (CLL1) CAR‐T cells, potentially reducing exhaustion and promoting a memory phenotype. (3) Activated endogenous T cells (primed by CAR‐DC) and CAR‐T cells work together to eliminate both CLL1‐positive and CLL1‐negative (via epitope spreading) AML clones.

TABLE 1.

Comparative characteristics of chimeric antigen receptor‐engineered dendritic cells (CAR‐DCs).

Target antigen CAR‐DC structure Delivery methods Animal models used Cytokine domains Antitumour outcomes
CD33 71 DC precursors were transduced with a CAR (pCCL‐anti‐CD33‐4‐1BB‐CD3ζ‐T2A‐GFP). The differentiation of DC in vitro employed Flt3L/GM‐CSF/IL‐4 Lentivirus transduction NSG AML mice model Differentiation of DC in vitro employed Flt3L/GM‐CSF/IL‐4 CAR‐DC and CAR‐T cells enhances cytotoxic cytokine production in response to DC‐derived IL‐12. These combined effects resulted in improved anti‐CD33 CAR‐T cytotoxicity in vitro and in vivo NSG AML mice model.
KRAS‐EphA‐2 [clinical trail: NCT05631899] KRAS‐EphA‐2‐CAR‐DC contain a scFv domain targeting EphA2 antigen and KRAS mutant peptide, CD8a transmembrane, tandem DC‐specific activation domains Vaccine Clinical trial for solid tumours IL‐2, IL‐6, IL‐8, IL‐10, IL‐12 (p70), TNF‐α KRAS‐EphA‐2‐CAR‐DC can suppress the growth of tumours expressing the correlated KRAS mutant in animal models. In addition, the combination of the immune checkpoint inhibitors could further reverse immunosuppressive TME and globally activate T‐cell responses.
P53‐EphA‐2 [clinical trail: NCT05631886] P53‐EphA‐2‐CAR‐DC contain a scFv domain targeting EphA2 antigen, CD8a transmembrane, tandem DC‐specific activation domains Vaccine Clinical trial for R/R lymphomas IL‐2, IL‐6, IL‐8, IL‐10, IL‐12 (p70), TNF‐α P53 (R273H, R175H, R248Q or R249S)‐EphA‐2‐CAR‐DC can suppress the growth of tumours expressing the correlated TP53 mutant in animal models. In addition, the combination of the immune checkpoint inhibitors could further reverse immunosuppressive TME and globally activate T‐cell responses.
BCMA 74 BCMA‐CAR‐DC: CD8a signal peptide, BCMA VHH antibodies, CD8a transmembrane, CD40 intracellular domain Lentiviral transduction Multiple myeloma IL‐12, TNF‐α CAR‐DC cells specifically target multiple myeloma cells overexpressing BCMA receptors. They selectively bind to these malignant cells and enhance TRAIL‐mediated apoptosis, thereby effectively eliminating multiple myeloma cells for therapeutic purposes.

CD123 75

CD33

scFv + CD8a transmembrane + intracellular domain (CD3ζ; 4‐1BB + CD3ζ; FLT3L + CD3ζ) Lentiviral transduction R/R AML IL‐12, TNF‐α Selecting the appropriate intracellular domain (e.g., 4‐1BB, FLT3 or CD40) of CAR can induce the mature phenotype of CAR‐DC when recognising the tumour, and overcome the acclimation of tolerant DC cells by the suppressive TME.

Abbreviations: AML, acute myeloid leukaemia; Flt3L, Fms‐like tyrosine kinase 3 ligand; IL, interleukin; R/R, relapsed/refractory; TME, tumour microenvironment; TNF, tumor necrosis factor.

Therefore, CAR‐DCs and CAR‐T‐cell therapy exhibit strong complementarity, and can enhance CAR‐T‐cell function while overcoming immune escape to reduce tumour recurrence. The limitations of the CAR‐DC and CAR‐T‐cell combination strategy can be summarised as follows: (1) early‐stage clinical evidence—the efficacy of this combination is primarily supported by preclinical data. Human clinical evidence remains limited to early‐phase pilot studies (e.g., NCT05631899 and NCT05631886), and definitive therapeutic value has not yet been established. (2) Logistical and manufacturing complexity—this therapeutic modality requires the production, quality control and administration of two advanced cellular products (CAR‐DCs and CAR‐T cells), substantially increasing complexity, cost and barriers to scalable manufacturing compared with single‐cell products. (3) Unverified synergy in human tumours—although animal models support the creation of an ‘immunological niche’ capable of reversing T‐cell exhaustion and overcoming the immunosuppressive TME, consistent confirmation in human tumours is still required. (4) Safety uncertainties—dual activation of engineered immune cell populations may amplify toxicity risks, including exacerbated CRS. The full safety profile of this combined approach remains insufficiently characterised. Table 2

TABLE 2.

Novel cellular immunotherapies: mechanisms and clinical status.

Therapeutic modality Mechanism of action Target antigen(s) Efficacy/response (clinical/preclinical) Key advantages Key limitations Clinical status
CLL1 CAR‐T CAR‐T cells directly kill CLL1+ AML cells CLL1 ∼70% CR (34/48 pts); ∼44% MRD‐ CR (21/48 pts) 26 , 27 Targets LSCs; clinically validated Antigen escape; TME suppression; high CRS/ICANS rates Phase I/II
CAR‐DC + CAR‐T CAR‐DCs phagocytose tumour, cross‐present antigens, secrete cytokines; synergise with CAR‐T CLL1 (CAR‐DC + CAR‐T) Preclinical: enhanced cytotoxicity and epitope spreading 78 , 80 Overcomes antigen escape; remodels TME; activates endogenous T cells Logistical complexity; unproven clinical synergy; potential CRS amplification Early‐phase trials (e.g., NCT05631899)
DC/AML fusion vaccine DCs loaded with AML antigens activate endogenous T cells Multiple tumour‐associated 73% 2‐year OS in elderly AML (phase II) 57 Broad antigen response; favourable safety Limited efficacy in elderly; variable response; DC dysfunction Phase II
Dual‐target CAR‐T (e.g., CD33 + CD123) CAR‐T cells co‐target two antigens to prevent escape CD33 and CD123 Preclinical: reduced escape and prolonged survival 99 Reduces antigen escape; improved coverage Potential increased toxicity, antigen co‐expression required Preclinical/phase I
iPSC‐derived CAR‐T Off‐the‐shelf CAR‐T from iPSC; scalable and uniform CD19, HER2 Early clinical responses (e.g., FT819:4/15 responders) 85 Scalable; uniform product; reduced variability Differentiation challenges; allo‐rejection risk; genomic instability Phase I
UCAR‐T Allogeneic γδ T cells with CAR; HLA‐independent killing CD20, NKG2D ligands 67% ORR in B‐cell malignancies (AD1‐001) 156 Low GvHD risk; intrinsic tumour sensing Rare cell source; expansion difficulties Phase I
Armoured CAR‐T (IL‐12) CAR‐T secreting IL‐12 to remodel TME and enhance persistence Tumour‐specific + IL‐12 Preclinical: enhanced efficacy; TME reprogramming 35 , 36 Counteracts TME; enhances T‐cell function Cytokine‐related toxicity (CRS/ICANS); narrow therapeutic window Early‐phase trials
Logic‐gated CAR‐T AND/NOT‐gates improve tumour specificity and safety MSLN + CDH3; CD33 + CD123 Preclinical: reduced on‐target, off‐tumour toxicity 101 , 108 Enhanced safety; precision targeting Design complexity; leaky activation; unproven in humans Preclinical/phase I
In vivo CAR‐T CAR gene delivered directly to T cells in vivo CD19, solid tumour antigens Mouse models: tumour regression 95 , 97 No ex vivo manufacturing; preserves T‐cell fitness Delivery specificity; safety control; transient expression Preclinical

Abbreviations: AML, acute myeloid leukaemia; CAR‐DC, chimeric antigen receptor‐engineered dendritic cell; CAR‐T, chimeric antigen receptor T; CLL1, C‐type lectin‐like molecule‐1; CRS, cytokine release syndrome; GvHD, graft‐versus‐host disease; HLA, human leukocyte antigen; ICANS, immune effector cell‐associated neurotoxicity syndrome; IL, interleukin; LSC, leukaemia stem cell; MSLN, mesothelin; ORR, overall response rate; OS, overall survival; TME, tumour microenvironment.

6. DESIGN AND FUNCTION OPTIMIZATION OF CAR‐DCS

The selection and design of the intracellular signalling region of CAR is critical in DC‐based therapy, as it enables DC differentiation, phagocytosis and antigen cross‐presentation following tumour antigen recognition (Figure 2):

  1. 4‐1BB signalling domain: CD33 CAR‐DCs containing a 4‐1BB‐CD3ζ intracellular region can activate CAR‐T cells and enhance their antitumour activity in AML co‐culture assays. 80

  2. Flt3L signalling domain: the University of Washington developed CAR‐DCs with an FLT3‐integrated domain, to support cDC differentiation and improve cross‐presentation of tumour antigen. 72 Flt3L‐secreting engineered T cells synergise with pattern recognition receptor poly(I:C) and 4‐1BB agonists to increase intratumoural DC accumulation and systemic antitumour responses, mitigating antigen‐negative tumour escape in solid tumours. 39 , 75

  3. CD40 signalling domain: CD40 signalling activation increases MHC‐II, co‐stimulatory molecules (CD86/CD70/CD80), and cytokines (IL‐12, TNF‐α), thereby strengthening cross‐presentation. 84 Preclinical evidence indicates that CD40‐containing CAR‐DCs combined with BCMA CAR‐T cells improve antimyeloma activity. 23

FIGURE 2.

FIGURE 2

Optimal intracellular signal of chimeric antigen receptor‐engineered dendritic cell (CAR‐DC): selecting the appropriate intracellular domain (CD3ζ; CD3ζ + 4‐1BB; FLT3L; CD40; and CD40 + FLT3L) of CAR can induce the mature phenotype of CAR‐DC.

Thus, appropriate intracellular domain selection (e.g., 4‐1BB, FLT3 or CD40) can promote maturation of CAR‐DCs following tumour encounter and counteract the suppression of the TME.

7. EMERGING TECHNOLOGIES AND FUTURE DIRECTIONS

7.1. iPSC‐derived allogeneic CAR products

iPSC‐derived allogeneic CAR platforms provide an off‐the‐shelf solution to reduce the cost, complexity and variability associated with autologous therapies. 85 FT819 (anti‐CD19 CAR‐T cells with a TRAC‐integrated CAR) showed no dose‐limiting toxicity, GVHD or severe CRS in a phase I trial, with responses in four of 15 patients, FT825/ONO‐8250 (anti‐HER2, seven edits) seeks to improve solid tumour targeting. iCAR‐NK programs, including FT596 (anti‐CD19 + hnCD16 + IL‐15RF), demonstrated responses in nine out of 17 patients, while FT522/FT576 include edits for durability and combination therapy. Century therapeutics CNTY‐101, a multi‐edited CD19‐targeting iCAR‐NK therapy, showed no significant toxicity and a 40% response rate in phase I. These findings demonstrate the potential for standardised, scalable and allogeneic immunotherapies. 86 The core advantage of iPSC platforms lies in generating unlimited, uniform CAR‐DCs and CAR‐T cells to overcome cost and manufacturing barriers inherent to autologous dual‐cell therapy.

The development of functionally mature iPSC‐derived CAR‐T (iCAR‐T) cells encounters significant technical constraints. Current differentiation methods primarily produce innate‐like T cells (iT cells) expressing CD8αα and CD56 but lacking CD8αβ, CD2, CD5 and CD28. 87 , 88 , 89 , 90 A major issue is inefficient progression through the double‐positive (DP) stage, essential for conventional αβ T‐cell development. Premature TCR or CAR expression, commonly observed with T‐cell‐derived iPSCs or constitutive CAR constructs, disrupts DP formation and biases differentiation towards innate CD8αα⁺ or γδ T‐like phenotypes by suppressing Notch signalling (reduced Notch1/3 and downstream effectors) and decreasing PTCRA expression, which is necessary for αβ T‐cell maturation. 89 , 91

Strategies to improve T‐lineage commitment include (a) removing double‐negative T cells to avoid DP progenitor depletion; (b) using OP9‐DLL4 or 3D thymic organoids to increase Notch signalling; (c) employing CAR designs with 4‐1BB instead of CD28 to enhance DP transition and CD8αβ⁺ development; (d) targeting TRAC insertion with attenuated ITAMs to limit tonic signalling and preserve Notch/PTCRA expression; and (e) supplying CAR antigen and 4‐1BBL during differentiation to support DP‐to‐SP maturation. Most systems predominantly yield CD8⁺ iCAR‐T cells, even when CD4⁺ starting populations are used. Co‐culture with artificial thymic organoids enables CD4⁺ iT‐cell production, and a recent feeder‐free approach using PMA/ionomycin to bias DP cells towards the CD4⁺ lineage offers a scalable alternative. Balanced CD4⁺/CD8⁺ ratios may improve persistence and antitumour efficacy.

These findings highlight the difficulty of dual genetic engineering (e.g., incorporating a CAR while maintaining DC differentiation potential in iPSCs) and underscore the necessity of eliminating alloreactivity, such as through human leukocyte antigen (HLA) editing, which further increases developmental complexity.

7.2. In vivo CAR cell programming

Direct in vivo CAR gene delivery can intrinsically activate T cells, for example, via innate immune sensing triggered by RNA in LNPs. Unlike strong artificial stimulation applied during ex vivo manufacturing, in vivo‐generated CAR‐T cells expand gradually under physiological antigen drive, preserve stem‐like characteristics, avoid exhaustion and exhibit sustained antitumour activity. 92 In vivo delivery may preferentially transduce less‐differentiated T cells, including naïve (Tn), stem‐like memory or central memory subsets, which possess superior proliferative capacity, persistence and antitumour potency. Reprogramming even a small number of these cells can result in durable responses. Local administration (e.g., intratumoural) can further enhance efficacy by engineering tissue‐resident memory T cells, enabling direct tumour targeting without reliance on trafficking. 93 Dynamic TME activation: in vivo‐generated CAR‐T cells undergo progressive activation within the TME. Synergy with the intact host immune system: because in vivo CAR‐T therapy often requires none or only limited lymphodepletion, host immunity remains largely preserved. 94

Conventional ex vivo CAR‐T‐cell products require complex, multi‐week manufacturing and are constrained by variability in patient T‐cell fitness. In vivo CAR‐T therapy instead administers CAR‐encoding vectors directly to patients. Two major systems are used: viral vectors (e.g., AAV and lentivirus), which support prolonged expression without integration but lack intrinsic T‐cell tropism. Lentiviruses can be pseudotyped with targeting ligands (e.g., anti‐CD3/CD8 scFv) to improve T‐cell specificity. Successful in vivo CAR‐T generation has eliminated CD19+ B‐cell malignancies in preclinical studies. 95 , 96 Challenges include insertional mutagenesis, off‐target transduction (e.g., germline or malignant clones) and pre‐existing immunity.

7.3. Non‐viral nanoparticle platforms

LNPs or polymer‐based nanocarriers primarily deliver CAR‐encoding mRNA for transient expression or DNA for potential stable integration (e.g., piggyBac transposon). Untargeted LNPs accumulate mainly in the liver and spleen, whereas targeted LNPs conjugated with T‐cell‐specific antibodies (e.g., anti‐CD5) improve delivery accuracy. Polymer nanoparticles encoding CAR mRNA against prostate or liver tumour antigens inhibited tumour growth in mice. 97 AAV‐mediated in vivo CAR‐T production induced tumour regression in murine models. 98 Loop33 and 123 CAR‐T cells targeting CD33 and CD123 eliminated AML cells and prolonged survival in leukaemia‐bearing mice, while mitigating immune escape. 99 Key challenges are transient expression, requiring repeated dosing, as well as the increased manufacturing complexity and potential immunogenicity associated with antibody conjugation.

Key limitations include: (1) delivery efficiency and specificity—achieving targeted gene transfer exclusively into desired T‐cell subsets remains difficult; (2) safety control—unlike ex vivo products, in vivo‐generated CAR‐T cells cannot undergo batch‐level phenotyping or safety‐switch activation prior to infusion; (3) transient expression risk—non‐viral systems may yield short‐lived CAR expression, necessitating repeat administration.

7.4. Logic‐gated and synthetic gene circuits

Boolean logic gates (e.g., ‘OR’, ‘AND’ and ‘NOT’) represent recent innovations for enhancing CAR‐T precision and safety.

The simplest AND‐gate strategy targets two tumour‐associate antigens (TAAs) co‐expressed on cancer cells but not normal cells, enabling cytotoxicity only when both antigens are engaged. 100 For TCEs, selective elimination of dual‐TAA⁺ cells results from cooperative binding (pseudo‐avidity). 101 A trispecific TCE against B7‐H4 and LY6E demonstrated preferential activity in colorectal cancer. 102 ISB2001 (anti‐BCMA/CD38) functions as a strict AND‐gate, showing a 100‐fold activity reduction after single‐antigen deletion. 103 Split haemibody TCEs position anti‐CD3 VH/VL domains on separate proteins that reassemble only on dual‐TAA⁺ cells. 104 The precision GATE platform incorporates extended‐half‐life haemibodies and protease masks to improve performance. 105 , 106 Split CAR systems segregate activation and co‐stimulation signals. A low‐affinity CD138 (stimulatory) CAR paired with a high‐affinity CD38 (co‐stimulatory) CAR distinguished myeloma tissue from healthy tissue. 107 Tousley et al. developed a leak‐resistant AND‐gate CAR incorporating LAT and SLP76 domains to require dual antigen input. 108 SynNotch CARs, such as anti‐EGFRvIII sensors driving EphA2/IL13Rα2‐CAR expression, reduce antigen escape in glioblastoma 109 and are in clinical evaluation (NCT06186401).

OR‐gate designs incorporate two or more binders into a single CAR 110 , 111 , 112 or express multiple mono‐specific CARs within a single vector. 113 , 114 , 115 , 116 Bicistronic CARs allow distinct co‐stimulatory domains per construct to broaden signalling diversity. 117 , 118 Careful antigen selection ensures binding to one target does not compromise recognition of the other, particularly in tandem designs. 111 Dual/triple antigen targeting has been widely applied in B‐cell lymphomas, 119 , 120 where antigen loss drives relapse. 121 Since B‐cell aplasia is manageable clinically, simultaneous targeting of CD19, CD20 and CD22 is common. 122 , 123 Tian et al. developed a bicistronic CAR against GPC2 and B7‐H3 for neuroblastoma, 124 addressing intratumoural heterogenicity. CITE‐Seq identified optimal CARs for T‐cell expansion and phenotype. Compared with single‐target CAR‐T cells, the bicistronic product eliminated single‐ and dual‐positive tumour cells in vitro and in vivo, and demonstrated enhanced persistence and reduced exhaustion in mixed xenografts. An alternative strategy engineers CAR‐T cells to secrete bispecific TCEs recognising a second antigen, creating a localised OR‐gate. 125

NOT‐gate logic relies on differential expression of a protective antigen present on normal cells but absent on tumour cells, analogous to NK‐cell inhibition via KIR receptors. 126 Inhibitory CARs (iCARs) contain ITIM motifs that suppress T‐cell activation upon binding to a protective antigen. Early iCARs incorporating PD‐1 or CTLA‐4 intracellular domains enabled rapid, reversible inhibition, outperforming irreversible kill switches. 127 , 128 Enhancing iCAR avidity and incorporating dual‐inhibitory domains (e.g., PD‐1 combined with LAIR‐1 or SIGLEC‐9) substantially improves Boolean precision and reduces escape signalling latency. 129 Key design considerations include iCAR avidity and expression relative to the activating CAR. Increasing affinity alone may not enhance inhibition, whereas increased avidity, such as through higher receptor density or target abundance, can markedly improve suppression. 130 Notably, iCARs often inhibit proliferation and cytokine release more efficiently than immediate cytotoxicity, which depends on pre‐formed granules. 131

A clinically promising strategy leverages tumour HLA loss of heterozygosity. The Tmod system pairs an inhibitory CAR recognising HLA‐A*02 (commonly lost in tumours) with an activating CAR targeting an antigen such as mesothelin or carcinoembryonic antigen. 132 , 133 Tumour cells that lose β2‐microglobulin (β2M) evade inhibitory signalling and remain susceptible to killing, whereas normal HLA‐A02+/β2M⁺ cells are protected. 134 Our group developed CD16‐CLL1 iCAR‐T cells that retain activity against leukaemia while sparing neutrophils, significantly reducing the incidence of granulocytopaenia during CAR‐T‐cell therapy. 135

Limitations include: (1) engineering complexity and off‐target risk—complex multi‐antigen logic designs (e.g., tandem CARs, split circuits) may cause scFv interference or ‘leaky’ signalling, increasing on‐target/off‐tumour toxicity. (2) Strict antigen‐profile requirements—AND‐gates require co‐expression of two tumour‐restricted antigens, whereas NOT‐gates depend on consistent absence of a protective marker on malignancies. Tumour heterogeneity and antigen loss remain major escape routes. (3) Kinetic and potency constraints—inhibitory signals may be too slow to prevent early cytotoxic damage to normal cells, and intricate signalling modules may reduce overall T‐cell vigor. (4) Limited clinical validation—most logic‐gated systems remain in preclinical or early clinical phases. Their performance in human TMEs is not definitively established and suppressive tumour niches may impair circuit fidelity.

Armoured CAR‐T designs aim to enhance antitumour function by enabling autocrine cytokine support. IL‐2: IL‐2 secretion promotes T‐cell proliferation and tumour regression in preclinical melanoma models, 136 , 137 , 138 but clinical trials revealed toxicity without efficacy improvement. 139 IL‐12: IL‐12 is highly potent yet systemically toxic. 140 Tumour‐localised IL‐12 from CAR‐T cells increases antitumour immunity and reprograms myeloid compartments in murine models. 141 , 142 , 143 Inducible systems (e.g., NFAT‐regulated IL‐12) improve safety, 144 , 145 and CD19‐CAR/IL‐12 T cells achieved tumour clearance and resistance to Treg‐mediated suppression in syngeneic settings. 146 IL‐15: IL‐15 enhances T‐ and NK‐cell persistence 147 , 148 , 149 ; constitutive expression improves CAR‐T durability in preclinical studies, 150 , 151 yet uncontrolled proliferation and leukemogenic events have been reported. 152 Incorporating inducible suicide switches (e.g., iCaspase9) improves safety. 153 IL‐21: ex vivo IL‐21 improves expansion and cytotoxicity of CAR‐T cells against Nalm6, 154 although the feasibility of IL‐21 transgenes requires further assessment.

Limitations of armoured CARs include: (1) cytokine‐linked toxicity—even restricted IL‐12 or IL‐15 secretion risks CRS and ICANS. (2) Control challenges—tight temporal regulation is difficult; constitutive cytokine production can drive uncontrolled proliferation and malignant transformation. (3) Narrow therapeutic index—clinical experience, especially with IL‐2, shows toxicity often rises faster than benefit. (4) Limited long‐term clinical evidence—safety and durability of inducible cytokine‐engineering systems remain insufficiently tested in large trials.

Multiple cell types for UCAR‐T‐cell manufacturing show natural resistance to graft‐versus‐host disease (GvHD). These include γδ T cells, invariant natural killer T cells (iNKTs), double‐negative T cells (DNTs) and virus‐specific T cells (VSTs), alongside engineered platforms such as iPSCs and placental circulating T (P‐T) cells. Their distinctive antigen‐recognition mechanisms offer potential advantages in solid tumours.

γδ T cells: γδ T cells (5% of peripheral CD3⁺ cells, dominated by Vγ9Vδ2) recognise ligands independent of HLA, minimising GvHD risk. 155 Their innate cytotoxicity persists even after CAR‐target antigen loss, addressing heterogeneity. 104 ADI‐001 (CD20‐targeted γδ UCAR‐T) achieved a 67% overall response rate in B‐cell malignancies, and dose‐dependent expansion despite HLA mismatch. 156 , 157 γδ T cells also exploit NKG2D‐mediated targeting, for instance, temozolomide upregulates NKG2DL in glioblastoma, enhancing γδ T‐cell tumour recognition. 158

iNKT cells: iNKT cells recognise glycolipids via CD1d, an HLA‐independent mechanism that reduces GvHD risk. 159 They preferentially home to tumours through chemokines such as CCL2 and CCL20, making them advantageous for solid tumour targeting. 160 CAR‐iNKTs have been investigated in both haematologic and solid malignancies. 161 , 162 An advanced allogeneic CD19‐CAR iNKT product co‐expressing IL‐15 and shRNAs against B2M/CD74 (to reduce HLA‐I/II expression) demonstrated efficacy in R/R non‐hodgkin lymphoma and acute lymphoblastic leukemia. 163 Autologous GD2‐CAR iNKTs expressing IL‐15 induced a complete response in neuroblastoma. 164 Beyond direct tumour killing, CAR‐iNKTs contribute to host immunity by cross‐priming CD8⁺ T cells, 107 depleting immunosuppressive CD1d⁺ TAMs and MDSCs, 165 , 166 activating DCs, and promoting epitope spreading. 108

DNTs: DNTs (CD3⁺CD4CD8) rarely cause GvHD. Allogeneic DNTs expanded from AML patients showed safety and antitumour activity via NKG2D and DNAM‐1 pathways. 109 , 167 , 168 , 169 , 170 CD19‐CAR‐DNTs effectively targeted B‐cell leukaemia and lung cancer without inducing GvHD. 171 CAR4‐DNTs targeting T‐cell malignancies demonstrated improved persistence with idelalisib. 172 A phase I trial of allogeneic CD19‐CAR‐DNTs (RJMty19) in B‐cell lymphoma reported no ≥G3 CRS, ICANS, GvHD or DLTs and all high‐dose participants achieved responses. 173

VSTs and engineered cell sources: VSTs have a restricted TCR repertoire, lowering GvHD risk. 174 , 175 They are clinically used to treat post‐HSCT viral infections, 176 although their cancer applications remain limited. 177 , 178 iPSCs offer a renewable CAR‐T source, CAR‐iPSCs differentiated via 3D organoids generate functional T cells with uniform TCRs and reduced MHC expression, minimising GvHD and rejection. 179 Inhibiting G9a/GLP enhances iPSC‐T‐cell maturation and CAR effector function. 180

Limitations of allogeneic UCAR‐T‐cell sources: (1) cell source scarcity and expansion difficulty—γδ T cells, iNKTs and DNTs are rare in peripheral blood, increasing the complexity and cost of isolation, engineering and large‐scale expansion. (2) Uncertain persistence and potency—some subsets, such as VSTs, may have limited in vivo persistence and expansion, reducing long‐term activity. (3) Allo‐rejection—host immunity may eliminate allogeneic CAR‐T cells due to HLA mismatch, limiting persistence. Although HLA editing in iPSCs can mitigate this, residual risk remains. (4) Technical complexity—iPSC‐derived CAR‐T manufacturing requires reprogramming, gene editing and controlled differentiation, posing challenges for scalable and consistent production. (5) Unconventional safety profiles—native reactivity of γδ T cells or iNKT may cause on‐target, off‐tumour effects in normal tissues and requires careful evaluation.

8. CURRENT CHALLENGES AND LIMITATIONS RELATED TO CAR‐DCS

8.1 Plasticity of DCs and intrinsic dysfunction in AML

AML disrupts normal DC development, characterised by an accumulation of arrested DC precursors (LinHLA‐DR⁺CD11c⁺CD123⁺) and a deficiency of terminal DC subsets (BDCA‐1⁺/BDCA‐3⁺mDCs; BDCA‐2⁺pDCs) in FLT3‐ITD⁺ patients at diagnosis. Impaired myeloid DC function persists even in remission, 181 suggesting intrinsic maturation defects that may limit CAR‐DC efficacy. Chemotherapeutic agents such as daunorubicin can exacerbate immunosuppression by inducing ATP release from dying blasts, activating the P2X7‐IDO1 axis in DCs and promoting Treg expansion. 182 AML‐educated DCs and CAR‐DCs may therefore retain aberrant plasticity or tolergenic traits.

Tumour‐induced tolerogenic DC phenotypes: (1) metabolite‐driven tolerance—mregDCs migrate to tumour dLNs, where they suppress cross‐presentation and induce Th2 and Treg differentiation. Tumour‐derived lactate activates sterol regulatory element‐binding protein 2 (SREBP2) in DCs, promoting mevalonate‐dependent mregDC differentiation that suppresses CD8⁺ T cells and enhances Th2/Treg responses. 64 (2) Stromal signalling—CAF‐secreted WNT2 inhibits DC maturation via SOCS3/p‐JAK2/p‐STAT3 signalling, weakening antitumour immunity. 65 These pathways suggest that infused CAR‐DCs may be ‘re‐educated’ by the TME into IDO1⁺CD39⁺DCs or mregDCs reducing synergy with CAR‐T cells. (3) Logistical and biological complexity—dual‐cell manufacturing significantly increases cost and complexity versus single‐cell therapies. Predictive biomarkers remain unclear and TP53/KRAS mutations alone may not identify optimal candidates.

Preclinical evidence indicates several theoretical advantages of CAR‐DCs: (1) antigen acquisition fidelity—cDC vaccines rely on tumour lysates or selected peptides, whereas CAR‐DCs actively locate tumour cells in vivo and acquire a diverse antigen repertoire directly from the patient's TME, a critical feature in heterogeneous diseases such as AML. (2) Overcoming DC dysfunction—given that endogenous DCs in AML are often impaired, engineered CAR‐DCs with activation domains (e.g., CD40 or 4‐1BB) can achieve rapid maturation upon CAR signalling and resist TME suppression more effectively than cDC vaccines. (3) Synergistic immuno‐orchestration—beyond functioning as an improved antigen‐presenting platform, CAR‐DCs act as an in situ immune organisers that support co‐administered CAR‐T cells through cytokine secretion (e.g., IL‐12) and induction of epitope spreading. CAR‐DCs combined with CLL1 CAR‐T cells represent a novel approach to overcoming current efficacy barriers in R/R AML. A deeper mechanistic understanding of CAR‐DC biology will support clinical translation and provide a scientific foundation for integrating CAR‐DCs with CLL1 CAR‐T therapy in refractory or relapsed AML.

CAR‐DCs present several challenges: (1) functional complexity—the efficacy of the indirect ‘bystander’ killing of antigen‐negative tumours depends entirely on efficient cross‐presentation and epitope spreading, which may be suboptimal in immunosuppressive settings. (2) Safety considerations—although the synergy between CAR‐DCs and CAR‐T cells offers a compelling strategy to enhance antitumour immunity, both modalities are engineered for robust immune activation, raising concerns regarding amplified immune‐related toxicities, particularly CRS. Preclinical studies of CAR‐DC and CAR‐T co‐administration have demonstrated increased cytokine release (e.g., IL‐6, IL‐12, IFN‐γ), which could heighten CRS risk. Existing preliminary clinical data (e.g., NCT05631899) have not reported severe CRS although sample sizes remain small. CAR‐DCs may support more controlled activation through localised cytokine release and epitope spreading, potentially lowering systemic toxicity compared to standalone CAR‐T therapy. Nonetheless, future clinical protocols should include stringent monitoring, prophylactic measures (e.g., tocilizumab, corticosteroids), and inducible safety switches such as caspase‐based suicide systems to allow rapid intervention in the event of excessive immune activation. (3) TME resistance—while CAR‐DCs can be engineered to secrete cytokines such as IL‐12, the long‐term maintenance of this function and their ability to resist tolerogenic reprogramming by the suppressive TME remain to be proven. (4) CAR design and patient selection—optimisation of intracellular signalling modules (e.g., FLT3 andCD40) should be prioritised and future studies must establish rational patient selection criteria and incorporate predictive biomarkers, including TME profiling.

The combination of CAR‐DCs and CLL1‐targeted CAR‐T cells constitutes a transformative therapeutic strategy to simultaneously address antigen escape and immunosuppression in patients with R/R AML. CAR‐DCs function as first‐wave ‘immune orchestrators’ by remodelling the TME, thereby facilitating a more effective second‐wave attack by CAR‐T cells. Through synergistic enhancement of antigen presentation and reversal of T‐cell exhaustion, this dual‐cell approach has the potential to induce deeper and more durable remissions. To translate this promising preclinical strategy into clinical practice, several critical areas require further investigation. First, in the engineering of CAR‐DCs, the optimal co‐stimulatory signalling domains—such as CD40 for DC maturation versus FLT3L for expansion—must be determined, along with the feasibility of dual‐targeting CARs. Second, clinical development must establish the safety profile of this combination therapy, particularly with respect to CRS, and optimise dosing regimens and routes of administration. The identification of predictive biomarkers will be essential for appropriate patient selection. Furthermore, the therapeutic efficacy may be enhanced through triple‐combination approaches, such as incorporating ICIs to prevent functional exhaustion of both endogenous and CAR‐T cells. While the path from a compelling preclinical concept to an established clinical therapy remains complex, the rationally designed synergy between these two cellular immunotherapies represents a promising advance for improving outcomes in R/R AML.

AUTHOR CONTRIBUTIONS

Conceptualisation of the project: Rui Zhang, Hongkai Zhang and Mingfeng Zhao. Investigation: Rui Zhang. Writing articles and revision of the manuscript: Rui Zhang and Jinlin Zhang. All the authors approved the final manuscript as submitted and agree to be accountable for all aspects of the work.

CONFLICT OF INTEREST STATEMENT

The authors declare no potential conflicts of interest concerning the research, authorship and/or publication of this article.

ETHICS STATEMENT

Ethics approval was provided from the Department of Hematology at Tianjin First Central Hospital (Tianjin, China) (2020N212KY; Tianjin First Central Hospital Medical Ethics Committee). Clinical trial number: ChiCTR2000041025.

Supporting information

Supporting Information

Supporting Information

Supporting Information

CTM2-15-e70536-s001.pdf (200.8KB, pdf)

Supporting Information

CTM2-15-e70536-s003.pdf (202.4KB, pdf)

ACKNOWLEDGEMENTS

This work is sponsored by Tianjin Health Science and Technology Projects (grant no. TJWJ2025MS011) and Tianjin Key Clinical Specialty Construction Project and Tianjin Key Medical Discipline Construction Project (grant No. TJYXZDXK‐3‐001A‐004).

Zhang R, Zhang J, Zhang H, Zhao M. CAR‐DC combined with CAR‐T therapy for relapsed/refractory acute myeloid leukaemia: Research progress and future perspectives. Clin Transl Med. 2025;15:e70536. 10.1002/ctm2.70536

DATA AVAILABILITY STATEMENT

All data generated or analysed during this study are included in this article.

REFERENCES

  • 1. Perl AE, Martinelli G, Cortes JE, et al. Gilteritinib or chemotherapy for relapsed or refractory FLT3‐mutated AML. N Engl J Med. 2019;381(18):1728‐1740. [DOI] [PubMed] [Google Scholar]
  • 2. Pollyea DA, Bixby D, Perl A, et al. NCCN guidelines insights: acute myeloid leukemia, version 2.2021. J Natl Compr Canc Netw. 2021;19(1):16‐27. [DOI] [PubMed] [Google Scholar]
  • 3. Kreidieh F, Abou Dalle I, Moukalled N, et al. Relapse after allogeneic hematopoietic stem cell transplantation in acute myeloid leukemia: an overview of prevention and treatment. Int J Hematol. 2022;116(3):330‐340. [DOI] [PubMed] [Google Scholar]
  • 4. Raje N, Berdeja J, Lin Y, et al. Anti‐BCMA CAR T‐cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med. 2019;380(18):1726‐1737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Grupp SA, Kalos M, Barrett D, et al. Chimeric antigen receptor‐modified T cells for acute lymphoid leukemia. N Engl J Med. 2013;368(16):1509‐1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Qi Y, Zhao M, Hu Y, et al. Efficacy and safety of CD19‐specific CAR‐T cell based therapy in B‐cell acute lymphoblastic leukemia patients with CNSL. Blood. 2022;139(23):3376‐3386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Bhagwat AS, Torres L, Shestova O, et al. Cytokine‐mediated CAR‐T therapy resistance in AML. Nat Med. 2024;30(12):3697‐3708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Ma YJ, Dai HP, Cui QY, et al. Successful application of PD‐1 knockdown CLL‐1 CAR‐T therapy in two AML patients with post‐transplant relapse and failure of anti‐CD38 CAR‐T cell treatment. Am J Cancer Res. 2022;12(2):615‐621. [PMC free article] [PubMed] [Google Scholar]
  • 9. Maciocia PM, Wawrzyniecka PA, Philip B, et al. Targeting the T cell receptor β‐chain constant region for immunotherapy of T cell malignancies. Nat Med. 2017;23(12):1416‐1423. [DOI] [PubMed] [Google Scholar]
  • 10. 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(4):318‐330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Scholler N, Perbost R, Locke FL, et al. Tumor immune contexture is a determinant of anti‐CD19 CAR‐T cell efficacy in large B cell lymphoma. Nat Med. 2022;28(9):1872‐1882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Chanukya KC, Erik LK, Cameron JT, et al. Evolving strategies to overcome barriers in CAR‐T cell therapy for acute myeloid leukemia. Expert Rev Hematol. 2024;17(11):797‐818. [DOI] [PubMed] [Google Scholar]
  • 13. Haubner S, Mansilla‐Soto J, Nataraj S, et al. Cooperative CAR targeting to selectively eliminate AML and minimize escape. Cancer Cell. 2023;41(11):1871‐1891.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Atar D, Ruoff L, Mast AS, et al. Rational combinatorial targeting by adapter CAR‐T cells (AdCAR‐T) prevents antigen escape in acute myeloid leukemia. Leukemia. 2024;38(10):2183‐2195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Zoine JT, Immadisetty K, Ibanez‐Vega J, et al. Peptide‐scFv antigen recognition domains effectively confer CAR‐T cell multiantigen specificity. Cell Rep Med. 2024;5(2):101422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. 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(6):1439‐1453.e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Appelbaum J, Price AE, Oda K, et al. Drug‐regulated CD33‐targeted CAR‐T cells control AML using clinically optimized rapamycin dosing. J Clin Invest. 2024;134(9):e162593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. 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(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Sommer C, Cheng HY, Nguyen D, et al. Allogeneic FLT3 CAR‐T cells with an off‐switch exhibit potent activity against AML and can be depleted to expedite bone marrow recovery. Mol Ther. 2020;28(10):2237‐2251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Baumeister SH, Murad J, Werner L, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Hu Y, Zhou Y, Zhang M, et al. Genetically modified CD7‐targeting allogeneic CAR‐T cell therapy with enhanced efficacy for relapsed/refractory CD7‐positive hematological malignancies: a phase I clinical study. Cell Res. 2022;32(11):995‐1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. 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(1):82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Jin X, Xie D, Sun R, et al. CAR‐T cells dual‐target CD123 and NKG2DLs to eradicate AML cells and selectively target immunosuppressive cells. Oncoimmunology. 2023;12(1):2248826. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. 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(9):2202‐2213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Zhang H, Gan WT, Hao WG, et al. Successful anti‐CLL1 CAR‐T cell therapy in secondary acute myeloid leukemia. Front Oncol. 2020;10:685. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Jin X, Zhang M, Sun R, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Zhang X, Lv H, Xiao X, et al. A phase I clinical trial of CLL‐1 CAR‐T cells for the treatment of relapsed/refractory acute myeloid leukemia in adults. Blood. 2023;142:2106. [Google Scholar]
  • 28. Pei K, Xu H, Wang P, et al. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Ng BD, Rajagopalan A, Kousa AI, et al. IL‐18‐secreting multiantigen targeting CAR‐T cells eliminate antigen‐low myeloma in an immunocompetent mouse model. Blood. 2024;144(2):171‐186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Pérez‐Amill L, Bataller À, Delgado J, et al. Advancing CAR‐T therapy for acute myeloid leukemia: recent breakthroughs and strategies for future development. Front Immunol. 2023;14:1260470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Ayyadurai VAS, Deonikar P, McLure KG, et al. Molecular systems architecture of interactome in the acute myeloid leukemia microenvironment. Cancers (Basel). 2022;14(3):756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Guo R, Lü M, Cao F, et al. Single‐cell map of diverse immune phenotypes in the acute myeloid leukemia microenvironment. Biomark Res. 2021;9(1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Benton A, Liu J, Poussin MA, et al. Mutant KRAS peptide targeted CAR‐T cells engineered for cancer therapy. Cancer Cell. 2025;43(7):1365‐1376.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Meister H, Look T, Roth P, et al. Multifunctional mRNA‐based CAR‐T cells display promising antitumor activity against glioblastoma. Clin Cancer Res. 2022;28(21):4747‐4756. [DOI] [PubMed] [Google Scholar]
  • 35. Lee EHJ, Murad JP, Christian L, et al. Antigen‐dependent IL‐12 signaling in CAR‐T cells promotes regional to systemic disease targeting. Nat Commun. 2023;14:4737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Agliardi G, Liuzzi AR, Hotblack A, et al. Intratumoral IL‐12 delivery empowers CAR‐T cell immunotherapy in a pre‐clinical model of glioblastoma. Nat Commun. 2021;12(1):444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Pittet MJ, Di Pilato M, Garris C, et al. Dendritic cells as shepherds of T cell immunity in cancer. Immunity. 2023;56(10):2218‐2230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Huang L, Rong Y, Tang X, et al. Engineered exosomes as an in situ DC‐primed vaccine to boost antitumor immunity in breast cancer. Mol Cancer. 2022;21(1):45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Ascic E, Åkerström F, Sreekumar Nair M, et al. In vivo dendritic cell reprogramming for cancer immunotherapy. Science. 2024;386(6719):eadn9083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Zagorulya M, Spranger S. Once upon a prime: DCs shape cancer immunity. Trends Cancer. 2023;9(2):172‐184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Zhivaki D, Kennedy SN, Park J, et al. Correction of age‐associated defects in dendritic cells enables CD4+ T cells to eradicate tumors. Cell. 2024;187(15):3888‐3903.e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Sprooten J, Vanmeerbeek I, Datsi A, et al. Lymph node and tumor‐associated PD‐L1+ macrophages antagonize dendritic cell vaccines by suppressing CD8+ T cells. Cell Rep Med. 2024;5(1):101377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Rodrigues PF, Trsan T, Cvijetic G, et al. Progenitors of distinct lineages shape the diversity of mature type 2 conventional dendritic cells. Immunity. 2024;57(7):1567‐1585.e5. [DOI] [PubMed] [Google Scholar]
  • 44. Bayerl F, Meiser P, Donakonda S, et al. Tumor‐derived prostaglandin E2 programs cDC1 dysfunction to impair intratumoral orchestration of anti‐cancer T cell responses. Immunity. 2023;56(6):1341‐1358.e11. [DOI] [PubMed] [Google Scholar]
  • 45. Ferris ST, Durai V, Wu R, et al. cDC1 prime and are licensed by CD4+ T cells to induce anti‐tumour immunity. Nature. 2020;584(7822):624‐629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Maier B, Leader AM, Chen ST, et al. A conserved dendritic‐cell regulatory program limits antitumour immunity. Nature. 2020;580(7802):257‐262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Zilionis R, Engblom C, Pfirschke C, et al. Single‐cell transcriptomics of human and mouse lung cancers reveals conserved myeloid populations across individuals and species. Immunity. 2019;50(5):1317‐1334.e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Villani AC, Satija R, Reynolds G, et al. Single‐cell RNA‐seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science. 2017;356(6335):eaah4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Cytlak U, Resteu A, Pagan S, et al. Differential IRF8 transcription factor requirement defines two pathways of dendritic cell development in humans. Immunity. 2020;53(2):353‐370. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Ma T, Chu X, Wang J, et al. Pan‐cancer analyses refine the single‐cell portrait of tumor‐infiltrating dendritic cells. Cancer Res. 2025;85(19):3596‐3613. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Tao Y, Ma Y, Gu L, et al. Single‐cell RNA sequencing reveals Shen‐Bai‐Jie‐Du decoction retards colorectal tumorigenesis by regulating the TMEM131‐TNF signaling pathway‐mediated differentiation of immunosuppressive dendritic cells. Acta Pharm Sin B. 2025;15(7):3545‐3560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Liu Z, Wang H, Li Z, et al. Dendritic cell type 3 arises from Ly6C+ monocyte‐dendritic cell progenitors. Immunity. 2023;56(8):1761‐1777.e6. [DOI] [PubMed] [Google Scholar]
  • 53. Yao RQ, Li ZX, Wang LX, et al. Single‐cell transcriptome profiling of the immune space‐time landscape reveals dendritic cell regulatory program in polymicrobial sepsis. Theranostics. 2022;12(10):4606‐4628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Hübbe ML, Jæhger DE, Andresen TL, et al. Leveraging endogenous dendritic cells to enhance the therapeutic efficacy of adoptive T‐cell therapy and checkpoint blockade. Front Immunol. 2020;11:578349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Theisen DJ, Davidson JT 4th, Briseño CG, et al. WDFY4 is required for cross‐presentation in response to viral and tumor antigens. Science. 2018;362(6415):694‐699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Sun S, Ding Z, Gao L, et al. A dendritic/tumor fusion cell vaccine enhances efficacy of nanobody‐based CAR‐T cells against solid tumor. Theranostics. 2023;13(14):5099‐5113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Pophali P, Cheloni G, Stone RM, et al. Randomized phase II trial of dendritic cell/AML fusion cell vaccination compared to standard of care therapy in AML CR1. Blood. 2024;144(1):4256. [Google Scholar]
  • 58. Wu M, Zhang L, Zhang H, et al. CD19 chimeric antigen receptor‐redirected T cells combined with epidermal growth factor receptor pathway substrate 8 peptide‐derived dendritic cell vaccine in leukemia. Cytotherapy. 2019;21(6):659‐670. [DOI] [PubMed] [Google Scholar]
  • 59. Perez CR, De Palma M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat Commun. 2019;100:5408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Ma L, Hostetler A, Morgan DM, et al. Vaccine‐boosted CAR‐T crosstalk with host immunity to reject tumors with antigen heterogeneity. Cell. 2023;186(15):3148‐3165.e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Zhang M, Wang Y, Chen X, et al. DC vaccine enhances CAR‐T cell antitumor activity by overcoming T cell exhaustion and promoting T cell infiltration in solid tumors. Clin Transl Oncol. 2023;25(10):2972‐2982. [DOI] [PubMed] [Google Scholar]
  • 62. Marciscano AE, Anandasabapathy N. The role of dendritic cells in cancer and anti‐tumor immunity. Semin Immunol. 2021;52:101481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Hegde S, Krisnawan VE, Herzog BH, et al. Dendritic cell paucity leads to dysfunctional immune surveillance in pancreatic cancer. Cancer Cell. 2020;37(3):289‐307.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Plebanek MP, Xue Y, Nguyen YV, et al. A lactate‐SREBP2 signaling axis drives tolerogenic dendritic cell maturation and promotes cancer progression. Sci Immunol. 2024;9(95):eadi4191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Huang TX, Tan XY, Huang HS, et al. Targeting cancer‐associated fibroblast‐secreted WNT2 restores dendritic cell‐mediated antitumour immunity. Gut. 2022;71(2):333‐344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Sathe A, Grimes SM, Lau BT, et al. Single‐cell genomic characterization reveals the cellular reprogramming of the gastric tumor microenvironment. Clin Cancer Res. 2020;26(11):2640‐2653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Devalaraja S, To TKJ, Folkert IW, et al. Tumor‐derived retinoic acid regulates intratumoral monocyte differentiation to promote immune suppression. Cell. 2020;180(6):1098‐1114.e16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Garris CS, Arlauckas SP, Kohler RH, et al. Successful anti‐PD‐1 cancer immunotherapy requires T cell‐dendritic cell crosstalk involving the cytokines IFN‐γ and IL‐12. Immunity. 2018;49(6):1148‐1161.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Ghasemi A, Martinez‐Usatorre A, Li L, et al. Cytokine‐armed dendritic cell progenitors for antigen‐agnostic cancer immunotherapy. Nat Cancer. 2024;5(2):240‐261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Lam KC, Araya RE, Huang A, et al. Microbiota triggers STING‐type I IFN‐dependent monocyte reprogramming of the tumor microenvironment. Cell. 2021;184(21):5338‐5356.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Chudnovskiy A, Castro TBR, Nakandakari‐Higa S, et al. Proximity‐dependent labeling identifies dendritic cells that drive the tumor‐specific CD4+ T cell response. Sci Immunol. 2024;9(100):eadq8843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Cabeza‐Cabrerizo M, Cardoso A, Minutti CM, et al. Dendritic cells revisited. Annu Rev Immunol. 2021;39:131‐166. [DOI] [PubMed] [Google Scholar]
  • 73. He F, Wu Z, Liu C, et al. Targeting BCL9/BCL9L enhances antigen presentation by promoting conventional type 1 dendritic cell (cDC1) activation and tumor infiltration. Signal Transduct Target Ther. 2024;9(1):139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Zhu W, Liu C, Xi K, et al. Discovery of novel 1‐phenylpiperidine urea‐containing derivatives inhibiting β‐catenin/BCL9 interaction and exerting antitumor efficacy through the activation of antigen presentation of cDC1 cells. J Med Chem. 2024;67(15):12485‐12520. [DOI] [PubMed] [Google Scholar]
  • 75. Lai J, Mardiana S, House IG, et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat Immunol. 2020;21(8):914‐926. [DOI] [PubMed] [Google Scholar]
  • 76. Gardner A, de Mingo Pulido Á, Hänggi K, et al. TIM‐3 blockade enhances IL‐12‐dependent antitumor immunity by promoting CD8+ T cell and XCR1+ dendritic cell spatial co‐localization. J Immunother Cancer. 2022;10(1):e003571. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Pfirschke C, Zilionis R, Engblom C, et al. Macrophage‐targeted therapy unlocks antitumoral cross‐talk between IFNγ‐secreting lymphocytes and IL12‐producing dendritic cells. Cancer Immunol Res. 2022;10(1):40‐55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Suh HC, Pohl K, Javier APL, Slamon DJ, Chute JP. Effect of dendritic cells (DC) transduced with chimeric antigen receptor (CAR) on CAR‐T cell cytotoxicity. JCO. 2017;35:144‐144. [Google Scholar]
  • 79. Suh HC, Pohl KA, Termini C, et al. Bioengineered autologous dendritic cells enhance CAR‐T cell cytotoxicity by providing cytokine stimulation and intratumoral dendritic cells. Blood. 2018;132:3693. [Google Scholar]
  • 80. Martinez‐Usatorre A, De Palma M. Dendritic cell cross‐dressing and tumor immunity. EMBO Mol Med. 2022;14(10):e16523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Ashour D, Arampatzi P, Pavlovic V, et al. IL‐12 from endogenous cDC1, and not vaccine DC, is required for Th1 induction. JCI Insight. 2020;5(10):e135143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Chimeric Antigen Receptor Dendritic Cells (CAR‐DCs) and Their Preparation and Usage Methods. Patent number: CN 115066248A.
  • 83. CAR‐DC Cells and Their Preparation Methods and Applications. Patent number: CN 117165531A.
  • 84. O'Sullivan BJ, Thomas R. CD40 ligation conditions dendritic cell antigen‐presenting function through sustained activation of NF‐kappaB. J Immunol. 2002;168(11):5491‐5498. [DOI] [PubMed] [Google Scholar]
  • 85. Guhr A, Kobold S, Seltmann S, Seiler Wulczyn AEM, Kurtz A, Löser P. Clinical Study Database for HPSC‐Based Cell Therapies . 2024.
  • 86. Alidadi M, Barzgar H, Zaman M, et al. Combining the induced pluripotent stem cell (iPSC) technology with chimeric antigen receptor (CAR)‐based immunotherapy: recent advances, challenges, and future prospects. Front Cell Dev Biol. 2024;12:1491282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Themeli M, Kloss CC, Ciriello G, et al. Generation of tumor‐targeted human T lymphocytes from induced pluripotent stem cells for cancer therapy. Nat Biotechnol. 2013;31(10):928‐933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Nishimura T, Kaneko S, Kawana‐Tachikawa A, et al. Generation of rejuvenated antigen‐specific T cells by reprogramming to pluripotency and redifferentiation. Cell Stem Cell. 2013;12(1):114‐126. [DOI] [PubMed] [Google Scholar]
  • 89. Vizcardo R, Masuda K, Yamada D, et al. Regeneration of human tumor antigen‐specific T cells from iPSCs derived from mature CD8(+) T cells. Cell Stem Cell. 2013;12(1):31‐36. [DOI] [PubMed] [Google Scholar]
  • 90. Ando M, Nishimura T, Yamazaki S, et al. A safeguard system for induced pluripotent stem cell‐derived rejuvenated T cell therapy. Stem Cell Rep. 2015;5(4):597‐608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. van der Stegen SJC, Lindenbergh PL, Petrovic RM, et al. Generation of T‐cell‐receptor‐negative CD8αβ‐positive CAR T cells from T‐cell‐derived induced pluripotent stem cells. Nat Biomed Eng. 2022;6(11):1284‐1297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Pinto E, Lione L, Compagnone M, et al. From ex vivo to in vivo chimeric antigen T cells manufacturing: new horizons for CAR T‐cell based therapy. J Transl Med. 2025;23(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Tao Z, Chyra Z, Kotulová J, et al. Impact of T cell characteristics on CAR‐T cell therapy in hematological malignancies. Blood Cancer J. 2024;14(1):213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Hunter TL, Bao Y, Zhang Y, et al. In vivo CAR T cell generation to treat cancer and autoimmune disease. Science. 2025;388(6753):1311‐1317. [DOI] [PubMed] [Google Scholar]
  • 95. Agarwal S, Hanauer JDS, Frank AM, Riechert V, Thalheimer FB, Buchholz CJ. In vivo generation of CAR T cells selectively in human CD4+ lymphocytes. Mol Ther. 2020;28(8):1783‐1794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Frank AM, Braun AH, Scheib L, et al. Combining T‐cell‐specific activation and in vivo gene delivery through CD3‐targeted lentiviral vectors. Blood Adv. 2020;4(22):5702‐5715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Xiao Y, Zhu T, Chen Z, Huang X. Lung metastasis and recurrence is mitigated by CAR macrophages, in‐situ‐generated from mRNA delivered by small extracellular vesicles. Nat Commun. 2025;16(1):7166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Nawaz W, Huang B, Xu S, et al. AAV‐mediated in vivo CAR gene therapy for targeting human T‐cell leukemia. Blood Cancer J. 2021;11(6):119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Ma H, Yan Z, Gu R, et al. Loop33 × 123 CAR‐T targeting CD33 and CD123 against immune escape in acute myeloid leukemia. Cancer Immunol Immunother. 2024;74(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Roybal KT, Rupp LJ, Morsut L, et al. Precision tumor recognition by T cells with combinatorial antigen‐sensing circuits. Cell. 2016;164(4):770‐779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Pham E, Lutterbuese P, Deegen P, et al. Abstract ND06: AMG 305, a dual targeting BiTE®molecule with selective activity for solid tumors that co‐express CDH3 and MSLN. Cancer Res. 2023;83(7):ND06. [Google Scholar]
  • 102. Dicara DM, Bhakta S, Go MA, et al. Development of T‐cell engagers selective for cells co‐expressing two antigens. MAbs. 2022;14(1):2115213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Carretero‐Iglesia L, Hall OJ, Berret J, et al. ISB 2001 trispecific T cell engager shows strong tumor cytotoxicity and overcomes immune escape mechanisms of multiple myeloma cells. Nat Cancer. 2024;5(10):1494‐1514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Banaszek A, Bumm TGP, Nowotny B, et al. On‐target restoration of a split T cell‐engaging antibody for precision immunotherapy. Nat Commun. 2019;10(1):5387. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Evolving T. Revitope—third‐generation T cell engager immunotherapies. Biopharma Dealmakers. 2021:B48‐B49. [Google Scholar]
  • 106. Minogue E, Millar D, Chuan  Y, et al. Redirecting T‐cells against AML in a multidimensional targeting space using T‐cell engaging antibody circuits (TEAC). Blood. 2019;134(1):2653. ISSN 0006‐4971. [Google Scholar]
  • 107. van der Schans JJ, Wang Z, van Arkel J, et al. Specific targeting of multiple myeloma by dual split‐signaling chimeric antigen receptor T cells directed against CD38 and CD138. Clin Cancer Res. 2023;29(20):4219‐4229. [DOI] [PubMed] [Google Scholar]
  • 108. Tousley AM, Rotiroti MC, Labanieh L, et al. Co‐opting signalling molecules enables logic‐gated control of CAR T cells. Nature. 2023;615(7952):507‐516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Choe JH, Watchmaker PB, Simic MS, et al. SynNotch‐CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 2021;13(591):eabe7378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Schneider D, Xiong Y, Wu D, et al. Trispecific CD19‐CD20‐CD22‐targeting duoCAR‐T cells eliminate antigen‐heterogeneous B cell tumors in preclinical models. Sci Transl Med. 2021;13(586):eabc6401. [DOI] [PubMed] [Google Scholar]
  • 111. Tong C, Zhang Y, Liu Y, et al. Optimized tandem CD19/CD20 CAR‐engineered T cells in refractory/relapsed B‐cell lymphoma. Blood. 2020;136(14):1632‐1644. Blood. 2023;141(15):1896. [DOI] [PubMed] [Google Scholar]
  • 112. Schneider D, Xiong Y, Wu D, et al. A tandem CD19/CD20 CAR lentiviral vector drives on‐target and off‐target antigen modulation in leukemia cell lines. J Immunother Cancer. 2017;5:42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Lam N, Finney R, Yang S, et al. Development of a bicistronic anti‐CD19/CD20 CAR construct including abrogation of unexpected nucleic acid sequence deletions. Mol Ther Oncolytics. 2023;30:132‐149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Wang Z, Lu Y, Liu Y, et al. Novel CD123×CD33 bicistronic chimeric antigen receptor (CAR)‐T therapy has potential to reduce escape from single‐target CAR‐T with no more hematotoxicity. Cancer Commun (Lond). 2023;43(10):1178‐1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Kokalaki E, Ma B, Ferrari M, et al. Dual targeting of CD19 and CD22 against B‐ALL using a novel high‐sensitivity aCD22 CAR. Mol Ther. 2023;31(7):2089‐2104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116. Roddie C, Lekakis LJ, Marzolini MAV, et al. Dual targeting of CD19 and CD22 with bicistronic CAR‐T cells in patients with relapsed/refractory large B‐cell lymphoma. Blood. 2023;141(20):2470‐2482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Wang Y, Zhong K, Ke J, et al. Combined 4‐1BB and ICOS co‐stimulation improves anti‐tumor efficacy and persistence of dual anti‐CD19/CD20 chimeric antigen receptor T cells. Cytotherapy. 2021;23(8):715‐723. doi: 10.1016/j.jcyt.2021.02.117 [DOI] [PubMed] [Google Scholar]
  • 118. Halim L, Das KK, Larcombe‐Young D, et al. Engineering of an avidity‐optimized CD19‐specific parallel chimeric antigen receptor that delivers dual CD28 and 4‐1BB co‐stimulation. Front Immunol. 2022;13:836549. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Yang J, Guo H, Han L, Song Y, Zhou K. Dual‐targeted CAR T‐cell immunotherapies for hematological malignancies: latest updates from the 2023 ASH annual meeting. Exp Hematol Oncol. 2024;13(1):25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Zah E, Lin MY, Silva‐Benedict A, Jensen MC, Chen YY. T cells expressing CD19/CD20 bispecific chimeric antigen receptors prevent antigen escape by malignant B cells. Cancer Immunol Res. 2016;4(6):498‐508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Sotillo E, Barrett DM, Black KL, et al. Convergence of acquired mutations and alternative splicing of CD19 enables resistance to CART‐19 immunotherapy. Cancer Discov. 2015;5(12):1282‐1295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Furqan F, Shah NN. Bispecific CAR T‐cells for B‐cell malignancies. Expert Opin Biol Ther. 2022;22(8):1005‐1015. [DOI] [PubMed] [Google Scholar]
  • 123. Wang L, Fang C, Kang Q, et al. Bispecific CAR‐T cells targeting CD19/20 in patients with relapsed or refractory B cell non‐Hodgkin lymphoma: a phase I/II trial. Blood Cancer J. 2024;14(1):130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Tian M, Cheuk AT, Wei JS, et al. An optimized bicistronic chimeric antigen receptor against GPC2 or CD276 overcomes heterogeneous expression in neuroblastoma. J Clin Invest. 2022;132(16):e155621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Choi BD, Gerstner ER, Frigault MJ, et al. Intraventricular CARv3‐TEAM‐E T cells in recurrent glioblastoma. N Engl J Med. 2024;390(14):1290‐1298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Anderson SK. Molecular evolution of elements controlling HLA‐C expression: adaptation to a role as a killer‐cell immunoglobulin‐like receptor ligand regulating natural killer cell function. HLA. 2018;92(5):271‐278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Fedorov VD, Themeli M, Sadelain M. PD‐1‐ and CTLA‐4‐based inhibitory chimeric antigen receptors (iCARs) divert off‐target immunotherapy responses. Sci Transl Med. 2013;5(215):215ra172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Shabaneh TB, Moffett HF, Stull SM, et al. Safety switch optimization enhances antibody‐mediated elimination of CAR T cells. Front Mol Med. 2022;2:1026474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Bangayan NJ, Wang L, Burton Sojo G, et al. Dual‐inhibitory domain iCARs improve the efficiency of the AND‐NOT gate CAR T strategy. Proc Natl Acad Sci U S A. 2023;120(47):e2312374120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Bangayan NJ, Wang L, Burton Sojo G, et al. Dual‐inhibitory domain iCARs improve the efficiency of the AND‐NOT gate CAR T strategy. Proc Natl Acad Sci U S A. 2023;120(47):e2312374120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Funk MA, Heller G, Waidhofer‐Söllner P, Leitner J, Steinberger P. Inhibitory CARs fail to protect from immediate T cell cytotoxicity. Mol Ther. 2024;32(4):982‐999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Manry D, Bolanos K, DiAndreth B, Mock JY, Kamb A. Robust in vitro pharmacology of Tmod, a synthetic dual‐signal integrator for cancer cell therapy. Front Immunol. 2022;13:826747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Sandberg ML, Wang X, Martin AD, et al. A carcinoembryonic antigen‐specific cell therapy selectively targets tumor cells with HLA loss of heterozygosity in vitro and in vivo. Sci Transl Med. 2022;14(634):eabm0306. [DOI] [PubMed] [Google Scholar]
  • 134. Wang H, Liu B, Wei J. Beta2‐microglobulin (B2M) in cancer immunotherapies: biological function, resistance and remedy. Cancer Lett. 2021;517:96‐104. [DOI] [PubMed] [Google Scholar]
  • 135. Zhang R, Zhao Y, Chai X, et al. Modified CD15/CD16‐CLL1 inhibitory CAR‐T cells for mitigating granulocytopenia toxicities in the treatment of acute myeloid leukemia. Transl Oncol. 2025;52:102225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Safran H, Druta  M, Morse M, et al. Abstract CT111: results of a phase 1 dose escalation study of ERY974, an anti‐glypican 3 (GPC3)/CD3 bispecific antibody, in patients with advanced solid tumors. Cancer Res. 2021;81(13):CT111. [Google Scholar]
  • 137. Qi C, Liu C, Gong J, et al. Claudin18.2‐specific CAR T cells in gastrointestinal cancers: phase 1 trial final results. Nat Med. 2024;30(8):2224‐2234. [DOI] [PubMed] [Google Scholar]
  • 138. Zhang Q, Fu Q, Cao W, et al. Phase I study of C‐CAR031, a GPC3‐specific TGFbRIIDN armored autologous CAR‐T, in patients with advanced hepatocellular carcinoma (HCC). J Clin Oncol. 2024;42(16):4019. [Google Scholar]
  • 139. Carvajal RD, Butler MO, Shoushtari AN, et al. Clinical and molecular response to tebentafusp in previously treated patients with metastatic uveal melanoma: a phase 2 trial. Nat Med. 2022;28(11):2364‐2373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Giffin MJ, Cooke K, Lobenhofer EK, et al. AMG 757, a half‐life extended, DLL3‐targeted bispecific T‐cell engager, shows high potency and sensitivity in preclinical models of small‐cell lung cancer. Clin Cancer Res. 2021;27(5):1526‐1537. [DOI] [PubMed] [Google Scholar]
  • 141. Klemm F, Joyce JA. Microenvironmental regulation of therapeutic response in cancer. Trends Cell Biol. 2015;25(4):198‐213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. van de Donk NWCJ, Zweegman S. T‐cell‐engaging bispecific antibodies in cancer. Lancet. 2023;402(10396):142‐158. [DOI] [PubMed] [Google Scholar]
  • 143. Savanur MA, Weinstein‐Marom H, Gross G. Implementing logic gates for safer immunotherapy of cancer. Front Immunol. 2021;12:780399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Arvedson T, Bailis JM, Britten CD, et al. Targeting solid tumors with bispecific T cell engager immune therapy. Annu Rev Cancer Biol. 2022;6:17‐34. [Google Scholar]
  • 145. Middelburg J, Kemper K, Engelberts P, Labrijn AF, Schuurman J, van Hall T. Overcoming challenges for CD3‐bispecific antibody therapy in solid tumors. Cancers (Basel). 2021;13(2):287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. DiAndreth B, Hamburger AE, Xu H, Kamb A. The Tmod cellular logic gate as a solution for tumor‐selective immunotherapy. Clin Immunol. 2022;241:109030. [DOI] [PubMed] [Google Scholar]
  • 147. Miyamoto T, Razavi S, DeRose R, Inoue T. Synthesizing biomolecule‐based Boolean logic gates. ACS Synth Biol. 2013;2(2):72‐82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Perna F, Berman SH, Soni RK, et al. Integrating proteomics and transcriptomics for systematic combinatorial chimeric antigen receptor therapy of AML. Cancer Cell. 2017;32(4):506‐519.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Haubner S, Perna F, Köhnke T, et al. Coexpression profile of leukemic stem cell markers for combinatorial targeted therapy in AML. Leukemia. 2019;33(1):64‐74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. Majzner RG, Mackall CL. Tumor antigen escape from CAR T‐cell therapy. Cancer Discov. 2018;8(10):1219‐1226. [DOI] [PubMed] [Google Scholar]
  • 151. Furqan F, Shah NN. Multispecific CAR T cells deprive lymphomas of escape via antigen loss. Annu Rev Med. 2023;74:279‐291. [DOI] [PubMed] [Google Scholar]
  • 152. Samur MK, Fulciniti M, Aktas Samur A, et al. Biallelic loss of BCMA as a resistance mechanism to CAR T cell therapy in a patient with multiple myeloma. Nat Commun. 2021;12(1):868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Cohen YC, Morillo D, Gatt ME, et al. First results from the RedirecTT‐1 study with teclistamab (tec)+ talquetamab (tal) simultaneously targeting BCMA and GPRC5D in patients (pts) with relapsed/refractory multiple myeloma (RRMM). J Clin Oncol. 2023;41(16):8002. [Google Scholar]
  • 154. Saber H, Del Valle P, Ricks TK, Leighton JK. An FDA oncology analysis of CD3 bispecific constructs and first‐in‐human dose selection. Regul Toxicol Pharmacol. 2017;90:144‐152. [DOI] [PubMed] [Google Scholar]
  • 155. Appelbaum J, Price AE, Oda K, et al. Drug‐regulated CD33‐targeted CAR T cells control AML using clinically optimized rapamycin dosing. J Clin Invest. 2024;134(9):e162593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156. Bouquet L, Bôle‐Richard E, Warda W, et al. RapaCaspase‐9‐based suicide gene applied to the safety of IL‐1RAP CAR‐T cells. Gene Ther. 2023;30(9):706‐713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Zheng Y, Nandakumar KS, Cheng K. Optimization of CAR‐T cell‐based therapies using small‐molecule‐based safety switches. J Med Chem. 2021;64(14):9577‐9591. [DOI] [PubMed] [Google Scholar]
  • 158. Wang Q, He F, He W, et al. A transgene‐encoded truncated human epidermal growth factor receptor for depletion of anti‐B‐cell maturation antigen CAR‐T cells. Cell Immunol. 2021;363:104342. [DOI] [PubMed] [Google Scholar]
  • 159. Ma L, Zhang K, Xu J, et al. Building a novel TRUCK by harnessing the endogenous IFN‐gamma promoter for cytokine expression. Mol Ther. 2024;32(8):2728‐2740. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Lanitis E, Poussin M, Klattenhoff AW, et al. Chimeric antigen receptor T cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol Res. 2013;1(1):43‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Smole A, Benton A, Poussin MA, et al. Expression of inducible factors reprograms CAR‐T cells for enhanced function and safety. Cancer Cell. 2022;40(12):1470‐1487.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Arcangeli S, Rotiroti MC, Bardelli M, et al. Balance of anti‐CD123 chimeric antigen receptor binding affinity and density for the targeting of acute myeloid leukemia. Mol Ther. 2017;25(8):1933‐1945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Roybal KT, Rupp LJ, Morsut L, et al. Precision tumor recognition by T cells with combinatorial antigen‐sensing circuits. Cell. 2016;164(4):770‐779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164. Han C, Sim SJ, Kim SH, et al. Desensitized chimeric antigen receptor T cells selectively recognize target cells with enhanced antigen expression. Nat Commun. 2018;9(1):468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Roybal KT, Lim WA. Synthetic immunology: hacking immune cells to expand their therapeutic capabilities. Annu Rev Immunol. 2017;35:229‐253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166. Chmielewski M, Hombach A, Heuser C, Adams GP, Abken H. T cell activation by antibody‐like immunoreceptors: increase in affinity of the single‐chain fragment domain above threshold does not increase T cell activation against antigen‐positive target cells but decreases selectivity. J Immunol. 2004;173(12):7647‐7653. [DOI] [PubMed] [Google Scholar]
  • 167. O'Rourke DM, Nasrallah MP, Desai A, et al. A single dose of peripherally infused EGFRvIII‐directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma. Sci Transl Med. 2017;9(399):eaaa0984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Bagley SJ, Binder ZA, Lamrani L, et al. Repeated peripheral infusions of anti‐EGFRvIII CAR T cells in combination with pembrolizumab show no efficacy in glioblastoma: a phase 1 trial. Nat Cancer. 2024;5(3):517‐531. [DOI] [PubMed] [Google Scholar]
  • 169. Wang Z, Wang F, Zhong J, et al. Using apelin‐based synthetic Notch receptors to detect angiogenesis and treat solid tumors. Nat Commun. 2020;11(1):2163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170. Sternjak A, Lee F, Thomas O, et al. Preclinical assessment of AMG 596, a bispecific T‐cell engager (BiTE) immunotherapy targeting the tumor‐specific antigen EGFRvIII. Mol Cancer Ther. 2021;20(5):925‐933. [DOI] [PubMed] [Google Scholar]
  • 171. Salzer B, Schueller CM, Zajc CU, et al. Engineering AvidCARs for combinatorial antigen recognition and reversible control of CAR function. Nat Commun. 2020;11(1):4166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172. Iwahori K, Kakarla S, Velasquez MP, et al. Engager T cells: a new class of antigen‐specific T cells that redirect bystander T cells. Mol Ther. 2015;23(1):171‐178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Velasquez MP, Torres D, Iwahori K, et al. T cells expressing CD19‐specific engager molecules for the immunotherapy of CD19‐positive malignancies. Sci Rep. 2016;6:27130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Rosenthal M, Curry R, Reardon DA, et al. Safety, tolerability, and pharmacokinetics of anti‐EGFRvIII antibody‐drug conjugate AMG 595 in patients with recurrent malignant glioma expressing EGFRvIII. Cancer Chemother Pharmacol. 2019;84(2):327‐336. [DOI] [PubMed] [Google Scholar]
  • 175. Bacac M, Fauti T, Sam J, et al. A novel carcinoembryonic antigen T‐cell bispecific antibody (CEA TCB) for the treatment of solid tumors. Clin Cancer Res. 2016;22(13):3286‐3297. [DOI] [PubMed] [Google Scholar]
  • 176. Segal NH, Melero I, Moreno V, et al. CEA‐CD3 bispecific antibody cibisatamab with or without atezolizumab in patients with CEA‐positive solid tumours: results of two multi‐institutional phase 1 trials. Nat Commun. 2024;15(1):4091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Yin Y, Rodriguez JL, Li N, et al. Locally secreted BiTEs complement CAR T cells by enhancing killing of antigen heterogeneous solid tumors. Mol Ther. 2022;30(7):2537‐2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Choi BD, Yu X, Castano AP, et al. CAR‐T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat Biotechnol. 2019;37(9):1049‐1058. [DOI] [PubMed] [Google Scholar]
  • 179. Schmidts A, Srivastava AA, Ramapriyan R, et al. Tandem chimeric antigen receptor (CAR) T cells targeting EGFRvIII and IL‐13Rα2 are effective against heterogeneous glioblastoma. Neurooncol Adv. 2022;5(1):vdac185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Mestermann K, Giavridis T, Weber J, et al. The tyrosine kinase inhibitor dasatinib acts as a pharmacologic on/off switch for CAR T cells. Sci Transl Med. 2019;11(499):eaau5907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. Rickmann M, Macke L, Sundarasetty BS, et al. Monitoring dendritic cell and cytokine biomarkers during remission prior to relapse in patients with FLT3‐ITD acute myeloid leukemia. Ann Hematol. 2013;92(8):1079‐1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Lecciso M, Ocadlikova D, Sangaletti S, et al. ATP release from chemotherapy‐treated dying leukemia cells elicits an immune suppressive effect by increasing regulatory T cells and tolerogenic dendritic cells. Front Immunol. 2017;8:1918. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supporting Information

Supporting Information

Supporting Information

CTM2-15-e70536-s001.pdf (200.8KB, pdf)

Supporting Information

CTM2-15-e70536-s003.pdf (202.4KB, pdf)

Data Availability Statement

All data generated or analysed during this study are included in this article.


Articles from Clinical and Translational Medicine are provided here courtesy of John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics

RESOURCES