Skip to main content

Current advances and challenges in CAR T-Cell therapy for solid tumors: tumor-associated antigens and the tumor microenvironment

Abstract

The past decade has witnessed ongoing progress in immune therapy to ameliorate human health. As an emerging technique, chimeric antigen receptor (CAR) T-cell therapy has the advantages of specific killing of cancer cells, a high remission rate of cancer-induced symptoms, rapid tumor eradication, and long-lasting tumor immunity, opening a new window for tumor treatment. However, challenges remain in CAR T-cell therapy for solid tumors due to target diversity, tumor heterogeneity, and the complex microenvironment. In this review, we have outlined the development of the CAR T-cell technique, summarized the current advances in tumor-associated antigens (TAAs), and highlighted the importance of tumor-specific antigens (TSAs) or neoantigens for solid tumors. We also addressed the challenge of the TAA binding domain in CARs to overcome off-tumor toxicity. Moreover, we illustrated the dominant tumor microenvironment (TME)-induced challenges and new strategies based on TME-associated antigens (TMAs) for solid tumor CAR T-cell therapy.

Background

In the last decade, scientists and clinicians have come to recognize the great value of chimeric antigen receptor (CAR) T-cell therapy in the fight against hematological malignancies. Since 2017, numerous CAR T-cell therapies have been approved and/or reached clinical validation trials for hematological tumors by targeting CD19, CD20, CD22, and the B-cell maturation antigen (BCMA) [1,2,3,4,5,6,7,8,9,10]. As a novel class of therapies, CAR T-cell therapy has helped transform the treatment landscape for people with hematological malignancies [11, 12]. Currently, to further optimize CAR T-cell systems, scientists are focusing on CAR structure design, transfection, and cell culture techniques. Many of these techniques have reached clinical validation trials [13,14,15,16].

Because of its high remission rates, rapid tumor eradication, and long-lasting response in patients, CAR T-cell therapy has shown great therapeutic potential not only for hematological malignancies but also for solid tumors [17, 18]. However, the use of CAR T cells for solid tumor therapy remains a challenge due to the lack of specific antigens, off-target effects, and the complex tumor microenvironment [19]. In this review, we have outlined the development of the CAR T-cell technique, summarized the current tumor-associated antigens (TAAs) for CAR T-cell therapy in solid tumors, and highlighted the tumor-specific antigens (TSAs) or neoantigens and the TAA binding domain in CARs to overcome the off-tumor toxicity challenge. Furthermore, we illustrated the dominant challenges caused by the solid tumor microenvironment, including the immunosuppressive microenvironment, T-cell infiltration, antigen heterogeneity, and chronic stimulation. Combination with targeting tumor microenvironment antigens (TMAs) was proposed as a new strategy.

Evolution of CAR design

Chimeric antigen receptors (CARs) are engineered proteins designed to enhance the tumor-targeting ability of immune cells. CARs have been used to generate various types of tumor-specific immune cells, such as T cells, NK cells, and macrophages. These immune cells, which are genetically modified through CAR transduction, are named CAR-T, CAR-NK, and CAR-M, respectively.

CAR is generally composed of an antigen-binding domain, a hinge region, a transmembrane region, one or more costimulatory domains, and an activation domain [11]. The extracellular antigen-binding (or recognition) domain consists of a single-chain variable fragment (scFv), which may also include peptides or other proteins that can recognize specific tumor antigens. The hinge region and the transmembrane region usually consist of either CD28 or IgG4. These proteins connect the intracellular and extracellular domains of the CAR. The costimulatory domains CD28 and 4-1BB (also known as CD137) are designed to increase T-cell proliferation, cytokine secretion, and the in vivo antitumor activity of CAR-T cells. The activation domain of CAR is made up of the CD3ζ domain, which transduces extracellular signals and activates T cells, leading to tumor lethality with cytolysis and cytokine release [20].

To date, the CAR T-cell technique has been developed for the fourth generation (Fig. 1). The first-generation CARs are formed from an scFv-based antigen-binding domain linked to a CD3ζ subunit of the T-cell receptor (TCR). Due to the lack of costimulatory signals, first-generation CARs are neither effective in amplification nor robust in antitumor activity, which limits their laboratory and clinical applications [21]. Compared with the first-generation CARs, the second-generation CARs include the whole structure of the first-generation CARs with the addition of costimulatory domains, such as CD28, 4-1BB, inducible T-cell costimulatory factor (ICOS), and DNA X-activating protein (DAP10) [21]. These modifications make CARs more favorable for inducing cytokine production and T-cell expansion, and some of these modifications also promote differentiation [22]. Third-generation CARs fuse CD137 or CD134 with CD28 simultaneously, which improves T-cell expansion and survival, promotes cytokine release, and enhances tumor killing [23]. Recently, the concept of fourth-generation CARs has been proposed. Fourth-generation CAR-T cells are further engineered to secrete universal cytokines, including IFN-γ, TNF-α, IL-2, and IL-8. The secretion of universal cytokines may enhance tumor-killing and immune cell recruitment effects [16, 21, 24,25,26,27].

Fig. 1
figure 1

Four generations of CAR T cells. ​An overview of the four generations of CARs displayed on the surface of a T-cell while contacting their antigens on a tumor cell. scFvs that act as ligand-binding domains in CARs mediating tumor cell recognition are shown in red, with VH and VL domains connected to intracellular signaling domains via a hinge and a transmembrane domain

TAAs for CAR T-cell therapy in solid tumors

TAAs, which are highly expressed in tumor tissue and low or absent in normal tissue, are prerequisites for the application of CAR T cells to the treatment of solid tumors. They are selected based on pathology tests and immunohistochemistry results. After much effort by scientists, some specific antigens against solid tumors have been found to be applied to CAR T cells for solid tumor treatment.

Mucin-1 (MUC1)

MUC1 is a tumor antigen closely related to tumorigenesis, invasion, and metastasis. A lentiviral vector containing the VH-based anti-MUC1 CAR gene is constructed by optimizing transient virus production followed by transfection into T cells to avoid immunogenic responses and signaling [28]. Transfected T cells were cocultured with MUC1-positive M47D or MCF-7 tumor cells or with MUC1-negative A431 tumor cells, and the cytotoxic activity of Th1 cytokines, including IL-2, TNF-α, and IFN-γ secreted by MUC1-recognizing CAR T cells, was significantly increased compared with that of untransduced T cells [28]. Heng Zhang et al. constructed enhanced CAR T cells, which not only targeted MUC1 but also activated JAK-STAT signaling induced by IL-12 and enhanced the proliferation of CAR T cells. The enhanced MUC1-CAR T cells possessed durable tumor-killing and proliferative capacity [29].

B7H3 (CD276)

B7H3 is an immune checkpoint molecule expressed on the cell surface that plays an immune-inhibitory role in downregulating the biological activity of T cells and the cytotoxic activity of natural killer cells [30]. Its low expression in normal cells and abnormally high expression in a variety of malignancies suggest that B7H3 can be used as a novel target for CAR T-cell therapy [31]. Du H et al. found that B7H3 CAR T cells significantly inhibited pancreatic ductal cancer cells in vitro, while mice achieved 100% survival and showed no side effects [32]. B7H3 CAR T-cell injection (TAA06 injection), which was developed by PersonGen Co., Ltd., was approved for testing in clinical trials by the Center for Drug Evaluation (CDE) of the State Drug Administration for relapsed/refractory neuroblastoma on July 4, 2022.

Human epidermal growth factor receptor 2 (HER2)

HER2 is a transmembrane glycoprotein that mediates cell proliferation and differentiation in both developing embryonic and adult tissues. HER2 can inhibit apoptosis, induce neovascularization, and enhance cell motility, thus promoting the rapid growth, proliferation, and metastasis of tumors [33, 34]. Sun et al. constructed humanized HER2-specific CAR T cells, which could react against HER2 + breast and ovarian cancer cells [35]. A phase I clinical trial (NCT01935843) demonstrated the safety and feasibility of HER2 CAR T cells in treating HER2-positive advanced biliary tract cancers and pancreatic cancers [36]. Moreover, HER2-targeted/4-1BB-costimulated CAR T cells could specifically recognize and kill synovial sarcoma cells by secreting IFN-γ and TNF-α [37]. Furthermore, HER2-specific CAR-T cells represent an effective immunotherapy for preventing CRC progression [38].

Epidermal growth factor receptor (EGFR)

EGFR is expressed in normal cells but overexpressed in tumor cells [39]. Lin et al. showed that third-generation EGFR CAR T cells effectively inhibited triple-negative breast cancer with limited cytotoxicity to normal breast epithelial cells or estrogen receptor-positive breast cancer cells in vitro, and they performed in vivo experiments. In addition, EGFR CAR T cells activated the interferon γ, granzyme-perforin-PARP, and Fas-FADD-caspase signaling pathways in triple-negative breast cancer [40]. Liu et al. treated 16 patients with anti-EGFR CAR T cells, and the median overall survival of all 14 evaluable patients was 4.9 months, suggesting that anti-EGFR CAR T cells were safe and effective for the treatment of metastatic pancreatic cancer [41].

Carcinoembryonic antigen (CEA)

CEA, a tumor-associated glycoprotein with a relative molecular mass of 180,000, is a tumor antigen produced continuously by progressive gastrointestinal malignancies. Investigators applied CAR T cells at five dose levels (1 × 105 to 1 × 108 CAR/kg cells) to 10 patients with colorectal cancer to demonstrate the safety and efficacy of treatment with CEA CAR T cells. Seven of these patients with progressive disease during previous treatments were stable after CAR T-cell therapy, and two were stable for over 30 weeks, showing tumor reduction by positron emission tomography (PET)/computed tomography (CT) and MRI analysis. The CAR T cells were observed to proliferate, especially after the second treatment [42]. Chi et al. designed CEA-specific CAR T cells in combination with rhIL-12 and evaluated their efficacy for treating different types of solid tumors. The in vivo results confirmed that the combination of CEA CAR T cells with rhIL-12 significantly enhanced their antitumor activity in terms of growth inhibition of the newly colonized colorectal cancer cell line HT-29, the pancreatic cancer cell line AsPC-1, and the gastric cancer cell line MGC803 compared with CEA CAR T-cell therapy alone. The combination of CEA CAR T cells and rhIL-12 overcame their separate defects to provide a new strategy for the treatment of solid cancer [43].

Mesothelin (MSLN)

MSLN is a cell surface antigen involved in tumor invasion and is expressed at low levels in mesothelial tissues (pleural, pericardial, and peritoneal mesothelial cells) but is highly expressed in mesothelioma, pancreatic cancer, ovarian adenocarcinoma (OVCA), lung cancer, breast cancer, and other cancers, so it can be used as a target for CAR T cells in treating solid tumors [44,45,46]. Haas et al. constructed a second-generation CAR targeting MSLN in transiently expressing autologous T cells and tested them in a phase I study of toxicity. A patient with malignant pleural mesothelioma developed anaphylaxis during the second infusion of CAR T cells, presumably due to immune rejection of the murine-derived scFv in the CAR. This approach was safe overall but had limited therapeutic efficacy for other patients. Given the lack of targeted toxicity by RNA CAR T cells, the team conducted a phase I trial using lentiviral vector transduction. Overall, all patients with CAR T-meso cells had successful amplification but with a limited duration, and no targeted toxicity was observed, including pleurisy, peritonitis, or pericarditis [47]. MSLN CAR T-cell therapy is feasible, and researchers have found good antitumor activity in preclinical studies, but clinical data to date indicate that mesothelin CAR T cells are not effective in patients or do not proliferate consistently. Thus, new solutions should be sought.

GD2

GD2, a disialoganglioside and acidic glycolipid found on the outer cell membrane, is a part of the immunological identity of mammalian cells and is generally nonimmunogenic. GD2 is expressed by a variety of embryonal cancers, including brain tumors, but it is barely expressed in normal cells [48], making it a target for CAR T therapy. Malvina et al. established a 3D culture glioblastoma model and reported that the intracerebral administration route significantly increased the survival rate in a dose-dependent manner without any side effects [49]. Researchers from Britain reported a phase 1 study (NCT02761915) in which 12 children with relapsed/refractory neuroblastoma were treated with escalating doses of second-generation GD2-directed CAR T cells. Their study showed that CAR T cells achieved rapid clearance of cancer. Although the beneficial effect lasted for only a short time, it provided important evidence that this particular CAR T-cell therapy could serve as a future treatment for solid cancers in children [50]. Recently, Majzner et al. carried out a clinical trial (NCT04196413) of autologous GD2 CAR T cells for the treatment of H3K27 M-mutated diffuse midline gliomas (DMGs) in children and young adults, identifying the maximally tolerated dose or recommended phase II dose. The tumor progressed in one of the four enrolled patients, and the patient died due to disease progression after three months. The other three patients derived radiographic and clinical benefits after i.v. administration of GD2 CAR T cells, although they developed CRS and tumor inflammation-associated neurotoxic reactions. This study provided valuable experience for further optimizing GD2 CAR T-cell therapy for spinal DMG [51].

Natural killer group 2D (NKG2D)

NKG2D-based CAR T cells have received much attention in the past few years because of their ability to recognize a panel of eight stress-associated ligands in the MIC and ULBP families, which are hardly expressed on normal tissues but are upregulated in malignant transformation, viral infection, and DNA damage. Tay et al. manufactured NKG2D CAR T cells with piggyBac transposon vectors and K562 artificial antigen-presenting cells instead of viral vectors. The results showed that small volumes of peripheral blood samples could produce many NKG2D CAR T cells, and they could be expanded to meet a clinical timeline designed for solid cancer therapy [52]. YanZhang designed an NKG2D CAR T-cell-fused NKG2D extracellular domain and 4-1BB and CD3z, which showed a strong antitumor effect against NKG2DL-positive cervical cancer cell lines in vitro as well as xenograft mice in vivo without toxicity [53]. Yu-Yang Ng constructed NKG2D CAR T cells with a DAP12 signaling domain that stimulated lower cytokine production to mitigate the risk of CRS compared with the CD3ζ activation domain but displayed similar antitumor activity, which completely eradicated established solid tumor xenografts in an NSG mouse model [54].

Epithelial cell adhesion molecule (EpCAM)

​EpCAM is a type I transmembrane glycoprotein that is frequently overexpressed in carcinomas, including colorectal, gastric, pancreatic, and endometrial cancers, in a heterogeneous manner [55, 56]. It was first found in colon cancer tissue and is a target of the Wnt/β-catenin pathway, the activation of which frequently contributes to poor infiltration of T cells across most human cancers [57]. Juan Fu found that the EpCAM target is highly upregulated in actual ovarian cancer samples and sought to construct 3rd -generation EpCAM CAR T cells and then verified their antitumor activities in vitro and in vivo. Their results showed that EpCAM CAR T cells could become a clinical therapeutic strategy against ovarian cancer [56]. There are an increasing number of clinical trials registered utilizing EpCAM CAR T cells (NCT04151186, NCT02915445, NCT03013712).

Numerous types of TAAs for CAR T targets are tested in clinical trials, not only those referenced above that have been tested in at least three trials thus far (MUC1, B7H3, HER2, EGFR, CEA, MSLN, GD2, NKG2D, and EpCAM) but also Nectin4, FAP, TM4SF1, LOH, CD22/TILs, claudin 18.2, ROR2, CLDN6, Lewis Y (LeY), glypican 3, PSCA, ROR1, CD7, IM96, IM92, IL13α2, GPC3, Mov19-BBz, CT041, PSMA, GUCY2C, and others. To date, 125 solid tumor CAR T clinical studies have been registered at ClinicalTrials.gov worldwide as of September 07, 2022 (Table 1). Moreover, hundreds of potential targets for CAR T-cell therapy are being applied to solid tumors in preclinical trials.

Table 1 The clinical trials of CAR T cell target to solid tumorsa

Tumor neoantigens for CAR T-cell therapy in solid tumors

Tumor neoantigens, which are tumor-specific antigens derived from somatic mutations that are expressed only on tumor cells and are more immunogenic and have less off-tumor toxicity than TAAs [58], may represent a new approach for CAR T-cell therapy.

Tumor neoantigens have been employed in the development of tumor vaccines for solid tumor treatment due to their specific immunogenicity. To date, hundreds of peptide vaccines for solid tumors based on tumor neoantigens have reached clinical validation trials. A typical example is the personalized neoantigen peptide vaccine for the treatment of EGFR-mutated non-small cell lung cancer, which has reached phase I clinical trials. Additionally, there are immune cells that are expanded in vitro targeting tumor neoantigens for solid tumor treatment, such as neoantigen-induced autologous DC-CIK cells (NCT05020119) and tumor-infiltrating lymphocytes (TILs) for terminal epithelial tumors (NCT05141474).

At present, CAR T cells targeting tumor neoantigens for solid tumor treatment have been developed. A case in point is that anti-EGFRvIII-CAR T cells can efficiently kill EGFRvIII cancer cells, which are high-grade glioblastoma cells with specific TSAs [59]. You Li’s team optimized an EGFRvIII-specific CAR construct with the TGFRII ectodomain as a TGFβ-resistant CAR-T for glioblastoma therapy, and the results showed that the architecture enhanced the antitumor efficacy of EGFRvIII-specific CAR-T cells and prolonged the survival of glioblastoma-bearing mice [60]. Bryan et al. developed bispecific CAR structures specifically targeting the glioblastoma tumor antigen EGFRvIII and bispecific T-cell engager (BiTE) against EGFR. BiTE cells secreted EGFR-specific BiTEs and recruited untransformed bystander T cells against wild-type EGFR. These CAR T BiTE cells eliminated heterogeneous tumors in a glioblastoma mouse model [61]. Several CAR T cells targeting EGFRvIII for recurrent glioblastoma treatment have reached clinical trials (NCT02844062 and NCT01454596).

Challenges of using TAAs for CAR-T-cell therapy in solid tumors

On target, off-tumor toxicity

Although many TAAs have been selected for CAR-T-cell therapy, and some of them have shown gratifying outcomes, off-tumor toxicity remains a major challenge because TAAs may also be expressed in normal tissues. For example, TAAs such as HER2 and EGFR are overexpressed in cancer cells, but they are also expressed at low density in normal epithelial tissues. Therefore, some experiments have shown that anti-HER2 CAR T cells are toxic to normal organs. This off-tumor target toxic effect is also observed in other CARs targeting the overexpression of TAAs, and severe off-tumor target effects may result in patient death [62]. Therefore, the design of CAR-T cells needs to be able to sense the density of antigens to distinguish between cancer cells and normal cells.

Lack of public tumor neoantigens

To date, although TSAs and tumor neoantigens are the ideal antigens for developing efficient CAR T cells, only a few TSAs in their true sense have been reported, such as BRAF, KRAS, TP53, HRAS/KRAS/NRAS, and BRAF [63]. These reported public TSAs can be used to generate TCR, CAR T cells, and dual-specific antibodies. Nevertheless, the downstream signals are different due to the differences in the formation of immune synapses, affinity, and structural dynamics of TCR, CAR T cells, and bispecific antibodies. ​In addition, these antigens are low in content, typically less than 100 copies per cell, whereas CAR T typically requires hundreds to thousands of copies for effective identification [63]. Therefore, it is urgent to establish prediction and identification methods for TSAs. Neoantigens can be discovered by genomic techniques such as next-generation sequencing and high-throughput single-cell sequencing, MHC-loaded peptide techniques such as mass spectrometry, and bioinformatics tools based on high-throughput sequencing data, mass spectrometry data, and biological databases [64,65,66]. The abovementioned techniques are essential for the discovery of TSAs, which may boost the development of CAR T-cell therapy for solid tumor treatment.

Tumor-antigen heterogeneity

The heterogeneity of TSAs is another key factor that leads to immunologic escape and tumor recurrence [67]. Approximately 30% of glioblastomas express mutant EGFRv III. A clinical trial demonstrated that the treatment of gliomas using anti-EGFRvIII CAR T-cell therapy leads to the growth of EGFRvIII-negative tumor cells [67]. Tumor-antigen heterogeneity is a challenge to overcome that results in unsatisfactory CAR T-cell therapy of solid tumors. To promote the effectiveness of CAR T-cell therapy, new strategies for tumor recognition are needed to cope with the challenges of tumor specificity and heterogeneity.

Strategies for overcoming the challenges of TAAs

Dual CAR and tandem CAR

The antigen of CARs requires specificity to recognize tumor cells, avoiding off-target effects. Additionally, to eliminate the possibility of tumor recurrence resulting from immunological escape, the antigen also needs to be universal to overcome the heterogeneity of tumor cells. Combining two or more antigens as targets and constructing multitarget CARs are potential approaches to solve the contradiction between specificity and heterogeneity.

Bispecific CARs have become an effective way to avoid tumor relapse due to antigen escape. They contain two different CARs coexpressed on one T cell, called dual CARs. Dual-targeting CAR T cells can be precisely positioned at the tumor site, have high cytotoxicity to tumor cells, alleviate off-target effects, and achieve precise treatment [68]. For example, the tumor-specific antigen EGFRvIII is activated by the SynNotch receptor to induce local CAR expression, which is then coupled with EphA2 and IL13Rα2 to induce tumor cell recognition and killing. This approach may avoid the off-target killing of the TAAs EphA2 and IL13Rα2 in distant normal tissues [69]. Peng Li’s team developed a DAP10 chimeric receptor with native NKG2D on T cells to target NKG2D ligand-expressing cancer cells. They tandemly incorporated it with anti-glypican 3 (GPC3) scFv to construct a dual-antigen-targeting system. This novel dual-targeting system enabled high efficacy in killing heterogeneous cancer tumors [70]. Bob S. Carter et al. designed EGFRvIII and BiTE bicistronic construct CAR T cells against EGFR to solve heterogeneous target antigen expression. Tumors lacking the antigen targeted by CAR T cells directed against single antigens grew outward [61]. Therefore, it is essential to recognize multiple antigens in tumor cells with a single CAR T-cell or to coexpress various CAR proteins and to achieve programmable CAR expression regulation with individual T cells.

Two CAR T-cell lines, each targeting a different antigen, were combined and named the CAR pool. T cells expressing a single-chain bispecific CAR are known as tandem CAR. HER2/MUC1 bispecific CAR for breast cancer cell therapy in vitro [71], an EGFR/EpCAM/HER2 triple-specific CAR against Raji lymphoma cells engineered to express TAAs [72], and a HER2/IL-13Ra2 bispecific CAR for the treatment of a glioma xenograft in vivo have shown promising results [73]. Dual GD2 and B7H3 CAR T cells provided optimal costimulation and T-cell metabolic fitness by independently activating the CD28 and 4-1BB pathways and fine-tuning CD3ζ chain-mediated signaling in a neuroblastoma model.

There are also studies of CEA plus MSLN dual-target CAR T cells expressing CEA-CD3ζ and the MSLN-4/1BB signaling domain. These dual-targeted CAR T cells became dCAR T cells, and dCAR T cells were significantly cytotoxic to two antigen-positive tumor cells but not to single antigen-positive tumor cells both in vivo and in vitro. The dual CAR T cells showed robust and sustained antitumor activity in vivo under stress conditions and prevented tumor escape due to heterogeneous antigen expression by tumor cells [74]. Sandra et al. constructed a dual-specific antibody, BiTE, which could target both EGFR of tumor cells and CD3 of T cells, and infused this BiTE into T cells, resulting in no attacks on normal tissues while locally killing tumor cells, indicating that this BiTE CAR T could well attenuate off-target effects [75].

Improved recognition ability of the TAA binding domain in CARs

Another key factor for the antitumor effect of CAR T cells is the recognition ability of the TAA binding domain in CARs. The interaction between the CAR and its binding target determines the tumor-killing effect and proliferation capacity of the constructed CAR-T cells and, to some extent, the persistence of the response [76]. ​ The TAA binding domain can be adjusted to enhance the scFv affinity, scFv steric hindrance, and stability of the TAAs or nanoantibodies can be used to enhance the ability of the TAA binding domain to better recognize and bind the TAAs (Fig. 2).

Fig. 2
figure 2

TAAs and TAA binding domain design strategy for developing multispecific CARs for solid tumors. TAAs and TSAs were selected as CAR T-cell targets. To overcome the challenges of tumor antigen off-target effects and heterogeneity, dual CARs or tandem CARs have been constructed to target dual TAAs. scFv affinity, steric hindrance, and stability were considered for the construction of CARs, and VHH nanobodies were applied as TAA binding domains to develop multispecific CARs for solid tumors

​Because of their small size, high affinity and antigen-specific recognition, scFvs are widely applicable to CAR-T therapy and bispecific antibody development. Modulating the affinity of scFv may improve the selective identification of target cells with higher ligand densities, thereby reducing off-target effects. Because TAAs are expressed not only in tumors but also to some extent in normal cells, modulating the affinity of scFvs can help to increase CAR specificity and reduce off-target effects [77, 78].

​Alternatively, adjusting the steric hindrance of scFvs based on the structure and function of target epitopes on TAAs may improve the antitumor effect. Researchers have used the piggyBac transposon system to construct two types of CAR, CAR-T cells targeting region I (meso1, residues 296–390) or region III (meso3, residues 487–598) of MSLN, and meso3 CAR-T cells were found to produce more CD107α, IL-2, TNF-α, and IFN-γ relative to meso1 CAR-T cells upon activation and showed an enhanced killing effect [79]. Therefore, meso3 has a better-targeted epitope than meso1, which may be related to the specific characteristics of the proximal membrane region. When the proximal membrane region bridges the extracellular and transmembrane domains of the MSLN, it is likely to have a rigid structure or be responsible for a specific functional region, and when the scFv on CAR-T binds to this region, it triggers more effective antitumor activity.

Moreover, precise control of the expression of scFvs on CAR may improve the intrinsic stability of the binding domains of TAAs. If the amount of scFv expression is too small, the amount of binding to the TAA antigen epitope would be too small to effectively capture tumor cells and exert CAR-T-cell antitumor effects. In turn, too high expression of scFvs can lead to scFv aggregation and ultimately to early depletion of T cells [80].

Nanobodies applied as the TAA binding domain of CAR-T cells

​Nanobodies, also known as VHH antibodies, are derived from the variable domain of a heavy chain antibody and can bind to antigen recognition sites. The nanobodies consist of a long CD3 sequence that accepts an adjusted flexible and extended conformation, capable of reaching certain epitopes that are inaccessible to conventional antibodies. In contrast to scFvs, VHH antibodies avoid potential disruption of interactions between variable (VH, VL) and constant (CH1, CL) regions and avoid exposure to hydrophobic regions, both of which can affect antibody solubility and stability [81]. Alternatively, nanotubes that act as TAA-binding domains for CARs do not produce surface aggregates and do not have target antigen-independent effects or cell activation limitations. Nanobodies with optimal stability and high affinity applied as the antigen binding domain of CAR-T cells may improve the specific binding of TAAs and CARs.

​As the extracellular binding domain of scFvs, it is difficult to balance optimal affinity, obtaining optimal steric hindrance, and optimal epitope and stability for targeting TAAs. The ability to generate functional multispecific CARs using nanobodies, D-domains, and other novel molecules with beneficial antitumor effects offers the opportunity to develop multispecific CARs that effectively address tumor recurrence due to antigen loss and provide a therapeutic approach to the intrinsic phenotypic diversity of tumors.

Challenges originating from the tumor microenvironment

The tumor microenvironment is a complex and evolving entity that contains immune cells, stromal cells, blood vessels, cytokines, and the extracellular matrix, and its features continuously promote tumor progression [82]. ​Early in tumor initiation, the tumor microenvironment promotes early cancer cell growth, supports tumor survival, invasion, and spread, and promotes angiogenesis to overcome hypoxic and acidic environments [83]. The immunosuppressive tumor microenvironment will cause CAR T cells to infiltrate into solid tumors inefficiently, resulting in inadequate activation and exhaustion of CAR T cells.

Tumor immunosuppressive microenvironment

​ Suppressive immune cells, such as regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), in the tumor microenvironment can secrete large amounts of PEG2, TGF-β, IL-10, nitric acid, and indoleamine 2, 3-dioxygenase (IDO) to inhibit T-cell proliferation. Immunosuppressive ligands, such as programmed cell death 1 ligand 1 (PD-L1), might induce intrinsic antitumor immune responses as well as CAR T-cell responses (Fig. 3) [84].

Fig. 3
figure 3

The immunosuppressive tumor microenvironment hinders CAR T-cell therapy for solid tumors. The solid tumor microenvironment is a complex and evolving entity containing immune-suppressing cells such as Tregs, MDSCs, TAMs and TANs; there are also CAFs, endothelial cells and extracellular matrix. The immunosuppressive ligands PD-L1, ARG-1, ARG-2, CTLA-4, and IDO secreted by those cells in the TME may all quell the intrinsic antitumor immune response, as well as the CAR T-cell response that helps tumor cells evade immune cell attack. Targeting those TMAs not only leads to a direct attack on the tumor cells but also modulates the tumor microenvironment, rendering it immunocompetent and tumor-hostile. Tregs regulatory T cells; TAM tumor-associated macrophages; TAN tumor-associated neutrophils; Anti-Tregs anti-regulatory T cells; MDSC myeloid-derived suppressor cells; TEff effector T cells; CAF cancer-associated fibroblasts. PD-L1 programmed cell death 1 ligand 1; IDO indoleamine 2,3-dioxygenase; ARG-1 arginase 1; ARG2 arginase 2; CTLA-4 cytotoxic T-lymphocyte-associated protein 4

Immune checkpoint inhibitors such as CTL4 and PD1 combined with CAR T cells have been used to target immune cells and weaken the role of immune cells in the immune microenvironment [85]. The T-cell immunoglobulin mucin 3 (Tim3) is an immune checkpoint receptor that plays a dominant role in T-cell depletion in the tumor microenvironment. The Hadjati team knocked down Tim3 in MSLN CAR T cells, and the knockout CAR T-cell lines had improved cytotoxic function, enhanced cytokine production and proliferation, and altered the immune microenvironment in response to the mitigated Tim3 signaling pathway [86].

Inefficient infiltration of CAR T cells in solid tumors

CAR-T cells have more opportunity to interact with blood tumor cells, whereas it is difficult for CAR-T cells to penetrate tumor tissues through the blood system in solid tumors. Furthermore, the lack of chemokine expression involved in T-cell infiltration of tumor tissues and the presence of dense fibrotic stroma in solid tumors result in a reduced ability of CAR-T cells to migrate and invade tumor cells [87]. Treatment with PARP inhibitors promoted CAR T-cell infiltration by stimulating a chemokine milieu and modulated the tumor microenvironment by activating the cGAS-STING pathway [88]. We can also eliminate poor tumor infiltration by decoding the complexities of IFN/PRR signaling and then engineering CAR T cells to deploy RN7SL1 to recode these signals in the tumor microenvironment. RN7SL1 CAR T cells exhibit good tumor infiltration, myeloid cell and DC production activation, a tumor microenvironment with antigen provision, and endogenous T cells to reject solid tumors under the threat of CAR antigen loss [89]. Furthermore, they can be reinfused within the tumor to improve CAR T-cell infiltration and the transshipment problem in some types of solid tumors.

Chronic antigen stimulation leads to CAR T-cell exhaustion

Limited persistence remains a significant hindrance to the advancement of effective CAR T-cell therapy. This cellular state is characterized by the overexpression of PD-L1 and other inhibitory factors. T-cell exhaustion, which can lead to treatment failure and cancer recurrence, is likely to play a major role in CAR T-cell therapy. Not only tonic signaling but also chronic and persistent antigen stimulation can lead to variable levels of T-cell exhaustion. Compared to hematological malignancies, solid tumor macroenvironments challenge CAR T cells with hypoxic, acidic, and nutrient-poor conditions that alter metabolism and accelerate exhaustion [90].

Perturbation of the INO80 and BAF chromatin remodeling complexes improves T-cell persistence in tumors. Depletion of Arid1a, a BAF complex member, resulted in the maintenance of an effector program and downregulation of exhaustion-related genes in tumor-infiltrating T cells [91]. Another study showed that RASA2 and SynNotch CAR T cells can overcome the challenges of specificity, heterogeneity, and persistence [69, 92].

The target of TMAs for CAR T-cell therapies in solid tumors

TMAs were first described by Andersen MH [93]. They include IDO, PD-L1, tryptophan 2,3-dioxygenase (TDO), arginase 1 and 2, CCL22, TGF β, and FoxP3, which are expressed by heterogeneous cell types in the tumor microenvironment, such as MDSCs, tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), tumor-associated dendritic cells (DCs), Treg cells and cancer-associated fibroblasts (CAFs), that help tumor cells escape immune cell attack (Fig. 3) [93].

As therapeutic targets, TMAs offer several advantages that differentiate them from more traditional tumor antigens. ​Combined targeted TMAs for antitumor immunotherapy can directly attack tumor cells, modulate the tumor microenvironment, establish a microenvironment that is not conducive to tumor growth, modulate immune cells in the microenvironment, restore their immune activity and tumor-killing effect, and inhibit tumor angiogenesis. Although the concept of TMAs has not yet been proposed in CAR T-cell therapy, the use of TMAs as binding domains for CARs has been applied in the study of CAR T-cell therapy. Tong Li constructed PD1-anti-MUC16 dual-CAR T cells that released many cytokines, such as IL-2, IFN-γ, and TNF-α, which demonstrated enhanced killing capacity of OVCAR-3 cells compared to single CAR T cells in vivo and survived significantly longer in vivo [94]. Wenzhen Li constructed a second-generation CAR targeting EpCAM plus hsBCL9CT-24, a peptide that inhibits the β-catenin/BCL9 interaction and exhibits potent antitumor effects. The results indicated that targeting EpCAM and Wnt/β-catenin can inhibit tumor growth and enhance tumor infiltration without affecting T-cell survival, promoting the efficacy of EpCAM CAR T-cell therapy [57]. Researchers have generated bispecific CAR T cells targeting EpCAM and ICAM-1 to increase resistance to antigen escape. ICAM-1 can induce the secretion of proinflammatory cytokines upon CAR interaction with the primary antigen EpCAM; thus, tumor cells are more vulnerable to dual CAR T cells [55].

A recent study showed that third-generation HER2 CAR T cells could efficiently eliminate glioblastoma cells in vitro and promote CAR T antitumor activity when combined with PD-1 blockade [94]. Shaw AR increased the breadth, potency, and duration of anti-PDAC HER2-specific CAR T-cell activity with oncolytic adeno-immunotherapy, which produced cytokines, achieved immune checkpoint blockade, and had a safety switch (CAdTrio). The combined use of CAdTrio and HER2 cured tumors in two PDAC xenograft models and produced a lasting tumor response in humanized mice, making it a potential treatment regimen for PDAC patients beyond surgical treatment [95]. Li et al. constructed HER2 CAR T cells secreting IL-2 and IFN, which bound to anti-PD1 antibodies, impaired the enhanced growth of HCC1954 tumors and overcame the resistance of trastuzumab [96]. He et al. constructed a hypoxia-inducible transcription amplification system (HiTA system) to control CAR expression in HiTA-CAR T cells. This HiTA system showed a significant improvement in hypoxia-restricted transgene expression over existing systems and showed significant antitumor activity in the absence of significant hepatic or systemic toxicity observed in vivo. This approach can also be applied to the design of CAR T cells directed against other tumor antigens [97]. Deepak et al. developed “switch” CAR systems, the activity of which could be controlled in vivo and which bound specific peptides; the specific peptides bound to Fab molecules, and the Fab bound to tumors. The “switch” acts as a bridge between the target and effector cells and controls antigen specificity, activation, and eventual tumor clearance. These switches have the advantage of a short half-life of the Fab fragment and limited immunogenicity of the peptide tag. This switchable CAR T is effective in preclinical models of leukemia and breast cancer cells and may allow the safe use of HER2 in clinical settings [98].

Local intrahepatic perfusion optimized the delivery of CAR T cells to liver metastases, but Burga et al. expected liver CD11b + Gr-1 + myeloid-derived suppressive cells (L-MDSCs) to inhibit CAR T-cell delivery to the liver. ​Experimental validation demonstrated the recovery of CAR T-cell efficacy when mice were treated with CEA CAR T cells in combination with L-MDSC depletion and GM-CSF neutralization by inhibition of L-MDSC expansion or PD-L1 blockade. In the future, inhibiting L-MDSC amplification after CEA CAR T-cell treatment will be a new research direction [99].

​At the same time, the challenges of insufficient T-cell penetration and weak effector function in the application of TMAs in CAR T-cell therapy should also be addressed. Therefore, TMAs should be combined with other immunotherapies to achieve better clinical efficacy, such as degradation of the tumor extracellular matrix using matrix metalloproteinase, which can help CAR T cells infiltrate tumors [100].

Conclusion

Humans have been fighting cancer for thousands of years, but the incidence of cancer continues to increase, and we are constantly searching for new cancer treatments. CAR T cells have occupied a leading position since their inception, from their application against hematological malignancies to many attempts to treat solid tumors. Scientists have identified several TAAs of solid tumors, some of which have been tested in phase I and II clinical trials. CAR T-cell treatment has encountered many problems in the treatment of solid tumors, and scientists have also worked out various solutions. In future studies, on the one hand, it is vital to increase multitarget CAR T-cell development to enhance their tumor-killing specificity and reduce off-target effects so that CAR T cells can simultaneously recognize the presence of two or more TAAs on tumors [61, 68, 75, 81, 101,102,103]. Dual CAR and tandem CAR can also be designed to enhance signaling by the 4-1BB and CD28 domains, optimized by the membrane-embedded proximal positioning of both signaling domains within separate, parallel CARs [104]. On the other hand, we developed a new strategy that combined targeting TMAs to create a tumor-hostile microenvironment and improve CAR T-cell infiltration into solid tumors. Third, it is necessary to construct a “suicide gene” system to destroy infused CAR T cells to regulate their proliferation and restrict their toxicity [72, 105, 106] or to construct a “device switch” to regulate the timing of T-cell activation to prevent emerging CRS and CERS and to moderate the apoptosis of CAR T cells [95, 98, 107,108,109,110].

Moreover, universal CAR T cells could be constructed for specific tumors by targeting highly expressed specific targets, which will shorten the waiting time, reduce the cost, broaden the cell source, strengthen the operability and quality control for patients, and make it easier to achieve widespread use. Nevertheless, universal CAR T cells should be given special attention to avoid GVHD compared with the side effects of autologous CAR T cells [111, 112]. The functions of solid tumor CAR T cells are different from those of hematological malignancy CAR T cells, implying that enhancing the binding interactions between CAR T cells and tumor cells may increase their responses in solid tumors [113]. New research has indicated that artificial thymic organoids allow the selective differentiation of CRT-transduced human iPSCs into CAR T cells and can have effective results in an animal model. This breakthrough may change the way T cells are obtained; it requires the patient’s blood to select CAR T cells [114, 115]. Researchers all over the world are trying to enhance the effectiveness and safety of CAR T-cell therapy, and we believe that it will be a big step toward curing cancer.

Availability of data and materials

Not applicable.

Abbreviations

CAR T:

Chimeric antigen receptor T

TAAs:

Tumor-associated antigens

TMAs:

Tumor microenvironment antigens

TSA:

Tumor-specific antigens

TCR:

T-cell receptor

scFv:

Single chain variable fragment

ICOS:

Inducible T-cell costimulatory factor

DAP10:

DNA X-activating protein

MUC1:

Mucin-1

CDE:

Center for Drug Evaluation

HER2:

Human epidermal growth factor receptor 2

EGFR:

Epidermal growth factor receptor

BiTE:

Bispecific T-cell engager

CEA:

Carcinoembryonic antigen

PET:

Positron emission tomography

CT:

Computed tomography

MSLN:

Mesothelin

OVCA:

Ovarian adenocarcinoma

NKG2D:

Natural killer group 2D

EpCAM:

Epithelial cell adhesion molecule

Tim3:

T-cell immunoglobulin mucin 3

GPC3:

Glypican 3

CRS:

Cytokine release syndrome

GM-CSF:

Granulocytic macrophage colony-stimulating factor

CRES:

CAR T-related encephalopathy syndrome

UCAR T:

Universal CAR T

HVGR:

Host versus graft reaction

GVHD:

graft-versus-host disease

ZFNs:

Zinc-finger nucleases

TALENs:

Transcription activator-like nucleases.

References

  1. Spiegel JY, Patel S, Muffly L, Hossain NM, Oak J, Baird JH, et al. CAR T cells with dual targeting of CD19 and CD22 in adult patients with recurrent or refractory B cell malignancies: a phase 1 trial. Nat Med. 2021;27(8):1419–31.

    Article  CAS  Google Scholar 

  2. Liu R, Cheng Q, Kang L, Wang E, Li Y, Zhang J, et al. CD19 or CD20 CAR T cell therapy demonstrates durable antitumor efficacy in patients with central nervous system lymphoma. Hum Gene Ther. 2022;33(5–6):318–29.

    Article  CAS  Google Scholar 

  3. Wu Y, Chen D, Lu Y, Dong S-C, Ma R, Tang W-y, et al. A new immunotherapy strategy targeted CD30 in peripheral T-cell lymphomas: CAR-modified T-cell therapy based on CD30 mAb. Cancer Gene Ther. 2022;29(2):167–77.

    Article  CAS  Google Scholar 

  4. Tambaro FP, Singh H, Jones E, Rytting M, Mahadeo KM, Thompson P, et al. Autologous CD33-CAR-T cells for treatment of relapsed/refractory acute myelogenous leukemia. Leukemia. 2021;35(11):3282–6.

    Article  CAS  Google Scholar 

  5. Qin H, Yang L, Chukinas JA, Shah NN, Tarun S, Pouzolles M, et al. Systematic preclinical evaluation of CD33-directed chimeric antigen receptor T cell immunotherapy for acute myeloid leukemia defines optimized construct design. J Immunother Cancer. 2021;9(9):e003149.

    Article  Google Scholar 

  6. Mei H, Li C, Jiang H, Zhao X, Huang Z, Jin D, et al. A bispecific CAR-T cell therapy targeting BCMA and CD38 in relapsed or refractory multiple myeloma. J Hematol Oncol. 2021;14(1):161.

    Article  CAS  Google Scholar 

  7. Cui Q, Qian C, Xu N, Kang L, Dai H, Cui W, 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.

    Article  CAS  Google Scholar 

  8. Feng Y, Liu X, Li X, Zhou Y, Song Z, Zhang J, et al. Novel BCMA-OR-CD38 tandem-dual chimeric antigen receptor T cells robustly control multiple myeloma. OncoImmunology. 2021;10(1):1959102.

    Article  Google Scholar 

  9. Luanpitpong S, Poohadsuan J, Klaihmon P, Issaragrisil S. Selective cytotoxicity of single and dual Anti-CD19 and Anti-CD138 chimeric Antigen receptor-natural killer cells against hematologic malignancies. J Immunol Res. 2021;2021:1–16.

    Article  Google Scholar 

  10. Wermke M, Kraus S, Ehninger A, Bargou RC, Goebeler M-E, Middeke JM, et al. Proof of concept for a rapidly switchable universal CAR-T platform with UniCAR-T-CD123 in relapsed/refractory AML. Blood. 2021;137(22):3145–8.

    Article  CAS  Google Scholar 

  11. Kang CH, Kim Y, Lee HK, Lee SM, Jeong HG, Choi SU, et al. Identification of potent CD19 scFv for CAR T cells through scFv Screening with NK/T-Cell line. Int J Mol Sci. 2020;21(23):E9163.

    Article  Google Scholar 

  12. Han D, Xu Z, Zhuang Y, Ye Z, Qian Q. Current progress in CAR-T cell therapy for hematological malignancies. J Cancer. 2021;12(2):326–34.

    Article  CAS  Google Scholar 

  13. Tong C, Zhang Y, Liu Y, Ji X, Zhang W, Guo Y, et al. Optimized tandem CD19/CD20 CAR-engineered T cells in refractory/relapsed B-cell lymphoma. Blood. 2020;136(14):1632–44.

    Google Scholar 

  14. Wang N, Hu X, Cao W, Li C, Xiao Y, Cao Y, et al. Efficacy and safety of CAR19/22 T-cell cocktail therapy in patients with refractory/relapsed B-cell malignancies. Blood. 2020;135(1):17–27.

    Article  Google Scholar 

  15. Morita D, Nishio N, Saito S, Tanaka M, Kawashima N, Okuno Y, et al. Enhanced expression of Anti-CD19 chimeric antigen receptor in piggyBac transposon-engineered T cells. Mol Ther Methods Clin Dev. 2018;8:131–40.

    Article  CAS  Google Scholar 

  16. Alizadeh D, Wong RA, Yang X, Wang D, Pecoraro JR, Kuo C-F, et al. IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol Res. 2019;7(5):759–72.

    Article  CAS  Google Scholar 

  17. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet. 2015;385(9967):517–28.

    Article  CAS  Google Scholar 

  18. Melenhorst JJ, Chen GM, Wang M, Porter DL, Chen C, Collins MA, et al. Decade-long leukaemia remissions with persistence of CD4 + CAR T cells. Nature. 2022;602(7897):503–9.

    Article  CAS  Google Scholar 

  19. Guerra E, Di Pietro R, Basile M, Trerotola M, Alberti S. Cancer-Homing CAR-T cells and endogenous immune population dynamics. Int J Mol Sci. 2021;23(1):405.

    Article  Google Scholar 

  20. Gross G, Eshhar Z. Therapeutic potential of T cell chimeric Antigen receptors (CARs) in cancer treatment: counteracting off-tumor toxicities for safe CAR T cell therapy. Annu Rev Pharmacol Toxicol. 2016;56:59–83.

    Article  CAS  Google Scholar 

  21. Ajina A, Maher J. Strategies to address chimeric antigen receptor tonic signaling. Mol Cancer Ther. 2018;17(9):1795–815.

    Article  CAS  Google Scholar 

  22. Gardner RA, Finney O, Annesley C, Brakke H, Summers C, Leger K, et al. Intent-to-treat leukemia remission by CD19 CAR T cells of defined formulation and dose in children and young adults. Blood. 2017;129(25):3322–31.

    Article  CAS  Google Scholar 

  23. Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR, et al. Phase I hepatic immunotherapy for metastases study of intra-arterial chimeric Antigen receptor-modified T-cell therapy for CEA + liver metastases. Clin Cancer Res. 2015;21(14):3149–59.

    Article  CAS  Google Scholar 

  24. Tokarew N, Ogonek J, Endres S, von Bergwelt-Baildon M, Kobold S. Teaching an old dog new tricks: next-generation CAR T cells. Br J Cancer. 2019;120(1):26–37.

    Article  CAS  Google Scholar 

  25. Kagoya Y, Tanaka S, Guo T, Anczurowski M, Wang C-H, Saso K, et al. A novel chimeric antigen receptor containing a JAK-STAT signaling domain mediates superior antitumor effects. Nat Med. 2018;24(3):352–9.

    Article  CAS  Google Scholar 

  26. Klampatsa A, Dimou V, Albelda SM. Mesothelin-targeted CAR-T cell therapy for solid tumors. Expert Opin Biol Ther. 2021;21(4):473–86.

    Article  CAS  Google Scholar 

  27. Poorebrahim M, Melief J, Pico de Coaña Y, Wickström L, Cid-Arregui S, Kiessling A. Counteracting CAR T cell dysfunction. Oncogene. 2021;40(2):421–35.

    Article  CAS  Google Scholar 

  28. Rajabzadeh A, Rahbarizadeh F, Ahmadvand D, Kabir Salmani M, Hamidieh AA. A VHH-Based Anti-MUC1 chimeric antigen receptor for specific retargeting of human primary T cells to MUC1-positive cancer cells. Cell J. 2021;22(4):502–13.

    Google Scholar 

  29. Li T, Wang J. Therapeutic effect of dual CAR-T targeting PDL1 and MUC16 antigens on ovarian cancer cells in mice. BMC Cancer. 2020;20(1):678.

    Article  CAS  Google Scholar 

  30. Long C, Li G, Zhang C, Jiang T, Li Y, Duan X, et al. B7-H3 as a target for CAR-T cell therapy in skull base chordoma. Front Oncol. 2021;11:659662.

    Article  Google Scholar 

  31. Maggs L, Cattaneo G, Dal AE, Moghaddam AS, Ferrone S. CAR T cell-based immunotherapy for the treatment of Glioblastoma. Front Neurosci. 2021;15:662064.

    Article  Google Scholar 

  32. Du H, Hirabayashi K, Ahn S, Kren NP, Montgomery SA, Wang X, et al. Antitumor responses in the absence of toxicity in solid tumors by targeting B7-H3 via chimeric antigen receptor T cells. Cancer Cell. 2019;35(2):221-37e8.

    Article  CAS  Google Scholar 

  33. Budi HS, Ahmad FN, Achmad H, Ansari MJ, Mikhailova MV, Suksatan W, et al. Human epidermal growth factor receptor 2 (HER2)-specific chimeric antigen receptor (CAR) for tumor immunotherapy; recent progress. Stem Cell Res Ther. 2022;13(1):40.

    Article  CAS  Google Scholar 

  34. Mitra S, Sydow S, Magnusson L, Piccinelli P, Törnudd L, Øra I, et al. Amplification of ERBB2 (HER2) in embryonal rhabdomyosarcoma: a potential treatment target in rare cases? Genes Chromosome Cancer. 2022;61(1):5–9.

    Article  CAS  Google Scholar 

  35. Sun M, Shi H, Liu C, Liu J, Liu X, Sun Y. Construction and evaluation of a novel humanized HER2-specific chimeric receptor. Breast Cancer Res. 2014;16(3):R61.

    Article  Google Scholar 

  36. Feng K, Liu Y, Guo Y, Qiu J, Wu Z, Dai H, et al. Phase I study of chimeric antigen receptor modified T cells in treating HER2-positive advanced biliary tract cancers and pancreatic cancers. Protein Cell. 2018;9(10):838–47.

    Article  CAS  Google Scholar 

  37. Murayama Y, Kawashima H, Kubo N, Shin C, Kasahara Y, Imamura M, et al. Effectiveness of 4-1BB-costimulated HER2-targeted chimeric antigen receptor T cell therapy for synovial sarcoma. Transl Oncol. 2021;14(12):101227.

    Article  CAS  Google Scholar 

  38. Xu J, Meng Q, Sun H, Zhang X, Yun J, Li B, et al. HER2-specific chimeric antigen receptor-T cells for targeted therapy of metastatic colorectal cancer. Cell Death Dis. 2021;12(12):1109.

    Article  CAS  Google Scholar 

  39. Zhu H, You Y, Shen Z, Shi L. EGFRvIII-CAR-T cells with PD-1 knockout have improved anti-glioma activity. Pathol Oncol Res. 2020;26(4):2135–41.

    Article  CAS  Google Scholar 

  40. Xia L, Zheng Z-Z, Liu J-Y, Chen Y-J, Ding J-C, Xia N-S, et al. EGFR-targeted CAR-T cells are potent and specific in suppressing triple-negative breast cancer both in vitro and in vivo. Clin Transl Immunology. 2020;9(5):e01135.

    Article  Google Scholar 

  41. Liu Y, Guo Y, Wu Z, Feng K, Tong C, Wang Y, et al. Anti-EGFR chimeric antigen receptor-modified T cells in metastatic pancreatic carcinoma: a phase I clinical trial. Cytotherapy. 2020;22(10):573–80.

    Article  Google Scholar 

  42. Zhang C, Wang Z, Yang Z, Wang M, Li S, Li Y, et al. Phase I escalating-dose trial of CAR-T therapy targeting CEA + metastatic colorectal cancers. Mol Ther. 2017;25(5):1248–58.

    Article  CAS  Google Scholar 

  43. Chi X, Yang P, Zhang E, Gu J, Xu H, Li M, et al. Significantly increased anti-tumor activity of carcinoembryonic antigen‐specific chimeric antigen receptor T cells in combination with recombinant human IL‐12. Cancer Med. 2019;8(10):4753–65.

    Article  CAS  Google Scholar 

  44. Sotoudeh M, Shirvani SI, Merat S, Ahmadbeigi N, Naderi M. MSLN (Mesothelin), ANTXR1 (TEM8), and MUC3A are the potent antigenic targets for CAR T cell therapy of gastric adenocarcinoma. J of Cellular Biochemistry. 2019;120(4):5010–7.

    Article  CAS  Google Scholar 

  45. Jafarzadeh L, Masoumi E, Alishah K, Mirzaei HR, Jamali A, Fallah-Mehrjardi K, et al. Construction and functional characterization of a fully human anti-mesothelin chimeric Antigen receptor (CAR)-expressing T cell. IJAAI. 2020;19(3):264–75.

    Google Scholar 

  46. Lv J, Zhao R, Wu D, Zheng D, Wu Z, Shi J, et al. Mesothelin is a target of chimeric antigen receptor T cells for treating gastric cancer. J Hematol Oncol. 2019;12(1):18.

    Article  Google Scholar 

  47. Haas AR, Tanyi JL, O’Hara MH, Gladney WL, Lacey SF, Torigian DA, et al. Phase I study of lentiviral-transduced chimeric antigen receptor-modified T cells recognizing mesothelin in advanced solid cancers. Mol Ther. 2019;27(11):1919–29.

    Article  CAS  Google Scholar 

  48. Sait S, Modak S. Anti-GD2 immunotherapy for neuroblastoma. Expert Rev Anticancer Ther. 2017;17(10):889–904.

    Article  CAS  Google Scholar 

  49. Prapa M, Chiavelli C, Golinelli G, Grisendi G, Bestagno M, Di Tinco R, et al. GD2 CAR T cells against human glioblastoma. npj Precis Onc. 2021;5(1):93.

    Article  Google Scholar 

  50. Straathof K, Flutter B, Wallace R, Jain N, Loka T, Depani S, et al. Antitumor activity without on-target off-tumor toxicity of GD2–chimeric antigen receptor T cells in patients with neuroblastoma. Sci Transl Med. 2020;12(571):eabd6169.

    Article  CAS  Google Scholar 

  51. Majzner RG, Ramakrishna S, Yeom KW, Patel S, Chinnasamy H, Schultz LM, et al. GD2-CAR T cell therapy for H3K27M-mutated diffuse midline gliomas. Nature. 2022;603(7903):934–41.

    Article  CAS  Google Scholar 

  52. Tay JCK, Wang J, Du Z, Ng YY, Li Z, Ren Y, et al. Manufacturing NKG2D CAR-T cells with piggyBac transposon vectors and K562 artificial antigen-presenting cells. Mol Ther Methods Clin Dev. 2021;21:107–20.

    Article  CAS  Google Scholar 

  53. Zhang Y, Li X, Zhang J, Mao L. Novel cellular immunotherapy using NKG2D CAR-T for the treatment of cervical cancer. Biomed Pharmacother. 2020;131:110562.

    Article  CAS  Google Scholar 

  54. Ng Y-Y, Tay JCK, Li Z, Wang J, Zhu J, Wang ST, Cells Expressing. NKG2D CAR with a DAP12 signaling domain stimulate lower cytokine production while effective in tumor eradication. Mol Ther. 2021;29(1):75–85.

    Article  CAS  Google Scholar 

  55. Yang Y, McCloskey JE, Yang H, Puc J, Alcaina Y, Vedvyas Y, et al. Bispecific CAR T cells against EpCAM and inducible ICAM-1 overcome Antigen heterogeneity and generate superior antitumor responses. Cancer Immunol Res. 2021;9(10):1158–74.

    Article  Google Scholar 

  56. Fu J, Shang Y, Qian Z, Hou J, Yan F, Liu G, et al. Chimeric Antigen receptor-T (CAR-T) cells targeting epithelial cell adhesion molecule (EpCAM) can inhibit tumor growth in ovarian cancer mouse model. J Vet Med Sci. 2021;83(2):241–7.

    Article  CAS  Google Scholar 

  57. Li W, Zhou Y, Wu Z, Shi Y, Tian E, Zhu Y, et al. Targeting wnt signaling in the tumor immune microenvironment to enhancing EpCAM CAR T-cell therapy. Front Pharmacol. 2021;12:724306.

    Article  CAS  Google Scholar 

  58. Zhang Z, Lu M, Qin Y, Gao W, Tao L, Su W, et al. Neoantigen: a new breakthrough in tumor immunotherapy. Front Immunol. 2021;12:672356.

    Article  CAS  Google Scholar 

  59. Ohno M, Ohkuri T, Kosaka A, Tanahashi K, June CH, Natsume A, et al. Expression of mir-17-92 enhances anti-tumor activity of T-cells transduced with the anti-EGFRvIII chimeric antigen receptor in mice bearing human GBM xenografts. J Immunother Cancer. 2013;1:21.

    Article  Google Scholar 

  60. Li Y, Wu H, Chen G, Wei X, Wang C, Zhou S, et al. Arming Anti-EGFRvIII CAR-T with TGFβ trap improves Antitumor efficacy in glioma mouse models. Front Oncol. 2020;10:1117.

    Article  Google Scholar 

  61. Choi BD, Yu X, Castano AP, Bouffard AA, Schmidts A, Larson RC, et al. CAR-T cells secreting BiTEs circumvent antigen escape without detectable toxicity. Nat Biotechnol. 2019;37(9):1049–58.

    Article  CAS  Google Scholar 

  62. Fisher J, Abramowski P, Wisidagamage Don ND, Flutter B, Capsomidis A, Cheung GW-K, et al. Avoidance of on-target off-tumor activation using a co-stimulation-only chimeric Antigen receptor. Mol Ther. 2017;25(5):1234–47.

    Article  CAS  Google Scholar 

  63. Pearlman AH, Hwang MS, Konig MF, Hsiue EH-C, Douglass J, DiNapoli SR, et al. Targeting public neoantigens for cancer immunotherapy. Nat Cancer. 2021;2(5):487–97.

    Article  CAS  Google Scholar 

  64. Zhang Q, Jia Q, Zhang J, Zhu B. Neoantigens in precision cancer immunotherapy: from identification to clinical applications. Chin Med J (Engl). 2022;135(11):1285–98.

    Article  Google Scholar 

  65. Gao J, Yuan X, Yuan J, Li L. Complete rejection of large established breast cancer by local immunochemotherapy with T cell activation against neoantigens. Cancer Immunol Immunother. 2021;70(11):3291–302.

    Article  CAS  Google Scholar 

  66. Okada M, Shimizu K, Fujii S-I. Identification of neoantigens in cancer cells as targets for immunotherapy. Int J Mol Sci. 2022;23(5):2594.

    Article  CAS  Google Scholar 

  67. O’Rourke DM, Nasrallah MP, Desai A, Melenhorst JJ, Mansfield K, Morrissette JJD, 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.

    Article  Google Scholar 

  68. Zhang E, Yang P, Gu J, Wu H, Chi X, Liu C, et al. Recombination of a dual-CAR-modified T lymphocyte to accurately eliminate pancreatic malignancy. J Hematol Oncol. 2018;11(1):102.

    Article  Google Scholar 

  69. Choe JH, Watchmaker PB, Simic MS, Gilbert RD, Li AW, Krasnow NA, et al. SynNotch-CAR T cells overcome challenges of specificity, heterogeneity, and persistence in treating glioblastoma. Sci Transl Med. 2021;13(591):eabe7378.

    Article  CAS  Google Scholar 

  70. Li S, Zhao R, Zheng D, Qin L, Cui Y, Li Y, et al. DAP10 integration in CAR-T cells enhances the killing of heterogeneous tumors by harnessing endogenous NKG2D. Mol Therapy - Oncolytics. 2022;26:15–26.

    Article  CAS  Google Scholar 

  71. Wilkie S, van Schalkwyk MCI, Hobbs S, Davies DM, van der Stegen SJC, Pereira ACP, et al. Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling. J Clin Immunol. 2012;32(5):1059–70.

    Article  CAS  Google Scholar 

  72. Mestermann K, Giavridis T, Weber J, Rydzek J, Frenz S, Nerreter T, 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.

    Article  Google Scholar 

  73. Hegde M, Mukherjee M, Grada Z, Pignata A, Landi D, Navai SA, et al. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J Clin Invest. 2016;126(8):3036–52.

    Article  Google Scholar 

  74. Hirabayashi K, Du H, Xu Y, Shou P, Zhou X, Fucá G, et al. Dual targeting CAR-T cells with optimal costimulation and metabolic fitness enhance antitumor activity and prevent escape in solid tumors. Nat Cancer. 2021;2(9):904–18.

    Article  CAS  Google Scholar 

  75. Ross SL, Sherman M, McElroy PL, Lofgren JA, Moody G, Baeuerle PA, et al. Bispecific T cell engager (BiTE®) antibody constructs can mediate bystander tumor cell killing. PLoS ONE. 2017;12(8):e0183390.

    Article  Google Scholar 

  76. Huang R, Li X, He Y, Zhu W, Gao L, Liu Y, et al. Recent advances in CAR-T cell engineering. J Hematol Oncol. 2020;13(1):86.

    Article  Google Scholar 

  77. Park S, Shevlin E, Vedvyas Y, Zaman M, Park S, Hsu Y-MS, et al. Micromolar affinity CAR T cells to ICAM-1 achieves rapid tumor elimination while avoiding systemic toxicity. Sci Rep. 2017;7(1):14366.

    Article  Google Scholar 

  78. Drent E, Themeli M, Poels R, de Jong-Korlaar R, Yuan H, de Bruijn J, et al. A rational strategy for reducing on-target off-tumor effects of CD38-chimeric antigen receptors by affinity optimization. Mol Ther. 2017;25(8):1946–58.

    Article  CAS  Google Scholar 

  79. Zhang Z, Jiang D, Yang H, He Z, Liu X, Qin W, et al. Modified CAR T cells targeting membrane-proximal epitope of mesothelin enhances the antitumor function against large solid tumor. Cell Death Dis. 2019;10(7):476.

    Article  Google Scholar 

  80. Calderon H, Mamonkin M, Guedan S. Analysis of CAR-mediated tonic signaling. In: Swiech K, Malmegrim KCR, Picanço-Castro V, editors. Chimeric Antigen receptor T cells. New York, NY: Springer US; 2020. p. 223–36.

    Chapter  Google Scholar 

  81. Bao C, Gao Q, Li L-L, Han L, Zhang B, Ding Y, et al. The application of nanobody in CAR-T therapy. Biomolecules. 2021;11(2):238.

    Article  CAS  Google Scholar 

  82. Jia Q, Wang A, Yuan Y, Zhu B, Long H. Heterogeneity of the tumor immune microenvironment and its clinical relevance. Exp Hematol Oncol. 2022;11(1):24.

    Article  CAS  Google Scholar 

  83. Liu G, Rui W, Zhao X, Lin X. Enhancing CAR-T cell efficacy in solid tumors by targeting the tumor microenvironment. Cell Mol Immunol. 2021;18(5):1085–95.

    Article  CAS  Google Scholar 

  84. Aparicio C, Belver M, Enríquez L, Espeso F, Núñez L, Sánchez A, et al. Cell therapy for colorectal cancer: the promise of chimeric antigen receptor (CAR)-T cells. Int J Mol Sci. 2021;22(21):11781.

    Article  CAS  Google Scholar 

  85. Marofi F, Achmad H, Bokov D, Abdelbasset WK, Alsadoon Z, Chupradit S, et al. Hurdles to breakthrough in CAR T cell therapy of solid tumors. Stem Cell Res Ther. 2022;13(1):140.

    Article  Google Scholar 

  86. Jafarzadeh L, Masoumi E, Mirzaei HR, Alishah K, Fallah-Mehrjardi K, Khakpoor-Koosheh M, et al. Targeted knockdown of Tim3 by short hairpin RNAs improves the function of anti-mesothelin CAR T cells. Mol Immunol. 2021;139:1–9.

    Article  CAS  Google Scholar 

  87. Li J, Li W, Huang K, Zhang Y, Kupfer G, Zhao Q. Chimeric antigen receptor T cell (CAR-T) immunotherapy for solid tumors: lessons learned and strategies for moving forward. J Hematol Oncol. 2018;11(1):22.

    Article  CAS  Google Scholar 

  88. Ji F, Zhang F, Zhang M, Long K, Xia M, Lu F, et al. Targeting the DNA damage response enhances CD70 CAR-T cell therapy for renal carcinoma by activating the cGAS-STING pathway. J Hematol Oncol. 2021;14(1):152.

    Article  CAS  Google Scholar 

  89. Johnson LR, Lee DY, Eacret JS, Ye D, June CH, Minn AJ. The immunostimulatory RNA RN7SL1 enables CAR-T cells to enhance autonomous and endogenous immune function. Cell. 2021;184(19):4981–95e14.

    Article  CAS  Google Scholar 

  90. Liu C, Qi T, Milner JJ, Lu Y, Cao Y. Speed and location both matter: Antigen stimulus dynamics controls CAR-T cell response. Front Immunol. 2021;12:748768.

    Article  CAS  Google Scholar 

  91. Belk JA, Yao W, Ly N, Freitas KA, Chen Y-T, Shi Q, et al. Genome-wide CRISPR screens of T cell exhaustion identify chromatin remodeling factors that limit T cell persistence. Cancer Cell. 2022;40(7):768–86e7.

    Article  CAS  Google Scholar 

  92. Carnevale J, Shifrut E, Kale N, Nyberg WA, Blaeschke F, Chen YY, et al. RASA2 ablation in T cells boosts antigen sensitivity and long-term function. Nature. 2022;609(7925):174–82.

    Article  CAS  Google Scholar 

  93. Andersen MH. Tumor microenvironment antigens. Semin Immunopathol. 2022. https://doi.org/10.1007/s00281-022-00966-0.

    Article  Google Scholar 

  94. Li P, Yang L, Li T, Bin S, Sun B, Huang Y, et al. The third generation Anti-HER2 chimeric Antigen receptor mouse T cells alone or together with Anti-PD1 antibody inhibits the growth of mouse breast tumor cells expressing HER2 in vitro and in immune competent mice. Front Oncol. 2020;10:1143.

    Article  Google Scholar 

  95. Rosewell Shaw A, Porter CE, Yip T, Mah W-C, McKenna MK, Dysthe M, et al. Oncolytic adeno-immunotherapy modulates the immune system enabling CAR T-cells to cure pancreatic tumors. Commun Biol. 2021;4(1):368.

    Article  CAS  Google Scholar 

  96. Li H, Yuan W, Bin S, Wu G, Li P, Liu M, et al. Overcome trastuzumab resistance of breast cancer using anti-HER2 chimeric antigen receptor T cells and PD1 blockade. Am J Cancer Res. 2020;10(2):688–703.

    CAS  Google Scholar 

  97. He H, Liao Q, Zhao C, Zhu C, Feng M, Liu Z, et al. Conditioned CAR-T cells by hypoxia-inducible transcription amplification (HiTA) system significantly enhances systemic safety and retains antitumor efficacy. J Immunother Cancer. 2021;9(10):e002755.

    Article  Google Scholar 

  98. Raj D, Yang M-H, Rodgers D, Hampton EN, Begum J, Mustafa A, et al. Switchable CAR-T cells mediate remission in metastatic pancreatic ductal adenocarcinoma. Gut. 2019;68(6):1052–64.

    Article  CAS  Google Scholar 

  99. Burga RA, Thorn M, Point GR, Guha P, Nguyen CT, Licata LA, et al. Liver myeloid-derived suppressor cells expand in response to liver metastases in mice and inhibit the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol Immunother. 2015;64(7):817–29.

    Article  CAS  Google Scholar 

  100. Zhang W, Liu L, Su H, Liu Q, Shen J, Dai H, et al. Chimeric antigen receptor macrophage therapy for breast tumours mediated by targeting the tumour extracellular matrix. Br J Cancer. 2019;121(10):837–45.

    Article  CAS  Google Scholar 

  101. Jiang W, Li T, Guo J, Wang J, Jia L, Shi X, et al. Bispecific c-Met/PD-L1 CAR-T cells have enhanced therapeutic effects on hepatocellular carcinoma. Front Oncol. 2021;11:546586.

    Article  Google Scholar 

  102. Evgin L, Kottke T, Tonne J, Thompson J, Huff AL, van Vloten J, et al. Oncolytic virus-mediated expansion of dual-specific CAR T cells improves efficacy against solid tumors in mice. Sci Transl Med. 2022;14(640):eabn2231.

    Article  CAS  Google Scholar 

  103. Zhang Y, Li S, Wang Y, Lu Y, Xu Y, Rao Q, et al. A novel and efficient CD22 CAR-T therapy induced a robust antitumor effect in relapsed/refractory leukemia patients when combined with CD19 CAR-T treatment as a sequential therapy. Exp Hematol Oncol. 2022;11(1):15.

    Article  CAS  Google Scholar 

  104. Muliaditan T, Halim L, Whilding LM, Draper B, Achkova DY, Kausar F, et al. Synergistic T cell signaling by 41BB and CD28 is optimally achieved by membrane proximal positioning within parallel chimeric antigen receptors. Cell Rep Med. 2021;2(12):100457.

    Article  CAS  Google Scholar 

  105. Minagawa K, Al-Obaidi M, Di Stasi A. Generation of suicide gene-modified chimeric antigen receptor-redirected T-cells for cancer immunotherapy. Methods Mol Biol. 2019;1895:57–73.

    Article  CAS  Google Scholar 

  106. Zhao Z, Chen Y, Francisco NM, Zhang Y, Wu M. The application of CAR-T cell therapy in hematological malignancies: advantages and challenges. Acta Pharm Sin B. 2018;8(4):539–51.

    Article  Google Scholar 

  107. Moghimi B, Muthugounder S, Jambon S, Tibbetts R, Hung L, Bassiri H, et al. Preclinical assessment of the efficacy and specificity of GD2-B7H3 SynNotch CAR-T in metastatic neuroblastoma. Nat Commun. 2021;12(1):511.

    Article  CAS  Google Scholar 

  108. Gill SI. How close are we to CAR T-cell therapy for AML? Best Pract Res Clin Haematol. 2019;32(4):101104.

    Article  Google Scholar 

  109. Loff S, Dietrich J, Meyer J-E, Riewaldt J, Spehr J, von Bonin M, et al. Rapidly switchable Universal CAR-T cells for treatment of CD123-positive leukemia. Mol Ther Oncolytics. 2020;17:408–20.

    Article  CAS  Google Scholar 

  110. Kobayashi A, Nobili A, Neier SC, Sadiki A, Distel R, Zhou ZS, et al. Light-controllable binary switch activation of CAR T cells. 2022;17(12): e202100722. 

    CAS  Google Scholar 

  111. Lin H, Cheng J, Mu W, Zhou J, Zhu L. Advances in Universal CAR-T cell therapy. Front Immunol. 2021;12:744823.

    Article  CAS  Google Scholar 

  112. Zhao J, Lin Q, Song Y, Liu D. Universal CARs, universal T cells, and universal CAR T cells. J Hematol Oncol. 2018;11(1):132.

    Article  CAS  Google Scholar 

  113. Larson RC, Kann MC, Bailey SR, Haradhvala NJ, Llopis PM, Bouffard AA, et al. CAR T cell killing requires the IFNγR pathway in solid but not liquid tumours. Nature. 2022;604(7906):563–70.

    Article  CAS  Google Scholar 

  114. Kaneko S. Successful organoid-mediated generation of iPSC-derived CAR-T cells. Cell Stem Cell. 2022;29(4):493–5.

    Article  CAS  Google Scholar 

  115. Wang Z, McWilliams-Koeppen HP, Reza H, Ostberg JR, Chen W, Wang X, et al. 3D-organoid culture supports differentiation of human CAR + iPSCs into highly functional CAR T cells. Cell Stem Cell. 2022;29(4):515–27e8.

    Article  Google Scholar 

Download references

Acknowledgements

We are all thankful to all researchers devoted to cancer therapy studies in the field. We are also thankful to Dr Weijia Wang for his professional English language editing.

Funding

This work was supported by grants from Key Research and Development Projects of Hainan Province of China (ZDYF2022SHFZ133), the Natural Science Foundation of Hainan Province of China (822MS179), the Natural Science Foundation of Fujian Province of China (2021J011266), and the National Natural Science Foundation of China (82260161).

Author information

Authors and Affiliations

Authors

Contributions

TY drafted the manuscript and visualized the tables and figures. LFZ and JC designed the review and critically revised the manuscript. All authors read and approved the final manuscript.

Corresponding authors

Correspondence to Lingfeng Zhu or Jin Chen.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yan, T., Zhu, L. & Chen, J. Current advances and challenges in CAR T-Cell therapy for solid tumors: tumor-associated antigens and the tumor microenvironment. Exp Hematol Oncol 12, 14 (2023). https://doi.org/10.1186/s40164-023-00373-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40164-023-00373-7

Keywords