Review Article

A Description of Chimeric Antigen ReceptorModified T Cells in Cancer Immunotherapy

by Ximei Xu*, Yin Xiang, Yang Yang, Zhiwei Cui, Maoqiang Wang, Yanhong Gu, Jing Huang

Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, Sichuan, China

*Corresponding author: Ximei Xu, Department of Medical Laboratory, People’s Hospital of Leshan, Leshan, Sichuan, 614000, China

Received Date: 04 October, 2023

Accepted Date: 11 October, 2023

Published Date: 16 October, 2023

Citation: Xu X, Xiang Y, Yang Y, Cui Z, Wang M, et al. (2023) A Description of Chimeric Antigen Receptor-Modified T Cells in Cancer Immunotherapy. J Oncol Res Ther 8: 10187. DOI: https://doi.org/10.29011/2574-710X.10187

Abstract

Chimeric Antigen Receptor (CAR)-modified T cells, or CAR-T cell therapy, is emerging as a promising new strategy for immunotherapy, and by modifying T cells in vitro to add a CAR that specifically recognizes tumor antigens, CAR-T cell immunotherapy improves tumor-specific killing. This immune cell therapy has the advantages of good targeting and killing power, and it enables effective activation and proliferation of T cells independent of MHC molecules, which is a very effective new strategy for immune killing. In this paper, we will introduce the composition of CAR, the basic principles of CAR-T cell immunotherapy and the preparation aspects of CAR-T cells, and highlight an overview of the use of CAR-T cell immunotherapy in various tumor treatments in recent years.

Keywords: Car-T therapy; Oncology; Immune cells; Targeted therapy; Immunotherapy

Introduction

Among the basic methods of treating tumors, radiotherapy and chemotherapy still occupy an important position, but the accompanying toxic side effects cannot be ignored, which seriously affect the daily life of patients. To solve these problems, researchers have gradually turned to research on the role of immune cells and inflammatory factors in the tumor microenvironment, hoping to use the normal body immune system to monitor and kill cancer cells. So immune cell therapy has begun to be used in the treatment of tumors [1].

Among them, chimeric antigen receptor T-cell immunotherapy (CAR-T), which introduces Chimeric Antigen Receptors (CARs) into T cells, has a specific killing effect on cancer cells by using modified CAR-T to specifically recognize and bind to Tumor Associated Antigens (TAAs) on the surface of cancer cells, thus achieving specific treatment for cancer [24]. This method can be used for specific treatment of cancer cells. At the same time, this method can show better anti-tumor effects because it does not require the participation of antigenpresenting cells (APCs) and is therefore not limited by the major histocompatibility complex (MHC) [5]. In the clinical treatment of hematologic tumors, CAR-T cell immunotherapy has achieved better results, especially in CD19-positive B-cell leukemia and lymphoma, with an overall remission rate of more than 80%, which has again increased the interest of researchers in modifying T-cell immunotherapy and gradually extending it to clinical treatment of various tumors [6,7].

In this paper, we first describe the basic components and principles of CAR-T, then review the progress of CAR-T cell immunotherapy for various tumors in recent years, and finally summarize the future of CAR-T cell immunotherapy.

CAR-T cell immunotherapy

Composition of CAR

The key site in CAR-T cell immunotherapy is the chimeric antigen receptor (CAR), which consists of three main components, namely the extracellular antigen-binding domain, the hinge region and the intracellular structural domain, and is a fusion protein expressed on T cells [8,9]. The extracellular antigen binding domain can be composed of a monoclonal antibody heavy chain and a light chain with a single variable fragment, and this binding domain can determine the target of CAR-T cell attack through its antigen specificity [10]. The hinge region, as a transmembrane spatial domain with a certain degree of toughness, mainly functions as a link between the extracellular antigen binding domain and the intracellular structural domain, which determines the stability and signalling function of CAR, and thus facilitates the binding of CAR-T cells to target cells [10,11]. The intracellular structural domains are mainly divided into co-stimulatory structural domains and T cell activation structural domains, which serve to trigger and activate antigens to generate specific immune responses and transmit first and second signals to influence the activation, proliferation and killing of T cells [12]. Currently, researchers have conducted research on the design of CARs for their structure and have reached the fifth generation. The first-generation CARs consist of an extracellular single-chain variable fragment scFv, a transmembrane region and a single intracellular activation signal molecule [13]. These CARs lack the second signal molecule to induce T-cell activation, and can only transmit the activation signal to the cell, causing a short period of T-cell proliferation and a small amount of cytokine secretion, which is not conducive to the long-term anti-tumor activity of CAR-T cells in patients [14]. The second-generation CARs add co-stimulatory molecules to the original ones, which can enhance the killing and toxic function of T cells on tumor cells by activating the second signal. The thirdgeneration CARs add multiple costimulatory molecules, which interact with each other to enhance multiple signaling pathways in T cells, resulting in more significant enhancement of T cells in various aspects of anti-tumor cell functions, prolonging T cell proliferation activity, survival cycle and promoting the release of multiple cytokines. The fourth-generation CAR, on the other hand, adds selectable markers to multiple co-stimulatory molecular structures, allowing T-cell activity to be enhanced by the genes encoding CARs, inducing them to secrete cytokines and chemokines, recruiting and activating their own intrinsic immune cells, and improving T-cell survival in the tumor microenvironment. Fifth-generation CART cells are general-purpose CAR-T cells designed on the basis of gene editing technology to recognize more target proteins and improve the flexibility of CART cells to treat different cancers. Its genes are designed to prevent rejection by the body and to pre-prepare allogeneic T cells for patients for ready supply, but so far, fifth-generation CAR-T cells have not been widely used [15] (Figure 1).

 

Figure 1: The way of Car-T cell therapy in human.

Principles of CAR-T cell immunotherapy

Activation signalling of T cells and tumor-specific recognition are the basic principles of CAR-T cell immunotherapy [16]. A dual signalling pathway induces T-cell activation, consisting of antigen-specific signals, i.e., the binding of T-cell receptors (TCRs) to major histocompatibility complexes (MHCs) - antigenic peptides, as the first signalling pathway, and antigennonspecific signals, consisting of the binding of T cells to CMs on the surface of antigen-presenting cells (APCs), as the second signalling pathway [17]. The activation and cascade of T cells are informed by both signalling pathways, with the ultimate goal of converting T cells into cytotoxic T cells (CLT). This binding can induce the production of perforin, which can lyse tumor cells, or release a large amount of cytokines, which can change the living environment of tumor cells, thereby directly killing them or inhibiting their growth [18-20]. CAR-T cell immunotherapy, on the other hand, utilizes the combination of the effect of CLT on target cell toxicity and the specific recognition of antigens by antibodies to enable CAR to kill tumor cells by generating activated T cells through the specific recognition of antigens that target tumor cells [21]. In this treatment, most of the T cells are derived from the peripheral blood of the patient, and by introducing the designed exogenous gene into the T cells, a stably expressed CAR-T cell can be obtained, then the stably expressed CAR-T cells are cultured and proliferated in vitro, and finally the proliferated CAR-T cells are injected back into the patient to kill the tumor cells.

Preparation of CAR-T

Firstly, T cells are isolated from the patient’s peripheral blood cells, and then the T cells are cultured and proliferated in vitro. And secondly, the CAR gene is introduced into the patient’s isolated T cells by electroporation, lentivirus or retrovirus transduction, so that the T cells are modified to express the CAR gene to obtain stably expressed CAR-T cells. The retroviral transduction method has a wide range of application and high transduction efficiency, but has a high risk of tumorigenicity due to the loss of the ability to infect non-dividing cells due to the small DNA fragments carried by the transduction method. For safety reasons, electroporation is safe, but has the disadvantage of transient expression, which requires multiple injections [22-24].

Current status of research on CAR-T therapy for various types of cancer treatment

Chimeric antigen receptor (CAR) T-cell technology was originally proposed by Eshhar and Gross in the 1980s with the idea of directing T-cell responses through genetic editing of T-cell receptors (TCRs) to enable them to play a greater role in the immune response and fight against tumors. In recent years, various gene editing techniques have enabled CAR-T cells to be targeted to different sites, thus greatly improving the therapeutic effect on various types of cancers [12].

CAR-T cell immunotherapy for head and neck squamous cell carcinoma

Head and neck squamous cell carcinomas (HNSCCs), as malignant lesions of the mucosal epithelium of the oral cavity, pharynx and larynx, account for 90% of all head and neck cancers and are among the sixth most common malignancies worldwide(25-27). Surgery with conventional radiotherapy, chemotherapy and immunotherapy is the basis of treatment for squamous cell carcinoma of the head and neck [28]. However, tumor recurrence and metastasis remain at a high level with these treatment modalities. There is growing evidence that chimeric antigen receptor T (CAR-T) cells have promising antitumor effects in the treatment of HNSCC [29,30].

Currently, investigators are approaching the treatment of squamous cell carcinoma of the head and neck through three main areas: preclinical studies, clinical studies, and the search for potential targets [30,31] (Table.1).

Preclinical study target

Reference

EGFR

[1]

MUCI

[2]

B7-H3

[3]

HER2

[4]

CD70

[5]

Clinical trial target

 Reference

ERBb2/HER2

[6]

NKG2DL

[6]

EpCAM

[6]

LMP1

 [6]

Potential target

Reference

NKGD2

[7]

HER3

 [8,9]

FAP

 [10-13]

Table 1: The treatment of squamous cell carcinoma of the head and neck.

In preclinical studies, investigators found that HER2 expression was detectable in 0-47% of HNSCC patient tissues and that HER2 overexpression was often associated with a poorer prognosis for HNSCC patients [44,45]. Warren et al. developed specific CAR-T cells targeting HER2 for the treatment of HNSCC and their results showed that anti-HER2 CAR-T cells resulted in a 56% reduction in tumor size, suggesting that HER2 may be a potential target for CAR-T cells to treat HERpositive HNSCC [38,46,47]. Similarly, overexpression of CD70, a ligand for tumor necrosis factor, which is highly expressed in HNSCC, is associated with a reduction in CD8+ T cells and can induce immunosuppression TME [48]. Park et al. analysed nine proteins highly expressed in HNSCC cells as potential CAR-T cell targets and demonstrated that compared to the untreated group, anti-CD70 CAR-T cells could effectively eradicate HNSCC cells [38,49]. In addition to this, EGFR was overexpressed in hypopharyngeal cancer, which accounts for about 5% of HNSCC [50]. Thus, Dong et al. developed CAR-T cells targeting EGFR to limit the growth of EGFR-positive hypopharyngeal cancer cells. The results showed that the cytokine secretion and lysis rates of hypopharyngeal cancer cells were significantly increased after coculture of hypopharyngeal cancer cells with CAR-T cells [32].

For clinical studies in the treatment of HNSCC, Sophie Papa et al. conducted a phase I clinical trial for the treatment of HNSCC in combination with intra-tumor injection of the binary lysing adenovirus CAdVEC to investigate the safety and cytotoxic effects of CAR-T cell immunotherapy targeting HER2 in the treatment of HNSCC [30].

In conclusion, in the treatment of HNSCC, researchers have been searching for its new targets to enhance the specificity of CAR-T cells, reduce the side effects of CAR-T cells, and continuously promote the transformation of CAR-T cell immunotherapy to clinical applications.

CAR-T cell immunotherapy for breast cancer

Breast cancer is the leading cancer in women. Approximately 20-30% of breast cancer patients develop invasive or metastatic disease after radical surgical resection and eventually die [51,52]. Despite advances in chemotherapy, endocrine therapy and molecular targeted therapy, there are still some breast cancer patients who are less sensitive to treatment. Treatment resistance and tumor recurrence or metastasis also develop as treatment proceeds [53]. Therefore, investigators need to explore more favourable treatment strategies. Chimeric antigen receptormodified T cells have made some progress in immunotherapy for breast cancer by exploring breast cancer-related targets [5456]. For example, for HER-2-positive breast cancer, a subtype that affects approximately one quarter of breast cancer patients, HER-2 gene amplification or protein overexpression underlies HER-2-positive breast cancer [57,58]. HER-2 can be used as a recognition molecule for constructing HER-2-specific CAR-T cells for immunotherapy of HER2-positive breast cancer [59]. li et al. used CAR-T cells targeting HER-2 cells in combination with PD-1 blockers to kill Herceptin-resistant breast cancer cells [60]. seyedmirzaei et al. used HER-2-specific CAR-T cells to eliminate tumors and improve survival of trastuzumab-resistant breast tumor cells in mice [61]. meili et al. prepared chA21 scFV-based HER-2specific CAR-T cells and it was shown to recognize and kill HER2-positive breast cancer cells in vitro, as well as to induce breast cancer regression in vivo [62].

In addition, CAR-T cell immunotherapy is being further investigated by researchers in triple-negative breast cancer (TNBC) and other types of breast cancer (Table 2).

Brest cancer subtype

Target

Intervention (CAR-T)

NCT Number

HER-2 positive

HER2

HER2-CAR T cells

NCT02547961, NCT03696030

Anti-HER2 CAR-T

NCT02713984

CAdVEC

NCT03740256

HER2-targeted dual-switch CAR-T cells

NCT04650451

HER2-negative

Mesothelin

Mesothelintargeted T cells

NCT02792114

TNBC

MUC1

anti-MUC1 CAR T Cells

NCT02587689

Mesothelin

CART-meso cells

NCT02580747

ROR1

ROR1 CAR-T cells

NCT02706392

ROR1-targeted CAR T cells

NCT05274451

NKG2DL

NKG2DL-targeting CAR- ch CAR-T

NCT04107142

c-Met

mRNA c-Met-CAR T cell

NCT01837602

Other types

CEA

Anti-CEA CAR T cells

NCT02349724, NCT03682744

CEA CAR-T cells

NCT04348643

Mesothelin

iCasp9M28z T cell infusions

NCT02414269

EpCAM

EpCAM CAR-T

NCT02915445

CD70

Anti-hCD70 CAR transduced PBL

NCT02830724

HER2/Gd2/ CD44v6

Multiple 4SCAR T cells

NCT04430595

Table 2: CAR-T cell immunotherapy in TNBC and other types of breast cancer.

CAR-T cell immunotherapy for HIV

CAR-T cell immunotherapy has been applied in clinical trials for a variety of cancers with promising results, which have demonstrated the safety, feasibility, efficacy and durability of CAR-T cell immunotherapy [63,64]. Based on these results, the HIV field has begun to view CAR-T cell immunotherapy as a promising HIV cure strategy [65,66]. Unlike other strategies for treating HIV, CAR-T cell immunotherapy would be a targeted, curative tool designed to completely eliminate the HIV reservoir from the patient’s organism [67-69]. Table 3 summarizes the clinical trials of investigators using CAR-T cell immunotherapy for HIV treatment over the last two decades.

Start Date

Study Title

NCT Number

Reference

2001

A phase I/II study of the safety, survival, and trafficking of autologous CD4-ζ genemodified T cells with and without extension Interleukin-2 in HIV infected patients

NCT01013415

-14

2017

The effect of CAR-T cell therapy on the reconstitution of HIV-specific immune function

NCT03240328

-15

2019

A pilot study of T cells genetically modified by Zinc Finger Nucleases SB-728mR and CD4 chimeric antigen receptor in HIV-infected subjects

NCT03617198

Ongoing

2021

Safety and anti-HIV activity of autologous CD4+ and CD8+ T cells transduced with a lentiviral vector encoding bispecific anti-gp120 CAR molecules

(LVgp120duoCAR-T) in anti-retroviral drug-treated HIV-1 infection

NCT04648046

Ongoing

Table 3: CAR-T cell immunotherapy for HIV treatment.

CAR-T cell immunotherapy for ovarian cancer

Ovarian cancer (OC), one of the most common gynecologic malignancies, has a poor prognosis and high mortality rate [72]. Most patients are diagnosed at advanced stages (stage III or IV), with a global 5-year survival rate of 25% to 47%. Surgical resection and first-line chemotherapy are the main treatment modalities for OC [73]. However, due to resistance to chemotherapy, patients usually relapse within a few years of initial treatment. Cell-based therapy, chimeric antigen receptor T (CAR-T) cell therapy, represents an alternative immunotherapeutic approach with great potential for the immunotherapy of ovarian cancer [74,75]. Ovarian tumors typically lack TSA, and therefore, the target of immunotherapy is usually one or more tumor-associated antigens [7]. Researchers are currently constructing CAR-T cells that target different targets on the surface of ovarian cancer to achieve ovarian cancer immunotherapy [5,19]. For example, erb-b2 receptor tyrosine kinase 2 (ERBB2) [62,76], programmed cell death-ligand 1 (CD274) [73,77], programmed cell death 1 (PDCD1) [78-80], epithelial cell adhesion molecule (Ep-CAM) [81], anti-Müllerian hormone receptor type 2 (AMHR2) [82], annexin A2 (ANXA2) [83], trophoblast glycoprotein (TPBG) [74], folate receptor alpha ( FOLR1) [84,85], mesothelin (MSLN) [72,8688], mucin 16 (MUC16) [89,90] and CD24 [75], among others (Figure 2).

 

Figure 2: Constructing CAR-T cells that target different targets on the surface of ovarian cancer to achieve ovarian cancer immunotherapy.

ERBB2, also known as human epidermal growth factor receptor 2 (HER2), is a proto-oncogene with gene amplification and overexpression associated with OC and negative or very low protein expression in normal tissues. Sun et al. [62] have developed a HER2-CAR-T-cell treatment and found that novel MSLN is a group of glycoproteins anchored to the plasma membrane by the phosphatidylinositol region (GPI) and is normally expressed in pleural, peritoneal, pericardial and mesothelial cells and is highly expressed in 30% of OCs are highly expressed. Due to the low non-specific toxicity of MSLN, researchers have identified it as a potential target for the treatment of OC [87,86,91]. Recently, Zhang et al.(88)developed MSLNCAR containing MSLN-scFv, CD8 transmembrane structural domain, CD28 and TNFRSF9 costimulatory structural domains, and activation structural domain CD247. In vitro experiments showed that MSLN-CAR was able to specifically kill tumor cells in OC cell lines and release cytokines.

Muc16, also known as cancer antigen 125 (CA125), is characterized by overexpression of MUC16 in more than 80% of OCs and serves as an important indicator for early diagnosis of OC [89]. Studies have shown that MUC16-CAR-T cells specifically kill MUC16+ OC cells in vitro. In mouse tumor models, intravenous or intraperitoneal injection of MUC16-CAR-T cells also delayed the progression of OC and even completely cleared tumors. Similarly, Ep-CAM is overexpressed in OC cells and can be targeted by CAR-T cells [90]. Fu et al. [81] developed a third-generation Ep-CAM- CAR containing an EpCAM- SCFV fragment, a CD8 transmembrane structural domain, stimulatory structural domains of CD28 and TNFRSF9, and a CD247 activation structural domain. The CAR was then transferred into T cells using lentivirus. Finally, it was also well demonstrated by in vivo and in vitro experiments that Ep-CAM-CAR-T cells could effectively inhibit OC tumor activity.

The use of cell-derived vesicles in cancer therapy

The selection and exploration of potential targets for CAR-T cell immunotherapy in targeting various types of cancers should be guided by the following two prerequisites: (1) proteins targeted by CAR-T cells have been reported for use in certain cancers, such as FAP and GD2; and (2) proteins that are highly expressed on the surface of the cancer cells of interest for treatment and little or no expression in normal tissues.

Mucin-1

An abnormal glycoform large-size protein, mucin-1 of the cell membrane (MUC1), capable of overexpression is present in a large number of adenocarcinomas [91,92]. Researchers designed a CAR based on mAb (5E5) that can effectively target the MUC1 glycopeptide to kill pancreatic tumor cells [93,94]. Meanwhile, because IL-4 is closely linked to the pathophysiology and treatment of cancer, the researchers used the IL-4 receptor outer structural domain to modify MUC1CAR-T cells, which allowed CAR-T cells to gain further viability in the treatment of cancer and significantly enhanced resistance to immunosuppressive cytokines, greatly improving the anti-tumor effect [95-97].

GD2

As a tumor-associated carbohydrate surface antigen, the ganglioside GD2, unlike other gangliosides expressed in most normal tissues, is preferentially overexpressed in the vast majority of neuroblastomas, melanomas, retinomas, and Ewing’s sarcomas [98-100]. GD2 not only promotes tumor development by inducing cell proliferation, migration, and anti-apoptosis, but also exhibits immunosuppressive properties when released into the circulation, it can impede T-cell activation and dendritic cell maturation [101103]. The researchers prepared Epstein-Barr virus (EBV) specific CAR T-cells targeting GD2 and delivered them to eight patients with neuroblastoma, showing necrosis and regression of tumor cells in four patients [104-106].

NKG2D

Natural killer group 2D (NKG2D), a key regulator of effector immune cell function, is able to activate a potent cytotoxic pathway against its target cells even in the presence of normal concentrations of inhibitory MHC-I molecules [107]. Since the ligand of NKG2D is overexpressed on tumor cells, the NKG2DCAR-T constructed using its ligand as a target has functional significance in innate and adaptive immunity against infected cells and malignant cells.

B7H3

B7H3 (CD276), an immune checkpoint molecule, enhances immune escape and metastasis of tumors and is associated with poor prognosis [108,109]. Recently, investigators have constructed B7H3-CAR-T cells and used them in preclinical models of various solid tumors, and the results of the study have demonstrated their potent anti-tumor effects, such as neuroblastoma, childhood malignancies and ovarian cancer [110,111].

GPC3

Glypian-3 (GPC3) belongs to the Glypian family of proteoglycans and is attached to the cell surface via glycosylphosphatidylinositol (GPI) anchors [112]. GPC3 is essential for cell differentiation, proliferation and migration and it has been shown that GPC3 is abundantly expressed in hepatocellular carcinoma HCC and is not expressed in normal tissues, so it can be used as a specific target for hepatocellular carcinoma [113]. The investigators infused the prepared GPC3-CAR-T cells back into 13 patients and studied their safety and preliminary efficacy [114116]. The results showed that GPC3-CAR-T cells were effective and safe for the treatment of GPC3-positive HCC patients, and when combined with lymphodepleting conditioning, GPC3- CAR-T cells showed strong anti-cancer potential.

Conclusion

CAR-T cell therapy can cause a range of immunopathological reactions due to its “targeted, off-tumor” toxicity, including fever, hypotension, hypoxia, neurotoxicity, skin toxicity, gastrointestinal toxicity, and multi-organ damage. The immunopathological reactions may include fever, hypotension, hypoxia, neurotoxicity, skin toxicity, gastrointestinal toxicity and multi-organ damage.

In severe cases such as cytokine release syndrome or tumor lysis syndrome, it may even cause death of the patient. Coupled with its high cost and complicated operation, the clinical application of CAR-T cell immunotherapy has been limited. Therefore, in addition to enhancing antitumor efficacy, methods to eliminate complications of CAR-T cell immunotherapy, reduce its cost, and optimize operational steps are also a major challenge for the clinical application of CAR-T cell immunotherapy. In summary, despite the many obstacles, with the study and in-depth exploration of precision medicine research, more and more researchers are joining the research on CAR-T cell immunotherapy, and CAR-T cell immunotherapy is likely to elevate the treatment of various types of cancer to a new level in the future and will play a more important role in cancer immunotherapy.

Disclosure

Conflict of Interest: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Funding: There is no fund.

Acknowledgments: Thank all authors.

References

  1. Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, et al. (2002) Cancer Regression and Autoimmunity in Patients after Clonal Repopulation with Antitumor Lymphocytes. Science 298: 850-4.
  2. Majzner RG, Mackall CL (2018) Tumor Antigen Escape from Car T-Cell Therapy. Cancer Discov  8: 1219-26.
  3. Rosenberg SA, Yang JC, Sherry RM, Kammula US, Hughes MS, Phan GQ, et al. (2011) Durable Complete Responses in Heavily Pretreated Patients with Metastatic Melanoma Using T-Cell Transfer Immunotherapy. Clin Cancer Res 17: 4550-7.
  4. Goff SL, Dudley ME, Citrin DE, Somerville RP, Wunderlich JR, Danforth DN, et al. (2016) Randomized, Prospective Evaluation Comparing Intensity of Lymphodepletion before Adoptive Transfer of TumorInfiltrating Lymphocytes for Patients with Metastatic Melanoma. J Clin Oncol  34: 2389-97.
  5. Miliotou AN, Papadopoulou LC (2018) Car T-Cell Therapy: A New Era in Cancer Immunotherapy. Curr Pharm Biotechnol 19: 5-18.
  6. Wagner J, Wickman E, DeRenzo C, Gottschalk S (2020) Car T Cell Therapy for Solid Tumors: Bright Future or Dark Reality? Mol Ther 28: 2320-39.
  7. Ma S, Li X, Wang X, Cheng L, Li Z, Zhang C, et al. (2019) Current Progress in Car-T Cell Therapy for Solid Tumors. Int J Biol Sci 15: 2548-60.
  8. Sadelain M, Brentjens R, Riviere I (2013) The Basic Principles of Chimeric Antigen Receptor Design. Cancer Discov 3: 388-98.
  9. Geldres C, Savoldo B, Dotti G (2016) Chimeric Antigen ReceptorRedirected T Cells Return to the Bench. Semin Immunol 28: 3-9.
  10. Dotti G, Gottschalk S, Savoldo B, Brenner MK (2014) Design and Development of Therapies Using Chimeric Antigen ReceptorExpressing T Cells. Immunol Rev 257: 107-26.
  11. Feins S, Kong W, Williams EF, Milone MC, Fraietta JA (2019) An Introduction to Chimeric Antigen Receptor (Car) T-Cell Immunotherapy for Human Cancer. Am J Hematol 94: S3-S9.
  12. June CH, O’Connor RS, Kawalekar OU, Ghassemi S, Milone MC (2018) Car T Cell Immunotherapy for Human Cancer. Science 359: 1361-5.
  13. Zhao J, Lin Q, Song Y, Liu D (2018) Universal Cars, Universal T Cells, and Universal Car T Cells. J Hematol Oncol 11: 132.
  14. Yu S, Yi M, Qin S, Wu K (2019) Next Generation Chimeric Antigen Receptor T Cells: Safety Strategies to Overcome Toxicity. Mol Cancer 18: 125.
  15. Feldmann A, Arndt C, Koristka S, Berndt N, Bergmann R, Bachmann MP (2019) Conventional Cars Versus Modular Cars. Cancer Immunol Immunother 68: 1713-9.
  16. Zhang H, Zhu S, Deng W, Li R, Zhou H, Xiong H (2022) The Landscape of Chimeric Antigen Receptor T Cell Therapy in Breast Cancer: Perspectives and Outlook. Front Immunol 13: 887471.
  17. Safarzadeh Kozani P, Naseri A, Mirarefin SMJ, Salem F, Nikbakht M, Evazi Bakhshi S, et al. (2022) Nanobody-Based Car-T Cells for Cancer Immunotherapy. Biomark Res 10: 24.
  18. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. (2014) Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N Engl J Med 371: 1507-17.
  19. Chimeric Antigen Receptor-Modified T Cells in Chronic Lymphoid Leukemia; Chimeric Antigen Receptor-Modified T Cells for Acute Lymphoid Leukemia; Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. N Engl J Med (2016) 374(10):998.
  20. Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, et al. (2015) Chimeric Antigen Receptor T Cells Persist and Induce Sustained Remissions in Relapsed Refractory Chronic Lymphocytic Leukemia. Sci Transl Med 7: 303ra139.
  21. Kakarla S, Gottschalk S (2014) Car T Cells for Solid Tumors: Armed and Ready to Go? Cancer J 20: 151-5.
  22. Brudno JN, Kochenderfer JN (2019) Recent Advances in Car T-Cell Toxicity: Mechanisms, Manifestations and Management. Blood Rev 34: 45-55.
  23. O’Leary K (2023) Base-Edited Car T Cells for Pediatric Leukemia. Nat Med.
  24. Lacan C, Caron J, Tarantino N, Fouquet B, Cherai M, Parizot C, et al. (2023) Car-T Cell Therapy for Central Nervous System Lymphomas: Blood and Cerebrospinal Fluid Biology, and Outcomes. Haematologica.
  25. Johnson DE, Burtness B, Leemans CR, Lui VWY, Bauman JE, Grandis JR (2020) Head and Neck Squamous Cell Carcinoma. Nat Rev Dis Primers 6: 92.
  26. Jemal A, Siegel R, Ward E, Murray T, Xu J, Thun MJ (2007) Cancer Statistics, 2007. CA Cancer J Clin 57: 43-66.
  27. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A (2018) Global Cancer Statistics 2018: Globocan Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 68: 394-424.
  28. Cramer JD, Burtness B, Ferris RL (2019) Immunotherapy for Head and Neck Cancer: Recent Advances and Future Directions. Oral Oncol 99: 104460.
  29. Haist C, Schulte E, Bartels N, Bister A, Poschinski Z, Ibach TC, et al. (2021) Cd44v6-Targeted Car T-Cells Specifically Eliminate Cd44 Isoform 6 Expressing Head/Neck Squamous Cell Carcinoma Cells. Oral Oncol 116: 105259.
  30. Papa S, Adami A, Metoudi M, Beatson R, George MS, Achkova D, et al. (2023) Intratumoral Pan-Erbb Targeted Car-T for Head and Neck Squamous Cell Carcinoma: Interim Analysis of the T4 Immunotherapy Study. J Immunother Cancer: 11.
  31. van Schalkwyk MC, Papa SE, Jeannon JP, Guerrero Urbano T, Spicer JF, et al. (2013) Design of a Phase I Clinical Trial to Evaluate Intratumoral Delivery of Erbb-Targeted Chimeric Antigen Receptor T-Cells in Locally Advanced or Recurrent Head and Neck Cancer. Hum Gene Ther Clin Dev 24: 134-42.
  32. Dong YH, Ding YM, Guo W, Huang JW, Yang Z, et al. (2018) The Functional Verification of Egfr-Car T-Cells Targeted to Hypopharyngeal Squamous Cell Carcinoma. Onco Targets Ther 11: 7053-9.
  33. Mei Z, Zhang K, Lam AK, Huang J, Qiu F, Qiao B, et al. (2020) Muc1 as a Target for Car-T Therapy in Head and Neck Squamous Cell Carinoma. Cancer Med 9: 640-52.
  34. Scribner JA, Brown JG, Son T, Chiechi M, Li P, Sharma S, et al. (2020) Preclinical Development of Mgc018, a Duocarmycin-Based AntibodyDrug Conjugate Targeting B7-H3 for Solid Cancer. Mol Cancer Ther 19: 2235-44.
  35. Rosewell Shaw A, Porter CE, Watanabe N, Tanoue K, Sikora A, Gottschalk S, et al. (2017) Adenovirotherapy Delivering Cytokine and Checkpoint Inhibitor Augments Car T Cells against Metastatic Head and Neck Cancer. Mol Ther 25: 2440-51.
  36. Park YP, Jin L, Bennett KB, Wang D, Fredenburg KM, et al. (2018) Cd70 as a Target for Chimeric Antigen Receptor T Cells in Head and Neck Squamous Cell Carcinoma. Oral Oncol 78: 145-50.
  37. Murad JM, Baumeister SH, Werner L, Daley H, Trebeden-Negre H, Reder J, et al. (2018) Manufacturing Development and Clinical Production of Nkg2d Chimeric Antigen Receptor-Expressing T Cells for Autologous Adoptive Cell Therapy. Cytotherapy 20: 952-63.
  38. Zuo BL, Yan B, Zheng GX, Xi WJ, Zhang X, Yang AG, et al. (2018) Targeting and Suppression of Her3-Positive Breast Cancer by T Lymphocytes Expressing a Heregulin Chimeric Antigen Receptor. Cancer Immunol Immunother 67: 393-401.
  39. Pollock NI, Wang L, Wallweber G, Gooding WE, Huang W, Chenna A, et al. (2015) Increased Expression of Her2, Her3, and Her2:Her3 Heterodimers in Hpv-Positive Hnscc Using a Novel Proximity-Based Assay: Implications for Targeted Therapies. Clin Cancer Res 21:4597606.
  40. Brown CE, Alizadeh D, Starr R, Weng L, Wagner JR, Naranjo A, et al. (2016) Regression of Glioblastoma after Chimeric Antigen Receptor T-Cell Therapy. N Engl J Med 375: 2561-9.
  41. Kakarla S, Chow KK, Mata M, Shaffer DR, Song XT, Wu MF, et al. (2013) Antitumor Effects of Chimeric Receptor Engineered Human T Cells Directed to Tumor Stroma. Mol Ther 21: 1611-20.
  42. Gulati P, Ruhl J, Kannan A, Pircher M, Schuberth P, Nytko KJ, et al.(2018) Aberrant Lck Signal Via Cd28 Costimulation Augments AntigenSpecific Functionality and Tumor Control by Redirected T Cells with Pd-1 Blockade in Humanized Mice. Clin Cancer Res 24: 3981-93.
  43. Schuberth PC, Hagedorn C, Jensen SM, Gulati P, van den Broek M, Mischo A, et al. (2013) Treatment of Malignant Pleural Mesothelioma by Fibroblast Activation Protein-Specific Re-Directed T Cells. J Transl Med 11: 187.
  44. Sankar S (2022) New Insights into Car T-Cell Therapy for Recurrent Head and Neck Squamous Cell Carcinoma. Oral Oncol 134: 106137.
  45. Wang HQ, Fu R, Man QW, Yang G, Liu B, Bu LL (2023) Advances in Car-T Cell Therapy in Head and Neck Squamous Cell Carcinoma. J Clin Med: 12.
  46. Pollock NI, Grandis JR (2015) Her2 as a Therapeutic Target in Head and Neck Squamous Cell Carcinoma. Clin Cancer Res 21: 526-33.
  47. Reynolds KL, Bedard PL, Lee SH, Lin CC, Tabernero J, Alsina M, et al. (2017) A Phase I Open-Label Dose-Escalation Study of the Anti-Her3 Monoclonal Antibody Ljm716 in Patients with Advanced Squamous Cell Carcinoma of the Esophagus or Head and Neck and Her2Overexpressing Breast or Gastric Cancer. BMC Cancer 17: 646.
  48. Warren EAK, Anil J, Castro PD, Kemnade J, Suzuki M, et al. (2021) Human Epidermal Growth Factor Receptor 2 Expression in Head and Neck Squamous Cell Carcinoma: Variation within and across Primary Tumor Sites, and Implications for Antigen-Specific Immunotherapy. Head Neck 43: 1983-94.
  49. Job S, Reynies A, Heller B, Weiss A, Guerin E, Macabre C, et al. (2019) Preferential Response of Basal-Like Head and Neck Squamous Cell Carcinoma Cell Lines to Egfr-Targeted Therapy Depending on EregDriven Oncogenic Addiction. Cancers (Basel): 11.
  50. Uribe ML, Marrocco I, Yarden Y (2021) Egfr in Cancer: Signaling Mechanisms, Drugs, and Acquired Resistance. Cancers (Basel): 13.
  51. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, et al. (2011) Identification of Human Triple-Negative Breast Cancer Subtypes and Preclinical Models for Selection of Targeted Therapies. J Clin Invest 121: 2750-67.
  52. Jiang YZ, Ma D, Suo C, Shi J, Xue M, Hu X, et al. (2019) Genomic and Transcriptomic Landscape of Triple-Negative Breast Cancers: Subtypes and Treatment Strategies. Cancer Cell 35: 428-40 e5.
  53. Dagogo-Jack I, Shaw AT (2018) Tumour Heterogeneity and Resistance to Cancer Therapies. Nat Rev Clin Oncol 15: 81-94.
  54. Tchou J, Zhao Y, Levine BL, Zhang PJ, Davis MM, Melenhorst JJ, et al. (2017) Safety and Efficacy of Intratumoral Injections of Chimeric Antigen Receptor (Car) T Cells in Metastatic Breast Cancer. Cancer Immunol Res 5: 1152-61.
  55. Ronchi A, Pagliuca F, Zito Marino F, Accardo M, Cozzolino I, Franco R (2021) Current and Potential Immunohistochemical Biomarkers for Prognosis and Therapeutic Stratification of Breast Carcinoma. Semin Cancer Biol 72: 114-22.
  56. Bozorgi A, Bozorgi M, Khazaei M (2022) Immunotherapy and Immunoengineering for Breast Cancer; a Comprehensive Insight into Car-T Cell Therapy Advancements, Challenges and Prospects. Cell Oncol (Dordr) 45: 755-77.
  57. Scholl S, Beuzeboc P, Pouillart P (2001) Targeting Her2 in Other Tumor Types. Ann Oncol 12: S81-7.
  58. Gerson JN, Skariah S, Denlinger CS, Astsaturov I (2017) Perspectives of Her2-Targeting in Gastric and Esophageal Cancer. Expert Opin Investig Drugs 26: 531-40.
  59. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, et al. (2001) Gene Expression Patterns of Breast Carcinomas Distinguish Tumor Subclasses with Clinical Implications. Proc Natl Acad Sci U S A 98: 10869-74.
  60. Li H, Yuan W, Bin S, Wu G, Li P, Liu M, et al. (2022) Erratum: Overcome Trastuzumab Resistance of Breast Cancer Using Anti-Her2 Chimeric Antigen Receptor T Cells and Pd1 Blockade. Am J Cancer Res 12: 2891-2.
  61. Seyedmirzaei H, Keshavarz-Fathi M, Razi S, Gity M, Rezaei N (2021) Recent Progress in Immunotherapy of Breast Cancer Targeting the Human Epidermal Growth Factor Receptor 2 (Her2). J Oncol Pharm Pract 27: 1235-44.
  62. Sun M, Shi H, Liu C, Liu J, Liu X, Sun Y (2014) Construction and Evaluation of a Novel Humanized Her2-Specific Chimeric Receptor. Breast Cancer Res 16: R61.
  63. Blanch-Lombarte O, Galvez C, Revollo B, Jimenez-Moyano E, Llibre JM, et al. (2019) Enhancement of Antiviral Cd8(+) T-Cell Responses and Complete Remission of Metastatic Melanoma in an Hiv-1-Infected Subject Treated with Pembrolizumab. J Clin Med: 8.
  64. Seif M, Einsele H, Loffler J (2019) Car T Cells Beyond Cancer: Hope for Immunomodulatory Therapy of Infectious Diseases. Front Immunol 10: 2711.
  65. Galvez C, Urrea V, Garcia-Guerrero MDC, Bernal S, Benet S, Mothe B, et al. (2022) Altered T-Cell Subset Distribution in the Viral Reservoir in Hiv-1-Infected Individuals with Extremely Low Proviral DNA (Lovirets). J Intern Med 292: 308-20.
  66. Salgado M, Kwon M, Galvez C, Badiola J, Nijhuis M, Bandera A, et al. (2018) Mechanisms That Contribute to a Profound Reduction of the Hiv-1 Reservoir after Allogeneic Stem Cell Transplant. Ann Intern Med 169: 674-83.
  67. Zhen A, Kamata M, Rezek V, Rick J, Levin B, Kasparian S, et al. (2015) Hiv-Specific Immunity Derived from Chimeric Antigen ReceptorEngineered Stem Cells. Mol Ther 23: 1358-67.
  68. York J, Gowrishankar K, Micklethwaite K, Palmer S, Cunningham AL, et al. (2022) Evolving Strategies to Eliminate the Cd4 T Cells Hiv Viral Reservoir Via Car T Cell Immunotherapy. Front Immunol 13: 873701.
  69. Hale M, Mesojednik T, Romano Ibarra GS, Sahni J, Bernard A, et al. (2017) Engineering Hiv-Resistant, Anti-Hiv Chimeric Antigen Receptor T Cells. Mol Ther 25: 570-9.
  70. Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM, et al. (2012) Decade-Long Safety and Function of Retroviral-Modified Chimeric Antigen Receptor T Cells. Sci Transl Med  4: 132ra53.
  71. Liu B, Zhang W, Xia B, Jing S, Du Y, Zou F, et al. (2021) Broadly Neutralizing Antibody-Derived Car T Cells Reduce Viral Reservoir in Individuals Infected with Hiv-1. J Clin Invest: 131.
  72. Okla K, Surowka J, Fraszczak K, Czerwonka A, Kalawaj K, Wawruszak A, et al. (2018) Assessment of the Clinicopathological Relevance of Mesothelin Level in Plasma, Peritoneal Fluid, and Tumor Tissue of Epithelial Ovarian Cancer Patients. Tumour Biol 40: 1010428318804937.
  73. Chardin L, Leary A (2021) Immunotherapy in Ovarian Cancer: Thinking Beyond Pd-1/Pd-L1. Front Oncol 11: 795547.
  74. Guo C, Dong E, Lai Q, Zhou S, Zhang G, Wu M, et al. (2020) Effective Antitumor Activity of 5t4-Specific Car-T Cells against Ovarian Cancer Cells in Vitro and Xenotransplanted Tumors in Vivo. MedComm 1: 338-50.
  75. Klapdor R, Wang S, Morgan M, Dork T, Hacker U, Hillemanns P, et al. (2019) Characterization of a Novel Third-Generation Anti-Cd24-Car against Ovarian Cancer. Int J Mol Sci: 20.
  76. Chang KL, Lee MY, Chao WR, Han CP (2016) The Status of Her2 Amplification and Kras Mutations in Mucinous Ovarian Carcinoma. Hum Genomics 10: 40.
  77. Cherkassky L, Morello A, Villena-Vargas J, Feng Y, Dimitrov DS, et al. (2016) Human Car T Cells with Cell-Intrinsic Pd-1 Checkpoint Blockade Resist Tumor-Mediated Inhibition. J Clin Invest 126: 313044.
  78. Moon EK, Ranganathan R, Eruslanov E, Kim S, Newick K, O’Brien S, et al. (2016) Blockade of Programmed Death 1 Augments the Ability of Human T Cells Engineered to Target Ny-Eso-1 to Control Tumor Growth after Adoptive Transfer. Clin Cancer Res 22: 436-47.
  79. Rafiq S, Yeku OO, Jackson HJ, Purdon TJ, van Leeuwen DG, et al. (2018) Targeted Delivery of a Pd-1-Blocking Scfv by Car-T Cells Enhances Anti-Tumor Efficacy in Vivo. Nat Biotechnol 36: 847-56.
  80. Fang J, Ding N, Guo X, Sun Y, Zhang Z, Xie B, et al. (2021) Alphapd1-Mesocar-T Cells Partially Inhibit the Growth of Advanced/Refractory Ovarian Cancer in a Patient Along with Daily Apatinib. J Immunother Cancer: 9.
  81. Fu J, Shang Y, Qian Z, Hou J, Yan F, Liu G, et al. (2021) 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 83: 241-7.
  82. Ramos CA, Rouce R, Robertson CS, Reyna A, Narala N, Vyas G, et al. (2018) In Vivo Fate and Activity of Second- Versus Third-Generation Cd19-Specific Car-T Cells in B Cell Non-Hodgkin’s Lymphomas. Mol Ther 26: 2727-37.
  83. Leong L, Tan HL, Cua S, Yong KSM, Chen Q, Choo A (2020) Preclinical Activity of Embryonic Annexin A2-Specific Chimeric Antigen Receptor T Cells against Ovarian Cancer. Int J Mol Sci: 21.
  84. Kurosaki A, Hasegawa K, Kato T, Abe K, Hanaoka T, Miyara A, et al. (2016) Serum Folate Receptor Alpha as a Biomarker for Ovarian Cancer: Implications for Diagnosis, Prognosis and Predicting Its Local Tumor Expression. Int J Cancer 138: 1994-2002.
  85. Zuo S, Wen Y, Panha H, Dai G, Wang L, Ren X, et al. (2017) Modification of Cytokine-Induced Killer Cells with Folate Receptor Alpha (Fralpha)-Specific Chimeric Antigen Receptors Enhances Their Antitumor Immunity toward Fralpha-Positive Ovarian Cancers. Mol Immunol 85: 293-304.
  86. Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, et al. (2014) Mesothelin-Specific Chimeric Antigen Receptor MrnaEngineered T Cells Induce Anti-Tumor Activity in Solid Malignancies. Cancer Immunol Res 2: 112-20.
  87. Adusumilli PS, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J, et al. (2014) Regional Delivery of Mesothelin-Targeted Car T Cell Therapy Generates Potent and Long-Lasting Cd4-Dependent Tumor Immunity. Sci Transl Med 6: 261ra151.
  88. Zhang Q, Liu G, Liu J, Yang M, Fu J, Liu G, et al. (2021) The Antitumor Capacity of Mesothelin-Car-T Cells in Targeting Solid Tumors in Mice. Mol Ther Oncolytics 20: 556-68.
  89. Rao TD, Tian H, Ma X, Yan X, Thapi S, Schultz N, et al. (2015) Expression of the Carboxy-Terminal Portion of Muc16/Ca125 Induces Transformation and Tumor Invasion. PLoS One 10: e0126633.
  90. Chekmasova AA, Rao TD, Nikhamin Y, Park KJ, Levine DA, Spriggs DR, et al. (2010) Successful Eradication of Established Peritoneal Ovarian Tumors in Scid-Beige Mice Following Adoptive Transfer of T Cells Genetically Targeted to the Muc16 Antigen. Clin Cancer Res 16: 3594-606.
  91. Morello A, Sadelain M, Adusumilli PS (2016) Mesothelin-Targeted Cars: Driving T Cells to Solid Tumors. Cancer Discov 6: 133-46.
  92. Finn OJ, Gantt KR, Lepisto AJ, Pejawar-Gaddy S, Xue J, Beatty PL (2011) Importance of Muc1 and Spontaneous Mouse Tumor Models for Understanding the Immunobiology of Human Adenocarcinomas. Immunol Res 50: 261-8.
  93. Tarp MA, Sorensen AL, Mandel U, Paulsen H, Burchell J, TaylorPapadimitriou J, et al. (2007) Identification of a Novel Cancer-Specific Immunodominant Glycopeptide Epitope in the Muc1 Tandem Repeat. Glycobiology 17: 197-209.
  94. Vickers NJ (2017) Animal Communication: When I’m Calling You, Will You Answer Too? Curr Biol 27: R713-R5.
  95. Posey AD, Jr., Schwab RD, Boesteanu AC, Steentoft C, Mandel U, Engels B, et al. (2016) Engineered Car T Cells Targeting the CancerAssociated Tn-Glycoform of the Membrane Mucin Muc1 Control Adenocarcinoma. Immunity 44: 1444-54.
  96. Wilkie S, Burbridge SE, Chiapero-Stanke L, Pereira AC, Cleary S, van der Stegen SJ, et al. (2010) Selective Expansion of Chimeric Antigen Receptor-Targeted T-Cells with Potent Effector Function Using Interleukin-4. J Biol Chem 285: 25538-44.
  97. Ramachandran M, Dimberg A, Essand M (2017) The Cancer-Immunity Cycle as Rational Design for Synthetic Cancer Drugs: Novel Dc Vaccines and Car T-Cells. Semin Cancer Biol 45: 23-35.
  98. You F, Jiang L, Zhang B, Lu Q, Zhou Q, Liao X, et al. (2016) Phase 1 Clinical Trial Demonstrated That Muc1 Positive Metastatic Seminal Vesicle Cancer Can Be Effectively Eradicated by Modified Anti-Muc1 Chimeric Antigen Receptor Transduced T Cells. Sci China Life Sci 59: 386-97.
  99. Nazha B, Inal C, Owonikoko TK (2020) Disialoganglioside Gd2 Expression in Solid Tumors and Role as a Target for Cancer Therapy. Front Oncol 10: 1000.
  100. Suzuki M, Cheung NK (2015) Disialoganglioside Gd2 as a Therapeutic Target for Human Diseases. Expert Opin Ther Targets 19: 349-62.
  101. 101.  Berois N, Pittini A, Osinaga E (2022) Targeting Tumor Glycans for Cancer Therapy: Successes, Limitations, and Perspectives. Cancers (Basel): 14.
  102. 102.  Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, et al. (2008) Virus-Specific T Cells Engineered to Coexpress TumorSpecific Receptors: Persistence and Antitumor Activity in Individuals with Neuroblastoma. Nat Med 14: 1264-70.
  103. 103.  Louis CU, Savoldo B, Dotti G, Pule M, Yvon E, Myers GD, et al. (2011) Antitumor Activity and Long-Term Fate of Chimeric Antigen ReceptorPositive T Cells in Patients with Neuroblastoma. Blood 118: 6050-6.
  104. 104.  Bocca P, Di Carlo E, Caruana I, Emionite L, Cilli M, De Angelis B, et al. (2017) Bevacizumab-Mediated Tumor Vasculature Remodelling Improves Tumor Infiltration and Antitumor Efficacy of Gd2-Car T Cells in a Human Neuroblastoma Preclinical Model. Oncoimmunology 7: e1378843.
  105. 105.  Yu L, Huang L, Lin D, Lai X, Wu L, Liao X, et al. (2022) Gd2-Specific Chimeric Antigen Receptor-Modified T Cells for the Treatment of Refractory and/or Recurrent Neuroblastoma in Pediatric Patients. J Cancer Res Clin Oncol 148: 2643-52.
  106. 106.  Stroncek DF, Ren J, Lee DW, Tran M, Frodigh SE, Sabatino M, et al. (2016) Myeloid Cells in Peripheral Blood Mononuclear Cell Concentrates Inhibit the Expansion of Chimeric Antigen Receptor T Cells. Cytotherapy 18: 893-901.
  107. 107.  Majzner RG, Ramakrishna S, Yeom KW, Patel S, Chinnasamy H, Schultz LM, et al. (2022) Gd2-Car T Cell Therapy for H3k27m-Mutated Diffuse Midline Gliomas. Nature 603: 934-41.
  108. 108.  Prajapati K, Perez C, Rojas LBP, Burke B, Guevara-Patino JA (2018) Functions of Nkg2d in Cd8(+) T Cells: An Opportunity for Immunotherapy. Cell Mol Immunol 15: 470-9.
  109. 109.  Picarda E, Ohaegbulam KC, Zang X (2016) Molecular Pathways: Targeting B7-H3 (Cd276) for Human Cancer Immunotherapy. Clin Cancer Res 22: 3425-31.
  110. 110.  Ye Z, Zheng Z, Li X, Zhu Y, Zhong Z, Peng L, et al. (2016) B7-H3 Overexpression Predicts Poor Survival of Cancer Patients: A MetaAnalysis. Cell Physiol Biochem 39: 1568-80.
  111. 111.  Majzner RG, Theruvath JL, Nellan A, Heitzeneder S, Cui Y, Mount CW, et al. (2019) Car T Cells Targeting B7-H3, a Pan-Cancer Antigen, Demonstrate Potent Preclinical Activity against Pediatric Solid Tumors and Brain Tumors. Clin Cancer Res 25: 2560-74.
  112. 112.  Du H, Hirabayashi K, Ahn S, Kren NP, Montgomery SA, Wang X, et al. (2019) Antitumor Responses in the Absence of Toxicity in Solid Tumors by Targeting B7-H3 Via Chimeric Antigen Receptor T Cells. Cancer Cell 35: 221-37 e8.
  113. 113.  Filmus J, Selleck SB (2001) Glypicans: Proteoglycans with a Surprise. J Clin Invest 108: 497-501.
  114. 114.  Baumhoer D, Tornillo L, Stadlmann S, Roncalli M, Diamantis EK, Terracciano LM (2008) Glypican 3 Expression in Human Nonneoplastic, Preneoplastic, and Neoplastic Tissues: A Tissue Microarray Analysis of 4,387 Tissue Samples. Am J Clin Pathol 129: 899-906.
  115. 115.  Shi D, Shi Y, Kaseb AO, Qi X, Zhang Y, Chi J, et al. (2020) Chimeric Antigen Receptor-Glypican-3 T-Cell Therapy for AdvancedHepatocellular Carcinoma: Results of Phase I Trials. Clin Cancer Res 26: 3979-89.
  116. 116.  Pang N, Shi J, Qin L, Chen A, Tang Y, Yang H, et al. (2021) Il-7 and Ccl19-Secreting Car-T Cell Therapy for Tumors with Positive Glypican-3 or Mesothelin. J Hematol Oncol 14: 118.

© by the Authors & Gavin Publishers. This is an Open Access Journal Article Published Under Attribution-Share Alike CC BY-SA: Creative Commons Attribution-Share Alike 4.0 International License. With this license, readers can share, distribute, download, even commercially, as long as the original source is properly cited. Read More.

Journal of Oncology Research and Therapy

cara menggunakan pola slot mahjongrtp tertinggi hari inislot mahjong ways 1pola gacor olympus hari inipola gacor starlight princessslot mahjong ways 2strategi olympustrik mahjong ways 2trik olympus hari inirtp koi gatertp pragmatic tertinggicheat jackpot mahjongpg soft link gamertp jackpotelemen sakti mahjongpola maxwin mahjongslot olympus mudah mainrtp live starlightrumus slot mahjongmahjong scatter hitamslot pragmaticjam gacor mahjongpola gacor mahjongstrategi maxwin olympusslot jamin menangrtp slot gacorscatter wild banditopola slot mahjongstrategi maxwin sweet bonanzartp slot terakuratkejutan scatter hitamslot88 resmimaxwin olympuspola mahjong pgsoftretas mahjong waystrik mahjongtrik slot olympusewallet modal recehpanduan pemula slotpg soft primadona slottercheat mahjong androidtips dewa slot mahjongslot demo mahjonghujan scatter olympusrtp caishen winsrtp sweet bonanzamahjong vs qilinmaxwin x5000 starlight princessmahjong wins x1000rtp baru wild scatterpg soft trik maxwinamantotorm1131