Logo-apb
Advanced pharmaceutical bulletin. 12(3):476-489. doi: 10.34172/apb.2022.051

Review Article

CAR T Cells: Cancer Cell Surface Receptors Are the Target for Cancer Therapy

Behrouz Shademan 1ORCID logo, Vahidreza Karamad 1, Alireza Nourazarian 2, Cigir Biray Avcı 1, *ORCID logo
1Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey.
2Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
*Corresponding Author: Cigir Biray Avci, Tel: +90232 390 2262, Email: cigir.biray@ege.edu.tr

Abstract

Immunotherapy has become a prominent strategy for the treatment of cancer. A method that improves the immune system’s ability to attack a tumor (Enhances antigen binding). Targeted killing of malignant cells by adoptive transfer of chimeric antigen receptor (CAR) T cells is a promising immunotherapy technique in the treatment of cancers. For this purpose, the patient’s immune cells, with genetic engineering aid, are loaded with chimeric receptors that have particular antigen binding and activate cytotoxic T lymphocytes. That increases the effectiveness of immune cells and destroying cancer cells. This review discusses the basic structure and function of CAR-T cells and how antigenic targets are identified to treat different cancers and address the disadvantages of this treatment for cancer.

Keywords: CAR-T cell, Immunotherapy, Surface receptors, Tumor microenvironment

Copyright

©2022 The Authors.
This is an Open Access article distributed under the terms of the Creative Commons Attribution (CC BY), which permits unrestricted use, distribution, and reproduction in any medium, as long as the original authors and source are cited. No permission is required from the authors or the publishers.


Introduction

Cancer is a multi-stage process in a cell for some causes, that begins with DNA damage, unregulated cell proliferation, and apoptosis. Statistics have shown that cancer is the second most severe and deadly disease. 1 In addition, cancers have the potential to develop resistance to traditional therapies. 2,3 Therefore It is essential to develop treatment alternatives. However, gene therapy, radiotherapy, chemotherapy, etc. are used to treat cancer. Immunotherapy has recently become the priority of researchers. Strategies that stimulate the immune system to kill tumor cells. Zelig Eshharand and colleagues have developed a hetero-dimer monoclonal antibody for the first time. If these antibodies are transferred to T cells, tumor cells can be detected and killed. In 2008, Malcolm Brenner and Houston made the first successful phase in the therapeutic application of CAR-T cells. 4 CAR-T cells consist of the target receptor segment (ScFv single-stranded variable fragment attached to the hinge), the membrane-permeable region, and the intracellular region. 5 The ScFv extracellular antigen recognition fragment was formed of a monoclonal antibody (mAb). That targets specific antigens but can contain ligands that bind to tumor cell surface antigens. 6,7 Various hinges have been found. These hinges can be short or long. Studies have shown that the hinge is a crucial structure for engineered cells and can significantly boost the efficiency of these cells. 8 There are structural variations in the intracellular region. Based on differences in intracellular messenger regions, CAR-T cells are categorized into the first, second, third, and fourth generations (). The first generation includes the cytoplasmic domain of CD3 ζ, which can initiate T-cell activation. Since the proliferation of T cells is low, they are less effective. Defects of the first generation have been overcome in the second and third generations due to the diversity of co-stimulatory molecules (CMs). This generation is called TRUCK (T cells redirected to universal cytokine killing). 9 TRUCK is a method of generating a particular CAR-T cell that stimulates an innate immune response to the destruction of tumor cells. These CAR-T cells have an engineered product, including IL-12. 10

apb-12-476-g001
Figure 1. The CAR-T cells are made up of three generations. The first generation of CAR-T cells is composed of scFv and CD3 immunoglobulin. Stimulating molecules such as CD28, CD134, and CD137 (4-1BB) have been attached to CAR-T cells, producing second and third generations.

The construction of CAR-T cells for therapeutic purposes includes five steps: T cell isolation, T cell activation, T cell engineering, CAR-T cell expansion, cell formulation, and cell storage (). Patients have prescribed cells developed for treatment. 11 The time needed for T cells’ genetic engineering is approximately two weeks. 12,13 The fourth generation of CAR-T cells performs better in TME than other generations due to their high ability to destroy cancer cells. TME decreases the potency of the immune system. These agents are secreted by tumor cells and are used to defend these cells against immune cells. The TRUCK cells are engineered to act better in TME by secreting inflammatory cytokines. 10

apb-12-476-g002
Figure 2. Engineering CAR-T cells for therapeutic purposes. 1) this section begins by obtaining blood from a cancer patient at the hospital. 2) T cells are separated from the blood during this process, and the remaining blood is returned to the body. 3) Following these steps, the T cells are appropriate for genetic modification in the laboratory. Relevant genetic modifications are made in the laboratory on cells isolated from cancer patients. 4) CAR-T cells are culture in the laboratory to produce more cells. 5) The cells produced are eventually transferred to the cancer patient by intravenous injection. 6) Modified T cells now have unique receptors on their surface that are considered chimeric antigen receptors (CAR). These receptors help T cells accurately detect and destroy cancer cells.


Functional mechanism of CAR-T

Cells are designed with specific antigenic receptors that enable modified T cells to acquire more advanced and directed anti-tumor properties. Auxiliary molecules can help T cells better fight cancer cells. Studies have shown that when a signaling molecule called “STAT5” is kept in CAR-T cells, CAR-T cells can stay active longer and help kill the tumor more effectively. 14 But, there is no requirement for auxiliary molecules to recognize cancer cells. 15 CAR-T cells can identify antigens of different structures, including proteins, carbohydrates, and glycolipids. 16 While designed, the genes encoding the CAR structure are transferred to the leukocytes through viral vectors. This intervention should be taken on autologous leukocytes due to the presence of self-identified molecules in T cells. This concern is overcome in patients such as lymphopenia that may not have sufficient lymphocytes to produce genetically engineered donor cells without autoimmune molecules such as class 1 HLA. 17,18 As CAR-T cells bind explicitly to tumor-associated antigens, T cells are triggered by phosphorylation that leads to the release of cytokines and cancer cells’ death. 19 Engineered T cells promote cell death through two main pathways: first, the release of perforin and granules, and second, through the triggering of the Fas or TNF signals. Cytotoxic T cells destroy tumor cells in two ways, so while T Helper cells kill cells by perforin/granzyme. 20,21


Tumor microenvironment

In solid tumors, the TME consists of various cellular and molecular components that reduce the immune response’s strength against the tumor. 22 TME prevents the proper function of CAR-T cells by decreasing penetration of CAR-T cells and compromising anti-tumor activity. 23,24 Cytokines such as TGF-β and IL-10 are developed by TME tumor cells. These agents can suppress a more robust immune response to the tumor and protect cancer cells. T lymphocytes such as TRUCKs are also used in these circumstances. Expressly, CAR-T cells release IL-12 and CAR-T cells, which induce tumor treatment due to micro-environmental tumor disruption. 11 Cancer cells evade the immune system with the aid of TME so that additional auxiliary agents would be required for long-term responses. Decreased immunosuppressive agents such as Tregs and TAM, enhance treatment efficacy with engineered cells. 25 Immunosuppressive factors abound in cancer, leading to over-proliferation of tumor cells and ineffective cancer therapy. 26,27 Drugs including anti-CCL2 (C-C motif chemokine ligand 2), CSF1R (colony stimulating factor 1 receptor) antibodies suppress the TAM microenvironment’s potency surrounding the tumor are under investigation. 28 Another way to improve the strength of CAR-T cells in solid tumors is to inhibit T-cell-mediated immune suppressor signals. 29 Checkpoint inhibitors are therapeutic antibodies that bind to T-cell receptors. These antibodies can be useful as therapeutic strategies by hindering checkpoint molecules (such as CTLA4, PD-1) that cause T cell apoptosis. Results of clinical studies have shown that the expression of checkpoint inhibitors in CAR-T cells can be beneficial. 30,31 A team of researchers fused MSLN-CAR-T cells with adenovirus expressing TNF-α (tumor necrosis factor-alpha) and IL-2 for the treatment of human and animal models that have improved the antitumor effects of engineered cells. 32 Due to the limitations of therapeutic alternatives to cancer therapy and the effectiveness of CAR-T cells in various cancers, the removal of TME components that suppress the immune system’s role in treating different cancers may be a good option.


Surface antigens

Several properties have been introduced to target suitable antigens for therapy, particularly its unique expression in tumor cells and the lack of antigens in normal cells. Abnormal expression of specific antigens in multiple cancers suggests these antigens’ role in the survival, invasion, and metastases of these cells (). Several CAR-T cell experiments targeting cancer cell antigen receptors, as shown in Table 1. Focusing on the high expression of these antigens in tumor cells over normal cells helps develop appropriate T cells with high affinity for cancer cells’ treatment. Several therapeutic target antigens have been studied for this purpose.

apb-12-476-g003
Figure 3. Targeting the right antigens to treat cancer. The expression of unique antigens in tumor cells and the lack of this antigen in normal cells reduces the extra-target toxicity and kills cancer cells.

Table 1. Several experiments have demonstrated that CAR-T cells are characteristic of solid tumors
Solid tumor Antigen Type of CAR-T cell Property conditions of research Reference
Pancreatic cancer Mesothelin meso-CAR-T cell Improved performance; increasing the effect time of engineered cells In vivo 32
Breast cancer HER2 HER2-CAR-T cell Contains CD28 or 4-1BB signal range for functional activity In vitro 33
Glioblastoma HER2 CAR-T Cell that target HER2 and IL-13Rα2 Improving performance with sequential expression; Cytokine secretion - 34
Non-Small Cell Lung Cancer EGFR CAR-T Cell that target EGFR Without severe toxicity, high tolerance for the patient clinical study 35
Neuroblastoma GD2
CD171
CAR-T Cell that target GD2
CE7-CAR T
Increase the anti-tumor effect; With central nervous system toxicity (CNS)
No out-of-target toxicity
In vivo
In vivo
36
37
Liver CEA CEA CAR-T No side effects of grade 3 or 4 clinical study 38
Head and neck cancer ErbB CAR-T Cell that target ErbB(T1E28z) Ability to target multiple antigens clinical study 39
Prostate cancer PSMA PSMA- CAR-T Cell :Negative TGF-β Receptor Increased cytokine secretion, resistance to burnout, long shelf life clinical study 40

HER2: human epidermal growth factor receptor 2; EGFR vIII: Epidermal growth factor receptor variant III; EGFR: Epidermal growth factor receptor; GD2: is a disialoganglioside expressed on tumors; CD171: neural cell adhesion molecule L1; CEA: Carcinoembryonic antigen; ErbB: family of receptor tyrosine kinases;

PSMA: prostate-specific membrane antigen.

Mesothelin (MSLN)

A receptor is mainly present in mesothelial cells but increases in some human cancers such as ovarian, lung, and pancreas. Mesothelin (MSLN) can play a significant function in cell adhesion such that it can be studied in cell invasion. 41 The expression of MSLN can be membrane or cytoplasmic in cancer cells. However, in mesothelial cancer cells, MSLN is also present in the cell membrane. 42 In lung adenocarcinoma, the expression pattern of the MSLN is cytoplasmic and heterogeneous. 43,44 Whereas cytoplasmic expression is more than membrane expression in gastric cancer. 45 Due to high expression in certain cancers and low expression in normal cells, mesothelin may be the right candidate for immune cell engineering therapies. 46 The concern about the MSLN CAR is the soluble MSLN, which could occupy the scFv segment. It should be noticed, however, that the activation of MSLN CAR-T cells depends on the presence of MSLNs in the cells. As a result, the toxicity of this treatment may be determined for normal cells expressing this receptor. 47,48 The presence of soluble mesothelin-related peptide in serum does not change the effectiveness of MSLN CAR-T cells. 48,49 The antigen deficiency of CAR-T cells is the target of the membrane due to the blockage of CAR by other serum proteins. This vulnerability can increase CAR-T cells by interacting with adhesion molecules and other by-products on T cells and tumor cells. 50 Diacylglycerol kinase (DGK) is one of the TME immune cell inhibitors. If this inhibitory factor is removed, it can increase T cells’ proliferation and improve T cells’ effectiveness against cancer cells. 51,52 Koretzky and colleagues have also demonstrated that DGKs elimination increases the activity of CAR-T cells against the tumor and stabilizes engineered cells targeting mesothelin receptors. 53 CAR-T cells against MSLN could be effective against adenocarcinoma of the pancreatic duct. Intravenous (IV) transfer of these cells to animal models. Promising results from tumor shrinkage and even tumor degradation indicate that this receptor is a possible target for therapeutic options. 49 In research performed by Beatty et al, due to high toxicity beyond the tumor to the production of CAR-T cells, mRNA-based methods have been used to reduce toxicity duration. Several injections were required to compensate for the short-term expression of CAR T. Out-of-tumor toxicity can cause adverse side effects through the use of engineered cells, as these target antigens are found at low levels in normal cells. 5 Around 90 % of the folate receptor (FRa) and more than 65 % of the mesothelin receptor are expressed in ovarian cancer. 54,55 Their pattern of expression in natural tissues is essentially non-overlapping. Therefore, Lanitis et al produced CAR-T cells that express all receptors simultaneously and reduce engineered cells’ potential toxicity to normal cells. 56 MSLN-CAR T structures developed for activation of DAP12 killer immunoglobulin show improved in vivo potency compared to second-generation MSLN-CAR T with signaling domains. 57 Adusumilli et al have demonstrated that intracellular administration of engineered immune cells displayed higher antitumor potency than intravenous injection. 48 CAR-T cells are actively halted before the tumor enters the lungs. In summary, local infusion of MSLN-CAR-T cells enhances antigen exposure and boosts engineered cell activity. 42

Epidermal growth factor receptor

An intermembrane protein with tyrosine kinase activity can cause cell differentiation and proliferation. Increased expression of these receptors has been reported in many cancer types. 58,59 Antibodies targeting EGFR include two groups: monoclonal antibodies targeting this receptor. These antibodies inhibit the action of the receptor by binding to the ligand-binding site. And the second category of inhibitors targeting tyrosine kinase activation in these receptors. These inhibitors prevent phosphorylation and prevent the transmission of the message to the cell. These antibodies were used to cure different forms of cancer, such as pancreatic cancer, kidney cancer, and colon cancer. 60,61 In some patients, antibody treatment is not successful due to the various mutations in this receptor. It also seems essential to find the right treatments for these defects. 62 One of the most common oncogenic mutations of EGFR is type VIII EGFR. This antigen tends to have all of the appropriate antigenic properties for CAR-T therapy since it is one of the most frequent EGFR mutations in cancers but has no expression in normal tissues. 63 Glioblastoma (GBM) is the fatal component in adult brain tumors, and GBM treatment does not improve life span. EGFR VIII has a high degree of expression in the GBM cells. The targeting of this receptor by engineered cells to treat this cancer will also benefit. 64 A team of researchers studied the ability of engineered cells to recognize EGFR VIII. With the help of CAR-T cells, they were able to target the EGFR VIII-containing GBM stem cells and identify the stem cells with this mutation. 65 Although they do not respond to cultured normal tissue cells. 66 EGFR-engineered immune cells have a significant anti-inflammatory effect in vitro and in vivo, but controlled and regulated toxicity such as skin, gastrointestinal, respiratory, and hypertension has been documented. 67

Human epidermal growth factor receptor

Glycoprotein is a member of the EGFR family. Overexpression of human epidermal growth factor 2 (HER2) has been shown in several cancers. This increased expression may mean that it is involved in the pathology of cancer. It is over-expressed in the breast and ovarian, and GBM cancers. However, this receptor is not present in normal brain cells and tissues. 68 The elevated level of HER2 in different cancers has made it an appropriate target for CAR T therapy. 69 Patients were treated with HER2 trastuzumab (herceptin) monoclonal antibody targeting the HER2 receptor. Observed in breast cancer treatment, Herceptin, which is also associated with chemotherapy, improves the lifespan of different stages of breast cancer. 70 Haggett and colleagues have developed a CAR bispecific molecule targeting HER2 and IL-13Rα2 antigens in cancer cells. These antigens are found in cancer cells of the glioma. These engineered cells had successive expressions (TanCAR). This consecutive expression increases the function of the engineered cells and stimulates the secretion of cytokines. TanCAR-T cell agents can be used as an effective treatment strategy to regulate tumor development, but the detection of limitations for therapeutic use seems to be necessary. 34 Clinical trials of tyrosine kinase HER2 inhibitors are progressing as low levels of HER2 are successfully identified in some cancers by Herceptin. This vulnerability has been used to treat some of these cancers. 71 Results have indicated that 50% of patients have not undergone successful treatment and have developed resistance. This aversion leads to inefficiency in treatment. 72


CAR-T cells in hematological malignancies

Cancer cells use a variety of strategies to overcome the immune response of the body’s antitumor system. Immune evasion mechanisms include changes in the expression levels of checkpoint proteins in the cell cycle, increased expression of inhibitory receptors in immune cells such as PD-L1 (programmed cell death receptor), and Treg cell development. 73,74 One of the defining characteristics of Treg cells is the aging of the immune cells, which in turn decreases the immune system’s capacity to fight cancer cells. 75 These findings are obtained in hematological malignancies such as leukemia, multiple myeloma (MM), and B-cell lymphoma. 76,77 They also provided different strategies to lead the immune system against malignant cells to evade cancer cells from the cell immune system (Table 2). For instance, T-cell injections that are genetically engineered and contain the expression of chimeric CD19-specific antigen receptors (CARs) could drive T-cells to CD19. Therapeutic experiments of CAR CD19 T cells were conducted with autologous patient cells involving substantial additional infrastructure. 78-80 These results show appropriate protective strategies to fight hematological cancers. 81 This treatment has undergone several trials and is considered an effective therapeutic regimen. 82

Table 2. A variety of studies have demonstrated that CAR-T cells are characteristic of leukemia
Disease Target antigen CAR features Percentage of total rate conditions of research Reference
DLBCL, MCL CD19 CD8-alpha hinge and transmembrane domains 70% clinical study 83
MCL, CLL CD19 and CD20 Local is produced < 60% clinical study 84
B-cell ALL CD19 and CD22 Bi-specific CAR; intracellular signaling domains 4-1BB and
CD3ζ
< 80% In vivo 85
DLBCL, MCL CD19 Number of T cells enriched for central memory (A) and aimless memory (B)after CAR T injection on day 2 A = 100%
B < 80%
clinical study 86
DLBCL, CLL CD19 Express 4-1BBL < 50% clinical study 87
DLBCL,PMBCL,
MCL, MALT
CD19alone(A) or
CD19 plus another target(B)
Tumor biopsies stained for CD19, CD20, CD22, CD30, CD38, CD70, and PMSA. Choice of CAR T target based
on staining results
Single A = 50%
Double B > 90%
- 88
AML CD123 and CD33 The presence of CD28OX40z increases the killing of CIK cells significantly N/A In vivo 89
AML FRβ Contains CD8a hinge domains and transmembrane domain with intracellular CD3z, alone or with CD28 signal range N/A In vivo
In vitro
90
MM BCMA t has a CD137 (4-1BB) excitation motif and a CD3-zeta signal range < 80% clinical study 91

DLBCL: Diffuse large B-cell lymphoma; MCL: Mantle cell lymphoma; CLL: Chronic lymphocytic leukemia; B-cell ALL: B-cell acute lymphoblastic leukemia;

PMBCL: Primary mediastinal B-cell lymphoma; MALT: mucosa-associated lymphoid tissue; AML: Acute myeloid leukemia; MM: Multiple myeloma.

Multiple myeloma

MM is a type of blood cell cancer in which the plasma cells multiply and are highly abundant in the bone marrow. It accounts for around 10% of all malignancies in the blood. However, there are numerous treatments to cure this condition; it is intolerable. 75 Engineered T-cell translocation is a therapeutic strategies approach to antigens. B-cell maturation antigen (BCMA) was a significant target for engineering cell studies. 92-94 No expression of this target antigen has been identified in normal cells, although it is slightly expressed in normal and differentiated B cells. BCMA antigen is thought to boost the lifespan of MM cells and drug resistance in these cells. 95,96 BCMA CAR-T cell therapy improves the intensity of response by more than 80%. However, the effect of treatment is temporary, and recovery has been documented after BCMA CAR-T cell therapy. The survival rate of BCMA CAR-T progression-free cells is approximately one year. Inhibition or suppression of BCMA expression may be an essential mechanism for disease progression that requires further investigation. 97,98 Different approaches to BCMA in cell therapy, engineering cells for MM, are also being studied. 94 Another target is CD138 (Syndecan 1), which is highly expressed in MM cells and is associated with disease pathogenesis. CD138 expression in MM cells in patients with progressive disease is higher than in MM cells in newly diagnosed patients. Implying the role of this antigen in the disease’s progression. 98 However, given the broad expression of CD138 in human tissues, including epithelial cells, the targeted CARS of this antigen can be used with caution. For instance, treatment with BT062 causes skin and mucosal toxicity 99 that shows the importance of integrating CD138 antigens with other target antigens in immune cell engineering. If we increase knowledge in this area, we are likely to discover more and better alternative approaches.

Lymphoma

Lymphoma is present in the lymph nodes and leukemia in the bone marrow and blood. CAR) binds to CD19-targeted T cells to efficiently cure leukemia/lymphoma, thus decreases normal B cells. As a result, immunoglobulin replacement was necessary for the patient is over a lifespan, so the determination of an appropriate target was necessary. 100 The CD30 antigen may serve as a proper target for engineered cells expressed in malignant lymphoid cells. It should be mentioned that while healthy lymphocytes still appear at certain stages. 101 Due to its high presence in malignant cells, this antigen is a significant target for immune cell engineering antitumor malignancies that appears to be safe. Engineered cells targeting CD30 antigen show considerable anticancer activity in various models. However, targeting normal lymphocytes with CD30 antigen can cause severe complications for patients undergoing these therapies. 102,103 During treatment with anti-CD30 CAR-T cells, the involuntary elimination of HSPCs resulted in unexpected outcomes. In addition to killing the lymphoma cells, the plasma of the blood cells has stabilized. This study observed that the best therapeutic index of anti-CD30 engineered T cells is appropriate for treating blood malignancies. They did not affect normal CD30 + HSPC. 100

Leukemia

More than 50 000 new leukemia patients and more than 20 000 leukemia-related deaths were documented in the United States in 2015. 104 The patient’s life span is approximately five years, based on the type of leukemia, with the lowest survival associated with acute myeloid leukemia (AML) and the highest survival for acute lymphoblastic leukemia (ALL). Treatment of various chemotherapy methods due to drug resistance was not very satisfactory. 105 Cancer immunotherapy has recently switched the therapeutic outlook as a useful clinical method for treating different types of cancer. Hematopoietic stem cell transplantation is among the immune-based treatments used for leukemia. Results in prolonged survival without disease symptoms in half of the patients with leukemia. 106 However, this approach still has its limitations; for example, many patients relapse after treatment, which is not a reasonable option for many patients. The design of new, efficient, and safe treatment strategies for treating leukemia patients is also sensed. That new therapies are not appropriate for them. 107,108

B-cell leukemia

Significant advances in the treatment of leukemia have been accomplished by engineered cells. These CAR-T cells do this by targeting the antigen CD19. 107,108 Even when antigens are found in both normal and cancer cells, most patients can tolerate long-term toxicity and survival 109 and They demonstrate long-lasting longevity in the blood and bone marrow after injection. 108,109 Injection of CD19-directed CAR-T cells to ALL patients who are resistant to treatment or relapse has a response rate of over 70%. 110,111 Although CD 19 acts as an alternative to the treatment of all engineered cells, research has shown that toxicity is beyond the scope of a potential barrier to engineered immune cells for treatment. As a result, additional optimization is necessary for the identification of targets. 112 The CD22 antigen was evaluated as a therapeutic target for CAR-T cell immunity to destroy leukemia cells. Lymphoblasts isolated from patients with precursor B cells all showed CD22 expression. 113 Cells engineered with CD22 antigens, such as CAR-T anti-TSLPR cells, repeatedly lyse target B cells under different environmental conditions. 114 The 4-1BB domain involvement causes the toxicity of engineered immune cells, while previous studies have documented the beneficial function of 4-1BB domains in CAR-T cells. This inconsistency may be attributable to discrepancies between the structure’s configuration and the tumor model. 115,116 Another research has shown that altering the position of immunoglobulins in engineered immune cells decreases the antitumor effect. 114,117

Myeloid leukemia

Lately, CAR-T therapeutic strategies have focused on AML leukemia, the most prevalent form of myeloid neoplasm in adults. 118,119 Hematopoietic stem cells (HSPCs) are thought to be an effective treatment for this disease. However, the mortality and other implications of this treatment indicate the vulnerability of this treatment. Detailed research towards using engineered immune cells is appropriate to solve problems in CAR T and offer appropriate therapeutic strategies. CD123 myeloid antigen is one of the potentially promising targets that are undergoing study. While CD123 antigen is present in HSPC cells to a limited extent, it has been well-tolerated when designed to target antigen for AML patients’ treatment. Treatment with engineered cells may decrease the burden of AML at the advanced stage of the disease and suggest that treatment may be effective at various stages of the disease. 89 The comparison between CD123 CAR-T and CD33 revealed that CD123 CAR-T was less toxic to regular hematopoietic stem cell activity. However, an independent clinical trial in xenograft models showed CD123-targeted CAR-T cells’ effect on normal bleeding. 120 The next target is the folate receptor (FR), a glycoprotein with a large cysteine amino acid. This receptor contains isoforms (FRa, FRb, and FRc) and facilitates folic acid absorption. The FRb isoform is often expressed in myeloid hematopoietic cells also expressed in different malignancies. 121 It has recently been shown that engineered cells targeting human FRb receptors can detect and kill target cells. 90 The combination of CARB-FR and FRb with trans-retinoic acid increases FRb levels and antitumor activity. Notably, FR-targeted cells do not have a detrimental effect on hematopoietic stem cells in vitro. The same group has demonstrated that HA-FRb engineered cells destroy cancer cells more quickly than previously engineered cells. 122

Chronic lymphocytic leukemia

CLL is a chronic malignancy of lymphocytes common in adults. 123 Allogeneic stem cell transplantation is the only current CLL therapy approach. 124 Patients with chronic and high-risk CLL are treated with CD19 CAR-T cells. 111 Several studies have been reviewed to evaluate the effect of engineered cells on individuals with CLL. Even though CLL pathogens contribute to premature immunodeficiency, the impact of CAR-T cells in CLL patients would be restricted. Appropriate approaches to the above problem need to be identified. 125

T-cell leukemia

Application of immunotherapy for T-cell neoplasms has been investigated. 126,127 Interestingly, the T lymphocytes’ cancer cells and regular lines have the same antigens, and no specific antigens have been identified to distinguish them. The use of CAR-T cells will also kill normal cells. Despite these issues, the evaluation of CAR-T cells designed to attack CD5 antigens was evaluated. This study’s findings revealed that engineered cells are attracted to cells that express CD5 and kill these cells. Normal T cells with unregulated expression of PI-9, cathepsin B, and BCL-2 have also been shown to prevent cell death against engineered immune cells. 126 They are also designed to attack CD4 antigens; CAR-T cells strongly destroy CD4 + leukemia cell lines in vitro lymphoma models and in vivo T cells. 127 These findings suggest that engineered cells can provide appropriate therapeutic approaches but need many experiments.


Limitations

Cancer treatment has achieved a new phase with the shift from chemotherapy to safety-based therapeutic approaches. In the meantime, CAR-T cells have demonstrated considerable therapeutic potential in cancer therapy. Although the treatment of CAR-T cells in cancer was very successful, it is still in its infancy, and the mechanisms of action of CAR-T cells are not fully understood. There are more limitations to CAR-T cell therapy that has not been removed. These limitations in the treatment of cancer CAR-T cells include (a) limitation of T cell function, proliferation, and persistence, (b) Restriction of CAR-T cell importation to the tumor site; (c) extra-target toxicity, and (4) loss of antigen to tumor cells, which causes the tumor to escape from CAR-T cells (). 29,128 ScFv-based CAR-T cells could be directed to cancer cell antigens if the antibody’s amino acid sequence with the target properties is identical. However, their use in CAR-T cells can be limited. 19 For instance, by comparing two anti-CD19 (FMC63-scFv) or GD2 (14g2a-scFv) CAR, it was found that 14g2a-scFv structure domains establish clusters independent of the anti-GD2-CAR antigen on the T cell surface. As a result, it promotes severe anti-GD2-CAR-T cells’ severe weakness and reduces their ability to act appropriately in vivo. Many CAR clinical trials have used scFv derived from mouse antibodies to increase the probability of anti-CAR-T cell response, which may cause toxicity or reduce T cells’ viability. 129,130 This problem can be overcome by humanizing mouse scFvs or removing scFv from fully human antibodies. 131,132 Human CAR-T cells can induce comparative recovery of ALL resistant to previous anti-CD19-CAR therapeutic cells based on scFv mice. 132 However, due to the receptors’ chemical existence, even human protein-derived structures can induce host immune response. 18 It is doubtful that the current CAR design template would enable T cells to resolve the multiple obstacles presented by tumor cells and the tumor’s microenvironment. The intrinsic defects of CAR-T cells must therefore be compensated for and novel techniques developed. Targeting more than one antigen on tumor cells at the same time can prevent the loss of the antigen and the tumor from escaping. 26 Due to tumorigenesis’s complex nature, even optimized cars cannot resolve all the various tumor entities’ obstacles. It is also essential to combine several modifications to compensate for some of the inherent defects of CAR-T cells and fulfill the specific needs of tumor cells and the microenvironment. To produce the best therapeutic outcomes, T cell trafficking, penetration, and persistence must be optimized.

apb-12-476-g004
Figure 4. Restrictions on the use of CAR-T. (A) (i) the cancer cells alter CD19 receptor expression so that other CAR-T cells are unable to identify and bind to the target, or (ii) they completely lose CD19 receptor expression. (B) Tumor cells can transform phenotypic deformation into a different breed that is inherently CD19 negative. (C) Cancer cells can cover the surface of the epitope and deprive them of access to CAR-T cell cells. (D) Cause off-target toxicity. (E) CAR-T cells have limited proliferation and persistence. And after a short time, they disappear.


Conclusion

TME significantly inhibits T-cells’ function and indicates that engineered cells may be less effective in targeting tumor cells. However, new therapeutic strategies for CAR-T cell repair and recovery and CAR-T cell activation also have promising approaches to modulate signaling at the tumor cells, transforming the immunosuppressive microenvironment into an efficient microenvironment. CAR-T cells in solid tumors require further investigation. Further research is needed for CAR-T cells in solid tumors. The potential insights into immunotherapy can be provided by understanding the cancer outbreak. Therapies are also different because cancer mechanisms differ.


Ethical Issues

This article does not contain any studies with human participants or animals performed by any of the authors.


Conflict of Interest

No potential conflict of interest was reported by the authors.


References

  1. Sleet DA. The global challenge of child injury prevention. Int J Environ Res Public Health 2018; 15(9):1921. doi: 10.3390/ijerph15091921 [Crossref]
  2. Isazadeh A, Hajazimian S, Garshasbi H, Shadman B, Baghbanzadeh A, Chavoshi R. Resistance mechanisms to immune checkpoints blockade by monoclonal antibody drugs in cancer immunotherapy: focus on myeloma. J Cell Physiol 2021; 236(2):791-805. doi: 10.1002/jcp.29905 [Crossref]
  3. Ozates NP, Soğutlu F, Lerminoglu F, Demir B, Gunduz C, Shademan B. Effects of rapamycin and AZD3463 combination on apoptosis, autophagy, and cell cycle for resistance control in breast cancer. Life Sci 2021; 264:118643. doi: 10.1016/j.lfs.2020.118643 [Crossref]
  4. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence and antitumor activity in individuals with neuroblastoma. Nat Med 2008; 14(11):1264-70. doi: 10.1038/nm.1882 [Crossref]
  5. Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol Res 2014; 2(2):112-20. doi: 10.1158/2326-6066.cir-13-0170 [Crossref]
  6. He Q, Liu Z, Liu Z, Lai Y, Zhou X, Weng J. TCR-like antibodies in cancer immunotherapy. J Hematol Oncol 2019; 12(1):99. doi: 10.1186/s13045-019-0788-4 [Crossref]
  7. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 2020; 17(3):147-67. doi: 10.1038/s41571-019-0297-y [Crossref]
  8. Hudecek M, Sommermeyer D, Kosasih PL, Silva-Benedict A, Liu L, Rader C. The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity. Cancer Immunol Res 2015; 3(2):125-35. doi: 10.1158/2326-6066.cir-14-0127 [Crossref]
  9. Smith AJ, Oertle J, Warren D, Prato D. Chimeric antigen receptor (CAR) T cell therapy for malignant cancers: summary and perspective. J Cell Immunother 2016; 2(2):59-68. doi: 10.1016/j.jocit.2016.08.001 [Crossref]
  10. Li H, Zhao Y. Increasing the safety and efficacy of chimeric antigen receptor T cell therapy. Protein Cell 2017; 8(8):573-89. doi: 10.1007/s13238-017-0411-9 [Crossref]
  11. Levine BL, Miskin J, Wonnacott K, Keir C. Global manufacturing of CAR T cell therapy. Mol Ther Methods Clin Dev 2017; 4:92-101. doi: 10.1016/j.omtm.2016.12.006 [Crossref]
  12. Zhang C, Liu J, Zhong JF, Zhang X. Engineering CAR-T cells. Biomark Res 2017; 5:22. doi: 10.1186/s40364-017-0102-y [Crossref]
  13. Gargett T, Yu W, Dotti G, Yvon ES, Christo SN, Hayball JD. GD2-specific CAR T cells undergo potent activation and deletion following antigen encounter but can be protected from activation-induced cell death by PD-1 blockade. Mol Ther 2016; 24(6):1135-49. doi: 10.1038/mt.2016.63 [Crossref]
  14. Ding ZC, Shi H, Aboelella NS, Fesenkova K, Park EJ, Liu Z, et al. Persistent STAT5 activation reprograms the epigenetic landscape in CD4(+) T cells to drive polyfunctionality and antitumor immunity. Sci Immunol 2020;5(52). 10.1126/sciimmunol.aba5962
  15. Sievers NM, Dörrie J, Schaft N. CARs: beyond T cells and T cell-derived signaling domains. Int J Mol Sci 2020; 21(10):3525. doi: 10.3390/ijms21103525 [Crossref]
  16. Abbott RC, Cross RS, Jenkins MR. Finding the keys to the CAR: identifying novel target antigens for T cell redirection immunotherapies. Int J Mol Sci 2020; 21(2):515. doi: 10.3390/ijms21020515 [Crossref]
  17. Torikai H, Reik A, Liu PQ, Zhou Y, Zhang L, Maiti S. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 2012; 119(24):5697-705. doi: 10.1182/blood-2012-01-405365 [Crossref]
  18. MacLeod DT, Antony J, Martin AJ, Moser RJ, Hekele A, Wetzel KJ. Integration of a CD19 CAR into the TCR alpha chain locus streamlines production of allogeneic gene-edited CAR T cells. Mol Ther 2017; 25(4):949-61. doi: 10.1016/j.ymthe.2017.02.005 [Crossref]
  19. Yu S, Li A, Liu Q, Li T, Yuan X, Han X. Chimeric antigen receptor T cells: a novel therapy for solid tumors. J Hematol Oncol 2017; 10(1):78. doi: 10.1186/s13045-017-0444-9 [Crossref]
  20. Volpe E, Sambucci M, Battistini L, Borsellino G. Fas-fas ligand: checkpoint of t cell functions in multiple sclerosis. Front Immunol 2016; 7:382. doi: 10.3389/fimmu.2016.00382 [Crossref]
  21. Backes CS, Friedmann KS, Mang S, Knörck A, Hoth M, Kummerow C. Natural killer cells induce distinct modes of cancer cell death: discrimination, quantification, and modulation of apoptosis, necrosis, and mixed forms. J Biol Chem 2018; 293(42):16348-63. doi: 10.1074/jbc.RA118.004549 [Crossref]
  22. Oliver AJ, Lau PKH, Unsworth AS, Loi S, Darcy PK, Kershaw MH. Tissue-dependent tumor microenvironments and their impact on immunotherapy responses. Front Immunol 2018; 9:70. doi: 10.3389/fimmu.2018.00070 [Crossref]
  23. Beavis PA, Slaney CY, Kershaw MH, Gyorki D, Neeson PJ, Darcy PK. Reprogramming the tumor microenvironment to enhance adoptive cellular therapy. Semin Immunol 2016; 28(1):64-72. doi: 10.1016/j.smim.2015.11.003 [Crossref]
  24. Scarfò I, Maus MV. Current approaches to increase CAR T cell potency in solid tumors: targeting the tumor microenvironment. J Immunother Cancer 2017; 5:28. doi: 10.1186/s40425-017-0230-9 [Crossref]
  25. Magalhaes I, Carvalho-Queiroz C, Hartana CA, Kaiser A, Lukic A, Mints M. Facing the future: challenges and opportunities in adoptive T cell therapy in cancer. Expert Opin Biol Ther 2019; 19(8):811-27. doi: 10.1080/14712598.2019.1608179 [Crossref]
  26. Cheng J, Zhao L, Zhang Y, Qin Y, Guan Y, Zhang T. Understanding the mechanisms of resistance to CAR T-cell therapy in malignancies. Front Oncol 2019; 9:1237. doi: 10.3389/fonc.2019.01237 [Crossref]
  27. Zhang E, Gu J, Xu H. Prospects for chimeric antigen receptor-modified T cell therapy for solid tumors. Mol Cancer 2018; 17(1):7. doi: 10.1186/s12943-018-0759-3 [Crossref]
  28. Cassetta L, Pollard JW. Targeting macrophages: therapeutic approaches in cancer. Nat Rev Drug Discov 2018; 17(12):887-904. doi: 10.1038/nrd.2018.169 [Crossref]
  29. Martinez M, Moon EK. CAR T cells for solid tumors: new strategies for finding, infiltrating, and surviving in the tumor microenvironment. Front Immunol 2019; 10:128. doi: 10.3389/fimmu.2019.00128 [Crossref]
  30. John LB, Devaud C, Duong CP, Yong CS, Beavis PA, Haynes NM. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin Cancer Res 2013; 19(20):5636-46. doi: 10.1158/1078-0432.ccr-13-0458 [Crossref]
  31. Moon EK, Ranganathan R, Eruslanov E, Kim S, Newick K, O’Brien S. 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 2016; 22(2):436-47. doi: 10.1158/1078-0432.ccr-15-1070 [Crossref]
  32. Watanabe K, Luo Y, Da T, Guedan S, Ruella M, Scholler J, et al. Pancreatic cancer therapy with combined mesothelin-redirected chimeric antigen receptor T cells and cytokine-armed oncolytic adenoviruses. JCI Insight 2018;3(7). 10.1172/jci.insight.99573
  33. Priceman SJ, Tilakawardane D, Jeang B, Aguilar B, Murad JP, Park AK. Regional delivery of chimeric antigen receptor–engineered T cells effectively targets HER2+ breast cancer metastasis to the brain. Clin Cancer Res 2018; 24(1):95-105. doi: 10.1158/1078-0432.ccr-17-2041 [Crossref]
  34. Hegde M, Mukherjee M, Grada Z, Pignata A, Landi D, Navai SA. Tandem CAR T cells targeting HER2 and IL13Rα2 mitigate tumor antigen escape. J Clin Invest 2016; 126(8):3036-52. doi: 10.1172/jci83416 [Crossref]
  35. Feng K, Guo Y, Dai H, Wang Y, Li X, Jia H. Chimeric antigen receptor-modified T cells for the immunotherapy of patients with EGFR-expressing advanced relapsed/refractory non-small cell lung cancer. Sci China Life Sci 2016; 59(5):468-79. doi: 10.1007/s11427-016-5023-8 [Crossref]
  36. Richman SA, Nunez-Cruz S, Moghimi B, Li LZ, Gershenson ZT, Mourelatos Z. High-affinity GD2-specific CAR T cells induce fatal encephalitis in a preclinical neuroblastoma model. Cancer Immunol Res 2018; 6(1):36-46. doi: 10.1158/2326-6066.cir-17-0211 [Crossref]
  37. Künkele A, Taraseviciute A, Finn LS, Johnson AJ, Berger C, Finney O. Preclinical assessment of CD171-directed CAR T-cell adoptive therapy for childhood neuroblastoma: CE7 epitope target safety and product manufacturing feasibility. Clin Cancer Res 2017; 23(2):466-77. doi: 10.1158/1078-0432.ccr-16-0354 [Crossref]
  38. Katz SC, Burga RA, McCormack E, Wang LJ, Mooring W, Point GR. 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. doi: 10.1158/1078-0432.ccr-14-1421 [Crossref]
  39. van Schalkwyk MC, Papa SE, Jeannon JP, Guerrero Urbano T, Spicer JF, Maher J. 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 2013; 24(3):134-42. doi: 10.1089/humc.2013.144 [Crossref]
  40. Kloss CC, Lee J, Zhang A, Chen F, Melenhorst JJ, Lacey SF. Dominant-negative TGF-β receptor enhances PSMA-targeted human CAR T cell proliferation and augments prostate cancer eradication. Mol Ther 2018; 26(7):1855-66. doi: 10.1016/j.ymthe.2018.05.003 [Crossref]
  41. Hassan R, Thomas A, Alewine C, Le DT, Jaffee EM, Pastan I. Mesothelin immunotherapy for cancer: ready for prime time?. J Clin Oncol 2016; 34(34):4171-9. doi: 10.1200/jco.2016.68.3672 [Crossref]
  42. Ali AI, Oliver AJ, Samiei T, Chan JD, Kershaw MH, Slaney CY. Genetic redirection of T cells for the treatment of pancreatic cancer. Front Oncol 2019; 9:56. doi: 10.3389/fonc.2019.00056 [Crossref]
  43. Kachala SS, Bograd AJ, Villena-Vargas J, Suzuki K, Servais EL, Kadota K. Mesothelin overexpression is a marker of tumor aggressiveness and is associated with reduced recurrence-free and overall survival in early-stage lung adenocarcinoma. Clin Cancer Res 2014; 20(4):1020-8. doi: 10.1158/1078-0432.ccr-13-1862 [Crossref]
  44. Thomas A, Chen Y, Steinberg SM, Luo J, Pack S, Raffeld M. High mesothelin expression in advanced lung adenocarcinoma is associated with KRAS mutations and a poor prognosis. Oncotarget 2015; 6(13):11694-703. doi: 10.18632/oncotarget.3429 [Crossref]
  45. Lv J, Li P. Mesothelin as a biomarker for targeted therapy. Biomark Res 2019; 7:18. doi: 10.1186/s40364-019-0169-8 [Crossref]
  46. Li S, Xie L, He L, Fan Z, Xu J, Xu K. Plasma mesothelin as a novel diagnostic and prognostic biomarker in colorectal cancer. J Cancer 2017; 8(8):1355-61. doi: 10.7150/jca.18014 [Crossref]
  47. Carpenito C, Milone MC, Hassan R, Simonet JC, Lakhal M, Suhoski MM. Control of large, established tumor xenografts with genetically retargeted human T cells containing CD28 and CD137 domains. Proc Natl Acad Sci U S A 2009; 106(9):3360-5. doi: 10.1073/pnas.0813101106 [Crossref]
  48. Adusumilli PS, Cherkassky L, Villena-Vargas J, Colovos C, Servais E, Plotkin J. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci Transl Med 2014; 6(261):261ra151. doi: 10.1126/scitranslmed.3010162 [Crossref]
  49. Lanitis E, Poussin M, Hagemann IS, Coukos G, Sandaltzopoulos R, Scholler N. Redirected antitumor activity of primary human lymphocytes transduced with a fully human anti-mesothelin chimeric receptor. Mol Ther 2012; 20(3):633-43. doi: 10.1038/mt.2011.256 [Crossref]
  50. González PA, Carreño LJ, Céspedes PF, Bueno SM, Riedel CA, Kalergis AM. Modulation of tumor immunity by soluble and membrane-bound molecules at the immunological synapse. Clin Dev Immunol 2013; 2013:450291. doi: 10.1155/2013/450291 [Crossref]
  51. Chen SS, Hu Z, Zhong XP. Diacylglycerol kinases in T cell tolerance and effector function. Front Cell Dev Biol 2016; 4:130. doi: 10.3389/fcell.2016.00130 [Crossref]
  52. Purow B. Molecular pathways: targeting diacylglycerol kinase alpha in cancer. Clin Cancer Res 2015; 21(22):5008-12. doi: 10.1158/1078-0432.ccr-15-0413 [Crossref]
  53. Riese MJ, Wang LC, Moon EK, Joshi RP, Ranganathan A, June CH. Enhanced effector responses in activated CD8+ T cells deficient in diacylglycerol kinases. Cancer Res 2013; 73(12):3566-77. doi: 10.1158/0008-5472.can-12-3874 [Crossref]
  54. Hassan R, Kreitman RJ, Pastan I, Willingham MC. Localization of mesothelin in epithelial ovarian cancer. Appl Immunohistochem Mol Morphol 2005; 13(3):243-7. doi: 10.1097/01.pai.00000141545.36485.d6 [Crossref]
  55. Song DG, Ye Q, Poussin M, Liu L, Figini M, Powell DJ, Jr Jr. A fully human chimeric antigen receptor with potent activity against cancer cells but reduced risk for off-tumor toxicity. Oncotarget 2015; 6(25):21533-46. doi: 10.18632/oncotarget.4071 [Crossref]
  56. Lanitis E, Poussin M, Klattenhoff AW, Song D, Sandaltzopoulos R, June CH. Chimeric antigen receptor T Cells with dissociated signaling domains exhibit focused antitumor activity with reduced potential for toxicity in vivo. Cancer Immunol Res 2013; 1(1):43-53. doi: 10.1158/2326-6066.cir-13-0008 [Crossref]
  57. Wang E, Wang LC, Tsai CY, Bhoj V, Gershenson Z, Moon E. Generation of potent T-cell immunotherapy for cancer using DAP12-based, multichain, chimeric immunoreceptors. Cancer Immunol Res 2015; 3(7):815-26. doi: 10.1158/2326-6066.cir-15-0054 [Crossref]
  58. Wee P, Wang Z. Epidermal growth factor receptor cell proliferation signaling pathways. Cancers (Basel) 2017; 9(5):52. doi: 10.3390/cancers9050052 [Crossref]
  59. Awasthi N, Scire E, Monahan S, Grojean M, Zhang E, Schwarz MA. Augmentation of response to nab-paclitaxel by inhibition of insulin-like growth factor (IGF) signaling in preclinical pancreatic cancer models. Oncotarget 2016; 7(30):46988-7001. doi: 10.18632/oncotarget.9029 [Crossref]
  60. Yewale C, Baradia D, Vhora I, Patil S, Misra A. Epidermal growth factor receptor targeting in cancer: a review of trends and strategies. Biomaterials 2013; 34(34):8690-707. doi: 10.1016/j.biomaterials.2013.07.100 [Crossref]
  61. Seshacharyulu P, Ponnusamy MP, Haridas D, Jain M, Ganti AK, Batra SK. Targeting the EGFR signaling pathway in cancer therapy. Expert OpinTher Targets 2012; 16(1):15-31. doi: 10.1517/14728222.2011.648617 [Crossref]
  62. Sakthianandeswaren A, Sabljak P, Elliott MJ, Palmieri M, Sieber OM. Predictive biomarkers for monoclonal antibody therapies targeting EGFR (cetuximab, panitumumab) in the treatment of metastatic colorectal cancer. In: Segelov E, ed. Advances in the Molecular Understanding of Colorectal Cancer. IntechOpen; 2018. 10.5772/intechopen.80690
  63. Maus MV. Designing CAR T cells for glioblastoma. Oncoimmunology 2015; 4(12):e1048956. doi: 10.1080/2162402x.2015.1048956 [Crossref]
  64. Eskilsson E, Rosland GV, Talasila KM, Knappskog S, Keunen O, Sottoriva A. EGFRvIII mutations can emerge as late and heterogenous events in glioblastoma development and promote angiogenesis through Src activation. Neuro Oncol 2016; 18(12):1644-55. doi: 10.1093/neuonc/now113 [Crossref]
  65. Song DG, Ye Q, Carpenito C, Poussin M, Wang LP, Ji C. In vivo persistence, tumor localization, and antitumor activity of CAR-engineered T cells is enhanced by costimulatory signaling through CD137 (4-1BB). Cancer Res 2011; 71(13):4617-27. doi: 10.1158/0008-5472.can-11-0422 [Crossref]
  66. Morgan RA, Johnson LA, Davis JL, Zheng Z, Woolard KD, Reap EA. Recognition of glioma stem cells by genetically modified T cells targeting EGFRvIII and development of adoptive cell therapy for glioma. Hum Gene Ther 2012; 23(10):1043-53. doi: 10.1089/hum.2012.041 [Crossref]
  67. Zhao L, Cao YJ. Engineered T cell therapy for cancer in the clinic. Front Immunol 2019; 10:2250. doi: 10.3389/fimmu.2019.02250 [Crossref]
  68. Wang Z, Chen W, Zhang X, Cai Z, Huang W. A long way to the battlefront: CAR T cell therapy against solid cancers. J Cancer 2019; 10(14):3112-23. doi: 10.7150/jca.30406 [Crossref]
  69. Kershaw MH, Westwood JA, Darcy PK. Gene-engineered T cells for cancer therapy. Nat Rev Cancer 2013; 13(8):525-41. doi: 10.1038/nrc3565 [Crossref]
  70. Pagani O, Klingbiel D, Ruhstaller T, Nolè F, Eppenberger S, Oehlschlegel C. Do all patients with advanced HER2 positive breast cancer need upfront-chemo when receiving trastuzumab? Randomized phase III trial SAKK 22/99. Ann Oncol 2017; 28(2):305-12. doi: 10.1093/annonc/mdw622 [Crossref]
  71. Pernas S, Tolaney SM. HER2-positive breast cancer: new therapeutic frontiers and overcoming resistance. Ther Adv Med Oncol 2019; 11:1758835919833519. doi: 10.1177/1758835919833519 [Crossref]
  72. Alexander PB, Chen R, Gong C, Yuan L, Jasper JS, Ding Y. Distinct receptor tyrosine kinase subsets mediate anti-HER2 drug resistance in breast cancer. J Biol Chem 2017; 292(2):748-59. doi: 10.1074/jbc.M116.754960 [Crossref]
  73. McGoverne I, Dunn J, Batham J, Tu WJ, Chrisp J, Rao S. Epitherapy and immune checkpoint blockade: using epigenetic reinvigoration of exhausted and dysfunctional T cells to reimburse immunotherapy response. BMC Immunol 2020; 21(1):22. doi: 10.1186/s12865-020-00353-0 [Crossref]
  74. Chaudhry A, Rudensky AY. Control of inflammation by integration of environmental cues by regulatory T cells. J Clin Invest 2013; 123(3):939-44. doi: 10.1172/jci57175 [Crossref]
  75. Pieren DKJ, Smits NAM, van de Garde MDB, Guichelaar T. Response kinetics reveal novel features of ageing in murine T cells. Sci Rep 2019; 9(1):5587. doi: 10.1038/s41598-019-42120-1 [Crossref]
  76. Sander FE, Nilsson M, Rydström A, Aurelius J, Riise RE, Movitz C. Role of regulatory T cells in acute myeloid leukemia patients undergoing relapse-preventive immunotherapy. Cancer Immunol Immunother 2017; 66(11):1473-84. doi: 10.1007/s00262-017-2040-9 [Crossref]
  77. Chung DJ, Pronschinske KB, Shyer JA, Sharma S, Leung S, Curran SA. T-cell exhaustion in multiple myeloma relapse after autotransplant: optimal timing of immunotherapy. Cancer Immunol Res 2016; 4(1):61-71. doi: 10.1158/2326-6066.cir-15-0055 [Crossref]
  78. Kantarjian H, Stein A, Gökbuget N, Fielding AK, Schuh AC, Ribera JM. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med 2017; 376(9):836-47. doi: 10.1056/NEJMoa1609783 [Crossref]
  79. Turtle CJ, Riddell SR, Maloney DG. CD19-Targeted chimeric antigen receptor-modified T-cell immunotherapy for B-cell malignancies. Clin PharmacolTher 2016; 100(3):252-8. doi: 10.1002/cpt.392 [Crossref]
  80. Candeias SM, Gaipl US. The immune system in cancer prevention, development and therapy. Anticancer Agents Med Chem 2016; 16(1):101-7. doi: 10.2174/1871520615666150824153523 [Crossref]
  81. Barrett DM, Grupp SA, June CH. Chimeric antigen receptor- and TCR-modified T cells enter main street and wall street. J Immunol 2015; 195(3):755-61. doi: 10.4049/jimmunol.1500751 [Crossref]
  82. Fujiwara H. Adoptive immunotherapy for hematological malignancies using T cells gene-modified to express tumor antigen-specific receptors. Pharmaceuticals (Basel) 2014; 7(12):1049-68. doi: 10.3390/ph7121049 [Crossref]
  83. Brudno J, Hartman S, Lam N, Stroncek DF, Rossi JM, Shen YW. Low levels of neurologic toxicity with retained anti-lymphoma activity in a phase I clinical trial of T cells expressing a novel anti-CD19 CAR. Blood 2018; 132(Suppl 1):697. doi: 10.1182/blood-2018-99-113731 [Crossref]
  84. Shah NN, Zhu F, Taylor C, Schneider D, Krueger W, Worden A. A phase 1 study with point-of-care manufacturing of dual targeted, tandem anti-CD19, anti-CD20 chimeric antigen receptor modified T (CAR-T) cells for relapsed, refractory, non-Hodgkin lymphoma. Blood 2018; 132(Suppl 1):4193. doi: 10.1182/blood-2018-99-110194 [Crossref]
  85. Hossain N, Sahaf B, Abramian M, Spiegel JY, Kong K, Kim S. Phase I experience with a bi-specific CAR targeting CD19 and CD22 in adults with B-cell malignancies. Blood 2018; 132(Suppl 1):490. doi: 10.1182/blood-2018-99-110142 [Crossref]
  86. Popplewell L, Wang X, Blanchard S, Wagner J, Naranjo A, Adria A. CD19-CAR therapy using naive/memory or central memory T cells integrated into the autologous stem cell transplant regimen for patients with B-NHL. Blood 2018; 132(Suppl 1):610. doi: 10.1182/blood-2018-99-119650 [Crossref]
  87. Park JH, Rivière I, Gonen M, Wang X, Sénéchal B, Curran KJ. Long-term follow-up of CD19 CAR therapy in acute lymphoblastic leukemia. N Engl J Med 2018; 378(5):449-59. doi: 10.1056/NEJMoa1709919 [Crossref]
  88. Chang L, Chang M, Chang HM, Chang F. Microsatellite instability: a predictive biomarker for cancer immunotherapy. Appl Immunohistochem Mol Morphol 2018; 26(2):e15-e21. doi: 10.1097/pai.0000000000000575 [Crossref]
  89. Pizzitola I, Anjos-Afonso F, Rouault-Pierre K, Lassailly F, Tettamanti S, Spinelli O. Chimeric antigen receptors against CD33/CD123 antigens efficiently target primary acute myeloid leukemia cells in vivo. Leukemia 2014; 28(8):1596-605. doi: 10.1038/leu.2014.62 [Crossref]
  90. Lynn RC, Poussin M, Kalota A, Feng Y, Low PS, Dimitrov DS. Targeting of folate receptor β on acute myeloid leukemia blasts with chimeric antigen receptor-expressing T cells. Blood 2015; 125(22):3466-76. doi: 10.1182/blood-2014-11-612721 [Crossref]
  91. Raje N, Berdeja J, Lin Y, Siegel D, Jagannath S, Madduri D. Anti-BCMA CAR T-cell therapy bb2121 in relapsed or refractory multiple myeloma. N Engl J Med 2019; 380(18):1726-37. doi: 10.1056/NEJMoa1817226 [Crossref]
  92. Danhof S, Hudecek M, Smith EL. CARs and other T cell therapies for MM: the clinical experience. Best Pract Res Clin Haematol 2018; 31(2):147-57. doi: 10.1016/j.beha.2018.03.002 [Crossref]
  93. Cho SF, Anderson KC, Tai YT. Targeting B cell maturation antigen (BCMA) in multiple myeloma: potential uses of BCMA-based immunotherapy. Front Immunol 2018; 9:1821. doi: 10.3389/fimmu.2018.01821 [Crossref]
  94. Cohen AD. CAR T cells and other cellular therapies for multiple myeloma: 2018 update. Am Soc Clin Oncol Educ Book 2018; 38:e6-e15. doi: 10.1200/edbk_200889 [Crossref]
  95. Tai YT, Acharya C, An G, Moschetta M, Zhong MY, Feng X. APRIL and BCMA promote human multiple myeloma growth and immunosuppression in the bone marrow microenvironment. Blood 2016; 127(25):3225-36. doi: 10.1182/blood-2016-01-691162 [Crossref]
  96. Ma T, Shi J, Liu H. Chimeric antigen receptor T cell targeting B cell maturation antigen immunotherapy is promising for multiple myeloma. Ann Hematol 2019; 98(4):813-22. doi: 10.1007/s00277-018-03592-9 [Crossref]
  97. Hamieh M, Dobrin A, Cabriolu A, van der Stegen SJC, Giavridis T, Mansilla-Soto J. CAR T cell trogocytosis and cooperative killing regulate tumour antigen escape. Nature 2019; 568(7750):112-6. doi: 10.1038/s41586-019-1054-1 [Crossref]
  98. Kawano Y, Fujiwara S, Wada N, Izaki M, Yuki H, Okuno Y. Multiple myeloma cells expressing low levels of CD138 have an immature phenotype and reduced sensitivity to lenalidomide. Int J Oncol 2012; 41(3):876-84. doi: 10.3892/ijo.2012.1545 [Crossref]
  99. Jagannath S, Chanan-Khan A, Heffner LT, Avigan D, Zimmerman TM, Lonial S. BT062, an antibody-drug conjugate directed against CD138, shows clinical activity in patients with relapsed or relapsed/refractory multiple myeloma. Blood 2011; 118(21):305. doi: 10.1182/blood.V118.21.305.305 [Crossref]
  100. Hombach AA, Görgens A, Chmielewski M, Murke F, Kimpel J, Giebel B. Superior therapeutic index in lymphoma therapy: CD30+ CD34+ hematopoietic stem cells resist a chimeric antigen receptor T-cell attack. Mol Ther 2016; 24(8):1423-34. doi: 10.1038/mt.2016.82 [Crossref]
  101. Muta H, Podack ER. CD30: from basic research to cancer therapy. Immunol Res 2013; 57(1-3):151-8. doi: 10.1007/s12026-013-8464-1 [Crossref]
  102. Li D, Li X, Zhou WL, Huang Y, Liang X, Jiang L. Genetically engineered T cells for cancer immunotherapy. Signal Transduct Target Ther 2019; 4:35. doi: 10.1038/s41392-019-0070-9 [Crossref]
  103. Wang D, Zeng C, Xu B, Xu JH, Wang J, Jiang LJ. Anti-CD30 chimeric antigen receptor T cell therapy for relapsed/refractory CD30+ lymphoma patients. Blood Cancer J 2020; 10(1):8. doi: 10.1038/s41408-020-0274-9 [Crossref]
  104. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 2015; 65(1):5-29. doi: 10.3322/caac.21254 [Crossref]
  105. Pulte D, Redaniel MT, Jansen L, Brenner H, Jeffreys M. Recent trends in survival of adult patients with acute leukemia: overall improvements, but persistent and partly increasing disparity in survival of patients from minority groups. Haematologica 2013; 98(2):222-9. doi: 10.3324/haematol.2012.063602 [Crossref]
  106. Ishii K, Barrett AJ. Novel immunotherapeutic approaches for the treatment of acute leukemia (myeloid and lymphoblastic). Ther Adv Hematol 2016; 7(1):17-39. doi: 10.1177/2040620715616544 [Crossref]
  107. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells in chronic lymphoid leukemia. N Engl J Med 2011; 365(8):725-33. doi: 10.1056/NEJMoa1103849 [Crossref]
  108. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014; 371(16):1507-17. doi: 10.1056/NEJMoa1407222 [Crossref]
  109. Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 2011; 118(18):4817-28. doi: 10.1182/blood-2011-04-348540 [Crossref]
  110. Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome associated with novel T cell-engaging therapies. Cancer J 2014; 20(2):119-22. doi: 10.1097/ppo.0000000000000035 [Crossref]
  111. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cells expressing an anti-CD19 chimeric antigen receptor. J Clin Oncol 2015; 33(6):540-9. doi: 10.1200/jco.2014.56.2025 [Crossref]
  112. Shalabi H, Angiolillo A, Fry TJ. Beyond CD19: opportunities for future development of targeted immunotherapy in pediatric relapsed-refractory acute leukemia. Front Pediatr 2015; 3:80. doi: 10.3389/fped.2015.00080 [Crossref]
  113. Haso W, Lee DW, Shah NN, Stetler-Stevenson M, Yuan CM, Pastan IH. Anti-CD22-chimeric antigen receptors targeting B-cell precursor acute lymphoblastic leukemia. Blood 2013; 121(7):1165-74. doi: 10.1182/blood-2012-06-438002 [Crossref]
  114. Qin H, Cho M, Haso W, Zhang L, Tasian SK, Oo HZ. Eradication of B-ALL using chimeric antigen receptor-expressing T cells targeting the TSLPR oncoprotein. Blood 2015; 126(5):629-39. doi: 10.1182/blood-2014-11-612903 [Crossref]
  115. Barrett DM, Singh N, Porter DL, Grupp SA, June CH. Chimeric antigen receptor therapy for cancer. Annu Rev Med 2014; 65:333-47. doi: 10.1146/annurev-med-060512-150254 [Crossref]
  116. Lim WA, June CH. The principles of engineering immune cells to treat cancer. Cell 2017; 168(4):724-40. doi: 10.1016/j.cell.2017.01.016 [Crossref]
  117. Zhang C, Zeng X, Li Z, Wang Z, Li S. Immunoglobulin A nephropathy: current progress and future directions. Transl Res 2015; 166(2):134-44. doi: 10.1016/j.trsl.2015.02.007 [Crossref]
  118. De Kouchkovsky I, Abdul-Hay M. ‘Acute myeloid leukemia: a comprehensive review and 2016 update’. Blood Cancer J 2016; 6(7):e441. doi: 10.1038/bcj.2016.50 [Crossref]
  119. Dombret H, Gardin C. An update of current treatments for adult acute myeloid leukemia. Blood 2016; 127(1):53-61. doi: 10.1182/blood-2015-08-604520 [Crossref]
  120. Gill S, Tasian SK, Ruella M, Shestova O, Li Y, Porter DL. Preclinical targeting of human acute myeloid leukemia and myeloablation using chimeric antigen receptor-modified T cells. Blood 2014; 123(15):2343-54. doi: 10.1182/blood-2013-09-529537 [Crossref]
  121. Chen C, Ke J, Zhou XE, Yi W, Brunzelle JS, Li J. Structural basis for molecular recognition of folic acid by folate receptors. Nature 2013; 500(7463):486-9. doi: 10.1038/nature12327 [Crossref]
  122. Lynn RC, Feng Y, Schutsky K, Poussin M, Kalota A, Dimitrov DS. High-affinity FRβ-specific CAR T cells eradicate AML and normal myeloid lineage without HSC toxicity. Leukemia 2016; 30(6):1355-64. doi: 10.1038/leu.2016.35 [Crossref]
  123. Enblad G, Karlsson H, Loskog AS. CAR T-cell therapy: the role of physical barriers and immunosuppression in lymphoma. Hum Gene Ther 2015; 26(8):498-505. doi: 10.1089/hum.2015.054 [Crossref]
  124. Mewawalla P, Nathan S. Role of allogeneic transplantation in patients with chronic lymphocytic leukemia in the era of novel therapies: a review. Ther Adv Hematol 2014; 5(5):139-52. doi: 10.1177/2040620714550773 [Crossref]
  125. Fraietta JA, Beckwith KA, Patel PR, Ruella M, Zheng Z, Barrett DM. Ibrutinib enhances chimeric antigen receptor T-cell engraftment and efficacy in leukemia. Blood 2016; 127(9):1117-27. doi: 10.1182/blood-2015-11-679134 [Crossref]
  126. Mamonkin M, Rouce RH, Tashiro H, Brenner MK. A T-cell-directed chimeric antigen receptor for the selective treatment of T-cell malignancies. Blood 2015; 126(8):983-92. doi: 10.1182/blood-2015-02-629527 [Crossref]
  127. Pinz K, Liu H, Golightly M, Jares A, Lan F, Zieve GW. Preclinical targeting of human T-cell malignancies using CD4-specific chimeric antigen receptor (CAR)-engineered T cells. Leukemia 2016; 30(3):701-7. doi: 10.1038/leu.2015.311 [Crossref]
  128. Han X, Wang Y, Wei J, Han W. Multi-antigen-targeted chimeric antigen receptor T cells for cancer therapy. J Hematol Oncol 2019; 12(1):128. doi: 10.1186/s13045-019-0813-7 [Crossref]
  129. Grupp SA, Maude SL, Shaw PA, Aplenc R, Barrett DM, Callahan C. Durable remissions in children with relapsed/refractory all treated with T cells engineered with a CD19-targeted chimeric antigen receptor (CTL019). Blood 2015; 126(23):681. doi: 10.1182/blood.V126.23.681.681 [Crossref]
  130. Cai B, Guo M, Wang Y, Zhang Y, Yang J, Guo Y. Co-infusion of haplo-identical CD19-chimeric antigen receptor T cells and stem cells achieved full donor engraftment in refractory acute lymphoblastic leukemia. J Hematol Oncol 2016; 9(1):131. doi: 10.1186/s13045-016-0357-z [Crossref]
  131. Wang CM, Wu ZQ, Wang Y, Guo YL, Dai HR, Wang XH. Autologous T cells expressing CD30 chimeric antigen receptors for relapsed or refractory Hodgkin lymphoma: an open-label phase I trial. Clin Cancer Res 2017; 23(5):1156-66. doi: 10.1158/1078-0432.ccr-16-1365 [Crossref]
  132. Wang Z, Wu Z, Liu Y, Han W. New development in CAR-T cell therapy. J Hematol Oncol 2017; 10(1):53. doi: 10.1186/s13045-017-0423-1 [Crossref]
Submitted: 16 Feb 2021
Revised: 12 Jun 2021
Accepted: 17 Aug 2021
First published online: 22 Aug 2021
EndNote EndNote

(Enw Format - Win & Mac)

BibTeX BibTeX

(Bib Format - Win & Mac)

Bookends Bookends

(Ris Format - Mac only)

EasyBib EasyBib

(Ris Format - Win & Mac)

Medlars Medlars

(Txt Format - Win & Mac)

Mendeley Web Mendeley Web
Mendeley Mendeley

(Ris Format - Win & Mac)

Papers Papers

(Ris Format - Win & Mac)

ProCite ProCite

(Ris Format - Win & Mac)

Reference Manager Reference Manager

(Ris Format - Win only)

Refworks Refworks

(Refworks Format - Win & Mac)

Zotero Zotero

(Ris Format - FireFox Plugin)

Abstract View: 824
PDF Download: 453
Full Text View: 192