CAR T Cell Immunotherapy That Revolutionary Breakthrough in Human Oncology Treatment: A Review

Abstract

The discovery of CAR T cell immunotherapy, also known as chimeric antigen receptor (CAR) T cell immunotherapy, has added a new dimension to the world of cancer treatment. This is a gene-based treatment in which T cells from the patient’s body are taken and genetically engineered in the lab to grow receptors. T cells containing this receptor are then injected into the patient’s body to bind to the antigen on the surface area of the cancer cell and kill the cancer cell. Structurally, the co-stimulatory domain added to CAR T cells has now reached the 5th GEN of chimeric antigen receptor T cells. Chimeric antigen T cell immunotherapy is the first FDA-approved treatment for hematological malignancies that is both safe and effective. However, due to some challenges such as a lack of safety control, an immunosuppressive tumor microenvironment, ineffective T cell trafficking, and so on, CAR-T immunotherapy treatment for solid malignancy is still in the clinical phase. In the result and discussion, we have presented a survey of CAR T cell therapy with a combination of pharmacological drugs. The things we mentioned are that CAR T cell immunotherapy is innovative, suitable, elegant, and also controls synergistic anti-cancer effects. A better understanding of combinatory CAR T cell therapies provides fundamental information for improvement of those therapies, in addition to the article highlighting future opportunities, commercial advancements, and various applications of CAR T cell therapy in different cancer cells. In the entire review article, we have highlighted the neck and crop of CAR T cell therapy, from which it is easy to understand the therapy and the need for this therapy in cancer prevention and its progress.

Share and Cite:

Bashar, S. , Akhter, Z. , Saha, B. , Himaloy, M. , Kulsum, U. and Rahman, M. (2022) CAR T Cell Immunotherapy That Revolutionary Breakthrough in Human Oncology Treatment: A Review. Pharmacology & Pharmacy, 13, 483-515. doi: 10.4236/pp.2022.1311036.

1. Introduction

In the rough, oncological treatments avowed as chemotherapy, surgery and radiation therapy are either unaccompanied or in combination. Though these schemes of treatment have exploited to adequate prosperous outcome including newer and more appropriate diagnostic tool, presage of most cancer is very ponderous. In this complexity more personalized treatment such as immunotherapy obligates for most cancers to cure or control. Currently treatment or management of cancer converted into more specific therapies with immunotherapy which corroborated better individualization care and feasibility of success for each cancer patient [1] .

Biotherapy is another name of immunotherapy which is naturally able to determine foreign pathogens or carcinogenic cells in the human body. Though some immunotherapy clinches the body’s immune system, some of their directly bind with cancer cells [3] [4] . In human immune way, T cells or T lymphocytes carry out an exigent role, those are work to detect or shatter potential malignant or tumor cells [5] . Adoptive immunotherapy based on T cell still now develops and deals with most hematologic cancers and other cancer cells [6] . Adoptively transferred T cell therapy (ATCT) is recognized as one kind of immunotherapy that proceeds against malignancies through the seclusion of lymphocytes by potent dynamic T cells, those are infused within a patient to cure a disease [1] . Adoptive T cell transfer (ACT) clovens into three forms for cancer therapy and these are TILs (infiltrating tumor lymphocyte), TCR and CAR cell [7] . T cell receptor (TCR) or antigen receptor of T cells detects tumor antigens which acquire intense immune response within a short time to banishing tumor cells [1] . In some clinical trials, tumor infiltrating lymphocytes (TILs) proved viable complete response against meta-static melanoma. Chimeric antigen receptor (CAR) is a pretended molecule which is a protein and it fuses an extracellular antigen to dig up domain with intracellular activating domain [1] . Here we showed a comparison between CAR and TCR (Table 1). This CAR T therapy belongs to cellular immunotherapy that reveals antibody based chimeric antigen receptor to target carcinogenic antigen which is very proficient therapy for different kinds of hematological malignancies and solid tumors [8] [9] . In Israel first chimeric antigen receptor was discovered by Eshhar’s group within 1989 at Weizmann Institute of Science. Through viral and non-viral transfection manner T cells are harvested and genetically modified and after modification these cells are patulous in culture [10] . CAR T cell is composed of two domains that deflected with TCR (T cell receptor) part of natural T cell and this domain termed as extracellular

Table 1. Comparison between CAR and TCR [2] .

and intracellular domain. Extracellular domain work against cell surface antigen which is an antibody single chain fragment (scFv) and intracellular domain is fused signaling domain which limns different generation of CAR T cell [11] [12] . CAR T cell production effectuate through which procedure that is complex [13] . At first without collection of granulocyte colony which is stimulating factor natural T cells collected by isolation of blood cells or phlebotomy from cellular components of blood [14] . Extension and perception of T cell may disrupt by granulocyte colony stimulating factors that’s why it excluded [15] [16] . In isolation of blood apheresis is broadly used because of it cut off from its components that also helps to collect platelet and another elements of blood from patient body through blood bank and prior re-injecting it into patient body this modified genetically [17] . By transfected with CAR retroviral or lentiviral in isolated T cells converted into CAR T cells were also occurring installation of genome DNA [14] [16] . To determine effectiveness of this adoptive immunotherapy through these newly designed T cells expansion and purification is a major step [14] . At last stage of modified T cell production it takes 2 - 4 weeks for quality control and sterility test which is necessary for safe and effective treatment [16] . If patient take a conditioning treatment such as lymphodepleting at least 2 days before administration of transduced T cells it may lead to huge number of T cells expansion [14] [16] . Composition of this T cell corresponded to an antibody which have detection and binding area upon T cell surface [18] . CAR T cell have antibody like recognition power which can activate T cell through a mechanism and for this reason it got very interest in immunotherapy [19] . The scFv part of this T cell composed of on moving light chain (VL) and heavy (VH) chain that attach with each other through a linker. CAR consist of activation domain (CD-3 zeta, transmembrane domain, co-incentive domain (CD28, CD137 (4-1BB)). Eventually CAR destroy tumor cell through T cell cytotoxic function after bind with carcinogens while T cell propagation commenced [8] . The scFv part of CAR obtain from antibody and have higher magnitude affinity than TCRs that’s why it is very advantageous for cancer therapy [20] . Another cause that responsible for this therapy effectiveness is surface protein of tumor cell easily detect by scFv process [21] .

2. CAR T Cell Profile

2.1. History of CAR T Cell

One of the oldest methods of cellular immunotherapy is allogenic bone marrow transplantation. Applying this immunotherapy to some patients in 1977, they found that Graft vs. Host disease emerged as a secondary disease in the patients. Later, scientists began to think about whether new immunotherapy could be invented that would not cause any secondary disease in the patient. Next, they think about T cell immunotherapy which they later genetically modify in the lab, expand and then infuse into the patient’s body [22] . From their thinking, this immunotherapy is experimented in different sites and modified after a certain time interval, and finally comes to today’s position, CAR T cell immunotherapy, which is accepted by the US FDA. The history of CAR T cell immunotherapy is presented in the historical diagram (Figure 1).

2.2. Generation of CAR

CAR Immunotherapy is one of the most common cancer treatments today. Chimeric Antigen Therapy (CAR) T is an effective treatment for a variety of hematological malignancies, such as lymphomas, acute lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), and solid tumors like breast cancer, liver cancer, and prostate cancer [8] . The four-generation summary of CAR T cells is provided in Figure 2.

2.3. CAR T Cell Structure

A typical CAR T cell is made up of many parts such as extracellular, spacer, transmembrane and intracellular domain etc.

2.3.1. Extracellular Domain

The extracellular domain component is composed of a single chain variable fragment designed with a light & heavy chain variable region with a coding section of monoclonal antibody that is capable of maintaining antibody affinity [24] . These variable regions are connected to each other with glycine serine residues which are able to minimize the risk of linker interference between the domains [25] . Antigen recognition domains in CAR are able to treat against autoimmune or infectious diseases [26] .

2.3.2. Spacer Domain

Spacer domains are usually bridges between extracellular domains and transmembrane domains. According to some reports, the activation of the carrier cell

Figure 1. History of CAR T cell immunotherapy [22] .

depends on the length of the spacer domain [27] . The smaller the length of the spacer domain, the greater the activation of the T cell. In many cases, the inability to bind the cell membrane to the target antigen plays an important role in binding to the cell membrane through the longer space domain Sc-Fv, although in many cases it becomes a risk because it depletes the carrier cell [27] [28] . In closing, I would say that based on the accessibility of the target antigen, I need to create an optimal length speacer domain that is able to ensure proper function of my CAR.

2.3.3. Transmembrane Domain

This transmembrane domain acts as a connector between the extracellular and

Figure 2. Four generation of CAR T cell [23] .

the intracellular domains. It is made up of a hydrophobic alpha helix which is able to span the membrane. This transmembrane region of CAR T cells plays a vital role in proper synapse formation or CAR surface expression [29] [30] .

2.3.4. Intracellular Domain

The intracellular domain basically activates the CAR T cell during signal passing [31] . In this study, we look at the development of a 4th generation CAR T cell which is able to overcome the restrictions of the 1st generation by adding mother domain CD3 zeta. Later, another co stimulatory domain, 4-1BB, was added to the 3rd generation to further improve the function of the CAR T cell. Finally, IL-2 cytokines were added to boost the activation of 4th generation CAR T cells.

2.4. CAR T Cell Manufacturing

In the case of CAR T cell manufacturing, first WBC has to be separated from the patient’s blood, which is called the leukapheresis process [32] . The leukapheresis product is enhanced for T cells once a sufficient number of leukocytes have been extracted. Subsequently, the cells are separated by centrifugal elutriation based on their size, and it’s activated in the presence of APC between monoclonal antibodies anti CD3/CD28, and the growth factor IL-2, and its growth increases within a few weeks. Earlier, the beads were removed from the T cells by magnetic separation [33] . The T cell is incubated with the viral vector and, after a few days, the vector is washed out. This viral vector induces genetic material in the T cell, which later turns into a chimeric antigen T cell. Finally, CAR T cells are manufactured in bioreactor devices on a large scale.

3. Present Circumstance of CAR Therapy

Lately health technology is growing up day by day with push ahead world. Still now in this antediluvian world those critical disease was coming cancer is one of them. Though many scientists try to bring out from this critical disease by severe methods and therapy in beginning of illness. This anti-cancer treatment proved as arresting invention within those treatments are established to cure. By this prodigious immunotherapy a satisfied number of cancer type treated such as R/R multiple myeloma, R/R large B cell cancer, diffuse large B cell lymphoma, R/R mantle cell lymphoma, B cell ALL, non-Hodgkin, R/R multiple myeloma, R/R follicular lymphoma. Against different kinds of tumor through this therapy proved 30% to 70% or greater than 90% potency. As much CTL019 a drug of CAR T therapy manufactured by Novartis that complete remission (CR) rate is 93%. At present clinical trials of this immunotherapy rise abruptly in the global [34] .

Record of clinical Trials.gov showed 836 clinical studies (Figure 3) are running currently on CAR T cell therapy in worldwide till 8th November, 2021.By US FDA (food and drug administration) till November, 2021 got approval five therapy of CAR T cell those are listed in Table 2 including their indications, yield institution and approval date.

Also exhibited in Figure 4 the OR rate and CR rate on clinical trials by each approved CAR therapy. There has a relation between some others disease along Covid-19 (listed in Figure 5) and CAR T cell such as allergic reaction exposed on mast cell including basophil, eosinophil while IgE bind with Fc𝗌RI receptor that can release inflammatory mediators’ results in type 1 hypersensitivity [26] .

Ads (allergic disease) caused by IgE derived from B cells that may target of this artificial T cells. Regulatory T cells (Tregs) able to control asthma with

Figure 3. Funnel graph on number of clinical studies about CAR T cell by worldwide [40] .

Table 2. List of currently existed CAR T therapy by FDA.

Abbreviation: R/R = Relapsed or Refractory, ALL = Acute Lymphoblastic Leukemia, MCL = Mantle Cell Lymphoma.

Figure 4. Efficacy rate of five approved CAR therapy [41] .

hypersensitivity that influenced by Th2 immune response [30] . To cure infectious disease CAR T cells constructs enchanting such as CD8+ efficient to remove antigens [35] . Cytotoxicity of CD8+ also proved its potency against infection caused by HIV-1. Consequently, CAR T cells used to treat HIV-1 and gives wonderful trace in HIV-1 treatment beyond oncology [36] [37] . Cardiac fibrosis is one condition of cardiac disease that have not any targeted treatment recently but an experiment on this re-designed T cell induced mice have cardiac fibrosis showed effective CAR T cell recognize FAP (fibroblast activation protein) and target on enabled heart fibroblast [38] [39] .

Figure 5. Relation between AIDs and viral infections with CAR T cells [2] .

4. CAR T Cell Therapy for Hematological & Solid Malignancy

4.1. CAR T Cell Therapy

Genetically modified chimeric antigen receptor (CAR) T cell selects the target tumor antigen then binds to it and kills the tumor cell in perforin. As CAR T therapy, we first collect blood from the patient’s body which is called leukapheresis then separate T cells from the blood. Later, by treating this T cell with a viral vector in the laboratory, we implant genetic materials in the T cell so that the receptors in the T cell grow. T cells containing this receptor are called Chimeric antigen receptor T cells. Next, we expand this generating CAR T cell then infuse this expanded CAR T cell into the patient’s body. Finally, this CAR T cell receptor binds to the antigen on the surface of the cancer cell in the patient’s body and kills the cancer cell. CAR T cell therapy is shown through Figure 6.

4.2. CAR-T Therapy for Hematological Malignancy

CAR-T cell immunotherapy is a significant treatment for hematological malignancies, which includes chronic lymphocytic leukemia (CLL), acute lymphoblastic leukemia (ALL), lymphoma, multiple myeloma (MM) etc.

4.2.1. Acute Lymphoblastic Leukemia (ALL)

The effective CAR for the treatment of acute lymphoblastic leukemia (ALL) is CD19. According to one clinical trial report, CD-19 infusions per 16 patients resulted in a complete remission (CR) rate of 88% among 15 patients [43] . Where CR is a detectable disease indicator that works based on PCR. The same trial applied to children and adult B-ALL patients showed that the CR rate is 70% [44] [45] . For ALL patients CD19 is an ideal target for CAR therapy because CD22 is a potential target, but this treatment shows anti-CD19 deficiency in the

Figure 6. CAR T cell therapy [42] .

patient’s body, which diminishes the response of B cells in the patient’s body [46] [47] [48] .

4.2.2. Chronic Lymphocytic Leukemia (CLL)

It is one kind of blood cancer. On this, CLL patients have some CAR T effects. In patients with chronic lymphocytic leukemia (CLL), there is an early immune deficiency that results in T-cell expansion not being properly due to which CAR T therapy efficacy is limited in these patients. To overcome this problem, a drug called Ibrutinib is used, which inhibits the proliferation of b-cells, increases T cell expansion, and increases antitumor capability [1] [49] . Other studies have shown that CAR T therapy can be used in cases of b-cell malignancy in patients with allogenic hematopoietic stem cell transplantation, but in this case a side-effect called graft-versus-host disease is seen in the patient’s body [50] [51] .

4.2.3. CAR T Therapy in Multiple Myeloma (mm)

This multiple myeloma is derived from bone marrow. Updated taken by Criteria for the Classification of Monoclonal Gammopathies, Multiple Myeloma and Related Disorders: A Report of the International Myeloma Working Group. Different types of treatment like autologous hematopoietic stem cell transplantation, chemotherapy, immune modulatory agents, etc. are not possible to treat this mm due to the abnormalities of myeloma cells [52] [53] . But according to some reports, CD19 has been shown to work on myeloma cells as well [54] [55] . But anti-CD19 may not play a vital role in myeloma cells, but it is harmful to more healthy tissue [56] . According to some clinical trials, CD138 CAR T cell therapy was used as the primary diagnostic marker in 5 multiple myeloma patients, resulting in stable condition in 4 patients, and in one patient, it was found that myeloma cells in the peripheral blood were reduced with advance plasma cell leukemia [57] .

4.3. CAR T Cell Therapy for Solid Malignancy

In the case of solid malignancy, the next step in genetically modifying the T cell is to infuse it into the body. But, due to the presence of some barriers such as ineffective T cell trafficking, immunosuppressive microenvironment, T cell infiltration, etc., this CAR T treatment is still in the clinical phase in the field of solid malignancy.

4.3.1. Ineffective T Cell Trafficking

The presence of tumor-infiltrating lymphocytes (TIL) in the field of solid cancer patients indicates a positive clinical outcome [58] [59] [60] [61] . The better the antitumor response in the patient, the more cytotoxic T lymphocytes it has. There must be proper trafficking of cytotoxic T lymphocytes. CTL is a type of immune cell that kills different types of foreign cells, cancer cells. Some factors work in the field of proper CTL trafficking, like mismatching of chemokine-chemokine receptor pairs, down regulation of adhesion molecules, aberrant vascular etc. which act as the main barriers to CTL trafficking [62] . Proper chemokine production plays an important role in this. Proper chemokine production from the cell will enable CAR T cells to migrate to myeloma cells, so CAR T will be able to show its function on myeloma cells [63] .

4.3.2. Immunosuppressive Microenvironment (IM)

Another barrier in the way of CAR T treatment in the field of solid malignancy is the tumor microenvironment. It is a key determinant of antitumor immunity with the capacity to inhibit the activation, infiltration, and effector activity of T cells. But immune suppressor, and some other obstacles like immunosuppression cytokines, inhibitory immune checkpoint which suppresses CAR T function from being present in the tumor microenvironment.

4.3.3. T Cell Infiltration

We use extracellular matrix, the main component of which is heparin sulfate proteoglycans (HSPGs), to help in the infiltration of T cells into the tumor microenvironment. HSPGs are produced by solid tumors, rather than by T cells produced by stem cells in the body [64] .

5. Result & Discussion

In treating malignant growths, CAR T cells have shown tremendous outcomes. But the therapy is still not optimal, because of the idea of the product, which is a “living medication,” proposing its starting point (patient wellness) and quality. It is important to note that, to date, all CAR studies have, in any case, identified extreme grade 3 or 4 clinical AEs and passed. In the present survey, novel, imaginative ways to deal with managing the flexibility of CAR-T cells by consolidating rather than with exogenous pharmacologic medication (see Table 3 for an outline) [65] .

Table 3. A study of selecting for combining CAR-T cells and small molecules/drugs.

The first apparent utility combines the effects of CAR therapy to sensitize tumor cells with drugs to further coordinate the effects of the two treatments. It demonstrates to the authorizing TME the strong tumor treatment mechanisms for relieving and non-prevention. This treatment also means more and often unexpected aspects, and patients will need careful observation and counseling during the effects and initial verification. Although it is elegant and able to protect patients from unwanted effects, it can be expensive and, when compared to patient-compatible CAR drug costs, may be a small amount of the total treatment cost [65] .

To summarize the discussion, the combination of pharmacological drugs with CAR treatment is innovative and elegant. A personalized adaptation of the “living drug” will be an important step in improving the effectiveness of CAR therapy. The FDA and EMA approved those drugs, so they can easily be transferred to use in the clinic. Hopefully, future CAR/drug trials will show efficient clinical results in the near future [65] .

*Just for the acknowledgment, stimulate, and signaling domains are depicted in order to grease the understanding. Fresh leader sequence, hinge/stalk, transmembrane domains, markers, and epitomes were removed when inapplicable. A rent (/) means that either one or further of the stimulate domains is present. N/A non-available. AML acute eyelid leukemia; B ALL precursor B acute lymphoid leukemia; Bcl-xL: Bcl extra-large; Bcl-2: B cell lymphoma 2; C2H2: Cys2-His2; CLL: chronic lymphoid leukemia; CAIX carbonic anhydrase IX; CRS cytokines release syndrome; CRBN: cerebelon; EGFR (epidermal growth factor receptor); FKBP: FK506 binding protein; FLT3: fms-like tyrosine kinase 3; FRB (FKBP-rapamycin-binding); GD2: disialoganglioside; GBM: globalism multiform; GM-CSF granulocyte-macrophage colony stimulating factor; HCV: Hepatitis C Virus; HER-2: epidermal growth factor receptor 2; hRBP4: human retinol retinal binding protein 4; IFN-γ: interferon gamma; iMC: ineducable MyD88/CD40; La/SS-B: human nuclear auto-antigen La/SS-B; LD3: human apolipoprotein E4 E; LID: ligand-induced dimerization; MDM2: mouse double minute 2 homolog; MDSC (myeloid-derived suppressor cells); Meso: mesothelin; NHL (non-Hodgkin lymphoma); Nos: nitric oxides; PD-1 (programmed cell death protein 1); PKC: protein kinase C; PSCA: prostate stem cell antigen; RCC: renal cell carcinoma; PSMA (prostate-specific membrane antigen); scFv: single-chain variable fragment; zip: leucine-zipper; SMAC (second mitochondria-derived activator of caspase); tCD34: truncated CD34; TNF-α (tumor necrosis factor alpha); tNGFR (truncated nerve growth factor receptor) [65] .

6. Toxicity Management

Though CAR T cell therapy proved its powerful efficacy against various malignancies but this therapy also represents some toxicities during therapy as this is cellular product such as infusion reaction toxicities, tumor lysis, graft vs host, on target off tumor, on target on tumor, hypogammaglobulinemia. In this therapy identify, schism, and regulation of those toxicities is very fateful action to perfectly complete whole therapy. Different modes of action are responsible for toxicities of CAR T cell therapy. If this cell work on normal tissue instead of aimed cell then those normal tissue may be destroyed as anti CD19 responsible for gulp normal B cells [97] [98] [99] . Another reason of normal tissue spoil is that protein is not revealed on cancer cell suddenly cross interaction by CAR T cell [100] [101] . CAR T cell infusion may cause of TLS (tumor lysis syndrome) and acute anaphylaxis [1] [102] [103] [104] [105] . CRS (cytokine release syndrome) with morbidities including hypertension and fever is the salient and elaborately explained (Figure 7) toxicity by infusion of CAR T cells [43] [106] - [111] . This immunotherapy may causes of neurological toxicities while it run with CRS or without CRS [106] [107] [112] . Usually ruined which tissues exposed antigen and recognized by CAR. During progression of this T cells the mechanism is liable for toxicity that can be repress but not terminated because of entire search for targeted antigen that revealed on normal tissues.

6.1. Acute Systemic Inflammatory Syndrome or CRS

In this therapy it’s a major enough dangerous adverse effect that treated by tisagenlecleucel is 77% [114] and 57% [115] gradually in patients who have NHL and ALL. In backward patient of NHL with CRS treated 93% by axicabtagene

Figure 7. CRS symptoms by different organs of human body [113] .

ciloleucil [116] Each grade of CRS has different symptoms and management system (Figure 8).

6.2. Neuropsychiatric Syndrome or ICANS

This kind of toxicity is another general side effect of CAR T cell therapy that’s symptoms are very apparently connected to CAR product, patient age, disease condition and ICANS may occurred during CRS or individually [114] [117] [118] . ASTCT also appending ICANS into consensus based grading system to clinical experiment and regular use [117] with ICE (Immune effector cell encephalopathy) score, some general recommendation, symptoms of cerebral oedema. For ICANS managements or prevention there have no approved treatment though it can be controlled initially by some supportive cares (Figure 9).

7. CAR Combining with Oncolytic Virus Therapy

Along this startling immunotherapy some other materials of treatment may use and this kind of therapy mentioned as combinational system. Solid tumors treated by this combinatory CAR method without exposing any heterogenic effect and obviously it succeeds to increase therapeutic window of this chimeric

Figure 8. Symptoms and Management of CRS as Grading [113] .

Figure 9. Symptoms and Management of ICANS grades [119] .

antigen receptor T cell therapy. One clinical trial on OVs (oncolytic) viruses proved as a plight product for treatment of solid tumor and FDA approved GM-CSF against advanced malignant melanoma that able to express OHVs (oncolytic herpes virus) [120] . This oncolytic virus therapy capable to work on specific target and destroy malignant cells omitted normal tissue. During need to active immune system for given risky signals of carcinogenic cells these virus results in lysis of cancer tumors through deliver of virus lineage and expansion of virus inoculum [121] . Yet now many clinical studies either in vitro or in vivo drive to determine synergism between OVs and CAR cell that listed in Table 5 Contemporary there may yield irrepressible synergic effect on CAR T cell while OVs engineered in such a way it pushes few immunomodulatory components to expose. But in case of better non-engineered OVs with CAR they may express different outcomes. Suchlike GD2 CAR T cell proliferation enhanced by HSV1716 in melanoma and rhabdomyosarcoma cancer [122] . By these experiments (Table 4) OVs therapy in single or combination recommended to curbed of tumor cell.

Table 4. Preclinical investigation of CAR combining with OVs.

8. Commercialization of CAR-T Cell

FDA-supported items are monetarily accessible for authorized signs altogether and DLBCL there is a quickly developing business in conveying CAR-T cell. CAR T cells are reasonable to expect to be that this extensive monetary motivating force is adding to current publicity concerning this treatment. Biotechnology organizations have drawn in interest in the request for billions of US dollars on the premise that CAR-T cell items will be industrially fruitful, yet this ought not to occupy from a target appraisal of the proof on which we base treatment choices. In the event that this treatment does satisfy its prospects, a political discussion will be required concerning admittance to therapeutic medicines either being an advantage for the rich or an appropriate for those out of luck [133] .

The tailor-made assembling processes presently utilized for profoundly customized designed T cell treatments bring about significantly expenses. The expense of manufacturing Car T cells is relied upon to diminish. A point-by-point examination of the general wellbeing consideration of the evaluation of quality changed cells is past the extent of this analysis. However, some factors have as of late been summed up [30] .

9. Application of CAR T Cell Therapy

The treatment methods of CAR-T cell therapy that are being used to treat cancer and in various clinical trials include hematological malignancies, which include ALL; acute lymphoblastic leukemia, CLL; chronic lymphoblastic leukemia, lymphoma and MM; multiple myeloma. Also, application of CAR therapy research development plan and development is underway for myeloma, sarcoma and breast cancer treatment which includes solid tumors. The use of CAR-T cell immunotherapy in the context of advances in the treatment of various cancers and its practical success is as follows: The use of CAR-T cell therapy in various clinical trials to treat ALL, CLL and its outcome is shown in Figure 10 and also treat MM, Lymphoma, solid tumor in various clinical trials their antigen uses or data information summarized in Tables 5-9.

9.1. CAR T Cell Therapy for ALL (Acute Lymphoblastic Leukemia)

Table 5. Effects of CAR therapy on ALL through selected clinical trials.

9.2. CAR-T Cell Therapy for CLL (Chronic Lymphocytic Leukemia)

Table 6. Effects of CAR therapy on CLL through selected clinical trials.

Figure 10. Outcomes of CR (Complete Remission) rate in CLL (chronic lymphocytic leukemia) and ALL (acute lymphoblastic leukemia) obtained by clinical trials using certain CAR antigens selected at different institutes [112] .

9.3. CAR-T Cell Therapy for Lymphoma

Table 7. Effects of CAR therapy on Lymphoma through selected clinical trials.

9.4. CAR-T Cell Therapy for MM (Multiple Myeloma)

Table 8. Effects of CAR therapy on MM through selected clinical trials.

Abbreviations: ALL (acute lymphoblastic leukemia); CAR t cell (chimeric antigen receptor T cell); BCMA (B cell maturation antigen); MM (multiple myeloma); CR (complete remission); M/F (male and female); PR (partial remission); Ref (reference); SD (stable disease); CLL ( chronic lymphocytic leukemia); aThe calculation is the total number of samples. bIn the trials has two groups; one contains 42 primary refractory/hematological relapsed and 9 refractory minimal residual disease by flow cytometry patients of B-ALL. cIn the trial two patients died from related treatment mortality. d4 CLL patients are including. eThere are CLL patients 8. fNo gender indicated. gThere are 11 lymphoma patients. hIn this clinical trial has two main groups, one is NHL (non-Hodgkin lymphoma) and the other one is CLL (chronic lymphocytic leukemia). iThere are 4 patients were not available, among which 2 died. jThis study apply the accurate value instead of rate, where the response rate is meaningless (sample size is less than 10). kBecause of screen fail (n = 2); five patients are not receiving treatment, rapid MM progression/renal failure (n = 2), self-choice (n = 1). Age of patients are expressed ± mean standard error of mean and whether the data are available [42] .

9.5. CAR-T Cell Therapy for Solid Tumors

Table 9. The application of some CAR antigen on different solid tumors.

10. Future Opportunity

The initiatory advancement of CAR-T cell immunotherapy treatments has concentrated amazingly on ALL, commonplace cancers in young people. Over 80% of kids are determined to have ALL that emerges in B cells. The dominating kind of pediatric ALL will be restored by serious chemotherapy therefore patients whose diseases return after chemotherapy or a foundational microorganism relocates, treatment choices are few and far between.

CAR-T cells have made significant progress in the treatment of hematological malignancies in recent years. However, it is still in the development stage and more advanced quality measures are being taken to prevent CRS with various adverse and side effects arising after providing CAR medical service to patients [42] . A review revealed that a bivalent tandem CAR (TanCAR) was intended to intervene in the bispecific initiation of immune system microorganisms [149] . And also proposed that tandem CAR (TanCAR) reactivated T cells against two unique antigens. Also, TanCAR delivered a synergistic improvement in T lymphocyte enactment [149] . Treatment for malignity in hematology, which might be possible Besides, with the advancement of universal CAR immunotherapy for hematological malignancies treatment, which can stay away from the issue that autologous T lymphocytes of patients are hard to acquire and diminish the expense of treatment, and then conjecture that the use of CRISPER innovation to alter allogeneic qualities gives potential to the progress of uniCAR T cell treatment [150] . Another technique called the “suicide gene system” is likewise commendable and merits consideration [151] . Artificially incited iCas9 dimerization is quite possibly the most noticeable self-destruction gene in lymphocytes based on immunotherapy, which has shown exceptional proficiency in clinical preliminaries of patients [152] . As a general rule, these treatments may stay away from the unfriendly occasions brought about via CAR-T cell immunotherapy for optional medication for cancer treatment, which additionally shows a new opportunity for the future [150] .

11. Conclusion

Chimeric antigen receptor T cells are creating the conversion from simply “favorable” to being “fruitful” for hematological virulence’s treatment. As we pursue to improve the ability of CAR T cells immunotherapy, that expresses chimeric tumor-targeting receptors and allows them to target in the lump micro-environment. We can anticipate broader application beyond hematological carcinomas and into solid tumors. With rising interest in the sphere from profit-oriented entities, we are optimistic that the evolution and clinical execution of this sensational perspective will now hasten.

Conflicts of Interest

The authors declare no conflicts of interest regarding the publication of this paper.

References

[1] Porter, D.L., et al. (2016) Chimeric Antigen Receptor T Cells Persist and Induce Sustained Remissions in Relapsed Refractory Chronic Lymphocytic Leukemia. Science Translational Medicine, 7, 303ra139.
https://doi.org/10.1126/scitranslmed.aac5415
[2] Zmievskaya, E., Valiullina, A., Ganeeva, I., Petukhov, A., Rizvanov, A. and Bulatov, E. (2021) Application of CAR-T Cell Therapy beyond Oncology: Autoimmune Diseases and Viral Infections. Biomedicines, 9, Article No. 59.
https://doi.org/10.3390/biomedicines9010059
[3] Sharpe, M. and Mount, N. (2015) Genetically Modified T Cells in Cancer Therapy: Opportunities and Challenges. Disease Models and Mechanisms, 8, 337-350.
https://doi.org/10.1242/dmm.018036
[4] McGuirk, J., et al. (2017) Building Blocks for Institutional Preparation of CTL019 Delivery. Cytotherapy, 19, 1015-1024.
https://doi.org/10.1016/j.jcyt.2017.06.001
[5] Houot, R., Schultz, L.M., Marabelle, A. and Kohrt, H. (2015) T-Cell-Based Immunotherapy: Adoptive Cell Transfer and Checkpoint Inhibition. Cancer Immunology Research, 3, 1115-1122.
https://doi.org/10.1158/2326-6066.CIR-15-0190
[6] Barrett, D.M., Grupp, S.A. and June, C.H. (2015) Chimeric Antigen Receptor- and TCR-Modified T Cells Enter Main Street and Wall Street. The Journal of Immunology, 195, 755-761.
https://doi.org/10.4049/jimmunol.1500751
[7] Verdegaal, E.M.E., et al. (2016) Neoantigen Landscape Dynamics during Human Melanoma-T Cell Interactions. Nature, 536, 91-95.
https://doi.org/10.1038/nature18945
[8] Maus, M.V., Grupp, S.A., Porter, D.L. and June, C.H. (2014) Antibody-Modified T Cells: CARs Take the Front Seat for Hematologic Malignancies. Blood, 123, 2625-2635.
https://doi.org/10.1182/blood-2013-11-492231
[9] Kakarla, S. and Gottschalk, S. (2014) CAR T Cells for Solid Tumors Armed and Ready to Go?
https://www.journalppo.com/
[10] Gross, G., Waks, T. and Eshhar, Z. (1989) Expression of Immunoglobulin-T-cell Receptor Chimeric Molecules as Functional Receptors with Antibody-Type Specificity. Proceedings of the National Academy of Sciences of the United States of America, 86, 10024-10028.
https://doi.org/10.1073/pnas.86.24.10024
[11] Enblad, G., Karlsson, H. and Loskog, A.S.I. (2015) CAR T-Cell Therapy: The Role of Physical Barriers and Immunosuppression in Lymphoma. Human Gene Therapy, 26, 498-505.
https://doi.org/10.1089/hum.2015.054
[12] Tang, H., Qiao, J. and Fu, Y.X. (2016) Immunotherapy and Tumor Microenvironment. Cancer Letters, 370, 85-90.
https://doi.org/10.1016/j.canlet.2015.10.009
[13] Levine, B.L. (2015) Performance-Enhancing Drugs: Design and Production of Redirected Chimeric Antigen Receptor (CAR) T Cells. Cancer Gene Therapy, 22, 79-84.
https://doi.org/10.1038/cgt.2015.5
[14] Maus, M.V. and Levine, B.L. (2016) Chimeric Antigen Receptor T-Cell Therapy for the Community Oncologist. The Oncologist, 21, 608-617.
https://doi.org/10.1634/theoncologist.2015-0421
[15] Tanaka, J., Mielcarek, M. and Torok-Storb, B. (1998) Impaired Induction of the CD28-Responsive Complex in Granulocyte Colony-Stimulating Factor Mobilized CD4 T Cells. Blood, 91, 347-352.
https://doi.org/10.1182/blood.V91.1.347
[16] Shank, B.R., Do, B., Sevin, A., Chen, S.E., Neelapu, S.S. and Horowitz, S.B. (2017) Chimeric Antigen Receptor T Cells in Hematologic Malignancies. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy, 37, Article ID: 334345.
https://doi.org/10.1002/phar.1900
[17] Vormittag, P., Gunn, R., Ghorashian, S. and Veraitch, F.S. (2018) A Guide to Manufacturing CAR T Cell Therapies. Current Opinion in Biotechnology, 53, 164-181.
https://doi.org/10.1016/j.copbio.2018.01.025
[18] Eshhar, Z., Waks, T., Gross, G. and Schindler, D.G. (1993) Specific Activation and Targeting of Cytotoxic Lymphocytes through Chimeric Single Chains Consisting of Antibody-Binding Domains and the γ or ζ Subunits of the Immunoglobulin and T-Cell Receptors. Proceedings of the National Academy of Sciences of the United States of America, 90, 720-724.
https://doi.org/10.1073/pnas.90.2.720
[19] Maher, J. (2012) Immunotherapy of Malignant Disease Using Chimeric Antigen Receptor Engrafted T Cells. International Scholarly Research Notices, 2012, Article ID: 278093.
https://doi.org/10.5402/2012/278093
[20] Stone J.D. and Kranz, D.M. (2013) Role of T Cell Receptor Affinity in the Efficacy and Specificity of Adoptive T Cell Therapies. Frontiers in Immunology, 4, Article No. 244.
https://doi.org/10.3389/fimmu.2013.00244
[21] Kershaw, M. H., et al. (2006) A Phase I Study on Adoptive Immunotherapy Using Gene-Modified T Cells for Ovarian Cancer. Clinical Cancer Research, 12, 6106-6115.
[22] Rodriguez-Cartagena, L.G., Bowles, B.S., Kurani, S.S., Windebank, A.J., Kenderian, S.S. and Greenberg-Worisek, A.J. (2018) Chimeric Antigen Receptor T-Cells: Successful Translation of the First Cell and Gene Therapy from Bench to Bedside. Clinical and Translational Science, 11, 537-539.
https://doi.org/10.1111/cts.12586
[23] Smith, A.J., Oertle, J., Warren, D. and Prato, D. (2016) Chimeric Antigen Receptor (CAR) T Cell Therapy for Malignant Cancers: Summary and Perspective. Journal of Cellular Immunotherapy, 2, 59-68.
https://doi.org/10.1016/j.jocit.2016.08.001
[24] Bird, R.E., Hardman, K.D., Jacobson, J.W., Johnson, S., Kaufman, B., et al. (1988) Single-Chain Antigen-Binding Proteins. Science, 242, 423-426.
https://doi.org/10.1126/science.3140379
[25] Chen, X., Zaro, J.L. and Shen, W.C. (2013) Fusion Protein Linkers: Property, Design and Functionality. Advanced Drug Delivery Reviews, 65, 1357-1369.
https://doi.org/10.1016/j.addr.2012.09.039
[26] Ward, D.E., Fay, B.L., Adejuwon, A., Han, H. and Ma, Z. (2018) Chimeric Antigen Receptors Based on Low Affinity Mutants of FcεRI Re-Direct T Cell Specificity to Cells Expressing Membrane IgE. Frontiers in Immunology, 9, Article No. 2231.
https://doi.org/10.3389/fimmu.2018.02231
[27] Hudecek, M., et al. (2015) The NonSignaling Extracellular Spacer Domain of Chimeric Antigen Receptors Is Decisive for in Vivo Antitumor Activity. Cancer Immunology Research, 3, 125-135.
https://doi.org/10.1158/2326-6066.CIR-14-0127
[28] Hombach, A.A., Schildgen, V., Heuser, C., Finnern, R., Gilham, D.E. and Abken, H. (2007) T Cell Activation by Antibody-Like Immunoreceptors: The Position of the Binding Epitope within the Target Molecule Determines the Efficiency of Activation of Redirected T Cells. The Journal of Immunology, 178, 4650-4657.
https://doi.org/10.4049/jimmunol.178.7.4650
[29] Zhang, T., Wu, M.-R. and Sentman, C.L. (2012) An NKp30-Based Chimeric Antigen Receptor Promotes T Cell Effector Functions and Antitumor Efficacy In Vivo. The Journal of Immunology, 189, 2290-2299.
https://doi.org/10.4049/jimmunol.1103495
[30] Guedan, S., et al. (2018) Enhancing CAR T Cell Persistence through ICOS and 4-1BB Costimulation. JCI Insight, 3, e96976.
https://doi.org/10.1172/jci.insight.96976
[31] Zabel, M., Tauber, P.A. and Pickl, W.F. (2019) The Making and Function of CAR Cells. Immunology Letters, 212, 53-69.
https://doi.org/10.1016/j.imlet.2019.06.002
[32] Smith, J.W. (1997) Apheresis Techniques and Cellular Immunomodulation. Therapeutic Apheresis, 1, 20-206.
https://doi.org/10.1111/j.1744-9987.1997.tb00137.x
[33] Suhoski, M.M., et al. (2007) Engineering Artificial Antigen-Presenting Cells to Express a Diverse Array of Co-Stimulatory Molecules. Molecular Therapy, 15, 981-988.
https://doi.org/10.1038/mt.sj.6300134
[34] Leyfman, Y. (2018) Chimeric Antigen Receptors: Unleashing a New Age of Anti-Cancer Therapy. Cancer Cell International, 18, Article No. 182.
https://doi.org/10.1186/s12935-018-0685-x
[35] Seif, M., Einsele, H. and Loffler, J. (2019) CAR T Cells beyond Cancer: Hope for Immunomodulatory Therapy of Infectious Diseases. Frontiers in Immunology, 10, Article No. 2711.
https://doi.org/10.3389/fimmu.2019.02711
[36] Masiero, S., et al. (2005) T-Cell Engineering by a Chimeric T-Cell Receptor with Antibody-Type Specificity for the HIV-1 gp120. Gene Therapy, 12, 299-310.
https://doi.org/10.1038/sj.gt.3302413
[37] Liu, B., Zhang, W. and Zhang, H. (2019) Development of CAR-T Cells for Long-Term Eradication and Surveillance of HIV-1 Reservoir. Current Opinion in Virology, 38, 21-30.
https://doi.org/10.1016/j.coviro.2019.04.004
[38] Vagnozzi, R.J., Johansen, A.K.Z. and Molkentin, J.D. (2019) CARdiac Immunotherapy: T Cells Engineered to Treat the Fibrotic Heart. Molecular Therapy, 27, 1869-1871.
https://doi.org/10.1016/j.ymthe.2019.09.021
[39] Aghajanian, H., et al. (2019) Targeting Cardiac Fibrosis with Engineered T Cells. Nature, 573, 430-433.
https://doi.org/10.1038/s41586-019-1546-z
[40] CAR T Cell immunotherapy—List Results. ClinicalTrials.gov.
https://clinicaltrials.gov/ct2/results?cond=CAR+T+Cell+immunotherapy&term=&cntry=&state=&city=&dist
[41] FDA.
https://www.fda.gov/search?s=CAR+T+cell+efficacy+rate
[42] Zhao, Z., Chen, Y., Francisco, N.M., Zhang, Y. and Wu, M. (2018) The Application of CAR-T Cell Therapy in Hematological Malignancies: Advantages and Challenges. Acta Pharmaceutica Sinica B, 8, 539-551.
https://doi.org/10.1016/j.apsb.2018.03.001
[43] Davila, M.L., et al. (2014) Efficacy and Toxicity Management of 19-28z CAR T Cell Therapy. Science Translational Medicine, 6, 224-225.
https://doi.org/10.1126/scitranslmed.3008226
[44] Davila, M.L. and Sadelain, M. (2016) Biology and Clinical Application of CAR T Cells for B Cell Malignancies. International Journal of Hematology, 104, 6-17.
https://doi.org/10.1007/s12185-016-2039-6
[45] Lee, D.W., et al. (2015) T Cells Expressing CD19 Chimeric Antigen Receptors for Acute Lymphoblastic Leukaemia in Children and Young Adults: A Phase 1 Dose-Escalation Trial. The Lancet, 385, 517-528.
https://doi.org/10.1016/S0140-6736(14)61403-3
[46] Shalabi, H., Angiolillo, A. and Fry, T.J. (2015) Beyond CD19: Opportunities for Future Development of Targeted Immunotherapy in Pediatric Relapsed-Refractory Acute Leukemia. Frontiers in Pediatrics, 3, Article No. 80.
https://doi.org/10.3389/fped.2015.00080
[47] Maude, S.L., Barrett, D., Teachey, D.T. and Grupp, S.A. (2014) Managing Cytokine Release Syndrome Associated with Novel T Cell-Engaging Therapies. The Cancer Journal, 20, 119-122.
https://doi.org/10.1097/PPO.0000000000000035
[48] Zah, E., Lin, M.Y., Anne, S.B., Jensen, M.C. and Chen, Y.Y. (2016) T Cells Expressing CD19/CD20 Bispecific Chimeric Antigen Receptors Prevent Antigen Escape by Malignant B Cells. Cancer Immunology Research, 4, 498-508.
https://doi.org/10.1158/2326-6066.CIR-15-0231
[49] Fraietta, J.A., et al. (2016) Ibrutinib Enhances Chimeric Antigen Receptor T-Cell Engraftment and Efficacy in Leukemia. Blood, 127, 1117-1127.
https://doi.org/10.1182/blood-2015-11-679134
[50] Roddie, C. and Peggs, K.S. (2011) Donor Lymphocyte Infusion following Allogeneic Hematopoietic Stem Cell Transplantation. Expert Opinion on Biological Therapy, 11, 473-487.
https://doi.org/10.1517/14712598.2011.554811
[51] Frey, N.V. and Porter, D.L. (2008) Graft-versus-Host Disease after Donor Leukocyte Infusions: Presentation and Management. Best Practice & Research Clinical Haematology, 21, 205-222.
https://doi.org/10.1016/j.beha.2008.02.007
[52] Chung, C. (2017) Role of Immunotherapy in Targeting the Bone Marrow Microenvironment in Multiple Myeloma: An Evolving Therapeutic Strategy. Pharmacotherapy: The Journal of Human Pharmacology and Drug Therapy, 37, 129-143.
https://doi.org/10.1002/phar.1871
[53] Zhang, K., Desai, A., Zeng, D., Gong, T., Lu, P. and Wang, M. (2017) Magic Year for Multiple Myeloma Therapeutics: Key Takeaways from the ASH 2015 Annual Meeting. Oncotarget, 8, 10748-10759.
https://doi.org/10.18632/oncotarget.13314
[54] Garfall, A.L., et al. (2015) Chimeric Antigen Receptor T Cells against CD19 for Multiple Myeloma. The New England Journal of Medicine, 373, 1040-1047.
https://doi.org/10.1056/NEJMoa1504542
[55] Hajek, R., Okubote, S.A. and Svachova, H. (2013) Myeloma Stem Cell Concepts, Heterogeneity and Plasticity of Multiple Myeloma. British Journal of Haematology, 163, 551-564.
https://doi.org/10.1111/bjh.12563
[56] Atanackovic, D., Radhakrishnan, S.V., Bhardwaj, N. and Luetkens, T. (2016) Chimeric Antigen Receptor (CAR) Therapy for Multiple Myeloma. British Journal of Haematology, 172, 685-698.
https://doi.org/10.1111/bjh.13889
[57] Guo, B., et al. (2016) CD138-Directed Adoptive Immunotherapy of Chimeric Antigen Receptor (CAR)-Modified T Cells for Multiple Myeloma. Journal of Cellular Immunotherapy, 2, 28-35.
https://doi.org/10.1016/j.jocit.2014.11.001
[58] Duong, C.P.M., Yong, C.S.M., Kershaw, M.H., Slaney, C.Y. and Darcy, P.K. (2015) Cancer Immunotherapy Utilizing Gene-Modified T Cells: From the Bench to the Clinic. Molecular Immunology, 67, 46-57.
https://doi.org/10.1016/j.molimm.2014.12.009
[59] Kim, S.T., et al. (2013) Tumor-Infiltrating Lymphocytes, Tumor Characteristics, and Recurrence in Patients with Early Breast Cancer. American Journal of Clinical Oncology: Cancer Clinical Trials, 36, 224-231.
https://doi.org/10.1097/COC.0b013e3182467d90
[60] Kmiecik, J., et al. (2013) Elevated CD3+ and CD8+ Tumor-Infiltrating Immune Cells Correlate with Prolonged Survival in Glioblastoma Patients despite Integrated Immunosuppressive Mechanisms in the Tumor Microenvironment and at the Systemic Level. Journal of Neuroimmunology, 264, 71-83.
https://doi.org/10.1016/j.jneuroim.2013.08.013
[61] Van der Burg, S.H. and Palefsky, J.M. (2009) Human Immunodeficiency Virus and Human Papilloma Virus—Why HPV-Induced Lesions Do Not Spontaneously Resolve and Why Therapeutic Vaccination Can Be Successful. Journal of Translational Medicine, 7, Article No. 108.
https://doi.org/10.1186/1479-5876-7-108
[62] Slaney, C.Y., Kershaw, M.H. and Darcy, P.K. (2014) Trafficking of T Cells into Tumors. Cancer Research, 74, 7168-7174.
https://doi.org/10.1158/0008-5472.CAN-14-2458
[63] Kershaw, M.H., et al. (2002) Redirecting Migration of T Cells to Chemokine Secreted from Tumors by Genetic Modification with CXCR2. Human Gene Therapy, 13, 1971-1980.
https://doi.org/10.1089/10430340260355374
[64] Caruana, I., et al. (2015) Heparanase Promotes Tumor Infiltration and Antitumor Activity of CAR-Redirected T Lymphocytes. Nature Medicine, 21, 524-529.
https://doi.org/10.1038/nm.3833
[65] Caulier, B., Enserink, J.M. and Walchli, S. (2021) Pharmacologic Control of CAR T Cells. International Journal of Molecular Sciences, 22, Article No. 4320.
https://doi.org/10.3390/ijms22094320
[66] Jan, M., et al. (2021) Reversible ON- and OFF-Switch Chimeric Antigen Receptors Controlled by Lenalidomide. Science Translational Medicine, 13, eabb6295.
https://doi.org/10.1126/scitranslmed.abb6295
[67] Richman, S.A., Wang, L.C., Moon, E.K., Khire, U.R., Albelda, S.M. and Milone, M.C. (2020) Ligand-Induced Degradation of a CAR Permits Reversible Remote Control of CAR T Cell Activity in Vitro and in Vivo. Molecular Therapy, 28, 1600-1613.
https://doi.org/10.1016/j.ymthe.2020.06.004
[68] Lee, S.M., et al. (2020) A Chemical Switch System to Modulate Chimeric Antigen Receptor T Cell Activity through Proteolysis-Targeting Chimaera Technology. ACS Synthetic Biology, 9, 987-992.
https://doi.org/10.1021/acssynbio.9b00476
[69] Giordano-Attianese, G., et al. (2020) A Computationally Designed Chimeric Antigen Receptor Provides a Small-Molecule Safety Switch for T-Cell Therapy. Nature Biotechnology, 38, 426-432.
https://doi.org/10.1038/s41587-019-0403-9
[70] Zajc, C.U., et al. (2020) A Conformation-Specific ON-Switch for Controlling CAR T Cells with an Orally Available Drug. Proceedings of the National Academy of Sciences of the United States of America, 117, 14926-14935.
https://doi.org/10.1073/pnas.1911154117
[71] Juillerat, A., et al. (2019) Modulation of Chimeric Antigen Receptor Surface Expression by a Small Molecule Switch. BMC Biotechnology, 19, Article No. 44.
https://doi.org/10.1186/s12896-019-0537-3
[72] Weber, E.W., Lynn, R.C., Sotillo, E., Lattin, J., Xu, P. and Mackall, C.L. (2019) Pharmacologic Control of CAR-T Cell Function Using Dasatinib. Blood Advances, 3, 711-717.
https://doi.org/10.1182/bloodadvances.2018028720
[73] Mestermann, K., et al. (2019) The Tyrosine Kinase Inhibitor Dasatinib Acts as a Pharmacologic on/off Switch for CAR T Cells. Science Translational Medicine, 11, eaau5907.
https://doi.org/10.1126/scitranslmed.aau5907
[74] Hill, Z.B., Martinko, A.J., Nguyen, D.P. and Wells, J.A. (2018) Human Antibody-Based Chemically Induced Dimerizers for Cell Therapeutic Applications. Nature Chemical Biology, 14, 112-117.
https://doi.org/10.1038/nchembio.2529
[75] Foster, A.E., et al. (2017) Regulated Expansion and Survival of Chimeric Antigen Receptor-Modified T Cells Using Small Molecule-Dependent Inducible MyD88/CD40. Molecular Therapy, 25, 2176-2188.
https://doi.org/10.1016/j.ymthe.2017.06.014
[76] Wu, C.Y., Roybal, K.T., Puchner, E.M., Onuffer, J. and Lim, W.A. (2015) Remote Control of Therapeutic T Cells through a Small Molecule-Gated Chimeric Receptor. Science, 350, aab4077.
https://doi.org/10.1126/science.aab4077
[77] Park, S., et al. (2021) Direct Control of CAR T Cells through Small Molecule-Regulated Antibodies. Nature Communications, 12, Article No. 710.
https://doi.org/10.1038/s41467-020-20671-6
[78] Cho, J.H., Collins, J.J. and Wong, W.W. (2018) Universal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell Responses. Cell, 173, 1426-1438.
https://doi.org/10.1016/j.cell.2018.03.038
[79] Cartellieri, M., et al. (2016) Switching CAR T Cells on and off: A Novel Modular Platform for Retargeting of T Cells to AML Blasts. Blood Cancer Journal, 6, e458.
https://doi.org/10.1038/bcj.2016.61
[80] Ma, J.S.Y., et al. (2016) Versatile Strategy for Controlling the Specificity and Activity of Engineered T Cells. Proceedings of the National Academy of Sciences of the United States of America, 113, E450-E458.
https://doi.org/10.1073/pnas.1524193113
[81] Dufva, O., et al. (2020) Integrated Drug Profiling and CRISPR Screening Identify Essential Pathways for CAR T-Cell Cytotoxicity. Blood, 135, 597-609.
https://doi.org/10.1182/blood.2019002121
http://ashpublications.org/blood/article-pdf/135/9/597/1717506/bloodbld2019002121.pdf
[82] Li, H., et al. (2019) CAIX-Specific CAR-T Cells and Sunitinib Show Synergistic Effects against Metastatic Renal Cancer Models. Journal of Immunotherapy, 43, 16-28.
https://doi.org/10.1097/CJI.0000000000000301
https://www.immunotherapy-journal.com/
[83] Torres-Collado, A.X. and Jazirehi, A.R. (2018) Overcoming Resistance of Human Non-Hodgkin’s Lymphoma to CD19-CAR CTL Therapy by Celecoxib and Histone Deacetylase Inhibitors. Cancers, 10, Article No. 200.
https://doi.org/10.3390/cancers10060200
[84] Jetani, H., et al. (2018) CAR T-Cells Targeting FLT3 Have Potent Activity against FLT3ITD+ AML and Act Synergistically with the FLT3-Inhibitor Crenolanib. Leukemia, 32, 1168-1179.
https://doi.org/10.1038/s41375-018-0009-0
[85] Urak, R., et al. (2017) Ex Vivo Akt Inhibition Promotes the Generation of Potent CD19CAR T Cells for Adoptive Immunotherapy. Journal for ImmunoTherapy of Cancer, 5, Article No. 26.
https://doi.org/10.1186/s40425-017-0227-4
[86] John, L.B., et al. (2013) Anti-PD-1 Antibody Therapy Potently Enhances the Eradication of Established Tumors by Gene-Modified T Cells. Clinical Cancer Research, 19, 5636-5646.
https://doi.org/10.1158/1078-0432.CCR-13-0458
[87] Karlsson, S.C.H., et al. (2013) Combining CAR T Cells and the Bcl-2 Family Apoptosis Inhibitor ABT-737 for Treating B-Cell Malignancy. Cancer Gene Therapy, 20, 386-393.
https://doi.org/10.1038/cgt.2013.35
[88] Sterner, R.M., et al. (2019) GM-CSF Inhibition Reduces Cytokine Release Syndrome and Neuroinflammation but Enhances CAR-T Cell Function in Xenografts. Blood, 133, 697-709.
https://doi.org/10.1182/blood-2018-10-881722
http://ashpublications.org/blood/article-pdf/133/7/697/1552534/blood881722.pdf
[89] Pennell, C.A., et al. (2018) Human CD19-Targeted Mouse T Cells Induce B Cell Aplasia and Toxicity in Human CD19 Transgenic Mice. Molecular Therapy, 26, 1423-1434.
https://doi.org/10.1016/j.ymthe.2018.04.006
[90] Norelli, M., et al. (2018) Monocyte-Derived IL-1 and IL-6 Are Differentially Required for Cytokine-Release Syndrome and Neurotoxicity Due to CAR T Cells. Nature Medicine, 24, 739-748.
https://doi.org/10.1038/s41591-018-0036-4
[91] Giavridis, T., van der Stegen, S.J.C., Eyquem, J., Hamieh, M., Piersigilli, A. and Sadelain, M. (2018) CAR T Cell-Induced Cytokine Release Syndrome Is Mediated by Macrophages and Abated by IL-1 Blockade. Nature Medicine, 24, 731-738.
https://doi.org/10.1038/s41591-018-0041-7
[92] Staedtke, V., et al. (2018) Disruption of a Self-Amplifying Catecholamine Loop Reduces Cytokine Release Syndrome. Nature, 564, 273-277.
https://doi.org/10.1038/s41586-018-0774-y
[93] Turtle, C.J., et al. (2016) CD19 CAR-T Cells of Defined CD4+:CD8+ Composition in Adult B Cell ALL Patients. Journal of Clinical Investigation, 126, 2123-2138.
https://doi.org/10.1172/JCI85309
[94] Stavrou, M., et al. (2018) A Rapamycin-Activated Caspase 9-Based Suicide Gene. Molecular Therapy, 26, 1266-1276.
https://doi.org/10.1016/j.ymthe.2018.03.001
[95] Diaconu, I., et al. (2017) Inducible Caspase-9 Selectively Modulates the Toxicities of CD19-Specific Chimeric Antigen Receptor-Modified T Cells. Molecular Therapy, 25, 580-592.
https://doi.org/10.1016/j.ymthe.2017.01.011
[96] Philip, B., et al. (2014) A Highly Compact Epitope-Based Marker/Suicide Gene for Easier and Safer T-Cell Therapy. Blood, 124, 1277-1287.
https://doi.org/10.1182/blood-2014-01-545020
[97] Grupp, S.A., et al. (2013) Chimeric Antigen Receptor-Modified T Cells for Acute Lymphoid Leukemia. The New England Journal of Medicine, 368, 1509-1518.
https://doi.org/10.1056/NEJMoa1215134
[98] Kochenderfer, J.N., et al. (2012) B-Cell Depletion and Remissions of Malignancy Along with Cytokine-Associated Toxicity in a Clinical Trial of Anti-CD19 Chimeric-Antigen-Receptor-Transduced T Cells. Blood, 119, 2709-2720.
https://doi.org/10.1182/blood-2011-10-384388
[99] Kochenderfer, J.N., et al. (2010) Eradication of B-Lineage Cells and Regression of Lymphoma in a Patient Treated with Autologous T Cells Genetically Engineered to Recognize CD19. Blood, 116, 4099-4102.
https://doi.org/10.1182/blood-2010-04-281931
[100] Morgan, R.A., Yang, J.C., Kitano, M., Dudley, M.E., Laurencot, C.M. and Rosenberg, S.A. (2010) Case Report of a Serious Adverse Event following the Administration of T Cells Transduced with a Chimeric Antigen Receptor Recognizing ERBB2. Molecular Therapy, 18, 843-851.
https://doi.org/10.1038/mt.2010.24
[101] Cameron, B.J., et al. (2013) Identification of a Titin-Derived HLA-A1-Presented Peptide as a Cross-Reactive Target for Engineered MAGE A3-Directed T Cells. Science Translational Medicine, 5, 197ra103.
https://doi.org/10.1126/scitranslmed.3006034
[102] Porter, D.L., Levine, B.L., Kalos, M., Bagg, A. and June, C.H. (2011) Chimeric Antigen Receptor-Modified T Cells in Chronic Lymphoid Leukemia. The New England Journal of Medicine, 365, 725-733.
https://doi.org/10.1056/NEJMoa1103849
[103] Brentjens, R.J., et al. (2011) Safety and Persistence of Adoptively Transferred Autologous CD19-Targeted T Cells in Patients with Relapsed or Chemotherapy Refractory B-Cell Leukemias. Blood, 118, 4817-4828.
https://doi.org/10.1182/blood-2011-04-348540
[104] Kochenderfer, J.N., et al. (2013) Donor-Derived CD19-Targeted T Cells Cause Regression of Malignancy Persisting after Allogeneic Hematopoietic Stem Cell Transplantation Key Points. Blood, 122, Article No. 151.
https://doi.org/10.1182/blood.V122.21.151.151
[105] Maus, M.V., et al. (2013) T Cells Expressing Chimeric Antigen Receptors Can Cause Anaphylaxis in Humans. Cancer Immunology Research, 1, 26-31.
https://doi.org/10.1158/2326-6066.CIR-13-0006
[106] Brentjens, R.J., et al. (2013) CD19-Targeted T Cells Rapidly Induce Molecular Remissions in Adults with Chemotherapy-Refractory Acute Lymphoblastic Leukemia. Science Translational Medicine, 5, 177ra38.
https://doi.org/10.1126/scitranslmed.3005930
[107] Maude, S.L., et al. (2014) Chimeric Antigen Receptor T Cells for Sustained Remissions in Leukemia. The New England Journal of Medicine, 371, 1507-1517.
https://doi.org/10.1056/NEJMoa1407222
[108] Barrett, D.M., Teachey, D.T. and Grupp, S.A. (2014) Toxicity Management for Patients Receiving Novel T-Cell Engaging Therapies. Current Opinion in Pediatrics, 26, 43-49.
https://doi.org/10.1097/MOP.0000000000000043
[109] Maus, M.V. and June, C.H. (2016) Making Better Chimeric Antigen Receptors for Adoptive T-Cell Therapy. Clinical Cancer Research, 22, 1875-1884.
https://doi.org/10.1158/1078-0432.CCR-15-1433
[110] Kalaitsidou, M., Kueberuwa, G., Schütt, A. and Gilham, D.E. (2015) CAR T-Cell Therapy: Toxicity and the Relevance of Preclinical Models. Immunotherapy, 7, 487-497.
https://doi.org/10.2217/imt.14.123
[111] Casucci, M., Hawkins, R.E., Dotti, G. and Bondanza, A. (2015) Overcoming the Toxicity Hurdles of Genetically Targeted T Cells. Cancer Immunology, Immunotherapy, 64, 123-130.
https://doi.org/10.1007/s00262-014-1641-9
[112] Kochenderfer, J.N., et al. (2015) 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. Journal of Clinical Oncology, 33, 540-549.
https://doi.org/10.1200/JCO.2014.56.2025
[113] Riegler, L.L., Jones, G.P. and Lee, D.W. (2019) Current Approaches in the Grading and Management of Cytokine Release Syndrome after Chimeric Antigen Receptor T-Cell Therapy. Therapeutics and Clinical Risk Management, 15, 323-335.
https://doi.org/10.2147/TCRM.S150524
[114] Maude, S.L., et al. (2018) Tisagenlecleucel in Children and Young Adults with B-Cell Lymphoblastic Leukemia. The New England Journal of Medicine, 378, 439-448.
https://doi.org/10.1056/NEJMoa1709866
[115] Schuster, S.J., et al. (2017) Chimeric Antigen Receptor T Cells in Refractory B-Cell Lymphomas. The New England Journal of Medicine, 377, 2545–2554.
https://doi.org/10.1056/NEJMoa1708566
[116] Neelapu, S.S., et al. (2017) Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B-Cell Lymphoma. The New England Journal of Medicine, 377, 2531-2544.
https://doi.org/10.1056/NEJMoa1707447
[117] Lee, D.W., et al. (2019) ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biology of Blood and Marrow Transplantation, 25, 625-638.
https://doi.org/10.1016/j.bbmt.2018.12.758
[118] Santomasso, B.D., Bachier, C., Westin, J., Rezvani, K. and Shpall, E.J. (2022) The Other Side of CAR T-Cell Therapy: Cytokine Release Syndrome, Neurologic Toxicity, and Financial Burden. American Society of Clinical Oncology-Educational Book, 39, 433-444.
https://doi.org/10.1200/EDBK_238691
[119] Garcia Borrega, J., et al. (2019) In the Eye of the Storm: Immune-Mediated Toxicities Associated with Car-T Cell Therapy. HemaSphere, 3, e191.
https://doi.org/10.1097/HS9.0000000000000191
[120] Andtbacka, R.H.I., et al. (2015) Talimogene Laherparepvec Improves Durable Response Rate in Patients with Advanced Melanoma. Journal of Clinical Oncology, 33, 2780-2788.
https://doi.org/10.1200/JCO.2014.58.3377
[121] Lichty, B.D., Breitbach, C.J., Stojdl, D.F. and Bell, J.C. (2014) Going Viral with Cancer Immunotherapy. Nature Reviews Cancer, 14, 559-567.
https://doi.org/10.1038/nrc3770
[122] Rezaei, R., et al. (2021) Combination Therapy with CAR T Cells and Oncolytic Viruses: A New Era in Cancer Immunotherapy. Cancer Gene Therapy, 29, 647-660.
https://doi.org/10.1038/s41417-021-00359-9
[123] Aalipour, A., et al. (2020) Viral Delivery of CAR Targets to Solid Tumors Enables Effective Cell Therapy. Molecular Therapy—Oncolytics, 17, 232-240.
https://doi.org/10.1016/j.omto.2020.03.018
[124] Li, Y., et al. (2020) Oncolytic Adenovirus Targeting TGF-β Enhances Anti-Tumor Responses of Mesothelin-Targeted Chimeric Antigen Receptor T Cell Therapy against Breast Cancer. Cellular Immunology, 348, Article ID: 104041.
https://doi.org/10.1016/j.cellimm.2020.104041
[125] Porter, C.E., et al. (2020) Oncolytic Adenovirus Armed with BiTE, Cytokine, and Checkpoint Inhibitor Enables CAR T Cells to Control the Growth of Heterogeneous Tumors. Molecular Therapy, 28, 1251-1262.
https://doi.org/10.1016/j.ymthe.2020.02.016
[126] Park, A.K., et al. (2020) Effective Combination Immunotherapy Using Oncolytic Viruses to Deliver CAR Targets to Solid Tumors. Science Translational Medicine, 11, eaaz1863.
https://doi.org/10.1126/scitranslmed.aaz1863
http://stm.sciencemag.org/
[127] Watanabe, K., et al. (2018) Pancreatic Cancer Therapy with Combined Mesothelin-Redirected Chimeric Antigen Receptor T Cells and Cytokine-Armed Oncolytic Adenoviruses. JCI Insight, 3, e99573.
https://doi.org/10.1172/jci.insight.99573
[128] Wing, A., et al. (2018) Improving CART-Cell Therapy of Solid Tumors with Oncolytic Virus-Driven Production of a Bispecific T-Cell Engager. Cancer Immunology Research, 6, 605-616.
https://doi.org/10.1158/2326-6066.CIR-17-0314
[129] Moon, E.K., et al. (2018) Intra-Tumoral Delivery of CXCL11 via a Vaccinia Virus, but Not by Modified T Cells, Enhances the Efficacy of Adoptive T Cell Therapy and Vaccines. Oncoimmunology, 7, e1395997.
https://doi.org/10.1080/2162402X.2017.1395997
[130] Shaw, A.R., et al. (2017) Adenovirotherapy Delivering Cytokine and Checkpoint Inhibitor Augments CAR T Cells against Metastatic Head and Neck Cancer. Molecular Therapy, 25, 2440-2451.
https://doi.org/10.1016/j.ymthe.2017.09.010
[131] Rosewell Shaw, A., et al. (2017) Adenovirotherapy Delivering Cytokine and Checkpoint Inhibitor Augments CAR T Cells against Metastatic Head and Neck Cancer. Molecular Therapy, 25, 2440-2451.
https://doi.org/10.1016/j.ymthe.2017.09.010
[132] Nishio, N., et al. (2014) Armed Oncolytic Virus Enhances Immune Functions of Chimeric Antigen Receptor-Modified T Cells in Solid Tumors. Cancer Research, 74, 5195-5205.
https://doi.org/10.1158/0008-5472.CAN-14-0697
[133] Charrot, S., Hallam, S. and Hallam, C.S. (2019) CAR-T Cells: Future Perspectives. HemaSphere, 2, e188.
https://doi.org/10.1097/HS9.0000000000000188
[134] Turtle, C.J., Riddell, S.R. and Maloney, D.G. (2016) CD19-Targeted Chimeric Antigen Receptor-Modified T-Cell Immunotherapy for B-Cell Malignancies. Clinical Pharmacology & Therapeutics, 100, 252-258.
https://doi.org/10.1002/cpt.392
[135] Pan, J., et al. (2017) High Efficacy and Safety of Low-Dose CD19-Directed CAR-T Cell Therapy in 51 Refractory or Relapsed B Acute Lymphoblastic Leukemia Patients. Leukemia, 31, 2587-2593.
https://doi.org/10.1038/leu.2017.145
[136] Brudno, J.N., et al. (2016) Allogeneic T Cells That Express an Anti-CD19 Chimeric Antigen Receptor Induce Remissions of B-Cell Malignancies That Progress after Allogeneic Hematopoietic Stem-Cell Transplantation without Causing Graft-versus-Host Disease. Journal of Clinical Oncology, 34, 1112-1121.
https://doi.org/10.1200/JCO.2015.64.5929
[137] Turtle, C.J., et al. (2017) Durable Molecular Remissions in Chronic Lymphocytic Leukemia Treated with CD19-Specific Chimeric Antigen Receptor-Modified T Cells after Failure of Ibrutinib. Journal of Clinical Oncology, 35, 3010-3020.
https://doi.org/10.1200/JCO.2017.72.8519
[138] Turtle, C.J., Robinson, E.M., Chaney, C. and Melville, K. (2015) Anti-CD19 Chimeric Antigen Receptor-Modified T Cell Therapy for B Cell Non-Hodgkin Lymphoma and Chronic Lymphocytic Leukemia: Fludarabine and Cyclophosphamide Lymphodepletion Improves in Vivo Expansion and Persistence of CAR-T Cells and Clinical Outcomes. Blood, 126, Article No. 184.
https://doi.org/10.1182/blood.V126.23.184.184
[139] Wang, C.M., et al. (2017) Autologous T Cells Expressing CD30 Chimeric Antigen Receptors for Relapsed or Refractory Hodgkin Lymphoma: An Open-Label Phase I Trial. Clinical Cancer Research, 23, 1156-1166.
https://doi.org/10.1158/1078-0432.CCR-16-1365
[140] Cohen, A.D., Garfall, A.L., Stadtmauer, E.A., Lacey, S.F., Ambrose, E. and Ferthio, R. (2016) B-Cell Maturation Antigen (BCMA)-Specific Chimeric Antigen Receptor T Cells (CART-BCMA) for Multiple Myeloma (MM): Initial Safety and Efficacy from a Phase I Study. Blood, 128, Article No. 1147.
https://doi.org/10.1182/blood.V128.22.1147.1147
[141] Ormhoj, M., Bedoya, F., Frigault, M.J. and Maus, M.V. (2017) CARs in the Lead against Multiple Myeloma. Current Hematologic Malignancy Reports, 12, 119-125.
https://doi.org/10.1007/s11899-017-0373-2
[142] Chekmasova, A.A., 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. Clinical Cancer Research, 16, 3594-3607.
https://doi.org/10.1158/1078-0432.CCR-10-0192
[143] Li, J., Li, W., Huang, K., Zhang, Y., Kupfer, G. and Zhao, Q. (2018) Chimeric Antigen Receptor T Cell (CAR-T) Immunotherapy for Solid Tumors: Lessons Learned and Strategies for Moving Forward. Journal of Hematology and Oncology, 11, Article No. 22.
https://doi.org/10.1186/s13045-018-0568-6
[144] Marofi, F., et al. (2021) CAR T Cells in Solid Tumors: Challenges and Opportunities. Stem Cell Research & Therapy, 12, Article No. 81.
https://doi.org/10.1186/s13287-020-02128-1
[145] Jung, M., et al. (2020) Chimeric Antigen Receptor T Cell Therapy Targeting ICAM-1 in Gastric Cancer. Molecular Therapy—Oncolytics, 18, 587-601.
https://doi.org/10.1016/j.omto.2020.08.009
[146] Wei, X., et al. (2017) PSCA and MUC1 in Non-Small-Cell Lung Cancer as Targets of Chimeric Antigen Receptor T Cells. Oncoimmunology, 6, e1284722.
https://doi.org/10.1080/2162402X.2017.1284722
[147] Transl, J., et al. (2020) 32A9, a Novel Human Antibody for Designing an Immunotoxin and CAR-T Cells against Glypican-3 in Hepatocellular Carcinoma. Journal of Translational Medicine, 18, Article No. 295.
https://doi.org/10.1186/s12967-020-02462-1
[148] Sureban, S.M., et al. (2020) DCLK1 Monoclonal Antibody-Based CAR-T Cells as a Novel Treatment Strategy against Human Colorectal Cancers. Cancers, 12, Article No. 54.
[149] Grada, Z., et al. (2013) TanCAR: A Novel Bispecific Chimeric Antigen Receptor for Cancer Immunotherapy. Molecular Therapy—Nucleic Acids, 2, E105.
https://doi.org/10.1038/mtna.2013.32
[150] Yan, W., Liu, Z., Liu, J., Xia, Y., Hu, K. and Yu, J. (2020) Application of Chimeric Antigen Receptor T Cells in the Treatment of Hematological Malignancies. BioMed Research International, 2020, Article ID: 4241864.
https://doi.org/10.1155/2020/4241864
[151] Jones, B.S., Lamb, L.S., Goldman, F. and Di Stasi, A. (2014) Improving the Safety of Cell Therapy Products by Suicide Gene Transfer. Frontiers in Pharmacology, 5, Article No. 254.
https://doi.org/10.3389/fphar.2014.00254
[152] Zhou, X., et al. (2015) Inducible Caspase-9 Suicide Gene Controls Adverse Effects from Alloreplete T Cells after Haploidentical Stem Cell Transplantation. Blood, 125, 4103-4113.
https://doi.org/10.1182/blood-2015-02-628354

Copyright © 2024 by authors and Scientific Research Publishing Inc.

Creative Commons License

This work and the related PDF file are licensed under a Creative Commons Attribution 4.0 International License.