Available online on 15.06.2024 at http://jddtonline.info

Journal of Drug Delivery and Therapeutics

Open Access to Pharmaceutical and Medical Research

Copyright  © 2024 The  Author(s): This is an open-access article distributed under the terms of the CC BY-NC 4.0 which permits unrestricted use, distribution, and reproduction in any medium for non-commercial use provided the original author and source are credited

Open Access   Full Text Article                                                                                                                           Review Article

Chimeric Antigen Receptor T-Cells (CAR T-Cells): An Engineered Targeted Therapy for Treatment of Cancer

Darshana Rathi *1, Nikita Patel 1, Trilochan Satapathy 2

1 M. Pharm Scholar, Columbia Institute of Pharmacy, Village Tekari, Near Vidhansabha, Raipur-493111, C.G., India

2 Professor, Department of Pharmacology, Columbia Institute of Pharmacy, Tekari, Near Vidhansabha. C.G., India.

Article Info:

___________________________________________

Article History:

Received 24 March 2024  

Reviewed 10 May 2024  

Accepted 03 June 2024  

Published 15 June 2024  

___________________________________________

Cite this article as: 

Rathi D, Patel N, Satapathy T, Chimeric Antigen Receptor T-Cells (CAR T-Cells): An Engineered Targeted Therapy for Treatment of Cancer, Journal of Drug Delivery and Therapeutics. 2024; 14(6):274-286

DOI: http://dx.doi.org/10.22270/jddt.v14i6.6601               ___________________________________________

*Address for Correspondence:  

Darshana Rathi, M. Pharm Scholar, Department of Pharmacology, Columbia Institute of Pharmacy, Tekari, Near Vidhansabha. C.G., India.

Abstract

___________________________________________________________________________________________________________________

We have undertaken this review to explore the various developments and insights of CAR-T cell therapy during 1989-2023 and its advantages in the treatment of cancer and immune modulation. It is a chimeric antigen receptor T-cell therapy, which is an innovative form of immunotherapy that harnesses the power of the immune system to fight cancer. At first, T cells are extracted from the patient’s blood through a process called leukapheresis. Then the modification has been done in T cells by genetically engineered to express chimeric antigen receptors (CARs) on their surface. These receptors are designed to recognize specific proteins, or antigens, that are found on the surface of cancer cells. Many conventional therapies available in the market for the treatment of cancer and Immuno modulation but most of them having Adverse Drug Reaction (ADR). But CAR-T cells possess upper hand on these conventional Formulations. Once a sufficient number of CAR-T cells have been produced, they are infused back into the patient’s bloodstream. Once reach inside the body, the CAR-T cells recognize and bind to the cancer cells that express the specific antigen targeted by the CAR. This triggers the destruction of the cancer cells by the immune system. CAR-T cell therapy has shown remarkable success in treating certain types of blood cancers, such as acute lymphoblastic leukaemia (ALL), chronic lymphocytic leukaemia (CLL), and certain types of lymphoma. The content of this review will pave the way to work on CAR-T cell therapy.

Keywords: CAR-T, Immunotherapy, cancer, antigen, leukemia, lymphoma.

 


 

1.INTRODUCTION:

Chimeric antigen receptors (CARs) are receptor proteins that have been modified to allow T cells to target a particular antigen. They are also referred to as chimeric immune receptors, chimeric T cell receptors, or artificial T cell receptors in the field of biology. The receptors are chimeric, combining into a single receptor the ability to attach to antigen and activate T cells.

Production and injection of T cells with chimeric antigen receptor-

1. A patient's blood is used to separate T cells. 

2. The T cells acquire a new gene that codes for a chimeric antigen receptor. 

3. Target antigen-specific engineered T cells are currently available. 

4. Tissue culture is used to grow engineered T cells. 

5. The patient receives a second infusion of engineering t cell.1


 

 

 image

Figure 1: Production and infusion of T-cell

Table 1: History of CAR T-cell

Year

Achievement

1989

production of effector T cells that express the receptor for chimeric T cells2,3

1993

CAR-T cells from the first generation are not therapeutically effective.4

2002

The first CAR-T cells that are efficient against the prostate cancer antigen in the lab.

2003

Second-generation CARs: CD19-directed CAR-T cells have the ability to eradicate mice leukaemia cells.

2009

Relapsed or resistant leukaemia is treated using CD19 CAR.

2011

Patients with chronic lymphocytic leukaemia with CD19 CAR.5

2013

Paediatric acute lymphoblastic leukaemia using CD19 CAR-T cells. 6

2013

Cancer immunotherapy was named the "Breakthrough of the Year" by Science magazine.

2014

Inducible caspase-9 suicide gene system as a "safety switch" to reduce on-target, off-tumour toxicities in third-generation CAR-T cells.7

2015

Fourth-generation CARs that generate different compounds being explored for ovarian cancer, such as armoured CARs or TRUCKs (CAR redirected T cells that deliver a transgenic product to the targeted tumour tissue).8

2015

The CAR-NK cell concept.9

2017

Clustered regularly interspaced short palindromic repeats (CRISPR) used to optimize CAR placement in T cells. 10

2017

For children and young adults with relapsed or resistant acute lymphoblastic leukaemia, the FDA has approved CD19-CAR-T cells.

2017

FDA approves CD19-CAR-T cells for adult DLBCL patients with relapses or resistance.

2018

For relapsed or resistant acute lymphoblastic leukaemia in children and young adults as well as relapsed or resistant DLBCL in adults, the EMA has approved CD19-CAR-T cells.

2019

In both adults and children with acute lymphoblastic leukaemia, dual CD19/CD22 CAR-T cells are present. 11

2023

CDSCO made NexCAR19, India’s first approved CAR T-cell therapy.

 


 

CAR T-Cells show some side effects which should be taken cared of. Some include: Cytokine Release Syndrome (CRS) is a common adverse reaction following CAR T-cell treatment, causing minor symptoms like headaches and fevers, or serious ones like hypotension, capillary leak syndrome, and organ malfunction12. Neurological toxicity, also known as ICANS, is an immune effector causing neurological symptoms like delirium, seizures, and disorientation, often occurring alongside CRS. Hypogammaglobulinemia is Immunoglobulin levels may drop as a result of CAR T-cell treatment, raising the possibility of infections. Tumour Lysis Syndrome is a condition where cancer cells are rapidly killed, causing metabolic abnormalities like hyperphosphatemia, hypocalcaemia, hyperkalaemia, and hyperuricemia13.CAR T-cell treatment, although rare, can occasionally lead to cardiac toxicity, causing symptoms like arrhythmias or reduced heart function14.

 

 

 

2.STRUCTURE OF CAR T CELLS:

CARs are divided into three main domains: 

1) The extracellular domain, or ecto-domain, is composed of a single peptide on the cell surface that is cleaved from the mature CAR cell and can be further subdivided into an antigen-recognition domain.15Aflexible linker divides the antigen-specific immune-globin segments that make up the antigen-recognition domain (scFV), which is primarily composed of heavy and variable light chain regions. A hinge connects the spacer (scFV) to the trans-membrane domain, which is in charge of transmitting receptor-binding signals.16 

2) The hydrophobic alpha helix known as the transmembrane domain, which extends throughout the cell membrane, is crucial for receptor stability and surface expression.17

3) the endo-domain, an intracellular domain that, when stimulated, clusters and alters its conformation, making it possible for downstream signalling proteins to be recruited and phosphorylated.18


 

 

image

Figure 2: Structure of Chimeric antigen receptor (CAR)


 

3. EVOLUTION:

Five generations of CARs are distinguished by their intracellular domain:

The first has a single activation domain, a cytoplasmic domain primarily consisting of CD3 zeta (CD3ζ), and some studies use the gamma chain (γ) of the Fc receptors.2.) The second has CD3ζ plus one costimulatory domain, obtained from costimulatory molecules like 4-1BB or CD28 connected to an activator domain (CD3ζ/γ chain of Fc receptor), to improve the cytotoxic and cell proliferative abilities of CAR T cells.19Though it possesses several costimulatory domains with CD3ζ, including 4-1BB and CD28, CD134, and CD13 20 7.3.)The third generation of CARs, T cells redirected for universal cytokine-mediated killing (TRUCKs), aims to overcome TME immune suppression and support strong therapeutic outcomes by releasing transgenic cytokine-like interleukin 12 (IL-12) upon CAR signalling in tumour tissue. IL-12 induces IFN-γ, perforin, and granzymes in T-cells, preventing Treg proliferation.4.) IL-15 and IL-18 are also examined in the fourth generation 21-,24 .IL-15, a γ-chain family member, is crucial for T cell survival and growth. When large pancreatic and lung tumours are treated with IL-18 CAR T-cells, the immune cell landscape changes, with an increase in macrophages and NKs and a decrease in Tregs. This suggests that "IL-18 TRUCKs" can sensitize large tumour lesions for effective immune destruction 25.5.) Currently under investigation, the fifth generation of CARs is primarily based on the second generation. Nonetheless, it has the transcription factor STAT3/5 binding motif, a β-chain domain (IL-2Rβ truncated intracellular interleukin 2β chain receptor), and a truncated cytoplasmic receptor (IL-12). 26

4. PRODUCTION

CAR T-cells are created by isolating T cells from human blood, either from the patient's own blood (autologous treatment) or a healthy donor (allogenic treatment). The process involves leukocyte apheresis, which separates PBMCs, and then being sent to a cell processing facility. T cells are stimulated by cytokine interleukin 2 (IL-2) and anti-CD3 antibodies. The enlarged T cells are purified and transduced with a modified CAR gene, such as integrated gamma-retrovirus (RV) or lentiviral vector. CRISPR/Cas9, a gene editing technique, is used to integrate the CAR gene into specific regions. Before the engineered CAR T-cells are introduced, the patient undergoes lymphodepletion chemotherapy, which increases cytokine production and reduces resource competition, promoting the proliferation of the altered CAR T-cells.27-34

5. CLINICAL PREPARATION OF CAR T CELLS:

Interest in CAR-T cell-based therapeutics has grown due to the success of CD19-targeted CAR-T cells in early phase clinical studies for hematologic malignancies. Current research focuses on targeting different cancer types using tumour-associated antigens like PSMA, mesothelin, GD2, HER2, and epidermal growth factor receptors. The production of CAR-T cells under cGMP is the central approach. The manufacturing process includes T-cell collection, processing, activation, genetic modification, large-scale expansion, and end-of-process formulation.

5.1 CAR T-Cell Source: CAR-T cells are produced through autologous treatment, involving peripheral blood mononuclear cells from patients through leukapheresis. Physicians determine the best collection time based on treatment plans. Different methods, such as Haemonetics Cell Saver 5+, COBE2991, and Fresenius Kabi LOVO, can be used to process collected apheresis products, eliminating platelet impurities and rough red blood cells. Terumo Elutra and Biosafe Sepax systems offer size-based cell fractionation for monocyte depletion and lymphocyte isolation.


 

 

image

Figure 3: Production of CAR T-cell


 

Clinical trials often use CAR-T cells from the CD3+ population, but research shows specific T cell subsets like naïve, memory stem cells, and central memory may have functional advantages. Clinical-scale procedures for growth, transduction, and selection have been developed for these subsets. Identifying the best therapeutic benefit and minimizing toxicity is crucial for a reliable manufacturing process. Cryo-preserved T-cell source material can be used for later use or directly for subsequent treatments, with cryo-preserving providing more flexibility for downstream processes and product assessment. 35-37


 

5.2 T-Cell Activation: T-cell growth in vivo requires specific and costimulatory signals from sources like CD28, 4-1BB, or OX40. Retroviral vector-mediated transduction of CAR cDNA also requires T-cell activation. Endogenous T-cell activators include dendritic cells (DCs), but their potency varies among patients, making them less reliable. Artificial antigen-presenting cells (AAPCs) are another cell-based T-cell activation strategy, induced by irradiated K562-derived AAPCs. However, generating and choosing GMP- grade HLA matched AAPC lines requires more work and resources. Biotech companies have produced clinical-grade T-cell activation reagents, such as Juno Stage Expamer technology, which significantly streamline the ex-vivo T-cell activation process.38-4

5.3 expamer Technology: Juno Therapeutics; Expamer is a recent advancement in T-cell activation reagent technology, using Streptamer technology to isolate lymphocytes specific to viruses. This soluble and dissociable reagent stimulates T cells to enable retroviral transduction and growth, inducing T cell receptor signalling. Despite functional assessment in vivo, Expamer is appealing for large-scale clinical manufacture due to ease of addition and removal from cell suspensions.41-43

 5.4 Genetic Modification of T-Cell: Stable CAR expression is crucial for current CAR-T cell treatments. There are three main categories of stable gene expression vectors: γ-retroviral vectors, lentiviral vectors, and transposon/transposase systems. γ-retroviral vectors are used in 25% of clinical trials due to their extensive tropism, stable packaging cell lines, and high gene expression. Studies have shown the safety profile of retroviral vectors when used with adoptive T-cell treatment. Research suggests growing retroviral stable packaging producer cell lines in scalable bioreactors could produce sufficient cGMP quality vector stocks for phase 3 trials and beyond. Retroviral and lentiviral vectors require extensive biosafety testing due to their complexity. The transposon/transposase system, a recent plasmid-based expression technology, offers straightforward release testing, low cost, and easy manufacturing. However, random integration may pose a secondary oncogenic risk due to mutagenesis. Low T-cell toxicity has been observed in an ongoing anti-CD19 CAR-T cell experiment, but its effectiveness remains unestablished.44-57

 5.5 Messenger RNA: Messenger RNA (mRNA) transfer allows transgenes to express themselves temporarily, unlike viral transduction or plasmid DNA transfection. This method eliminates genomic integration events, genotoxicity, and replication-competent retrovirus production. RNA transfection is used to transfer mRNA for TCR/CAR, chemokine receptors, and cytokines, allowing transgene expression for about one week. A recent study showed that “off the shelf” CAR-T cells can be produced from unrelated healthy donors using electroporation of transcription activator-like effector nucleases.58-62

Off-the-shelf CAR T-cell therapies: CRISPR, natural killers, and mRNA: Scientists are exploring the origins of CAR T-cell treatments, using T cells from healthy donors instead of patients. The goal is to provide off-the-shelf CAR T-cell therapies that can be used immediately. FDA-approved therapies use a disarmed virus to transfer genetic material into T cells, but gene-editing technologies like CRISPR and TALON are being used to stimulate donated T cells. Natural killer (NK) cells are also being explored as an alternative form of immune cell. The therapeutic manufacturing process is also being re-examined, with research teams using mRNA-based strategies and nanotechnology to create CAR T cells inside the body. 63

6.DEVELOPMENT OF CAR ENGINEERED T-CELL THERAPIES:

6.1. The Emergence of Chimeric T-Cell Receptor: The concept of a chimeric T cell receptor was first documented in 1987 by Dr. Yoshikazu Kurosawa and his colleagues at the Institute for Comprehensive Medical Science. They demonstrated that anti-phosphorylcholine chimeric receptors in murine T-cell lymphomaEL4 cells caused a calcium influx, indicating that the receptor could activate T cells in response to antigens. In 1989, Israeli immunologist Dr. Zelig Eshhar and his colleagues reported that T cells can recognize antigens in a non-major histocompatibility complex (MHC)-restricted manner using a similar strategy. The resulting chimeric T-cell receptor (cTCR) was made up of the constant sections of alpha and beta TCR chains fused with the variable heavy and light chains of the anti-2,4,6-trinitrophenyl (TNP) antibody Sp6. Functional cTCRs were expressed on the cell surface and able to bind to TNP antigen upon co-transfection into murine MD.45 cyto-toxic T lymphocyte hybridoma cells, resulting in T cell activation and the generation of interleukin-2 (IL-2) and the destruction of target cells. 64-69

6.2. The First Generation CAR: In mouse models, the modified T cells containing the first-generation CAR, which had scFv fused to CD3ζ or FcϵRIγ, demonstrated anti-cancer activity. For example, in athymic BALB/c mice, the growth of subcutaneous tumours was reduced by a scFvR including anti-ERBB2/HER2 scFv and a murine CD3β signalling endo-domain. In a different investigation, syngeneic C57BL/6 mice, athymic nude mice, and murine tumour-infiltrating T cells expressing a scFvR called MOv-γ—a combination of anti-α-folate receptor (FR) scFv and the Fc receptor γ chain—exhibited anticancer efficacy in vitro. It is significant to remember that following the infusion of the gene-modified T cells in these in vivo studies, mice got systemic high-dose IL-2 therapy 70-72.Two human clinical trials of CAR T cell therapy were conducted in ovarian cancer patients and metastatic renal cell carcinoma patients. Despite promising anticancer results in vitro and in mice, no patient showed a decrease in tumour amount and most patients' genetically modified T cells rapidly decreased to undetectable levels. In another study, modified T cells expressing CE7R(huCD3ζ) were durable in vivo for up to six weeks in children with recurrent neuroblastoma. However, drug-selection genes were produced by modified T cells, potentially reducing their ability to persist in vivo. Patients with neuroblastoma showed some durability and antitumor activity from first-generation GD2-specific CAR T cells. Efforts to improve first-generation CAR design have been prompted by their inefficiency in treating cancer in humans 73-77

6.3. The Second Generation-CAR: Two signals are normally needed for T cell activation: the first is produced when the TCR engages with peptide-loaded major histo-compatibility complex (pMHC), and the second is produced by costimulatory receptors like CD2878. Therefore, it was suggested that adding a costimulatory endo-domain to T cells that have been modified could improve the cells' ability to proliferate and remain in place. A chimeric receptor that combines the CD3ζ and CD28 endo-domains was created by Dr. Michel Sadelain's lab at Memorial Sloan Kettering Cancer Centre (MSKCC). This receptor produces both activation and co-stimulatory signals, and it improves antigen-dependent proliferation, interleukin-2 production, and in vitro cancer cell death. Moreover, compared to T cells expressing chimeric receptors containing only the CD3ζ endo-domain, T cells expressing chimeric receptors containing both the CD3ζ and CD28 endo-domains demonstrated noticeably greater growth and durability in human patients. These findings unequivocally showed how crucial it is for the chimeric receptor to contain a costimulatory endo-domain like CD28 79-81 .Another important development in the field is the inclusion of the 4-1BB/CD137 signal transduction domain in the CAR design, which was pioneered by Dr Dario Campana at St Jude Children's Research Hospital. The longevity and anticancer activity of CAR-engineered T cells in mice were markedly enhanced by the addition of 4-1BB costimulatory endo-domain. Furthermore, while employing the elongation factor-1α promoter (EF-1α) in lentiviral vector, T cells have expressed chimeric receptors more steadily and permanently than when using promoters from ubiquitin, phosphoglycerate kinase (PGK), and cytomegalovirus (CMV). A noteworthy advancement in the field of CAR T cell therapy was reached when a second-generation CAR construct incorporating both a costimulatory endo-domain (like CD28 or 4-1BB) and the CD3ζ signalling endo-domain demonstrated remarkable success in human clinical trials. The comparison between the second-generation CAR that recognises tumour-associated antigen and the standard TCR that recognises peptide-major histocompatibility complex (pMHC). It also demonstrates the extraction of the single-chain variable fragment (scFv) domain from a monoclonal antibody.82-84

6.4.The Clinical Success of CAR-T Cell Therapy: A patient at NCI and MSKCC showed promise with second-generation CAR T cell therapy for advanced follicular lymphoma and relapsed B-cell ALL and CLL. The treatment involved expressing a CD19-specific CAR using a retroviral vector called MSGV. The patient experienced selective B-lineage cell elimination and partial remission of the lymphoma after two doses of CAR T cells and eight doses of IL-2. Autologous CD19-targeted CAR T cells were assessed for safety and durability in the MSKCC Phase 1 study. Individuals treated without prior conditioning did not show objective disease responses 85,86.

7. CAR T-CELL THERAPY:

Chimeric antigen receptor (CAR) T-cell treatment is used in immunotherapy for certain blood cancers. This treatment alters T-cells by inserting a new gene, increasing their ability to identify and eliminate malignant cells. CAR T-cell therapy can sometimes cure blood cancer and prolong patient survival 87.


 

 

image

Figure 4: Diagrammatic representation of CAR T-cell Therapy


 

7.1 How Does It Works: Formation of tumours include the mechanism: Viral genetic information disrupts the sequence of cellular oncogenes by integrating into the cellular genome. Viral gene products interact with tumour suppressor gene products, leading to a transformation process. Viral proteins can also interact with basic regulatory proteins of the cell cycle, affecting transcription and expression of certain genes. They promote cell proliferation and have an anti-apoptotic effect. Some viruses contribute to this transformation by increasing telomerase activity in cells, which plays a crucial role in chromosome stability during the cell cycle. Telomerase activity is present in prenatal development, isolated groups, and individuals lives. Recent studies have found an association between increased telomerase activity and infection with viruses like HPV 16, herpesvirus HHV8, and EB virus.

T-cells are white blood cells in the immune system that track antigens on the surface of intruder cells, including cancer. They system for aberrant cells, activating when a receptor identifies one. CAR T-cells are modified T-cells that identify a specific antigen on the surface of cancerous cells. Scientists introduce a synthetic gene for a chimeric antigen receptor, stimulating the T-cells to proliferate and develop until an adequate number of cells exist to efficiently target malignant cells.

7.2 Who Can Have CAR T-Cell Therapy: The FDA has approved CAR- T cell therapy for various types of B-cell cancers, including acute lymphoblastic leukaemia, Large-cell diffuse B-cell lymphoma (DLBCL), primary mediastinal lymphoma, follicle lymphoma, high-grade B cell lymphoma, activating B-cell lymphoma without specific indication, mantle cell lymphoma, and follicular lymphoma tumours. 

7.3 Therapy process: CAR T cell treatment is a complex procedure requiring skilled professionals. It involves collecting T-cells from an arm vein, engineering them in a lab using synthetic CAR, and infusion into the patient arm. To increase treatment efficacy, chemotherapy may be advised before the infusion. The procedure can occur at a hospital or outpatient infusion facility. It typically takes a few weeks to complete.88

 

8. FDA APPROVED CAR T-CELLS THERAPIES

The US Food and Drug Administration (FDA) has authorized CAR T-cell therapy for the treatment of multiple myeloma and certain types of leukaemia and lymphomas. Usually, CAR T-cell therapy is employed following the failure of conventional forms of treatment. Currently authorized CAR T-cell treatment include the following: 89


 

 

Table no.2: FDA approved CAR T-cell therapies

GENERIC NAME

BRAND NAME

TARGET ANTIGEN

TARGETED DISEASE

PATIENT POPULATION

Tisagenlecleucel

Kymriah 

CD19

B-cell acute lymphoblastic leukaemia (ALL)

B-cell non-Hodgkin lymphoma(NHL) 

Children and young adults with refractory or relapsed B-cell ALL

Adults with refractory or relapsed B-cell NHL

Axicabtagene ciloleucel

Yescarta 

CD19

B-cell non-Hodgkin lymphoma(NHL)

 

Follicular lymphoma

Adults with refractory or relapsed B-cell NHL

Adults with refractory or relapsed follicular lymphoma

Brexucabtagene autoleucel

Tecartus 

CD19

Mantle cell lymphoma(MCL)

B-cell acute lymphoblastic leukaemia (ALL)

Adults with refractory or relapsed MCL

Adults with refractory or relapsed B-cell ALL

Lisocabtagene maraleucel

Breyanzi 

CD19

B-cell non-Hodgkin lymphoma(NHL)

Adults with refractory or relapsed B-cell NHL

Idecabtagene vicleucel

Abecma

BCMA

Multiple myeloma 

Adults with refractory or relapsed multiple myeloma

Ciltacabtegene autoleucel

carvykti

BCMA

Multiple myeloma 

Adults with refractory or relapsed multiple myeloma

 


 

9. DIFFERENT TYPES OF CANCER ON WHICH CAR T-CELL THERAPY ACTS:

Since the U.S. Food and Drug Administration (FDA) initially authorised CAR T-cell therapy in 2017, the treatment has become accessible. Six iterations of CAR T-cell therapy have been authorised by the FDA thus far to treat various forms of blood cancer, including:

Leukaemia acute lymphoblastic-Acute lymphoblastic leukaemia (ALL) occurs when white blood cells from lymphoid progenitors aggregate in the bone marrow. T-cells and B-cells, two subtypes of lymphoid cells, are involved in ALL. B-cells are often linked to ALL and work to keep the body free of pathogens and illnesses by eliminating diseased cells.

Multiple myeloma- Multiple myeloma is a blood cancer that originates from the bone marrow's plasma cells, which produce antibody proteins to aid the body's immune system in fighting infections. As the cancer spreads, an overabundance of aberrant plasma cells forms, causing tumours to develop throughout the bone marrow, affecting the formation of healthy red blood cells and weakening the body's defences against infection. This type of cancer is usually uncontrollable and develops into multiple tumours, often not being detected until it has spread.

Hodgkin lymphoma- Non-Hodgkin lymphoma is a cancer that develops in the lymph system, which transports immune cells and filters wastes and poisons. It affects areas other than lymph nodes, such as the stomach, Waldeyer ring, central nervous system, lung, bone, and skin. The disease can also affect the spleen. B cells and T cells are affected by lymphoma, which helps produce antibodies against pathogens. CAR T-cell therapy targets specific cancer cell types, allowing treatment for follicular lymphoma, high-grade B-cell lymphoma, and mediastinal large B-cell lymphoma.90

10. PROCEDURE DETAILS:

10.1 What happens before CAR T-cell therapy?

Prior to receiving CAR T-cell therapy, patients usually follow these procedures:

1. Consultation and Evaluation: Patients consult with a haematologist or oncologist to talk about possible courses of treatment, such as CAR T-cell therapy. To decide if CAR T-cell therapy is appropriate, the medical team considers the patient's past medical history, present health, and response to prior therapies.

2. Pre-Treatment Testing: A battery of tests is administered to patients in order to evaluate their general health and eligibility for CAR T-cell treatment. This might involve heart evaluations, imaging studies, and blood testing.

3. T Cell Collection: Leukapheresis is the procedure used to obtain the patient's immune cells (T cells), should they be found to be candidates for CAR T-cell treatment. Leukapheresis involves drawing blood from the patient, then using machine to separate the white blood cells—including T cells—out of the blood. After that, a lab receives these T cells to be genetically altered.

4. Genetic Modification: In a lab setting, the patient's T cells undergo genetic modification to produce chimeric antigen receptors (CARs) on their surface. The T cells are now able to identify and attack cancer cells because to this alteration.

5. Preparatory Treatment: Patients frequently go through a conditioning programme prior to getting the modified T cells. Chemotherapy or other therapies to get the body ready for the CAR T cell injection may be part of this. The conditioning regimen assists in reducing the number of immune cells already in place and improves the conditions for the proliferation and assault of cancer cells by the modified T cells.

6. CAR T-cell Infusion: Following their preparation, the altered T cells are reinfused into the patient's circulation. Usually, this infusion happens in a hospital environment so that the patient may be thoroughly watched for any negative responses.

7. Monitoring and Follow-Up: CAR T-cell therapy patients undergo regular monitoring and assessments to ensure effectiveness and avoid adverse effects. A multidisciplinary team, including nurses, supportive care providers, haematologists, and oncologists, supports their safety and wellbeing throughout the treatment.

10.2 What happens during CAR T-cell therapy?

Several crucial processes take place throughout the CAR T-cell treatment procedure:

1. Preparation and Conditioning: Patients frequently go through a conditioning programme prior to receiving a CAR T cell injection. Chemotherapy or other therapies to get the body ready for the injection of T cells that have been changed may be part of this. The conditioning regimen aids in fostering an environment that is more conducive to the proliferation and assault of cancer cells by the modified T cells.

2. CAR T-cell Infusion: The keystone of the process is the CAR T-cell infusion. The patient receives the altered T cells intravenously. These cells are designed to express chimeric antigen receptors (CARs) on their surface. Typically, the infusion happens in a hospital environment so that the patient may be continuously watched for any negative responses.

3. Monitoring: Patients are constantly watched for any adverse responses or acute side effects following the infusion. Vital signs are monitored often, and the patient's health is continuously watched for indications of neurotoxicity or cytokine release syndrome (CRS), two possible side effects of CAR T-cell treatment.

4. Hospitalization: Depending on their unique response and any potential consequences, patients usually stay in the hospital for a while after receiving a CAR T-cell infusion. This duration might range from a few days to a few weeks. They get expert attention and supervision throughout this period to handle any negative effects and guarantee their safety.

5. Supportive treatment: Patients get supportive treatment to manage side effects and maximise their comfort and well-being during the surgery and hospital stay. This might involve supportive therapy to address certain issues like CRS or neurotoxicity, as well as drugs to reduce symptoms like fever, nausea, and discomfort.

6. Follow-Up: Patients undergo CAR T-cell therapy, a complex procedure requiring careful monitoring and supportive care to optimize outcomes. The treatment is tailored to each patient's individual needs, aiming to achieve a durable, effective anti-cancer response while minimizing risks and complications.

10.3 What happen after CAR T-cell therapy?

Following CAR T-cell treatment, patients move into an important phase of care that involves follow-up surveillance. This is what usually occurs: 

1. Acute Monitoring: After receiving a CAR T-cell infusion, patients are promptly watched for any negative responses or acute side effects. Based on the patient's condition and reaction to therapy, this monitoring typically takes place in a hospital environment for a few days to a few weeks.

2. Management of adverse Effects: Cytokine release syndrome (CRS) and neurotoxicity are two adverse effects that patients may encounter as a result of CAR T-cell treatment. Medical professionals give the proper care, which may include drugs to reduce fever, inflammation, and other symptoms. In order to minimise side effects and maximise comfort and well-being, patients get supportive care.

3. Hospital Discharge: Patients may be allowed to leave the hospital if their side effects have been managed and they are considered stable. Close observation is still necessary, though, and patients usually need to make follow-up consultations with their doctors on a regular basis.

4. Long-Term Follow-Up: Patients receive long-term follow-up to track how well CAR T-cell treatment is working and to look for any problems or late-onset adverse effects. In order to monitor the course of the disease and general health, this follow-up may involve routine blood tests, imaging scans, and clinical assessments.

5. Supportive Care: To address any residual side effects and enhance their quality of life, patients get continuous supportive care. In addition to supportive therapies including physical therapy, dietary support, and mental support, this may involve prescription drugs to treat persistent symptoms.

6. Survivorship Care Planning: Medical professionals create a survivorship care plan specific to each patient as they go from current treatment to survivorship. The purpose of this plan is to address any potential side effects of therapy and to promote long-term wellbeing by including suggestions for supportive care, health maintenance, and continuing monitoring.
7. Education and Resources: 
Post-CAR T-cell treatment, patients receive resources for side effects management, maintaining a healthy lifestyle, seeking help, and managing psychological and emotional aspects. Continuous monitoring, supportive care, and survivorship planning are essential for optimal outcomes, requiring close coordination between patients, carers, and healthcare professionals.91

11. CLINICAL APPLICATIONS OF CAR T-CELLS

Chimeric antigen receptor T-cell therapy (CAR T-cell therapy) is an immunotherapy method that uses the patient's immune system to target and eliminate cancer cells. It requires genetic modification to express chimeric antigen receptors on cancer cells' surface. CAR T-cell therapy has shown remarkable results in treating certain cancers, particularly those with haematological malignancies. The following are some important clinical uses:

11.1 Acute lymphoblastic leukaemia (ALL): CAR-T cells are used in treating haematological malignancies like Acute lymphoblastic leukaemia (ALL), which is characterized by rapid proliferation of naïve cells in the bone marrow. They have shown efficacy in treating ALL, especially when engineered T cells target CD19, a highly expressed biomarker of the B-cell lineage. However, T-cell malignancy of ALL showed limited efficacy when engineered CAR T-cells targeted CD19. Anti-CD4 CAR-T cells show promising results against T-cell lymphoma models. Clinical trials have shown promising results with multitargeted CAR-T cell therapy.92-95

11.2 Chronic Lymphocytic Leukaemia (CLL): Chronic lymphocytic leukaemia (CLL) is a condition characterized by excessive mature lymphocytes in the blood, bone marrow, and lymphoid tissue. The use of CD19 as a target for CLL treatment has shown promising results in patients with complete remission and minimal residual disease. Pharmacokinetics play a crucial role in enhancing the outcomes and safety of CAR-T cell treatments. A recent study showed that higher doses effectively induce complete remission without excessive toxicitiesHowever, antigen-negative relapse poses a threat to CAR T-cell therapy's success. Current data suggests that ibrutinib administration and partial reversing of exhausted CAR T-cells in CLL patients are promising. Genetic modification and bispecific CAR targeting antigens on tumour cell surfaces could also improve treatment efficacy.96-102

 11.3 Non-Hodgkin Lymphoma (NHL): Non-Hodgkin's lymphoma (NHL) is a group of neoplasms with varying degrees of malignancy. These malignancies are less predictable and have a higher risk of spreading to extranodal sites. Anti-CD19 CAR-T cells have shown success in treating chemo-resistant lymphomas, while CD22 has shown promising results in patients with diffuse large B-cell lymphoma. In 2017, the FDA approved YESCARTA, a CD19 directed second-generation CAR-T cell product for NHL treatment.103-108

11.4 Multiple Myeloma: Multiple myeloma (MM) is a B-cell malignancy involving long-lived plasma cells that produce specific immunoglobulins. Disease management has been compromised due to the lack of an ideal target. Syndecan 1 (CD138) was used as a target for MM treatment, but it caused "on-target-off-tumour" toxicity. BCMA, a highly expressed B-cell maturation antigen, is considered a better target in CAR-T cell therapy. A phase 1 clinical trial showed preliminary results, with one patient achieving complete remission for over three months. BCMA CAR T-cells have shown minor cytotoxicity and an objective response rate (ORR) of 20%. Bispecific CAR T-cell (LCAR-B38M) targets VHH1 and VHH2 epitopes of BCMA, with an ORR of 88% and a 68% CR. In 2021, the FDA approved ABECMA (idecabtagene vicleucel) for MM, a second-generation CAR-T cell product directed against the BCMA tumour antigen.109,110

11.5 Solid Tumours: CAR-T cell therapies targeting specific antigens have shown poor results in solid tumours, despite the unmet clinical need for these therapies. The biological complexity of this field and the lack of evidence supporting any single CAR-T cell product against specific solid tumours have contributed to these unsatisfactory results. Common factors contributing to these unsatisfactory results include difficulty accessing target cells in poorly vascularized tumour masses, exposure to a hostile tumour microenvironment, and a scarcity of appropriate single-antigen targets. Improvements in CAR-T cell design have the potential to infiltrate and counteract immunosuppressive milieu, and experiments are underway to promote tumour-associated antigen expression via radiation or epigenetic treatment 111. The most practical and successful use for CAR-T cells in solid tumours is to deepen or sustain chemotherapy, radiation, or surgically induced remission in diseases with a high risk of recurrence. Replicating techniques used in blood cancers is unlikely to yield clinical benefits, and finding the right single-antigen target for third-generation CAR-T cells will require more complicated, new techniques. Addressing these concerns could provide fascinating outcomes for solid malignancies, representing a significant unmet need and a potentially profitable market for investors.

11.6 HIV- Infection: CAR T-cell therapy, while effective in treating some cancers, is still experimental and faces challenges in treating HIV infection. However, studies are exploring its potential as an HIV/AIDS therapeutic strategy.

CAR T-cell therapy is a promising and rapidly developing cancer treatment method, providing hope to patients with limited treatment options. Current studies aim to expand cancer types and improve safety and effectiveness of this treatment.112,113

12. CAR T-CELLS: FUTURE PERSPECTIVES

CD19-expressing blood cancers are most suitable for CAR-T cell therapy due to their high tumour expression, easy access, and tolerability of B cell aplasia. However, only 5% of new cancer diagnoses are CD19-targetable by licensed products. Innovative strategies to improve tumour killing efficacy, CAR-T cell persistence, and activity control are being pursued to bring CAR-T cell therapies to other diseases. 

The future of CAR T-cell therapy is promising, with ongoing research and development aimed at overcoming current challenges and enhancing its therapeutic potential. Some potential prospects for CAR T-cell treatment include:

12.1 Advancements in Target Identification: Researchers are discovering new tumour-specific antigens for CAR T-cell therapy, expanding the target repertoire to treat a wider range of malignancies and reducing antigen escape.

12.2 Next-Generation CAR Designs: Future CAR designs may enhance T-cell persistence, invasion, and specificity by developing dual-targeted and switchable CARs that target multiple antigens simultaneously114.

12.3 Allogeneic CAR T-cells: Off-the-shelf universal CAR T-cells from healthy donors may address manufacturing issues in patient-specific treatment, while strategies to reduce graft versus-host disease risk and improve allogeneic CAR T-cell durability are being developed.

12.4 Gene Editing Technologies: Advances in gene editing technologies likeCRISPR-Cas9 show promise for enhancing the functionality and safety of CART cells by disrupting fatigue-related genes or boosting T-cell activity115.

12.5 Combination Treatments: CAR T-cell therapy is being combined with other therapeutic options like checkpoint inhibitors, targeted treatments, and chemotherapy to prevent resistance, enhance antitumor immunity, and enhance

treatment outcomes.

12.6 Expanded Applications Beyond Oncology: CAR T-cell therapy, primarily used in cancer treatment, is gaining interest for potential applications in autoimmune, infectious, and transplantation diseases, despite challenges and promising therapeutic innovation.

12.7 Personalized Medicine Approaches: Advances in biomarker identification and patient classification may enable more personalized CAR T-cell treatment, focusing on specific patient features like tumour genetics, immunology, and microenvironmental variables.

12.8 Improved Manufacturing and Accessibility: The CAR T-cell treatment manufacturing process is being improved through automation, closed-system production platforms, and alternative cell sources to reduce costs, enhances calability, and enhance patient access.

CAR T-cell therapy’s future is marked by continuous innovation, with potential to transform cancer treatment and expand its use to other diseases.

Collaboration between academics, industry, and regulatory authorities is needed 115-118.

DISCUSSION AND CONCLUSION

CAR T-cell therapy (Chimeric Antigen Receptor T-cell therapy) is a form of immunotherapy that involves modifying a patient's T cells to attack cancer cells. Here are some advantages includes that the therapy has shown remarkable efficacy, particularly in hematologic malignancies like certain types of leukaemia and lymphoma. It provides long-lasting remissions, potentially resulting to cure different types of cancer. The disadvantages include that CRS is a potentially life-threatening adverse effect of CAR T-cell treatment produced by cytokine production in response to T cell activation. It can cause fever, hypotension, and organ malfunction, some people may develop neurotoxicity, which can cause disorientation, seizures, or other neurological symptoms. This adverse effect is frequently connected with CRS.CD19-expressing blood malignancies are most suited for CAR-T cell treatment due to their high tumour expression, ease of access, and tolerance of B cell aplasia. Only 5% of new cancer diagnoses may be treated with approved drugs that target CD19. Innovative ways for improving tumour killing effectiveness, CAR-T cell persistence, and activity control are being developed in order to introduce CAR-T cell therapy to various disorders. The future of CAR T-cell therapy is bright, with continuing research and development targeted at overcoming present obstacles and increasing therapeutic potential. 

Conflict of interest: The authors declare no conflict of interest.

Acknowledgement: The authors are thankful to the management and principal, Columbia Institute of Pharmacy, Raipur (C.G.) for providing necessary facilities to complete this review article.

Funding: We do not receive any kind of funding from any source for this review article.

Author’s contribution:

Darshana Rathi: Diagram, figures and overall correction 

Nikita Patel: Preparation of content   

Trilochan Satapathy: Proof reading 

REFERENCES

1. Dardamani DG. Modern Biotechnological Approaches in Diagnosis and Treatment of Lung Cancer.

2. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules asfunctional receptors with antibody-type specificity. Proceedings of the National Academy ofSciences. 1989 Dec;86(24):10024-8. https://doi.org/10.1073/pnas.86.24.10024 PMid:2513569 PMCid:PMC298636

3. Gross G, Gorochov G, Waks T, Eshhar Z. Generation of effector T-cells expressing chimeric T-cellreceptor with antibody type-specificity. InTransplantation proceedings 1989;21(1):127-130

4. Porter DL, Levine BL, Kalos M, Bagg A, June CH. Chimeric antigen receptor-modified T cells inchronic lymphoid leukemia. New England Journal of Medicine. 2011 Aug 25;365(8):725-33. https://doi.org/10.1056/NEJMoa1103849 PMid:21830940 PMCid:PMC3387277

5. Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B,Wright JF, Milone MC. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia.New England Journal of Medicine. 2013 Apr 18;368(16):1509-18. https://doi.org/10.1056/NEJMoa1215134 PMid:23527958 PMCid:PMC4058440

6. Gargett T, Brown MP. The inducible caspase-9 suicide gene system as a "safety switch" to limit on-target, off-tumor toxicities of chimeric antigen receptor T cells. Frontiers in pharmacology. 2014 Oct28;5:117198. https://doi.org/10.3389/fphar.2014.00235 PMid:25389405 PMCid:PMC4211380

7. Chmielewski M, Abken H. TRUCKs: the fourth generation of CARs. Expert opinion on biologicaltherapy. 2015 Aug 3;15(8):1145-54. https://doi.org/10.1517/14712598.2015.1046430 PMid:25985798

8. Rezvani K, Rouce RH. The application of natural killer cell immunotherapy for the treatment ofcancer. Frontiers in immunology. 2015 Nov 17;6:169989.. https://doi.org/10.3389/fimmu.2015.00578

9. Ren J, Zhao Y. Advancing chimeric antigen receptor T cell therapy with CRISPR/Cas9. Protein &cell. 2017 Sep;8(9):634-43. https://doi.org/10.1007/s13238-017-0410-x PMid:28434148 PMCid:PMC5563282

10. Schultz LM, Muffly LS, Spiegel JY, Ramakrishna S, Hossain N, Baggott C, Sahaf B, Patel S, Craig J,Yoon J, Kadapakkam M. Phase I trial using CD19/CD22 bispecific CAR T cells in pediatric and adultacute lymphoblastic leukemia (ALL). Blood. 2019 Nov 13;134:744. https://doi.org/10.1182/blood-2019-129411

11. Lee S, Kim TD. Breakthroughs in cancer immunotherapy: an overview of T cell, NK cell, Mφ, and DC-based treatments. International Journal of Molecular Sciences. 2023 Dec 18;24(24):17634. https://doi.org/10.3390/ijms242417634 PMid:38139461 PMCid:PMC10744055

12.CAR T. cells: engineering patients' immune cells to treat their cancers. Originally published by National Cancer Institute. 2022.

13.Passweg JR, Baldomero H, Chabannon C, Basak GW, Corbacioglu S, Duarte R, Dolstra H, Lankester AC, Mohty M, Montoto S, Peffault de Latour R. The EBMT activity survey on hematopoietic-cell transplantation and cellular therapy 2018: CAR-T's come into focus. Bone marrow transplantation. 2020 Aug;55(8):1604-13. https://doi.org/10.1038/s41409-020-0826-4 PMid:32066864 PMCid:PMC7391287

14. Ricardo Goulart L, Souza Santos P, Paula Carneiro A, Brasil Santana B, C Vallinoto A, GoncalvesAraujo T. Unraveling antibody display: systems biology and personalized medicine. CurrentPharmaceutical Design. 2016 Dec 1;22(43):6560-76. https://doi.org/10.2174/1381612822666160923112816 PMid:27669968

15. Zhang C, Liu J, Zhong JF, Zhang X. Engineering car-t cells. Biomarker research. 2017 Dec;5:1-6. https://doi.org/10.1186/s40364-017-0102-y PMid:28652918 PMCid:PMC5482931

16. Ramos CA, Dotti G. Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer therapy.Expert opinion on biological therapy. 2011 Jul 1;11(7):855-73. https://doi.org/10.1517/14712598.2011.573476 PMid:21463133 PMCid:PMC3107373

17. Cantrell DA. T-cell antigen receptor signal transduction. Immunology. 2002 Apr;105(4):369. https://doi.org/10.1046/j.1365-2567.2002.01391.x PMid:11985657 PMCid:PMC1782684

18. Finney HM, Lawson AD, Bebbington CR, Weir AN. Chimeric receptors providing both primary andcostimulatory signaling in T cells from a single gene product. The Journal of Immunology. 1998 Sep15;161(6):2791-7. https://doi.org/10.4049/jimmunol.161.6.2791 PMid:9743337

19. Sadelain M, Brentjens R, Rivière I. The basic principles of chimeric antigen receptor design. Cancerdiscovery. 2013 Apr 1;3(4):388-98. https://doi.org/10.1158/2159-8290.CD-12-0548 PMid:23550147 PMCid:PMC3667586

20. Chmielewski M, Hombach AA, Abken H. Of CAR s and TRUCK s: chimeric antigen receptor (CAR)T cells engineered with an inducible cytokine to modulate the tumor stroma. Immunological reviews.2014 Jan;257(1):83-90. https://doi.org/10.1111/imr.12125 Mid:24329791

21. Kubin M, Kamoun M, Trinchieri G. Interleukin 12 synergizes with B7/CD28 interaction in inducingefficient proliferation and cytokine production of human T cells. The Journal of experimentalmedicine. 1994 Jul 1;180(1):211 https://doi.org/10.1084/jem.180.1.211 PMid:7516408 PMCid:PMC2191554

22. Hurton LV, Singh H, Najjar AM, Switzer KC, Mi T, Maiti S, Olivares S, Rabinovich B, Huls H,Forget MA, Datar V. Tethered IL-15 augments antitumor activity and promotes a stem-cell memorysubset in tumor-specific T cells. Proceedings of the National Academy of Sciences. 2016 Nov29;113(48):E7788-97. https://doi.org/10.1073/pnas.1610544113 PMid:27849617 PMCid:PMC5137758

23. Klebanoff CA, Finkelstein SE, Surman DR, Lichtman MK, Gattinoni L, Theoret MR, Grewal N,Spiess PJ, Antony PA, Palmer DC, Tagaya Y. IL-15 enhances the in vivo antitumor activity of tumor-reactive CD8+ T cells. Proceedings of the National Academy of Sciences. 2004 Feb 17;101(7):1969-74. https://doi.org/10.1073/pnas.0307298101 PMid:14762166 PMCid:PMC357036

24. Chmielewski M, Abken H. CAR T cells releasing IL-18 convert to T-Bethigh FoxO1low effectorsthat exhibit augmented activity against advanced solid tumors. Cell reports. 2017 Dec12;21(11):3205-19. https://doi.org/10.1016/j.celrep.2017.11.063 PMid:29241547

25. Tokarew N, Ogonek J, Endres S, von Bergwelt-Baildon M, Kobold S. Teaching an old dog newtricks: next-generation CAR T cells. British journal of cancer. 2019 Jan 8;120(1):26-37. https://doi.org/10.1038/s41416-018-0325-1 PMid:30413825 PMCid:PMC6325111

26. Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C,Olszewska M, Borquez-Ojeda O. CD19-targeted T cells rapidly induce molecular remissions in adultswith chemotherapy-refractory acute lymphoblastic leukemia. Science translational medicine. 2013Mar 20;5(177):177ra38-. https://doi.org/10.1126/scitranslmed.3005930 PMid:23515080 PMCid:PMC3742551

27.Hartmann J, SchüßlerLenz M, Bondanza A, Buchholz CJ. Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts. EMBO molecular medicine. 2017Sep;9(9):1183-97. https://doi.org/10.15252/emmm.201607485 PMid:28765140 PMCid:PMC5582407

28.Jin C, Yu D, Hillerdal V, Wallgren A, Karlsson-Parra A, Essand M.Allogeneic lymphocyte-licensed DCs expand T cells with improved antitumoractivity and resistance to oxidative stress and immunosuppressive factors.Molecular Therapy-Methods & Clinical Development. 2014 Jan 1;1. https://doi.org/10.1038/mtm.2014.1 PMid:26015949 PMCid:PMC4362340

29.Li N, Ho M. Development of Glypican-2 Targeting Single-DomainAntibody CAR T Cells for Neuroblastoma. In Single-Domain Antibodies:Methods and Protocols 2022 Feb 14 (pp. 451-468). New York, NY: SpringerUS. https://doi.org/10.1007/978-1-0716-2075-5_23 PMid:35157288

30.Makita S, Yoshimura K, Tobinai K. Clinical development of antiCD 19chimeric antigen receptor Tcell therapy for Bcell nonHodgkin lymphoma.Cancer science. 2017 Jun;108(6):1109-18. https://doi.org/10.1111/cas.13239 PMid:28301076 PMCid:PMC5480083

31.Jin C, Fotaki G, Ramachandran M, Nilsson B, Essand M, Yu D. Safeengineering of CAR T cells for adoptive cell therapy of cancer using longtermepisomal gene transfer. EMBO molecular medicine. 2016 Jul;8(7):702-11. https://doi.org/10.15252/emmm.201505869 PMid:27189167 PMCid:PMC4931286

32. Jensen TI, Axelgaard E, Bak RO. Therapeutic gene editing inhaematological disorders with CRISPR/Cas9. British journal of haematology.2019 Jun;185(5):821-35. https://doi.org/10.1111/bjh.15851 PMid:30864164

33. Muranski P, Boni A, Wrzesinski C, Citrin DE, Rosenberg SA, Childs R,Restifo NP. Increased intensity lymphodepletion and adoptiveimmunotherapy-how far can we go?. Nature clinical practice Oncology. 2006Dec 1;3(12):668-81. https://doi.org/10.1038/ncponc0666 PMid:17139318 PMCid:PMC1773008

34.Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A, Hauck B,Wright JF, Milone MC. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia.New England Journal of Medicine. 2013 Apr 18;368(16):1509-18. https://doi.org/10.1056/NEJMoa1215134 PMid:23527958 PMCid:PMC4058440

35.Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, Chung SS, Stefanski J, Borquez-OjedaO, Olszewska M, Qu J. Efficacy and toxicity management of 19-28z CAR T cell therapy in B cellacute lymphoblastic leukemia. Science translational medicine. 2014 Feb 19;6(224):224ra25-.

36.Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M,Yang JC, Phan GQ, Hughes MS, Sherry RM, Raffeld M. Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cell malignancies can be effectively treated with autologous T cellsexpressing an anti-CD19 chimeric antigen receptor. Journal of clinical oncology. 2015 Feb2;33(6):540. https://doi.org/10.1200/JCO.2014.56.2025 PMid:25154820 PMCid:PMC4322257

37.Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fučíková J, Cremer I, Galon J, Tartour E, ZitvogelL, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based interventions for cancer therapy.Oncoimmunology. 2013 Oct 1;2(10):e25771. https://doi.org/10.4161/onci.25771 PMid:24286020 PMCid:PMC3841205

38.Kim JV, Latouche JB, Rivière I, Sadelain M. The ABCs of artificial antigen presentation. Naturebiotechnology. 2004 Apr 1;22(4):403-10. https://doi.org/10.1038/nbt955 PMid:15060556

39.Singh H, Huls H, Kebriaei P, Cooper LJ. A new approach to gene therapy using Sleeping Beauty togenetically modify clinicalgrade T cells to target CD 19. Immunological reviews. 2014Jan;257(1):181-90. Odendahl, M, Grigoleit, GU, Bönig, H, Neuenhahn, M, Albrecht, J, Anderl, Fet al. (2014). Clinicalscale isolation of 'minimally manipulated' cytomegalovirus-specific donorlymphocytes for the treatment of refractory cytomegalovirus disease. Cytotherapy16: 1245-1256. https://doi.org/10.1016/j.jcyt.2014.05.023 PMid:25108651

40. Freimüller, C, Stemberger, J, Artwohl, M, Germeroth, L, Witt, V, Fischer, G et al. (2015). Selectionof adenovirus-specific and Epstein-Barr virus-specific T cells with major h

41.Freimüller C, Stemberger J, Artwohl M, Germeroth L, Witt V, Fischer G, Tischer S, Eiz-Vesper B,Knippertz I, Dörrie J, Schaft N. Selection of adenovirus-specific and Epstein-Barr virus-specific T-cells with major histocompatibility class I streptamers under Good Manufacturing Practice(GMP)-compliant conditions. Cytotherapy. 2015 Jul 1;17(7):989-1007. https://doi.org/10.1016/j.jcyt.2015.03.613 PMid:25866178

42. Bashour KT, Graef P, Stemberger C, Lothar G, Odegard V and Ramsborg CG (2015). Functionalcharacterization of a T cell stimulation reagent for the production of therapeutic chimeric antigenreceptor T cells. ASH 57th Annual Meeting & Exposition, Orlando, FL. https://doi.org/10.1182/blood.V126.23.1901.1901

43. Brentjens RJ, Latouche JB, Santos E, Marti F, Gong MC, Lyddane C, King PD, Larson S, Weiss M,Rivière I, Sadelain M. Eradication of systemic B-cell tumors by genetically targeted human Tlymphocytes co-stimulated by CD80 and interleukin-15. Nature medicine. 2003 Mar 1;9(3):279-86. https://doi.org/10.1038/nm827 PMid:12579196

44. Deichmann A, Schmidt M. Biosafety considerations using gamma-retroviral vectors in gene therapy.Current gene therapy. 2013 Dec 1;13(6):469-77. https://doi.org/10.2174/15665232113136660004 PMid:24195605

45. Ghani K, Wang X, de Campos-Lima PO, Olszewska M, Kamen A, Riviere I, Caruso M. Efficienthuman hematopoietic cell transduction using RD114-and GALV-pseudotyped retroviral vectorsproduced in suspension and serum-free media. Human gene therapy. 2009 Sep 1;20(9):966-74. https://doi.org/10.1089/hum.2009.001 PMid:19453219 PMCid:PMC2861952

46. Miller AD, Garcia JV, Von Suhr N, Lynch CM, Wilson C, Eiden MV. Construction and properties ofretrovirus packaging cells based on gibbon ape leukemia virus. Journal of virology. 1991May;65(5):2220-4. https://doi.org/10.1128/jvi.65.5.2220-2224.1991 PMid:1850008 PMCid:PMC240569

47. Bonini C, Grez M, Traversari C, Ciceri F, Marktel S, Ferrari G, Dinauer M, Sadat M, Aiuti A, DeolaS, Radrizzani M. Safety of retroviral gene marking with a truncated NGF receptor. Nature medicine.2003 Apr 1;9(4):367-9. https://doi.org/10.1038/nm0403-367 PMid:12669036

48. Brenner MK, Heslop HE. Is retroviral gene marking too dangerous to use?. Cytotherapy. 2003 Jan1;5(3):190-3. https://doi.org/10.1080/14653240310001307 PMid:12850996

49. Macpherson JL, Boyd MP, Arndt AJ, Todd AV, Fanning GC, Ely JA, Elliott F, Knop A, Raponi M,Murray J, Gerlach W. Longterm survival and concomitant gene expression of ribozymetransducedCD4+ Tlymphocytes in HIVinfected patients. The Journal of Gene Medicine: A crossdisciplinaryjournal for research on the science of gene transfer and its clinical applications. 2005 May;7(5):552-64. https://doi.org/10.1002/jgm.705 PMid:15655805

50. Muul LM, Tuschong LM, Soenen SL, Jagadeesh GJ, Ramsey WJ, Long Z, Carter CS, GarabedianEK, Alleyne M, Brown M, Bernstein W. Persistence and expression of the adenosine deaminase genefor 12 years and immune reaction to gene transfer components: long-term results of the first clinicalgene therapy trial. Blood, The Journal of the American Society of Hematology. 2003 Apr1;101(7):2563-9. https://doi.org/10.1182/blood-2002-09-2800 PMid:12456496

51. Scholler J, Brady TL, Binder-Scholl G, Hwang WT, Plesa G, Hege KM, Vogel AN, Kalos M, RileyJL, Deeks SG, Mitsuyasu RT. Decade-long safety and function of retroviral-modified chimericantigen receptor T cells. Science translational medicine. 2012 May 2;4(132):132ra53-. https://doi.org/10.1126/scitranslmed.3003761 PMid:22553251 PMCid:PMC4368443

52. Wang X, Olszewska M, Qu J, Wasielewska T, Bartido S, Hermetet G, Sadelain M, Riviere I. Large-scale clinical-grade retroviral vector production in a fixed-bed bioreactor. Journal of Immunotherapy.2015 Apr 1;38(3):127-35. https://doi.org/10.1097/CJI.0000000000000072 PMid:25751502 PMCid:PMC4353472

53. Naldini L, Blömer U, Gallay P, Ory D, Mulligan R, Gage FH, Verma IM, Trono D. In vivo genedelivery and stable transduction of nondividing cells by a lentiviral vector. Science. 1996 Apr12;272(5259):263-7. https://doi.org/10.1126/science.272.5259.263 PMid:8602510

54. Vannucci L, Lai M, Chiuppesi F, Ceccherini-Nelli L, Pistello M. Viral vectors: a look back andahead on gene transfer technology. New Microbiologica. 2013;36(1):1-22.

55. Ni Y, Sun S, Oparaocha I, Humeau L, Davis B, Cohen R, Binder G, Chang YN, Slepushkin V,Dropulic B. Generation of a packaging cell line for prolonged largescale production of hightiterHIV1based lentiviral vector. The Journal of Gene Medicine: A crossdisciplinary journal forresearch on the science of gene transfer and its clinical applications. 2005 Jun;7(6):818-34. https://doi.org/10.1002/jgm.726 PMid:15693055

56. Throm RE, Ouma AA, Zhou S, Chandrasekaran A, Lockey T, Greene M, De Ravin SS, Moayeri M,Malech HL, Sorrentino BP, Gray JT. Efficient construction of producer cell lines for a SIN lentiviralvector for SCID-X1 gene therapy by concatemeric array transfection. Blood, The Journal of theAmerican Society of Hematology. 2009 May 21;113(21):5104-10. https://doi.org/10.1182/blood-2008-11-191049 PMid:19286997 PMCid:PMC2686181

57. Zhao Y, Zheng Z, Cohen CJ, Gattinoni L, Palmer DC, Restifo NP, Rosenberg SA, Morgan RA. High-efficiency transfection of primary human and mouse T lymphocytes using RNA electroporation.Molecular therapy. 2006 Jan 1;13(1):151-9. https://doi.org/10.1016/j.ymthe.2005.07.688 PMid:16140584 PMCid:PMC1473967

58. Yoon SH, Lee JM, Cho HI, Kim EK, Kim HS, Park MY, Kim TG. Adoptive immunotherapy usinghuman peripheral blood lymphocytes transferred with RNA encoding Her-2/neu-specific chimericimmune receptor in ovarian cancer xenograft model. Cancer gene therapy. 2009 Jun;16(6):489-97. https://doi.org/10.1038/cgt.2008.98 PMid:19096447

59. Rowley J, Monie A, Hung CF, Wu TC. Expression of IL15RA or an IL15/IL15RA fusion on CD8+T cells modifies adoptively transferred Tcell function in cis. European journal of immunology. 2009Feb;39(2):491-506. https://doi.org/10.1002/eji.200838594

PMid:19180469 PMCid:PMC3004157

60. Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, Chew A, Zhao Y, Levine BL,Albelda SM, Kalos M. Mesothelin-specific chimeric antigen receptor mRNA-engineered T cellsinduce antitumor activity in solid malignancies. Cancer immunology research. 2014 Feb 1;2(2):112-20. https://doi.org/10.1158/2326-6066.CIR-13-0170 PMid:24579088 PMCid:PMC3932715

61. Poirot L, Philip B, Schiffer-Mannioui C, Le Clerre D, Chion-Sotinel I, Derniame S, Potrel P, Bas C,Lemaire L, Galetto R, Lebuhotel C. Multiplex genome-edited T-cell manufacturing platform for "off-the-shelf" adoptive T-cell immunotherapies. Cancer research. 2015 Sep 15;75(18):3853-64. https://doi.org/10.1158/0008-5472.CAN-14-3321 PMid:26183927

62. https://www.cancer.gov/about-cancer/treatment/research/car-t-cells 

63. Kuwana Y, Asakura Y, Utsunomiya N, Nakanishi M, Arata Y, Itoh S, Nagase F, Kurosawa Y.Expression of chimeric receptor composed of immunoglobulin-derived V resions and T-cell receptor-derived C regions. Biochemical and biophysical research communications. 1987 Dec 31;149(3):960-8. https://doi.org/10.1016/0006-291X(87)90502-X PMid:3122749

64. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell receptor chimeric molecules asfunctional receptors with antibody-type specificity. Proceedings of the National Academy ofSciences. 1989 Dec;86(24):10024-8. https://doi.org/10.1073/pnas.86.24.10024 PMid:2513569 PMCid:PMC298636

65. Eshhar Z, Waks T, Gross G, Schindler DG. Specific activation and targeting of cytotoxiclymphocytes through chimeric single chains consisting of antibody-binding domains and the gammaor zeta subunits of the immunoglobulin and T-cell receptors. Proceedings of the National Academy ofSciences. 1993 Jan 15;90(2):720-4. https://doi.org/10.1073/pnas.90.2.720 PMid:8421711 PMCid:PMC45737

66. Bird RE, Hardman KD, Jacobson JW, Johnson S, Kaufman BM, Lee SM, Lee T, Pope SH, RiordanGS, Whitlow M. Single-chain antigen-binding proteins. Science. 1988 Oct 21;242(4877):423-6. https://doi.org/10.1126/science.3140379 PMid:3140379

67. Huston JS, Levinson D, Mudgett-Hunter M, Tai MS, Novotný J, Margolies MN, Ridge RJ,Bruccoleri RE, Haber E, Crea R. Protein engineering of antibody binding sites: recovery of specificactivity in an anti-digoxin single-chain Fv analogue produced in Escherichia coli. Proceedings of theNational Academy of Sciences. 1988 Aug;85(16):5879-83. https://doi.org/10.1073/pnas.85.16.5879 PMid:3045807 PMCid:PMC281868

68. Eshhar Z. Tumor-specific T-bodies: towards clinical application. Cancer Immunology,Immunotherapy. 1997 Nov;45:131-6. https://doi.org/10.1007/s002620050415 PMid:9435856 PMCid:PMC11037636

69. Moritz D, Wels W, Mattern J, Groner B. Cytotoxic T lymphocytes with a grafted recognitionspecificity for ERBB2-expressing tumor cells. Proceedings of the National Academy of Sciences.1994 May 10;91(10):4318-22. https://doi.org/10.1073/pnas.91.10.4318 PMid:7910405 PMCid:PMC43776

70. Hwu P, Shafer GE, Treisman J, Schindler DG, Gross G, Cowherd R, Rosenberg SA, Eshhar Z. Lysisof ovarian cancer cells by human lymphocytes redirected with a chimeric gene composed of anantibody variable region and the Fc receptor gamma chain. The Journal of experimental medicine.1993 Jul 1;178(1):361-6. https://doi.org/10.1084/jem.178.1.361 PMid:8315392 PMCid:PMC2191075

71. Hwu P, Yang JC, Cowherd R, Treisman J, Shafer GE, Eshhar Z, Rosenberg SA. In vivo antitumoractivity of T cells redirected with chimeric antibody/T-cell receptor genes. Cancer research. 1995 Aug1;55(15):3369-73.

72. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, White DE,Wunderlich JR, Canevari S, Rogers-Freezer L, Chen CC. A phase I study on adoptiveimmunotherapy using gene-modified T cells for ovarian cancer. Clinical cancer research. 2006 Oct15;12(20):6106-15. https://doi.org/10.1158/1078-0432.CCR-06-1183 PMid:17062687 PMCid:PMC2154351

73. Lamers CH, Sleijfer S, Vulto AG, Kruit WH, Kliffen M, Debets R, Gratama JW, Stoter G,Oosterwijk E. Treatment of metastatic renal cell carcinoma with autologous T-lymphocytesgenetically retargeted against carbonic anhydrase IX: first clinical experience. Journal of clinicaloncology: official journal of the American Society of Clinical Oncology. 2006 May 1;24(13):e20-2. https://doi.org/10.1200/JCO.2006.05.9964 PMid:16648493

74. Till BG, Jensen MC, Wang J, Chen EY, Wood BL, Greisman HA, Qian X, James SE, Raubitschek A,Forman SJ, Gopal AK. Adoptive immunotherapy for indolent non-Hodgkin lymphoma and mantlecell lymphoma using genetically modified autologous CD20-specific T cells. Blood, The Journal ofthe American Society of Hematology. 2008 Sep 15;112(6):2261-71. https://doi.org/10.1182/blood-2007-12-128843 PMid:18509084 PMCid:PMC2532803

75. Park JR, DiGiusto DL, Slovak M, Wright C, Naranjo A, Wagner J, Meechoovet HB, Bautista C,Chang WC, Ostberg JR, Jensen MC. Adoptive transfer of chimeric antigen receptor re-directedcytolytic T lymphocyte clones in patients with neuroblastoma. Molecular therapy. 2007 Apr1;15(4):825-33. https://doi.org/10.1038/sj.mt.6300104 PMid:17299405

76. Pule MA, Savoldo B, Myers GD, Rossig C, Russell HV, Dotti G, Huls MH, Liu E, Gee AP, Mei Z,Yvon E. Virus-specific T cells engineered to coexpress tumor-specific receptors: persistence andantitumor activity in individuals with neuroblastoma. Nature medicine. 2008 Nov;14(11):1264-70. https://doi.org/10.1038/nm.1882 PMid:18978797 PMCid:PMC2749734

77. Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7 system of T cell costimulation. Annual reviewof immunology. 1996 Apr;14(1):233-58. https://doi.org/10.1146/annurev.immunol.14.1.233 PMid:8717514

78. Krause A, Guo HF, Latouche JB, Tan C, Cheung NK, Sadelain M. Antigen-dependent CD28signaling selectively enhances survival and proliferation in genetically modified activated humanprimary T lymphocytes. The Journal of experimental medicine. 1998 Aug 17;188(4):619-26. https://doi.org/10.1084/jem.188.4.619 PMid:9705944 PMCid:PMC2213361

79. Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M. Human T-lymphocyte cytotoxicity andproliferation directed by a single chimeric TCRζ/CD28 receptor. Nature biotechnology. 2002 Jan1;20(1):70-5. https://doi.org/10.1038/nbt0102-70 PMid:11753365

80. Savoldo B, Ramos CA, Liu E, Mims MP, Keating MJ, Carrum G, Kamble RT, Bollard CM, Gee AP,Mei Z, Liu H. CD28 costimulation improves expansion and persistence of chimeric antigenreceptor-modified T cells in lymphoma patients. The Journal of clinical investigation. 2011 May2;121(5):1822-6. https://doi.org/10.1172/JCI46110 PMid:21540550 PMCid:PMC3083795

81. Finney HM, Akbar AN, Lawson AD. Activation of resting human primary T cells with chimericreceptors: costimulation from CD28, inducible costimulator, CD134, and CD137 in series withsignals from the TCRζ chain. The Journal of Immunology. 2004 Jan 1;172(1):104-13. https://doi.org/10.4049/jimmunol.172.1.104 PMid:14688315

82. Imai CM, Mihara K, Andreansky M, Nicholson IC, Pui CH, Geiger TL, Campana D. Chimericreceptors with 4-1BB signaling capacity provoke potent cytotoxicity against acute lymphoblasticleukemia. Leukemia. 2004 Apr;18(4):676-84. https://doi.org/10.1038/sj.leu.2403302 PMid:14961035

83. Milone MC, Fish JD, Carpenito C, Carroll RG, Binder GK, Teachey D, Samanta M, Lakhal M, GlossB, Danet-Desnoyers G, Campana D. Chimeric receptors containing CD137 signal transductiondomains mediate enhanced survival of T cells and increased antileukemic efficacy in vivo. Moleculartherapy. 2009 Aug 1;17(8):1453-64. https://doi.org/10.1038/mt.2009.83 PMid:19384291 PMCid:PMC2805264

84. Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, Maric I,Raffeld M, Nathan DA, Lanier BJ, Morgan RA. Eradication of B-lineage cells and regression oflymphoma in a patient treated with autologous T cells genetically engineered to recognize CD19.Blood, The Journal of the American Society of Hematology. 2010 Nov 18;116(20):4099-102. https://doi.org/10.1182/blood-2010-04-281931 PMid:20668228 PMCid:PMC2993617

85. Brentjens RJ, Rivière I, Park JH, Davila ML, Wang X, Stefanski J, Taylor C, Yeh R, Bartido S,Borquez-Ojeda O, Olszewska M. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood, TheJournal of the American Society of Hematology. 2011 Nov 3;118(18):4817-28. https://doi.org/10.1182/blood-2011-04-348540 PMid:21849486 PMCid:PMC3208293

86. Sahu M, Satapathy T, Bahadur S, Saha S, Purabiya P, Kaushik S, Netam AK, Prasad J. Preparation methods for nanoparticle: A smart carrier system for treatment of cancer. March. 2018 Mar 19;7(19):216-6.

87. Satapathy T, kumar Panda P. Gastric Cancer: An Overview. Journal of Advanced Pharmaceutical Research. 2012;3(3):49-57.

88.Khan MA, Satapathy T, Sen K, Sahu S, Pradhan B, Gupta A, Satapathy A, Satapathy A. Pharmacological Targeting of Ferroptosis in Cancer Treatment. JDDT. 2024;14(2):205-21. https://doi.org/10.22270/jddt.v14i2.6371

89. https://www.cancer.gov/about-cancer/treatment/research/car-t-cells 

90.Satapathy T, Kaushik S, Netam AK, Prasad J, Rao SP, Sahu MK, Baghel P. NANO DELIVERY: A SMART CARRIER FOR TREATMENT OF OVARIAN CANCER.

91. Wang K, Wei G, Liu D. CD19: a biomarker for B cell development, lymphomadiagnosis and therapy. Experimental hematology & oncology. 2012 Dec;1:1-7. https://doi.org/10.1186/2162-3619-1-36 PMid:23210908 PMCid:PMC3520838

92. Gill S, Maus MV, Porter DL. Chimeric antigen receptor T cell therapy: 25 yearsin the making. Blood reviews. 2016 May 1;30(3):157-67. https://doi.org/10.1016/j.blre.2015.10.003 PMid:26574053

93. Mamonkin M, Rouce RH, Tashiro H, Brenner MK. A T-cell-directed chimericantigen receptor for the selective treatment of T-cell malignancies. Blood, TheJournal of the American Society of Hematology. 2015 Aug 20;126(8):983-92. https://doi.org/10.1182/blood-2015-02-629527 PMid:26056165 PMCid:PMC4543231

94. Pinz K, Liu H, Golightly M, Jares A, Lan F, Zieve GW, Hagag N, Schuster M,Firor AE, Jiang X, Ma Y. Preclinical targeting of human T-cell malignanciesusing CD4-specific chimeric antigen receptor (CAR)-engineered T cells.Leukemia. 2016 Mar;30(3):701-7. https://doi.org/10.1038/leu.2015.311 PMid:26526988

95. Nanda BL, Antioxidant and anticancer activity of edible flowers, Journal of Drug Delivery and Therapeutics. 2019;9(3-s)290-295.

96. Porter DL, Hwang WT, Frey NV, Lacey SF, Shaw PA, Loren AW, Bagg A,Marcucci KT, Shen A, Gonzalez V, Ambrose D. Chimeric antigen receptor Tcells persist and induce sustained remissions in relapsed refractory chroniclymphocytic leukemia. Science translational medicine. 2015 Sep2;7(303):303ra139-. https://doi.org/10.1126/scitranslmed.aac5415

97. Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO,Stetler-Stevenson M, Yang JC, Phan GQ, Hughes MS, Sherry RM, Raffeld M.Chemotherapy-refractory diffuse large B-cell lymphoma and indolent B-cellmalignancies can be effectively treated with autologous T cells expressing ananti-CD19 chimeric antigen receptor. Journal of clinical oncology. 2015 Feb2;33(6):540. https://doi.org/10.1200/JCO.2014.56.2025 PMid:25154820 PMCid:PMC4322257

98. Norelli M, Casucci M, Bonini C, Bondanza A. Clinical pharmacology of CAR-T cells: Linking cellular pharmacodynamics to pharmacokinetics and antitumoreffects. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer. 2016 Jan1;1865(1):90-100. https://doi.org/10.1016/j.bbcan.2015.12.001 PMid:26748354

99. Frey NV, Gill S, Hexner EO, Schuster S, Nasta S, Loren A, Svoboda J,Stadtmauer E, Landsburg DJ, Mato A, Levine BL. Long-term outcomes from arandomized dose optimization study of chimeric antigen receptor modified Tcells in relapsed chronic lymphocytic leukemia. Journal of Clinical Oncology.2020 Sep 9;38(25):2862. https://doi.org/10.1200/JCO.19.03237 PMid:32298202 PMCid:PMC8265376

100. Mancikova V, Smida M. Current state of CAR T-cell therapy in chroniclymphocytic leukemia. International Journal of Molecular Sciences. 2021 May24;22(11):5536. https://doi.org/10.3390/ijms22115536 PMid:34073911 PMCid:PMC8197365

101. Parajapati SK, Maurya SD, Das MK, Tilak VK, Verma KK, Dhakar RC, Potential application of dendrimers in drug delivery: A concise review and update, Journal of Drug Delivery and Therapeutics, 2016;6(2):71-88 https://doi.org/10.22270/jddt.v6i2.1195

102. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA,Braunschweig I, Oluwole OO, Siddiqi T, Lin Y, Timmerman JM. Axicabtageneciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. NewEngland Journal of Medicine. 2017 Dec 28;377(26):2531-44.

103. Schuster SJ, Svoboda J, Chong EA, Nasta SD, Mato AR, Anak Ö, Brogdon JL,Pruteanu-Malinici I, Bhoj V, Landsburg D, Wasik M. Chimeric antigenreceptor T cells in refractory B-cell lymphomas. New England Journal ofMedicine. 2017 Dec 28;377(26):2545-54. https://doi.org/10.1056/NEJMoa1708566 PMid:29226764 PMCid:PMC5788566

104. Abramson JS, Chen YB. More on anti-CD19 CAR T cells in CNS diffuse large-B-cell lymphoma. The New England Journal of Medicine. 2017 Nov1;377(21):2102-. https://doi.org/10.1056/NEJMc1712460

105. Fry TJ, Stetler-Stevenson M, Shah NN, Yuan CM, Yates B, Delbrook C, ZhangL, Lee III DW, Stroncek D, Mackall CL. Clinical activity and persistence ofanti-CD22 chimeric antigen receptor in children and young adults withrelapsed/refractory acute lymphoblastic leukemia (ALL). Blood. 2015 Dec3;126(23):1324. https://doi.org/10.1182/blood.V126.23.1324.1324

106. Till BG, Jensen MC, Wang J, Qian X, Gopal AK, Maloney DG, Lindgren CG,Lin Y, Pagel JM, Budde LE, Raubitschek A. CD20-specific adoptiveimmunotherapy for lymphoma using a chimeric antigen receptor with bothCD28 and 4-1BB domains: pilot clinical trial results. Blood, The Journal of theAmerican Society of Hematology. 2012 Apr 26;119(17):3940-50. https://doi.org/10.1182/blood-2011-10-387969 PMid:22308288 PMCid:PMC3350361

107. Bird SA, Boyd K. Multiple myeloma: an overview of management.Palliative care and social practice. 2019 Sep;13:1178224219868235. https://doi.org/10.1177/1178224219868235 PMid:32215370 PMCid:PMC7065505

108. Heffner LT, Jagannath S, Zimmerman TM, Lee KP, Rosenblatt J, Lonial S,Lutz RJ, Czeloth N, Osterroth F, Ruehle M, Beelitz MA. BT062, an antibody-drug conjugate directed against CD138, given weekly for 3 weeks in each 4week cycle: safety and further evidence of clinical activity. Blood. 2012 Nov16;120(21):4042. https://doi.org/10.1182/blood.V120.21.4042.4042

109. Ali SA, Shi V, Maric I, Wang M, Stroncek DF, Rose JJ, Brudno JN, Stetler-Stevenson M, Feldman SA, Hansen BG, Fellowes VS. T cells expressing ananti-B-cell maturation antigen chimeric antigen receptor cause remissions ofmultiple myeloma. Blood, The Journal of the American Society of Hematology.2016 Sep 29;128(13):1688-700. https://doi.org/10.1182/blood-2016-04-711903 PMid:27412889 PMCid:PMC5043125

110. Garfall AL, Maus MV, Hwang WT, Lacey SF, Mahnke YD, Melenhorst JJ,Zheng Z, Vogl DT, Cohen AD, Weiss BM, Dengel K. Chimeric antigenreceptor T cells against CD19 for multiple myeloma. New England Journal ofMedicine. 2015 Sep 10;373(11):1040-7. https://doi.org/10.1056/NEJMoa1504542 PMid:26352815 PMCid:PMC4646711

111. Okoye AA, Picker LJ. CD 4+ Tcell depletion in HIV infection: mechanismsof immunological failure. Immunological reviews. 2013 Jul;254(1):54-64. https://doi.org/10.1111/imr.12066 PMid:23772614 PMCid:PMC3729334

112. Charrot S, Hallam S. CAR-T cells: future perspectives. Hemasphere. 2019Apr 1;3(2):e188. https://doi.org/10.1097/HS9.0000000000000188 PMid:31723827 PMCid:PMC6746028

113. Zhao WH, Liu J, Wang BY, Chen YX, Cao XM, Yang Y, Zhang YL, WangFX, Zhang PY, Lei B, Gu LF. A phase 1, open-label study of LCAR-B38M, achimeric antigen receptor T cell therapy directed against B cell maturationantigen, in patients with relapsed or refractory multiple myeloma. Journal ofhematology & oncology. 2018 Dec;11:1-8. https://doi.org/10.1186/s13045-018-0681-6 PMid:30572922 PMCid:PMC6302465

114. Churchill MJ, Deeks SG, Margolis DM, Siliciano RF, Swanstrom R. HIVreservoirs: what, where and how to target them. Nature Reviews Microbiology.2016 Jan;14(1):55-60. https://doi.org/10.1038/nrmicro.2015.5 PMid:26616417

115. Kim Y, Anderson JL, Lewin SR. Getting the "kill" into "shock and kill":strategies to eliminate latent HIV. Cell host & microbe. 2018 Jan 10;23(1):14-26. https://doi.org/10.1016/j.chom.2017.12.004 PMid:29324227 PMCid:PMC5990418

116. Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. Tcells with chimeric antigen receptors have potent antitumor effects and canestablish memory in patients with advanced leukemia. Science translationalmedicine. 2011 Aug 10;3(95):95ra73-. https://doi.org/10.1126/scitranslmed.3002842 PMCid:PMC3393096

117. Spivak AM, Andrade A, Eisele E, Hoh R, Bacchetti P, Bumpus NN, EmadF, Buckheit III R, McCance-Katz EF, Lai J, Kennedy M. A pilot studyassessing the safety and latency-reversing activity of disulfiram in HIV-1-infected adults on antiretroviral therapy. Clinical infectious diseases. 2014Mar 15;58(6):883-90. https://doi.org/10.1093/cid/cit813 PMid:24336828 PMCid:PMC3935499

118. Zhen A, Peterson CW, Carrillo MA, Reddy SS, Youn CS, Lam BB, ChangNY, Martin HA, Rick JW, Kim J, Neel NC. Long-term persistence and functionof hematopoietic stem cell-derived chimeric antigen receptor T cells in anonhuman primate model of HIV/AIDS. PLoS pathogens. 2017 Dec28;13(12):e1006753. https://doi.org/10.1371/journal.ppat.1006753 PMid:29284044 PMCid:PMC5746250