fbpx
Wikipedia

CAR T cell

In biology, chimeric antigen receptors (CARs)—also known as chimeric immunoreceptors, chimeric T cell receptors or artificial T cell receptors—are receptor proteins that have been engineered to give T cells the new ability to target a specific antigen. The receptors are chimeric in that they combine both antigen-binding and T cell activating functions into a single receptor.

Chimeric antigen receptor T cell production and infusion:
1. T cells are isolated from a patient's blood
2. A new gene encoding a chimeric antigen receptor is incorporated into the T cells
3. Engineered T cells are now specific to a desired target antigen
4. Engineered T cells are expanded in tissue culture
5. Engineered T cells are infused back into the patient

CAR T cell therapy uses T cells engineered with CARs to treat cancer. T cells are modified to recognize cancer cells and destroy them. The standard approach is to harvest T cells from patients, genetically alter them, then infuse the resulting CAR T cells into patients to attack their tumors.[1]

CAR T cells can be derived either autologously from T cells in a patient's own blood or allogeneically from those of a donor. Once isolated, these T cells are genetically engineered to express a specific CAR, using a vector derived from an engineered lentivirus such as HIV (see Lentiviral vector in gene therapy). The CAR programs the T cells to target an antigen present on the tumor cell surface. For safety, CAR T cells are engineered to be specific to an antigen that is expressed on a tumor cell but not on healthy cells.[2]

After the modified T cells are infused into a patient, they act as a "living drug" against cancer cells.[3] When they come in contact with their targeted antigen on a cell's surface, T cells bind to it and become activated, then proceed to proliferate and become cytotoxic.[4] CAR T cells destroy cells through several mechanisms, including extensive stimulated cell proliferation, increasing the degree to which they are toxic to other living cells (cytotoxicity), and by causing the increased secretion of factors that can affect other cells such as cytokines, interleukins and growth factors.[5]

The surface of CAR T cells can bear either of two types of co-receptors, CD4 and CD8. These two cell types, called CD4+ and CD8+, respectively, have different and interacting cytotoxic effects. Therapies employing a 1-to-1 ratio of the cell types apparently provide synergistic antitumor effects.[6]

History edit

The first chimeric receptors containing portions of an antibody and the T cell receptor was described in 1987 by Yoshihisa Kuwana et al.[7] at Fujita Health University and Kyowa Hakko Kogyo, Co. Ltd. in Japan, and independently in 1989 by Gideon Gross and Zelig Eshhar[8][9] at the Weizmann Institute in Israel.[10] Originally termed "T-bodies", these early approaches combined an antibody's ability to specifically bind to diverse targets with the constant domains of the TCR-α or TCR-β proteins.[11]

In 1991, chimeric receptors containing the intracellular signaling domain of CD3ζ were shown to activate T cell signaling by Arthur Weiss at the University of California, San Francisco.[12] This work prompted CD3ζ intracellular domains to be added to chimeric receptors with antibody-like extracellular domains, commonly single-chain fraction variable (scFv) domains, as well as proteins such as CD4, subsequently termed first generation CARs.[13][14]

A first generation CAR containing a CD4 extracellular domain and a CD3ζ intracellular domain was used in the first clinical trial of chimeric antigen receptor T cells by the biotechnology company Cell Genesys in the mid 1990s, allowing adoptively transferred T cells to target HIV infected cells, although it failed to show any clinical improvement.[13] Similar early clinical trials of CAR T cells in solid tumors in the 1990s using first generation CARs targeting a solid tumor antigens such as MUC1 did not show long-term persistence of the transferred T cells or result in significant remissions.[15]

In the early 2000s, co-stimulatory domains such as CD28 or 4-1BB were added to first generation CAR's CD3ζ intracellular domain. Termed second generation CARs, these constructs showed greater persistence and improved tumor clearance in pre-clinical models.[16] Clinical trials in the early 2010s using second generation CARs targeting CD19, a protein expressed by normal B cells as well as B-cell leukemias and lymphomas, by investigators at the NCI, University of Pennsylvania, and Memorial Sloan Kettering Cancer Center demonstrated the clinical efficacy of CAR T cell therapies and resulted in complete remissions in many heavily pre-treated patients.[15] These trials ultimately led in the US to the FDA's first two approvals of CAR T cells in 2017, those for tisagenlecleucel (Kymriah), marketed by Novartis originally for B-cell precursor acute lymphoblastic leukemia (B-ALL), and axicabtagene ciloleucel (Yescarta), marketed by Kite Pharma originally for diffuse large B-cell lymphoma (DLBCL).[15] There are now six FDA-approved CAR T therapies.[17]

Production edit

 
Depiction of adoptive cell transfer therapy with CAR-engineered T cells

The first step in the production of CAR T-cells is the isolation of T cells from human blood. CAR T-cells may be manufactured either from the patient's own blood, known as an autologous treatment, or from the blood of a healthy donor, known as an allogeneic treatment. The manufacturing process is the same in both cases; only the choice of initial blood donor is different.[citation needed]

First, leukocytes are isolated using a blood cell separator in a process known as leukocyte apheresis. Peripheral blood mononuclear cells (PBMCs) are then separated and collected.[18][19] The products of leukocyte apheresis are then transferred to a cell-processing center. In the cell processing center, specific T cells are stimulated so that they will actively proliferate and expand to large numbers. To drive their expansion, T cells are typically treated with the cytokine interleukin 2 (IL-2) and anti-CD3 antibodies.[20] Anti-CD3/CD28 antibodies are also used in some protocols.[19]

The expanded T cells are purified and then transduced with a gene encoding the engineered CAR via a retroviral vector, typically either an integrating gammaretrovirus (RV) or a lentiviral (LV) vector.[19] These vectors are very safe in modern times due to a partial deletion of the U3 region.[21] The new gene editing tool CRISPR/Cas9 has recently been used instead of retroviral vectors to integrate the CAR gene into specific sites in the genome.[22]

The patient undergoes lymphodepletion chemotherapy prior to the introduction of the engineered CAR T-cells.[4] The depletion of the number of circulating leukocytes in the patient upregulates the number of cytokines that are produced and reduces competition for resources, which helps to promote the expansion of the engineered CAR T-cells.[23]

Clinical applications edit

As of March 2019, there were around 364 ongoing clinical trials happening globally involving CAR T cells.[24] The majority of those trials target blood cancers: CAR T therapies account for more than half of all trials for hematological malignancies.[24] CD19 continues to be the most popular antigen target,[25] followed by BCMA (commonly expressed in multiple myeloma).[24][26] In 2016, studies began to explore the viability of other antigens, such as CD20.[27] Trials for solid tumors are less dominated by CAR T, with about half of cell therapy-based trials involving other platforms such as NK cells.[24]

Cancer edit

T cells are genetically engineered to express chimeric antigen receptors specifically directed toward antigens on a patient's tumor cells, then infused into the patient where they attack and kill the cancer cells.[28] Adoptive transfer of T cells expressing CARs is a promising anti-cancer therapeutic, because CAR-modified T cells can be engineered to target potentially any tumor associated antigen.[29][30]

Early CAR T cell research has focused on blood cancers. The first approved treatments use CARs that target the antigen CD19, present in B-cell-derived cancers such as acute lymphoblastic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL).[31][32] There are also efforts underway to engineer CARs targeting many other blood cancer antigens, including CD30 in refractory Hodgkin's lymphoma; CD33, CD123, and FLT3 in acute myeloid leukemia (AML); and BCMA in multiple myeloma.[33]

Solid tumors have presented a more difficult target.[34] Identification of good antigens has been challenging: such antigens must be highly expressed on the majority of cancer cells, but largely absent on normal tissues.[35][36][37][30] CAR T cells are also not trafficked efficiently into the center of solid tumor masses, and the hostile tumor microenvironment suppresses T cell activity.[33]

Autoimmune disease edit

While most CAR T cell studies focus on creating a CAR T cell that can eradicate a certain cell population (for instance, CAR T cells that target lymphoma cells), there are other potential uses for this technology. T cells can also mediate tolerance to antigens.[38] A regulatory T cell outfitted with a CAR could have the potential to confer tolerance to a specific antigen, something that could be utilized in organ transplantation or rheumatologic diseases like lupus.[39][40]

Approved therapies edit

CAR T Cell Therapies with Regulatory Approval
CAR T cell (Brand name) Company Approval Agency: Date Target Antigen recognition domain Intracellular signaling domain Indication (Targeted disease / Line of Therapy) Agency Product Number, Drug Label
tisagenlecleucel

(Kymriah)

Novartis FDA: 08/30/2017 [41]

EMA: 08/22/2018 [42]

MHLW: 05/15/2019 [43]

CD19 scFV 41BB - CD3ζ B-cell precursor ALL (Third Line)[41][42][43]

Diffuse large B-cell lymphoma (Third Line)[44] [42][43]

Follicular Lymphoma (Third Line)[45] [46]

FDA:125646, Label

EMA:004090, Label

axicabtagene ciloleucel

(Yescarta)

Kite Pharma / Gilead FDA: 10/18/2017 [47]

EMA: 08/27/2018 [48]

NMPA: 06/23/2021 [49]

MHLW: 12/22/2022 [50]

CD19 scFV CD28 - CD3ζ Diffuse large B-cell lymphoma (Second Line)[51] [52] [49][50]

Follicular lymphoma (Third Line) [53] [54] [49][50]

Primary mediastinal large B-cell lymphoma (Third Line) [48][49][50]

FDA:125643, Label

EMA:004480, Label

brexucabtagene autoleucel

(Tecartus)

Kite Pharma / Gilead FDA: 07/24/2020 [55]

EMA: 12/14/2020 [56]

CD19 scFV CD28 - CD3ζ Mantle cell lymphoma (Third Line) [57][56]

B-cell precursor ALL (Third Line)[57] [56]

FDA:125703, Label

EMA:005102, Label

lisocabtagene maraleucel

(Breyanzi)

Juno Therapeutics / BMS FDA: 02/05/2021[58]

EMA: 04/04/2022 [59]

MHLW: 12/20/2022 [60]

CD19 scFV 41BB - CD3ζ Diffuse large B-cell lymphoma (Second Line)[61] [59][60] FDA: 25714, Label

EMA:004731, Label

idecabtagene vicleucel

(Abecma)

Bluebird Bio / BMS FDA: 03/26/2021 [62]

EMA: 08/18/2021 [63]

BCMA scFV 41BB - CD3ζ Multiple myeloma (Fourth Line),[63] (Fifth Line)[62] FDA:125736, Label

EMA:004662, Label

ciltacabtagene autoleucel

(Carvykti)

Janssen / J&J FDA: 02/28/2022 [64]

EMA: 05/25/2022 [65]

BCMA VHH 41BB - CD3ζ Multiple myeloma (Fourth Line),[65] (Fifth Line)[64] FDA:125746, Label

EMA:005095, Label

Safety edit

There are serious side effects that result from CAR T-cells being introduced into the body, including cytokine release syndrome and neurological toxicity.[4] Because it is a relatively new treatment, there are few data about the long-term effects of CAR T-cell therapy. There are still concerns about long-term patient survival, as well as pregnancy complications in female patients treated with CAR T-cells.[66] Anaphylaxis may be a side effect, as the CAR is made with a foreign monoclonal antibody, and as a result provokes an immune response.[citation needed]

On-target/off-tumor recognition occurs when the CAR T-cell recognizes the correct antigen, but the antigen is expressed on healthy, non-pathogenic tissue. This results in the CAR T-cells attacking non-tumor tissue, such as healthy B cells that express CD19 causing B-cell aplasia. The severity of this adverse effect can vary but the combination of prior immunosuppression, lymphodepleting chemotherapy and on-target effects causing hypogammaglobulinaemia and prolonged cytopenias places patients at increased risk of serious infections.[20][67]

There is also the unlikely possibility that the engineered CAR T-cells will themselves become transformed into cancerous cells through insertional mutagenesis, due to the viral vector inserting the CAR gene into a tumor suppressor or oncogene in the host T cell's genome. Some retroviral (RV) vectors carry a lower risk than lentiviral (LV) vectors. However, both have the potential to be oncogenic. Genomic sequencing analysis of CAR insertion sites in T cells has been established for better understanding of CAR T-cell function and persistence in vivo.[35]

Cytokine release syndrome edit

The most common issue after treatment with CAR T-cells is cytokine release syndrome (CRS), a condition in which the immune system is activated and releases an increased number of inflammatory cytokines. The clinical manifestation of this syndrome resembles sepsis with high fever, fatigue, myalgia, nausea, capillary leakages, tachycardia and other cardiac dysfunction, liver failure, and kidney impairment.[68] CRS occurs in almost all patients treated with CAR T-cell therapy; in fact, the presence of CRS is a diagnostic marker that indicates the CAR T-cells are working as intended to kill the cancer cells.[66] The severity of CRS does not correlate with an increased response to the treatment, but rather higher disease burden.[66] Severe cytokine release syndrome can be managed with immunosuppressants such as corticosteroids, and with tocilizumab, an anti-IL-6 monoclonal antibody.[69] Early intervention using tocilizumab was shown to reduce the frequency of severe CRS in multiple studies[70][71] without affecting the therapeutic effect of the treatment.

Immune effector cell-associated neurotoxicity edit

Neurological toxicity is also often associated with CAR T-cell treatment.[72] The underlying mechanism is poorly understood, and may or may not be related to CRS. Clinical manifestations include delirium, the partial loss of the ability to speak coherently while still having the ability to interpret language (expressive aphasia), lowered alertness (obtundation), and seizures.[66] During some clinical trials, deaths caused by neurotoxicity have occurred. The main cause of death from neurotoxicity is cerebral edema. In a study carried out by Juno Therapeutics, Inc., five patients enrolled in the trial died as a result of cerebral edema. Two of the patients were treated with cyclophosphamide alone and the remaining three were treated with a combination of cyclophosphamide and fludarabine.[73] In another clinical trial sponsored by the Fred Hutchinson Cancer Research Center, there was one reported case of irreversible and fatal neurological toxicity 122 days after the administration of CAR T-cells.[74]

Hypokinetic movement disorder (parkinsonism, or movement and neurocognitive treatment emergent adverse events) has been observed with BCMA-chimeric antigen receptor (CAR) T-cell treatment for multiple myeloma.[75]

Chimeric antigen receptor structure edit

Chimeric antigen receptors combine many facets of normal T cell activation into a single protein. They link an extracellular antigen recognition domain to an intracellular signalling domain, which activates the T cell when an antigen is bound. CARs are composed of four regions: an antigen recognition domain, an extracellular hinge region, a transmembrane domain, and an intracellular T cell signaling domain.[76][77]

 
Different components of a chimeric antigen receptor

Antigen recognition domain edit

The antigen recognition domain is exposed to the outside of the cell, in the ectodomain portion of the receptor. It interacts with potential target molecules and is responsible for targeting the CAR T cell to any cell expressing a matching molecule.[citation needed]

The antigen recognition domain is typically derived from the variable regions of a monoclonal antibody linked together as a single-chain variable fragment (scFv).[77] An scFv is a chimeric protein made up of the light (VL) and heavy (VH) chains of immunoglobins, connected with a short linker peptide.[78] These VL and VH regions are selected in advance for their binding ability to the target antigen (such as CD19). The linker between the two chains consists of hydrophilic residues with stretches of glycine and serine in it for flexibility as well as stretches of glutamate and lysine for added solubility.[79] Single domain antibodies (e.g. VH, VHH, VNAR) have been engineered and developed as antigen recognition domains in the CAR format due to their high transduction efficiency in T cells.[80][35][81][82][83]

In addition to antibody fragments, non‐antibody‐based approaches have also been used to direct CAR specificity, usually taking advantage of ligand/receptor pairs that normally bind to each other.[76] Cytokines, innate immune receptors, TNF receptors, growth factors, and structural proteins have all been successfully used as CAR antigen recognition domains.[76]

Hinge region edit

The hinge, also called a spacer, is a small structural domain that sits between the antigen recognition region and the cell's outer membrane. An ideal hinge enhances the flexibility of the scFv receptor head, reducing the spatial constraints between the CAR and its target antigen. This promotes antigen binding and synapse formation between the CAR T cells and target cells.[84] Hinge sequences are often based on membrane-proximal regions from other immune molecules including IgG, CD8, and CD28.[76][85][81][82]

Transmembrane domain edit

The transmembrane domain is a structural component, consisting of a hydrophobic alpha helix that spans the cell membrane. It anchors the CAR to the plasma membrane, bridging the extracellular hinge and antigen recognition domains with the intracellular signaling region.[76] This domain is essential for the stability of the receptor as a whole. Generally, the transmembrane domain from the most membrane-proximal component of the endodomain is used, but different transmembrane domains result in different receptor stability. The CD28 transmembrane domain is known to result in a highly expressed, stable receptor.[82]

Using the CD3-zeta transmembrane domain is not recommended, as it can result in incorporation of the artificial TCR into the native TCR.[86]

Intracellular T cell signaling domain edit

 
Depiction of first, second, and third generation chimeric antigen receptors with the scFv segments in green and the various intracellular TCR signaling components in red, blue and yellow[87]

The intracellular T cell signaling domain lies in the receptor's endodomain, inside the cell.[76] After an antigen is bound to the external antigen recognition domain, CAR receptors cluster together and transmit an activation signal. Then the internal cytoplasmic end of the receptor perpetuates signaling inside the T cell.[78]

Normal T cell activation relies on the phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) present in the cytoplasmic domain of CD3-zeta. To mimic this process, CD3-zeta's cytoplasmic domain is commonly used as the main CAR endodomain component. Other ITAM-containing domains have also been tried, but are not as effective.[77]

T cells also require co-stimulatory molecules in addition to CD3 signaling in order to persist after activation. For this reason, the endodomains of CAR receptors typically also include one or more chimeric domains from co-stimulatory proteins.[3] Signaling domains from a wide variety of co-stimulatory molecules have been successfully tested, including CD28, CD27, CD134 (OX40), and CD137 (4‐1BB).[76]

The intracellular signaling domain used defines the generation of a CAR T cell.[4] First generation CARs include only a CD3-zeta cytoplasmic domain.[4] Second generation CARs add a co‐stimulatory domain, like CD28 or 4‐1BB. The involvement of these intracellular signaling domains improve T cell proliferation, cytokine secretion, resistance to apoptosis, and in vivo persistence.[4] Third generation CARs combine multiple co-stimulatory domains, such as CD28-41BB or CD28-OX40, to augment T cell activity. Preclinical data show the third-generation CARs exhibit improved effector functions and better in vivo persistence as compared to second‐generation CARs.[4]

Research directions edit

Antigen recognition edit

Although the initial clinical remission rates after CAR T cell therapy in all patients are as high as 90%,[88] long-term survival rates are much lower. The cause is typically the emergence of leukemia cells that do not express CD19 and so evade recognition by the CD19–CAR T cells, a phenomenon known as antigen escape.[33] Preclinical studies developing CAR T cells with dual targeting of CD19 plus CD22 or CD19 plus CD20 have demonstrated promise, and trials studying bispecific targeting to circumvent CD19 down-regulation are ongoing.[33]

In 2018, a version of CAR was developed that is referred to as SUPRA CAR, or split, universal, and programmable.[89] Multiple mechanisms can be deployed to finely regulate the activity of SUPRA CAR, which limits overactivation. In contrast to the traditional CAR design, SUPRA CAR allows targeting of multiple antigens without further genetic modification of a person's immune cells.[90]

SMDCs (small molecule drug conjugates) platform in immuno-oncology is an experimental approach that makes possible the engineering of a single universal CAR T cell, which binds with extraordinarily high affinity to a benign molecule designated as fluorescein isothiocyanate (FITC). These cells are then used to treat various cancer types when co-administered with bispecific SMDC adaptor molecules. These unique bispecific adaptors are constructed with a FITC molecule and a tumor-homing molecule to precisely bridge the universal CAR T cell with the cancer cells, which causes localized T cell activation. Anti-tumor activity in mice is induced only when both the universal CAR T cells plus the correct antigen-specific adaptor molecules are present. Anti-tumor activity and toxicity can be controlled by adjusting the administered adaptor molecule dosing. Treatment of antigenically heterogeneous tumors can be achieved by administration of a mixture of the desired antigen-specific adaptors.[91][92]

CAR T function edit

Fourth generation CARs (also known as TRUCKs or armored CARs) further add factors that enhance T cell expansion, persistence, and anti‐tumoral activity. This can include cytokines, such is IL-2, IL-5, IL-12 and co‐stimulatory ligands.[93][94]

Control mechanisms edit

Adding a synthetic control mechanism to engineered T cells allows doctors to precisely control the persistence or activity of the T cells in the patient's body, with the goal of reducing toxic side effects.[95] The major control techniques trigger T cell death or limit T cell activation, and often regulate the T cells via a separate drug that can be introduced or withheld as needed.[citation needed]

Suicide genes: Genetically modified T cells are engineered to include one or more genes that can induce apoptosis when activated by an extracellular molecule. Herpes simplex virus thymidine kinase (HSV-TK) and inducible caspase 9 (iCasp9) are two types of suicide genes that have been integrated into CAR T cells.[95][96][97] In the iCasp9 system, the suicide gene complex has two elements: a mutated FK506-binding protein with high specificity to the small molecule rimiducid/AP1903, and a gene encoding a pro-domain-deleted human caspase 9. Dosing the patient with rimiducid activates the suicide system, leading to rapid apoptosis of the genetically modified T cells. Although both the HSV-TK and iCasp9 systems demonstrate a noticeable function as a safety switch in clinical trials, some defects limit their application. HSV-TK is virus-derived and may be immunogenic to humans.[95][98] It is also currently unclear whether the suicide gene strategies will act quickly enough in all situations to halt dangerous off-tumor cytotoxicity.[citation needed]

Dual-antigen receptor: CAR T cells are engineered to express two tumor-associated antigen receptors at the same time, reducing the likelihood that the T cells will attack non-tumor cells. Dual-antigen receptor CAR T cells have been reported to have less intense side effects.[99] An in vivo study in mice shows that dual-receptor CAR T cells effectively eradicated prostate cancer and achieved complete long-term survival.[100]

ON-switch and OFF-switch: In this system, CAR T cells can only function in the presence of both tumor antigen and a benign exogenous molecule. To achieve this, the CAR T cell's engineered chimeric antigen receptor is split into two separate proteins that must come together in order to function. The first receptor protein typically contains the extracellular antigen binding domain, while the second protein contains the downstream signaling elements and co-stimulatory molecules (such as CD3ζ and 4-1BB). In the presence of an exogenous molecule (such as a rapamycin analog), the binding and signaling proteins dimerize together, allowing the CAR T cells to attack the tumor.[101] Human EGFR truncated form (hEGFRt) has been used as an OFF-switch for CAR T cells using cetuximab.[35][37][81]

Bispecific molecules as switches: Bispecific molecules target both a tumor-associated antigen and the CD3 molecule on the surface of T cells. This ensures that the T cells cannot become activated unless they are in close physical proximity to a tumor cell.[102] The anti-CD20/CD3 bispecific molecule shows high specificity to both malignant B cells and cancer cells in mice.[103] FITC is another bifunctional molecule used in this strategy. FITC can redirect and regulate the activity of the FITC-specific CAR T cells toward tumor cells with folate receptors.[104]

Advances in CAR T cell manufacturing. edit

Due to the high costs of CAR T cell therapy,[105] a number of alternative efforts are being investigated to improve CAR T cell manufacturing and reduce costs. In vivo CAR T cell manufacturing strategies[106][107] are being tested. In addition, bioinstructive materials have been developed for CAR T cell generation.[108] Rapid CAR T cell generation is also possible through shortening or eliminating the activation and expansion steps.[109]

In situ modification edit

Another approach is to modify T cells and/or B cells still in the body using viral vectors.[110]

Economics edit

The cost of CAR T cell therapies has been criticized, with the initial costs of tisagenlecleucel (Kymriah) and axicabtagene ciloleucel (Yescarta) being $375,000 and $475,000 respectively.[105] The high cost of CAR T therapies is due to complex cellular manufacturing in specialized good manufacturing practice (GMP) facilities as well as the high level of hospital care necessary after CAR T cells are administered due to risks such as cytokine release syndrome.[105] In the United States, CAR T cell therapies are covered by Medicare and by many but not all private insurers.[111][112] Manufacturers of CAR T cells have developed alternative payment programs due to the high cost of CAR T therapy, such as by requiring payment only if the CAR T therapy induces a complete remission by a certain time point after treatment.[113]

Additionally, CAR T cell therapies are not available worldwide yet. CAR T cell therapies have been approved in China, Australia, Singapore, the United Kingdom, and some European countries.[114] In February 2022 Brazil approved tisagenlecleucel (Kymriah) treatment.[115]

See also edit

References edit

  1. ^ Fox M (July 12, 2017). "New Gene Therapy for Cancer Offers Hope to Those With No Options Left". NBC News.
  2. ^ Srivastava S, Riddell SR (August 2015). "Engineering CAR-T cells: Design concepts". Trends in Immunology. 36 (8): 494–502. doi:10.1016/j.it.2015.06.004. PMC 4746114. PMID 26169254.
  3. ^ a b Sadelain M, Brentjens R, Rivière I (April 2013). "The basic principles of chimeric antigen receptor design". Cancer Discovery. 3 (4): 388–398. doi:10.1158/2159-8290.CD-12-0548. PMC 3667586. PMID 23550147.
  4. ^ a b c d e f g Hartmann J, Schüßler-Lenz M, Bondanza A, Buchholz CJ (September 2017). "Clinical development of CAR T cells-challenges and opportunities in translating innovative treatment concepts". EMBO Molecular Medicine. 9 (9): 1183–1197. doi:10.15252/emmm.201607485. PMC 5582407. PMID 28765140.
  5. ^ Tang XJ, Sun XY, Huang KM, Zhang L, Yang ZS, Zou DD, et al. (December 2015). "Therapeutic potential of CAR-T cell-derived exosomes: a cell-free modality for targeted cancer therapy". Oncotarget. 6 (42): 44179–44190. doi:10.18632/oncotarget.6175. PMC 4792550. PMID 26496034.
  6. ^ Zhang H, Zhao P, Huang H (December 2020). "Engineering better chimeric antigen receptor T cells". Experimental Hematology & Oncology. 9 (1): 34. doi:10.1186/s40164-020-00190-2. PMC 7709221. PMID 33292660.
  7. ^ Kuwana Y, Asakura Y, Utsunomiya N, Nakanishi M, Arata Y, Itoh S, et al. (December 1987). "Expression of chimeric receptor composed of immunoglobulin-derived V regions and T-cell receptor-derived C regions". Biochemical and Biophysical Research Communications. 149 (3): 960–968. doi:10.1016/0006-291x(87)90502-x. PMID 3122749.
  8. ^ Gross G, Gorochov G, Waks T, Eshhar Z (February 1989). "Generation of effector T cells expressing chimeric T cell receptor with antibody type-specificity". Transplantation Proceedings. 21 (1 Pt 1): 127–130. PMID 2784887.
  9. ^ Rosenbaum L (October 2017). "Tragedy, Perseverance, and Chance - The Story of CAR-T Therapy". The New England Journal of Medicine. 377 (14): 1313–1315. doi:10.1056/NEJMp1711886. PMID 28902570. S2CID 205114161.
  10. ^ Gross G, Waks T, Eshhar Z (December 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 (24): 10024–10028. Bibcode:1989PNAS...8610024G. doi:10.1073/pnas.86.24.10024. JSTOR 34790. PMC 298636. PMID 2513569.
  11. ^ Eshhar Z, Bach N, Fitzer-Attas CJ, Gross G, Lustgarten J, Waks T, Schindler DG (1996). "The T-body approach: potential for cancer immunotherapy". Springer Seminars in Immunopathology. 18 (2): 199–209. doi:10.1007/BF00820666. PMID 8908700. S2CID 19872173.
  12. ^ Irving BA, Weiss A (March 1991). "The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor-associated signal transduction pathways". Cell. 64 (5): 891–901. doi:10.1016/0092-8674(91)90314-o. PMID 1705867. S2CID 23466990.
  13. ^ a b Hege KM, Roberts MR (December 1996). "T-cell gene therapy". Current Opinion in Biotechnology. 7 (6): 629–634. doi:10.1016/s0958-1669(96)80074-7. PMID 8939644.
  14. ^ June CH, Sadelain M (July 2018). "Chimeric Antigen Receptor Therapy". The New England Journal of Medicine. 379 (1): 64–73. doi:10.1056/NEJMra1706169. PMC 7433347. PMID 29972754.
  15. ^ a b c Braendstrup P, Levine BL, Ruella M (February 2020). "The long road to the first FDA-approved gene therapy: chimeric antigen receptor T cells targeting CD19". Cytotherapy. 22 (2): 57–69. doi:10.1016/j.jcyt.2019.12.004. PMC 7036015. PMID 32014447.
  16. ^ Sadelain M, Rivière I, Brentjens R (January 2003). "Targeting tumours with genetically enhanced T lymphocytes". Nature Reviews. Cancer. 3 (1): 35–45. doi:10.1038/nrc971. PMID 12509765. S2CID 33707802.
  17. ^ Center for Biologics Evaluation and Research (2022-03-01). "Approved Cellular and Gene Therapy Products". FDA.
  18. ^ Jin C, Yu D, Hillerdal V, Wallgren A, Karlsson-Parra A, Essand M (2014-03-05). "Allogeneic lymphocyte-licensed DCs expand T cells with improved antitumor activity and resistance to oxidative stress and immunosuppressive factors". Molecular Therapy: Methods & Clinical Development. 1: 14001. doi:10.1038/mtm.2014.1. PMC 4362340. PMID 26015949.
  19. ^ a b c Li, Nan; Ho, Mitchell (2022). "Development of Glypican-2 Targeting Single-Domain Antibody CAR T Cells for Neuroblastoma". Single-Domain Antibodies. Methods in Molecular Biology. Vol. 2446. pp. 451–468. doi:10.1007/978-1-0716-2075-5_23. ISBN 978-1-0716-2074-8. ISSN 1940-6029. PMID 35157288. S2CID 246813053.
  20. ^ a b Makita S, Yoshimura K, Tobinai K (June 2017). "Clinical development of anti-CD19 chimeric antigen receptor T-cell therapy for B-cell non-Hodgkin lymphoma". Cancer Science. 108 (6): 1109–1118. doi:10.1111/cas.13239. PMC 5480083. PMID 28301076.
  21. ^ Jin C, Fotaki G, Ramachandran M, Nilsson B, Essand M, Yu D (July 2016). "Safe engineering of CAR T cells for adoptive cell therapy of cancer using long-term episomal gene transfer". EMBO Molecular Medicine. 8 (7): 702–711. doi:10.15252/emmm.201505869. PMC 4931286. PMID 27189167.
  22. ^ Jensen TI, Axelgaard E, Bak RO (June 2019). "Therapeutic gene editing in haematological disorders with CRISPR/Cas9". British Journal of Haematology. 185 (5): 821–835. doi:10.1111/bjh.15851. PMID 30864164.
  23. ^ Muranski P, Boni A, Wrzesinski C, Citrin DE, Rosenberg SA, Childs R, Restifo NP (December 2006). "Increased intensity lymphodepletion and adoptive immunotherapy--how far can we go?". Nature Clinical Practice. Oncology. 3 (12): 668–681. doi:10.1038/ncponc0666. PMC 1773008. PMID 17139318.
  24. ^ a b c d Xin Yu J, Hubbard-Lucey VM, Tang J (October 2019). "The global pipeline of cell therapies for cancer". Nature Reviews. Drug Discovery. 18 (11): 821–822. doi:10.1038/d41573-019-00090-z. PMID 31673124. S2CID 190862546.
  25. ^ Brudno and Kochenderfer. Chimeric antigen receptor T cell therapies for lymphoma. Nature Reviews Clinical Oncology. 2018. 15: 31-46.
  26. ^ Mikkilineni and Kochenderfer. Chimeric antigen receptor T-cell therapies for multiple myeloma. Blood. 2017. 130: 2594-602
  27. ^ Almåsbak H, Aarvak T, Vemuri MC (2016). "CAR T Cell Therapy: A Game Changer in Cancer Treatment". Journal of Immunology Research. 2016: 5474602. doi:10.1155/2016/5474602. PMC 4889848. PMID 27298832.
  28. ^ Jacobson CA, Ritz J (November 2011). "Time to put the CAR-T before the horse". Blood. 118 (18): 4761–4762. doi:10.1182/blood-2011-09-376137. PMID 22053170.
  29. ^ Li, Nan; Spetz, Madeline R.; Li, Dan; Ho, Mitchell (July 2021). "Advances in immunotherapeutic targets for childhood cancers: A focus on glypican-2 and B7-H3". Pharmacology & Therapeutics. 223: 107892. doi:10.1016/j.pharmthera.2021.107892. ISSN 1879-016X. PMC 8202769. PMID 33992682.
  30. ^ a b Li, Dan; Lin, Shaoli; Hong, Jessica; Ho, Mitchell (2022). "Immunotherapy for hepatobiliary cancers: Emerging targets and translational advances". Advances in Cancer Research. 156: 415–449. doi:10.1016/bs.acr.2022.01.013. ISBN 9780323983921. ISSN 2162-5557. PMID 35961708. S2CID 246978004.
  31. ^ Kochenderfer JN, Wilson WH, Janik JE, Dudley ME, Stetler-Stevenson M, Feldman SA, et al. (November 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 (20): 4099–4102. doi:10.1182/blood-2010-04-281931. PMC 2993617. PMID 20668228.
  32. ^ Kochenderfer JN, Dudley ME, Kassim SH, Somerville RP, Carpenter RO, Stetler-Stevenson M, et al. (February 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 (6): 540–549. doi:10.1200/JCO.2014.56.2025. PMC 4322257. PMID 25154820.
  33. ^ a b c d Schultz L, Mackall C (February 2019). "Driving CAR T cell translation forward". Science Translational Medicine. 11 (481): eaaw2127. doi:10.1126/scitranslmed.aaw2127. PMID 30814337.
  34. ^ Lim WA, June CH (February 2017). "The Principles of Engineering Immune Cells to Treat Cancer". Cell. 168 (4): 724–740. doi:10.1016/j.cell.2017.01.016. PMC 5553442. PMID 28187291.
  35. ^ a b c d Li D, Li N, Zhang YF, Fu H, Feng M, Schneider D, et al. (June 2020). "Persistent Polyfunctional Chimeric Antigen Receptor T Cells That Target Glypican 3 Eliminate Orthotopic Hepatocellular Carcinomas in Mice". Gastroenterology. 158 (8): 2250–2265.e20. doi:10.1053/j.gastro.2020.02.011. PMC 7282931. PMID 32060001.
  36. ^ Li, Nan; Fu, Haiying; Hewitt, Stephen M.; Dimitrov, Dimiter S.; Ho, Mitchell (2017-08-08). "Therapeutically targeting glypican-2 via single-domain antibody-based chimeric antigen receptors and immunotoxins in neuroblastoma". Proceedings of the National Academy of Sciences of the United States of America. 114 (32): E6623–E6631. Bibcode:2017PNAS..114E6623L. doi:10.1073/pnas.1706055114. ISSN 1091-6490. PMC 5559039. PMID 28739923.
  37. ^ a b Li, Nan; Torres, Madeline B.; Spetz, Madeline R.; Wang, Ruixue; Peng, Luyi; Tian, Meijie; Dower, Christopher M.; Nguyen, Rosa; Sun, Ming; Tai, Chin-Hsien; de Val, Natalia; Cachau, Raul; Wu, Xiaolin; Hewitt, Stephen M.; Kaplan, Rosandra N. (2021-06-15). "CAR T cells targeting tumor-associated exons of glypican 2 regress neuroblastoma in mice". Cell Reports. Medicine. 2 (6): 100297. doi:10.1016/j.xcrm.2021.100297. ISSN 2666-3791. PMC 8233664. PMID 34195677.
  38. ^ Sakaguchi S, Yamaguchi T, Nomura T, Ono M (May 2008). "Regulatory T cells and immune tolerance". Cell. 133 (5): 775–787. doi:10.1016/j.cell.2008.05.009. PMID 18510923. S2CID 2315895.
  39. ^ Zhang Q, Lu W, Liang CL, Chen Y, Liu H, Qiu F, Dai Z (2018). "Chimeric Antigen Receptor (CAR) Treg: A Promising Approach to Inducing Immunological Tolerance". Frontiers in Immunology. 9: 2359. doi:10.3389/fimmu.2018.02359. PMC 6194362. PMID 30369931.
  40. ^ Mougiakakos D, Krönke G, Völkl S, Kretschmann S, Aigner M, Kharboutli S, et al. (August 2021). "CD19-Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus". The New England Journal of Medicine. 385 (6): 567–569. doi:10.1056/NEJMc2107725. PMID 34347960. S2CID 236927691.
  41. ^ a b "Novartis receives first ever FDA approval for a CAR-T cell therapy, Kymriah(TM) (CTL019), for children and young adults with B-cell ALL that is refractory or has relapsed at least twice". www.novartis.com. 2017-08-30.
  42. ^ a b c "Novartis receives European Commission approval of its CAR-T cell therapy, Kymriah® (tisagenlecleucel)". www.novartis.com. Retrieved 2023-11-18.
  43. ^ a b c "Novartis gets approval to sell Kymriah in Japan for $306,000". www.reuters.com. 2019-05-15.
  44. ^ "KYMRIAH (tisagenlecleucel)". US Food and Drug Administration. 2019-04-05.
  45. ^ "FDA approves Novartis Kymriah® CAR-T cell therapy for adult patients with relapsed or refractory follicular lymphoma". Novartis. Retrieved 2022-06-05.
  46. ^ "Novartis Kymriah® receives EC approval as first CAR-T cell therapy for adults with relapsed or refractory follicular lymphoma". www.novartis.com. Retrieved 2023-11-18.
  47. ^ Center for Biologics Evaluation and Research (2017-10-18). "FDA approves CAR-T cell therapy to treat adults with certain types of large B-cell lymphoma". FDA.
  48. ^ a b "Yescarta® (Axicabtagene Ciloleucel) Receives European Marketing Authorization for the Treatment of Relapsed or Refractory DLBCL and PMBCL, After Two or More Lines of Systemic Therapy". www.gilead.com. Retrieved 2023-11-18.
  49. ^ a b c d "Kite Joint Venture - Fosun Kite - Gains the First CAR T-cell Therapy Approval in China". www.gilead.com. Retrieved 2023-11-13.
  50. ^ a b c d "Yescarta® Now Approved in Japan for Initial Treatment of Relapsed/Refractory Large B-Cell Lymphoma". www.gilead.com. 2022-12-22.
  51. ^ Center for Biologics Evaluation and Research (2020-05-28). "YESCARTA (axicabtagene ciloleucel)". FDA.
  52. ^ "Kite's CAR T-cell Therapy Yescarta® First in Europe to Receive Positive CHMP Opinion for Use in Second-line Diffuse Large B-cell Lymphoma and High-grade B-cell Lymphoma". www.gilead.com. Retrieved 2023-11-18.
  53. ^ "U.S. FDA Approves Yescarta® for Relapsed or Refractory Follicular Lymphoma After Two or More Lines of Systemic Therapy". www.gilead.com. Retrieved 2021-03-15.
  54. ^ "Kite's CAR T-cell Therapy Yescarta® Granted European Marketing Authorization for the Treatment of Relapsed or Refractory Follicular Lymphoma". www.gilead.com. Retrieved 2023-11-18.
  55. ^ Center for Biologics Evaluation and Research (2021-03-04). "TECARTUS (brexucabtagene autoleucel)". FDA.
  56. ^ a b c European Medicines Agency (2023-01-30). "TECARTUS (brexucabtagene autoleucel)". EMA.
  57. ^ a b Center for Biologics Evaluation and Research (2021-03-04). "TECARTUS (brexucabtagene autoleucel)". FDA.
  58. ^ "U.S. Food and Drug Administration Approves Bristol Myers Squibb's Breyanzi (lisocabtagene maraleucel), a New CAR T Cell Therapy for Adults with Relapsed or Refractory Large B-cell Lymphoma". news.bms.com. Retrieved 2023-11-21.
  59. ^ a b European Medicines Agency (2023-11-10). "Breyanzi". EMA.
  60. ^ a b "CAR T Cell Therapy Breyanzi® Approved as Relapsed or Refractory Large B-cell Lymphoma Second-Line Therapy in Japan". new.bms.com.
  61. ^ Center for Biologics Evaluation and Research (2022-06-24). "FDA D.I.S.C.O. Burst Edition: FDA approval of Breyanzi (lisocabtagene maraleucel) for second-line treatment of large B-cell lymphoma". FDA.
  62. ^ a b Center for Biologics Evaluation and Research (2021-03-27). "ABECMA (idecabtagene vicleucel)". FDA.
  63. ^ a b European Medicines Agency (2023-07-27). "Abecma". EMA.
  64. ^ a b Center for Biologics Evaluation and Research (2022-03-21). "CARVYKTI". FDA.
  65. ^ a b European Medicines Agency (2023-07-27). "Carvykti". EMA.
  66. ^ a b c d Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ (2016). "Toxicity and management in CAR T-cell therapy". Molecular Therapy: Oncolytics. 3: 16011. doi:10.1038/mto.2016.11. PMC 5008265. PMID 27626062.
  67. ^ Bupha-Intr O, Haeusler G, Chee L, Thursky K, Slavin M, Teh B (June 2021). "CAR-T cell therapy and infection: a review". Expert Review of Anti-Infective Therapy. 19 (6): 749–758. doi:10.1080/14787210.2021.1855143. PMID 33249873. S2CID 227235627.
  68. ^ Breslin S (February 2007). "Cytokine-release syndrome: overview and nursing implications". Clinical Journal of Oncology Nursing. 11 (1 Suppl): 37–42. doi:10.1188/07.CJON.S1.37-42. PMID 17471824. S2CID 35773028.
  69. ^ Lee DW, Gardner R, Porter DL, Louis CU, Ahmed N, Jensen M, et al. (July 2014). "Current concepts in the diagnosis and management of cytokine release syndrome". Blood. 124 (2): 188–195. doi:10.1182/blood-2014-05-552729. PMC 4093680. PMID 24876563.
  70. ^ Berg P, Schönefeld S, Ruppert-Seipp G, Funk MB (2022-11-29). "Regulatory Measures to Improve the Safety of CAR-T-Cell Treatment". Transfusion Medicine and Hemotherapy. 50 (3): 218–225. doi:10.1159/000526786. ISSN 1660-3796. PMC 10331154. PMID 37435000.
  71. ^ Gardner RA, Ceppi F, Rivers J, Annesley C, Summers C, Taraseviciute A, et al. (December 2019). "Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without attenuation of antileukemic efficacy". Blood. 134 (24): 2149–2158. doi:10.1182/blood.2019001463. PMC 6908832. PMID 31697826.
  72. ^ Brudno JN, Kochenderfer JN (June 2016). "Toxicities of chimeric antigen receptor T cells: recognition and management". Blood. 127 (26): 3321–3330. doi:10.1182/blood-2016-04-703751. PMC 4929924. PMID 27207799.
  73. ^ "Study Evaluating the Efficacy and Safety of JCAR015 in Adult B-cell Acute Lymphoblastic Leukemia (B-ALL)". ClinicalTrials.gov. Retrieved 2018-02-21.
  74. ^ Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, et al. (June 2016). "CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients". The Journal of Clinical Investigation. 126 (6): 2123–2138. doi:10.1172/JCI85309. PMC 4887159. PMID 27111235.
  75. ^ Gust, Juliane (October 5, 2023). "BCMA-CAR T-cell treatment–associated parkinsonism". ashpublications.org. doi:10.1182/blood.2023021860. Retrieved April 4, 2024.
  76. ^ a b c d e f g Chandran SS, Klebanoff CA (July 2019). "T cell receptor-based cancer immunotherapy: Emerging efficacy and pathways of resistance". Immunological Reviews. 290 (1): 127–147. doi:10.1111/imr.12772. PMC 7027847. PMID 31355495.   Material was copied from this source, which is available under a Creative Commons Attribution 4.0 International License.
  77. ^ a b c Dotti G, Gottschalk S, Savoldo B, Brenner MK (January 2014). "Design and development of therapies using chimeric antigen receptor-expressing T cells". Immunological Reviews. 257 (1): 107–126. doi:10.1111/imr.12131. PMC 3874724. PMID 24329793.
  78. ^ a b Zhang C, Liu J, Zhong JF, Zhang X (2017-06-24). "Engineering CAR-T cells". Biomarker Research. 5: 22. doi:10.1186/s40364-017-0102-y. PMC 5482931. PMID 28652918.
  79. ^ Baldo BA (May 2015). "Chimeric fusion proteins used for therapy: indications, mechanisms, and safety". Drug Safety. 38 (5): 455–479. doi:10.1007/s40264-015-0285-9. PMID 25832756. S2CID 23852865.
  80. ^ Li N, Fu H, Hewitt SM, Dimitrov DS, Ho M (August 2017). "Therapeutically targeting glypican-2 via single-domain antibody-based chimeric antigen receptors and immunotoxins in neuroblastoma". Proceedings of the National Academy of Sciences of the United States of America. 114 (32): E6623–E6631. Bibcode:2017PNAS..114E6623L. doi:10.1073/pnas.1706055114. PMC 5559039. PMID 28739923.
  81. ^ a b c Li, Nan; Quan, Alex; Li, Dan; Pan, Jiajia; Ren, Hua; Hoeltzel, Gerard; de Val, Natalia; Ashworth, Dana; Ni, Weiming; Zhou, Jing; Mackay, Sean; Hewitt, Stephen M.; Cachau, Raul; Ho, Mitchell (2023-04-08). "The IgG4 hinge with CD28 transmembrane domain improves VHH-based CAR T cells targeting a membrane-distal epitope of GPC1 in pancreatic cancer". Nature Communications. 14 (1): 1986. Bibcode:2023NatCo..14.1986L. doi:10.1038/s41467-023-37616-4. ISSN 2041-1723. PMC 10082787. PMID 37031249.
  82. ^ a b c Kolluri, Aarti; Li, Dan; Li, Nan; Duan, Zhijian; Roberts, Lewis R.; Ho, Mitchell (2023-02-01). "Human VH-based chimeric antigen receptor T cells targeting glypican 3 eliminate tumors in preclinical models of HCC". Hepatology Communications. 7 (2): e0022. doi:10.1097/HC9.0000000000000022. ISSN 2471-254X. PMC 9851680. PMID 36691969.
  83. ^ Li, Dan; English, Hejiao; Hong, Jessica; Liang, Tianyuzhou; Merlino, Glenn; Day, Chi-Ping; Ho, Mitchell (2022-03-17). "A novel PD-L1-targeted shark VNAR single-domain-based CAR-T cell strategy for treating breast cancer and liver cancer". Molecular Therapy: Oncolytics. 24: 849–863. doi:10.1016/j.omto.2022.02.015. ISSN 2372-7705. PMC 8917269. PMID 35317524.
  84. ^ Hudecek M, Sommermeyer D, Kosasih PL, Silva-Benedict A, Liu L, Rader C, et al. (February 2015). "The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity". Cancer Immunology Research. 3 (2): 125–135. doi:10.1158/2326-6066.CIR-14-0127. PMC 4692801. PMID 25212991.
  85. ^ Qin L, Lai Y, Zhao R, Wei X, Weng J, Lai P, et al. (March 2017). "Incorporation of a hinge domain improves the expansion of chimeric antigen receptor T cells". Journal of Hematology & Oncology. 10 (1): 68. doi:10.1186/s13045-017-0437-8. PMC 5347831. PMID 28288656.
  86. ^ Bridgeman JS, Hawkins RE, Bagley S, Blaylock M, Holland M, Gilham DE (June 2010). "The optimal antigen response of chimeric antigen receptors harboring the CD3zeta transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR/CD3 complex". Journal of Immunology. 184 (12): 6938–6949. doi:10.4049/jimmunol.0901766. PMID 20483753.
  87. ^ Casucci M, Bondanza A (2011). "Suicide gene therapy to increase the safety of chimeric antigen receptor-redirected T lymphocytes". Journal of Cancer. 2: 378–382. doi:10.7150/jca.2.378. PMC 3133962. PMID 21750689.
  88. ^ "A Cure for Cancer? How CAR-T Therapy is Revolutionizing Oncology" (Press release). labiotech. March 8, 2018. Retrieved April 19, 2018.
  89. ^ Choe JH, Williams JZ, Lim WA (2020). "Engineering T Cells to Treat Cancer: The Convergence of Immuno-Oncology and Synthetic Biology". Annual Review of Cancer Biology. 4: 121–139. doi:10.1146/annurev-cancerbio-030419-033657.
  90. ^ Cho JH, Collins JJ, Wong WW (May 2018). "Universal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell Responses". Cell. 173 (6): 1426–1438.e11. doi:10.1016/j.cell.2018.03.038. PMC 5984158. PMID 29706540.
  91. ^ SMDC technology. 2016-03-27 at the Wayback Machine ENDOCYTE
  92. ^ (Press release). Endocyte. April 19, 2016. Archived from the original on July 30, 2017. Retrieved December 20, 2017.
  93. ^ Kueberuwa G, Kalaitsidou M, Cheadle E, Hawkins RE, Gilham DE (March 2018). "CD19 CAR T Cells Expressing IL-12 Eradicate Lymphoma in Fully Lymphoreplete Mice through Induction of Host Immunity". Molecular Therapy: Oncolytics. 8: 41–51. doi:10.1016/j.omto.2017.12.003. PMC 5772011. PMID 29367945.
  94. ^ Chmielewski M, Abken H (2015). "TRUCKs: the fourth generation of CARs". Expert Opinion on Biological Therapy. 15 (8): 1145–1154. doi:10.1517/14712598.2015.1046430. PMID 25985798. S2CID 42535203.
  95. ^ a b c Zhang E, Xu H (January 2017). "A new insight in chimeric antigen receptor-engineered T cells for cancer immunotherapy". Journal of Hematology & Oncology. 10 (1): 1. doi:10.1186/s13045-016-0379-6. PMC 5210295. PMID 28049484.
  96. ^ Bonini C, Ferrari G, Verzeletti S, Servida P, Zappone E, Ruggieri L, et al. (June 1997). "HSV-TK gene transfer into donor lymphocytes for control of allogeneic graft-versus-leukemia". Science. 276 (5319): 1719–1724. doi:10.1126/science.276.5319.1719. PMID 9180086.
  97. ^ Quintarelli C, Vera JF, Savoldo B, Giordano Attianese GM, Pule M, Foster AE, et al. (October 2007). "Co-expression of cytokine and suicide genes to enhance the activity and safety of tumor-specific cytotoxic T lymphocytes". Blood. 110 (8): 2793–2802. doi:10.1182/blood-2007-02-072843. PMC 2018664. PMID 17638856.
  98. ^ Riddell SR, Elliott M, Lewinsohn DA, Gilbert MJ, Wilson L, Manley SA, et al. (February 1996). "T-cell mediated rejection of gene-modified HIV-specific cytotoxic T lymphocytes in HIV-infected patients". Nature Medicine. 2 (2): 216–223. doi:10.1038/nm0296-216. PMID 8574968. S2CID 35503876.
  99. ^ Maher J, Brentjens RJ, Gunset G, Rivière I, Sadelain M (January 2002). "Human T-lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta /CD28 receptor". Nature Biotechnology. 20 (1): 70–75. doi:10.1038/nbt0102-70. PMID 11753365. S2CID 20302096.
  100. ^ Wilkie S, van Schalkwyk MC, Hobbs S, Davies DM, van der Stegen SJ, Pereira AC, et al. (October 2012). "Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling". Journal of Clinical Immunology. 32 (5): 1059–1070. doi:10.1007/s10875-012-9689-9. PMID 22526592. S2CID 17660404.
  101. ^ Wu CY, Roybal KT, Puchner EM, Onuffer J, Lim WA (October 2015). "Remote control of therapeutic T cells through a small molecule-gated chimeric receptor". Science. 350 (6258): aab4077. Bibcode:2015Sci...350.4077W. doi:10.1126/science.aab4077. PMC 4721629. PMID 26405231.
  102. ^ Frankel SR, Baeuerle PA (June 2013). "Targeting T cells to tumor cells using bispecific antibodies". Current Opinion in Chemical Biology. 17 (3): 385–392. doi:10.1016/j.cbpa.2013.03.029. PMID 23623807.
  103. ^ Sun LL, Ellerman D, Mathieu M, Hristopoulos M, Chen X, Li Y, et al. (May 2015). "Anti-CD20/CD3 T cell-dependent bispecific antibody for the treatment of B cell malignancies". Science Translational Medicine. 7 (287): 287ra70. doi:10.1126/scitranslmed.aaa4802. PMID 25972002. S2CID 24939667.
  104. ^ Kim CH, Axup JY, Lawson BR, Yun H, Tardif V, Choi SH, et al. (October 2013). "Bispecific small molecule-antibody conjugate targeting prostate cancer". Proceedings of the National Academy of Sciences of the United States of America. 110 (44): 17796–17801. Bibcode:2013PNAS..11017796K. doi:10.1073/pnas.1316026110. PMC 3816437. PMID 24127589.
  105. ^ a b c Lyman GH, Nguyen A, Snyder S, Gitlin M, Chung KC (April 2020). "Economic Evaluation of Chimeric Antigen Receptor T-Cell Therapy by Site of Care Among Patients With Relapsed or Refractory Large B-Cell Lymphoma". JAMA Network Open. 3 (4): e202072. doi:10.1001/jamanetworkopen.2020.2072. PMC 7136832. PMID 32250433.
  106. ^ Smith, Tyrel T.; Stephan, Sirkka B.; Moffett, Howell F.; McKnight, Laura E.; Ji, Weihang; Reiman, Diana; Bonagofski, Emmy; Wohlfahrt, Martin E.; Pillai, Smitha P. S.; Stephan, Matthias T. (2017-04-17). "In situ programming of leukaemia-specific T cells using synthetic DNA nanocarriers". Nature Nanotechnology. 12 (8): 813–820. doi:10.1038/nnano.2017.57. ISSN 1748-3387. PMC 5646367. PMID 28416815.
  107. ^ Agarwal, Shiwani; Weidner, Tatjana; Thalheimer, Frederic B.; Buchholz, Christian J. (2019-10-10). "In vivo generated human CAR T cells eradicate tumor cells". OncoImmunology. 8 (12): e1671761. doi:10.1080/2162402x.2019.1671761. ISSN 2162-402X. PMC 6844313. PMID 31741773.
  108. ^ Agarwalla, Pritha; Ogunnaike, Edikan A.; Ahn, Sarah; Froehlich, Kristen A.; Jansson, Anton; Ligler, Frances S.; Dotti, Gianpietro; Brudno, Yevgeny (2022-03-24). "Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR-T cells". Nature Biotechnology. 40 (8): 1250–1258. doi:10.1038/s41587-022-01245-x. ISSN 1087-0156. PMC 9376243. PMID 35332339.
  109. ^ Ghassemi, Saba; Durgin, Joseph S.; Nunez-Cruz, Selene; Patel, Jai; Leferovich, John; Pinzone, Marilia; Shen, Feng; Cummins, Katherine D.; Plesa, Gabriela; Cantu, Vito Adrian; Reddy, Shantan; Bushman, Frederic D.; Gill, Saar I.; O'Doherty, Una; O'Connor, Roddy S. (February 2022). "Rapid manufacturing of non-activated potent CAR T cells". Nature Biomedical Engineering. 6 (2): 118–128. doi:10.1038/s41551-021-00842-6. ISSN 2157-846X. PMC 8860360. PMID 35190680.
  110. ^ Ledford, Heidi (2023-12-20). "Cancer-fighting CAR-T cells could be made inside body with viral injection". Nature. 625 (7994): 225–226. doi:10.1038/d41586-023-03969-5. PMID 38129613.
  111. ^ "Decision Memo for Chimeric Antigen Receptor (CAR) T-cell Therapy for Cancers (CAG-00451N)". www.cms.gov. Retrieved 2021-03-22.
  112. ^ . www.hematology.org. Archived from the original on 2022-01-24. Retrieved 2021-03-22.
  113. ^ Fiorenza S, Ritchie DS, Ramsey SD, Turtle CJ, Roth JA (September 2020). "Value and affordability of CAR T-cell therapy in the United States". Bone Marrow Transplantation. 55 (9): 1706–1715. doi:10.1038/s41409-020-0956-8. PMID 32474570. S2CID 218987876.
  114. ^ Eder M (25 November 2021). "Which countries is CAR T-cell therapy available in? | SingleUseSupport". Retrieved 13 May 2022.
  115. ^ "Anvisa aprova produto de terapia avançada para tratamento de câncer". Agência Nacional de Vigilância Sanitária - Anvisa (in Brazilian Portuguese). 2022-02-23. Retrieved 2022-06-07.

External links edit

  • CAR T Cells: Engineering Patients' Immune Cells to Treat Their Cancers. National Cancer Institute July 2019

cell, biology, chimeric, antigen, receptors, cars, also, known, chimeric, immunoreceptors, chimeric, cell, receptors, artificial, cell, receptors, receptor, proteins, that, have, been, engineered, give, cells, ability, target, specific, antigen, receptors, chi. In biology chimeric antigen receptors CARs also known as chimeric immunoreceptors chimeric T cell receptors or artificial T cell receptors are receptor proteins that have been engineered to give T cells the new ability to target a specific antigen The receptors are chimeric in that they combine both antigen binding and T cell activating functions into a single receptor Chimeric antigen receptor T cell production and infusion 1 T cells are isolated from a patient s blood2 A new gene encoding a chimeric antigen receptor is incorporated into the T cells3 Engineered T cells are now specific to a desired target antigen4 Engineered T cells are expanded in tissue culture5 Engineered T cells are infused back into the patient CAR T cell therapy uses T cells engineered with CARs to treat cancer T cells are modified to recognize cancer cells and destroy them The standard approach is to harvest T cells from patients genetically alter them then infuse the resulting CAR T cells into patients to attack their tumors 1 CAR T cells can be derived either autologously from T cells in a patient s own blood or allogeneically from those of a donor Once isolated these T cells are genetically engineered to express a specific CAR using a vector derived from an engineered lentivirus such as HIV see Lentiviral vector in gene therapy The CAR programs the T cells to target an antigen present on the tumor cell surface For safety CAR T cells are engineered to be specific to an antigen that is expressed on a tumor cell but not on healthy cells 2 After the modified T cells are infused into a patient they act as a living drug against cancer cells 3 When they come in contact with their targeted antigen on a cell s surface T cells bind to it and become activated then proceed to proliferate and become cytotoxic 4 CAR T cells destroy cells through several mechanisms including extensive stimulated cell proliferation increasing the degree to which they are toxic to other living cells cytotoxicity and by causing the increased secretion of factors that can affect other cells such as cytokines interleukins and growth factors 5 The surface of CAR T cells can bear either of two types of co receptors CD4 and CD8 These two cell types called CD4 and CD8 respectively have different and interacting cytotoxic effects Therapies employing a 1 to 1 ratio of the cell types apparently provide synergistic antitumor effects 6 Contents 1 History 2 Production 3 Clinical applications 3 1 Cancer 3 2 Autoimmune disease 4 Approved therapies 5 Safety 5 1 Cytokine release syndrome 5 2 Immune effector cell associated neurotoxicity 6 Chimeric antigen receptor structure 6 1 Antigen recognition domain 6 2 Hinge region 6 3 Transmembrane domain 6 4 Intracellular T cell signaling domain 7 Research directions 7 1 Antigen recognition 7 2 CAR T function 7 3 Control mechanisms 7 4 Advances in CAR T cell manufacturing 7 5 In situ modification 8 Economics 9 See also 10 References 11 External linksHistory editThe first chimeric receptors containing portions of an antibody and the T cell receptor was described in 1987 by Yoshihisa Kuwana et al 7 at Fujita Health University and Kyowa Hakko Kogyo Co Ltd in Japan and independently in 1989 by Gideon Gross and Zelig Eshhar 8 9 at the Weizmann Institute in Israel 10 Originally termed T bodies these early approaches combined an antibody s ability to specifically bind to diverse targets with the constant domains of the TCR a or TCR b proteins 11 In 1991 chimeric receptors containing the intracellular signaling domain of CD3z were shown to activate T cell signaling by Arthur Weiss at the University of California San Francisco 12 This work prompted CD3z intracellular domains to be added to chimeric receptors with antibody like extracellular domains commonly single chain fraction variable scFv domains as well as proteins such as CD4 subsequently termed first generation CARs 13 14 A first generation CAR containing a CD4 extracellular domain and a CD3z intracellular domain was used in the first clinical trial of chimeric antigen receptor T cells by the biotechnology company Cell Genesys in the mid 1990s allowing adoptively transferred T cells to target HIV infected cells although it failed to show any clinical improvement 13 Similar early clinical trials of CAR T cells in solid tumors in the 1990s using first generation CARs targeting a solid tumor antigens such as MUC1 did not show long term persistence of the transferred T cells or result in significant remissions 15 In the early 2000s co stimulatory domains such as CD28 or 4 1BB were added to first generation CAR s CD3z intracellular domain Termed second generation CARs these constructs showed greater persistence and improved tumor clearance in pre clinical models 16 Clinical trials in the early 2010s using second generation CARs targeting CD19 a protein expressed by normal B cells as well as B cell leukemias and lymphomas by investigators at the NCI University of Pennsylvania and Memorial Sloan Kettering Cancer Center demonstrated the clinical efficacy of CAR T cell therapies and resulted in complete remissions in many heavily pre treated patients 15 These trials ultimately led in the US to the FDA s first two approvals of CAR T cells in 2017 those for tisagenlecleucel Kymriah marketed by Novartis originally for B cell precursor acute lymphoblastic leukemia B ALL and axicabtagene ciloleucel Yescarta marketed by Kite Pharma originally for diffuse large B cell lymphoma DLBCL 15 There are now six FDA approved CAR T therapies 17 Production edit nbsp Depiction of adoptive cell transfer therapy with CAR engineered T cells The first step in the production of CAR T cells is the isolation of T cells from human blood CAR T cells may be manufactured either from the patient s own blood known as an autologous treatment or from the blood of a healthy donor known as an allogeneic treatment The manufacturing process is the same in both cases only the choice of initial blood donor is different citation needed First leukocytes are isolated using a blood cell separator in a process known as leukocyte apheresis Peripheral blood mononuclear cells PBMCs are then separated and collected 18 19 The products of leukocyte apheresis are then transferred to a cell processing center In the cell processing center specific T cells are stimulated so that they will actively proliferate and expand to large numbers To drive their expansion T cells are typically treated with the cytokine interleukin 2 IL 2 and anti CD3 antibodies 20 Anti CD3 CD28 antibodies are also used in some protocols 19 The expanded T cells are purified and then transduced with a gene encoding the engineered CAR via a retroviral vector typically either an integrating gammaretrovirus RV or a lentiviral LV vector 19 These vectors are very safe in modern times due to a partial deletion of the U3 region 21 The new gene editing tool CRISPR Cas9 has recently been used instead of retroviral vectors to integrate the CAR gene into specific sites in the genome 22 The patient undergoes lymphodepletion chemotherapy prior to the introduction of the engineered CAR T cells 4 The depletion of the number of circulating leukocytes in the patient upregulates the number of cytokines that are produced and reduces competition for resources which helps to promote the expansion of the engineered CAR T cells 23 Clinical applications editAs of March 2019 there were around 364 ongoing clinical trials happening globally involving CAR T cells 24 The majority of those trials target blood cancers CAR T therapies account for more than half of all trials for hematological malignancies 24 CD19 continues to be the most popular antigen target 25 followed by BCMA commonly expressed in multiple myeloma 24 26 In 2016 studies began to explore the viability of other antigens such as CD20 27 Trials for solid tumors are less dominated by CAR T with about half of cell therapy based trials involving other platforms such as NK cells 24 Cancer edit T cells are genetically engineered to express chimeric antigen receptors specifically directed toward antigens on a patient s tumor cells then infused into the patient where they attack and kill the cancer cells 28 Adoptive transfer of T cells expressing CARs is a promising anti cancer therapeutic because CAR modified T cells can be engineered to target potentially any tumor associated antigen 29 30 Early CAR T cell research has focused on blood cancers The first approved treatments use CARs that target the antigen CD19 present in B cell derived cancers such as acute lymphoblastic leukemia ALL and diffuse large B cell lymphoma DLBCL 31 32 There are also efforts underway to engineer CARs targeting many other blood cancer antigens including CD30 in refractory Hodgkin s lymphoma CD33 CD123 and FLT3 in acute myeloid leukemia AML and BCMA in multiple myeloma 33 Solid tumors have presented a more difficult target 34 Identification of good antigens has been challenging such antigens must be highly expressed on the majority of cancer cells but largely absent on normal tissues 35 36 37 30 CAR T cells are also not trafficked efficiently into the center of solid tumor masses and the hostile tumor microenvironment suppresses T cell activity 33 Autoimmune disease edit While most CAR T cell studies focus on creating a CAR T cell that can eradicate a certain cell population for instance CAR T cells that target lymphoma cells there are other potential uses for this technology T cells can also mediate tolerance to antigens 38 A regulatory T cell outfitted with a CAR could have the potential to confer tolerance to a specific antigen something that could be utilized in organ transplantation or rheumatologic diseases like lupus 39 40 Approved therapies editThe examples and perspective in this section may not represent a worldwide view of the subject You may improve this section discuss the issue on the talk page or create a new section as appropriate November 2023 Learn how and when to remove this message CAR T Cell Therapies with Regulatory Approval CAR T cell Brand name Company Approval Agency Date Target Antigen recognition domain Intracellular signaling domain Indication Targeted disease Line of Therapy Agency Product Number Drug Label tisagenlecleucel Kymriah Novartis FDA 08 30 2017 41 EMA 08 22 2018 42 MHLW 05 15 2019 43 CD19 scFV 41BB CD3z B cell precursor ALL Third Line 41 42 43 Diffuse large B cell lymphoma Third Line 44 42 43 Follicular Lymphoma Third Line 45 46 FDA 125646 Label EMA 004090 Label axicabtagene ciloleucel Yescarta Kite Pharma Gilead FDA 10 18 2017 47 EMA 08 27 2018 48 NMPA 06 23 2021 49 MHLW 12 22 2022 50 CD19 scFV CD28 CD3z Diffuse large B cell lymphoma Second Line 51 52 49 50 Follicular lymphoma Third Line 53 54 49 50 Primary mediastinal large B cell lymphoma Third Line 48 49 50 FDA 125643 Label EMA 004480 Label brexucabtagene autoleucel Tecartus Kite Pharma Gilead FDA 07 24 2020 55 EMA 12 14 2020 56 CD19 scFV CD28 CD3z Mantle cell lymphoma Third Line 57 56 B cell precursor ALL Third Line 57 56 FDA 125703 Label EMA 005102 Label lisocabtagene maraleucel Breyanzi Juno Therapeutics BMS FDA 02 05 2021 58 EMA 04 04 2022 59 MHLW 12 20 2022 60 CD19 scFV 41BB CD3z Diffuse large B cell lymphoma Second Line 61 59 60 FDA 25714 Label EMA 004731 Label idecabtagene vicleucel Abecma Bluebird Bio BMS FDA 03 26 2021 62 EMA 08 18 2021 63 BCMA scFV 41BB CD3z Multiple myeloma Fourth Line 63 Fifth Line 62 FDA 125736 Label EMA 004662 Label ciltacabtagene autoleucel Carvykti Janssen J amp J FDA 02 28 2022 64 EMA 05 25 2022 65 BCMA VHH 41BB CD3z Multiple myeloma Fourth Line 65 Fifth Line 64 FDA 125746 Label EMA 005095 LabelSafety editThere are serious side effects that result from CAR T cells being introduced into the body including cytokine release syndrome and neurological toxicity 4 Because it is a relatively new treatment there are few data about the long term effects of CAR T cell therapy There are still concerns about long term patient survival as well as pregnancy complications in female patients treated with CAR T cells 66 Anaphylaxis may be a side effect as the CAR is made with a foreign monoclonal antibody and as a result provokes an immune response citation needed On target off tumor recognition occurs when the CAR T cell recognizes the correct antigen but the antigen is expressed on healthy non pathogenic tissue This results in the CAR T cells attacking non tumor tissue such as healthy B cells that express CD19 causing B cell aplasia The severity of this adverse effect can vary but the combination of prior immunosuppression lymphodepleting chemotherapy and on target effects causing hypogammaglobulinaemia and prolonged cytopenias places patients at increased risk of serious infections 20 67 There is also the unlikely possibility that the engineered CAR T cells will themselves become transformed into cancerous cells through insertional mutagenesis due to the viral vector inserting the CAR gene into a tumor suppressor or oncogene in the host T cell s genome Some retroviral RV vectors carry a lower risk than lentiviral LV vectors However both have the potential to be oncogenic Genomic sequencing analysis of CAR insertion sites in T cells has been established for better understanding of CAR T cell function and persistence in vivo 35 Cytokine release syndrome edit Main article Cytokine release syndrome The most common issue after treatment with CAR T cells is cytokine release syndrome CRS a condition in which the immune system is activated and releases an increased number of inflammatory cytokines The clinical manifestation of this syndrome resembles sepsis with high fever fatigue myalgia nausea capillary leakages tachycardia and other cardiac dysfunction liver failure and kidney impairment 68 CRS occurs in almost all patients treated with CAR T cell therapy in fact the presence of CRS is a diagnostic marker that indicates the CAR T cells are working as intended to kill the cancer cells 66 The severity of CRS does not correlate with an increased response to the treatment but rather higher disease burden 66 Severe cytokine release syndrome can be managed with immunosuppressants such as corticosteroids and with tocilizumab an anti IL 6 monoclonal antibody 69 Early intervention using tocilizumab was shown to reduce the frequency of severe CRS in multiple studies 70 71 without affecting the therapeutic effect of the treatment Immune effector cell associated neurotoxicity edit Neurological toxicity is also often associated with CAR T cell treatment 72 The underlying mechanism is poorly understood and may or may not be related to CRS Clinical manifestations include delirium the partial loss of the ability to speak coherently while still having the ability to interpret language expressive aphasia lowered alertness obtundation and seizures 66 During some clinical trials deaths caused by neurotoxicity have occurred The main cause of death from neurotoxicity is cerebral edema In a study carried out by Juno Therapeutics Inc five patients enrolled in the trial died as a result of cerebral edema Two of the patients were treated with cyclophosphamide alone and the remaining three were treated with a combination of cyclophosphamide and fludarabine 73 In another clinical trial sponsored by the Fred Hutchinson Cancer Research Center there was one reported case of irreversible and fatal neurological toxicity 122 days after the administration of CAR T cells 74 Hypokinetic movement disorder parkinsonism or movement and neurocognitive treatment emergent adverse events has been observed with BCMA chimeric antigen receptor CAR T cell treatment for multiple myeloma 75 Chimeric antigen receptor structure editChimeric antigen receptors combine many facets of normal T cell activation into a single protein They link an extracellular antigen recognition domain to an intracellular signalling domain which activates the T cell when an antigen is bound CARs are composed of four regions an antigen recognition domain an extracellular hinge region a transmembrane domain and an intracellular T cell signaling domain 76 77 nbsp Different components of a chimeric antigen receptor Antigen recognition domain edit The antigen recognition domain is exposed to the outside of the cell in the ectodomain portion of the receptor It interacts with potential target molecules and is responsible for targeting the CAR T cell to any cell expressing a matching molecule citation needed The antigen recognition domain is typically derived from the variable regions of a monoclonal antibody linked together as a single chain variable fragment scFv 77 An scFv is a chimeric protein made up of the light VL and heavy VH chains of immunoglobins connected with a short linker peptide 78 These VL and VH regions are selected in advance for their binding ability to the target antigen such as CD19 The linker between the two chains consists of hydrophilic residues with stretches of glycine and serine in it for flexibility as well as stretches of glutamate and lysine for added solubility 79 Single domain antibodies e g VH VHH VNAR have been engineered and developed as antigen recognition domains in the CAR format due to their high transduction efficiency in T cells 80 35 81 82 83 In addition to antibody fragments non antibody based approaches have also been used to direct CAR specificity usually taking advantage of ligand receptor pairs that normally bind to each other 76 Cytokines innate immune receptors TNF receptors growth factors and structural proteins have all been successfully used as CAR antigen recognition domains 76 Hinge region edit The hinge also called a spacer is a small structural domain that sits between the antigen recognition region and the cell s outer membrane An ideal hinge enhances the flexibility of the scFv receptor head reducing the spatial constraints between the CAR and its target antigen This promotes antigen binding and synapse formation between the CAR T cells and target cells 84 Hinge sequences are often based on membrane proximal regions from other immune molecules including IgG CD8 and CD28 76 85 81 82 Transmembrane domain edit The transmembrane domain is a structural component consisting of a hydrophobic alpha helix that spans the cell membrane It anchors the CAR to the plasma membrane bridging the extracellular hinge and antigen recognition domains with the intracellular signaling region 76 This domain is essential for the stability of the receptor as a whole Generally the transmembrane domain from the most membrane proximal component of the endodomain is used but different transmembrane domains result in different receptor stability The CD28 transmembrane domain is known to result in a highly expressed stable receptor 82 Using the CD3 zeta transmembrane domain is not recommended as it can result in incorporation of the artificial TCR into the native TCR 86 Intracellular T cell signaling domain edit nbsp Depiction of first second and third generation chimeric antigen receptors with the scFv segments in green and the various intracellular TCR signaling components in red blue and yellow 87 The intracellular T cell signaling domain lies in the receptor s endodomain inside the cell 76 After an antigen is bound to the external antigen recognition domain CAR receptors cluster together and transmit an activation signal Then the internal cytoplasmic end of the receptor perpetuates signaling inside the T cell 78 Normal T cell activation relies on the phosphorylation of immunoreceptor tyrosine based activation motifs ITAMs present in the cytoplasmic domain of CD3 zeta To mimic this process CD3 zeta s cytoplasmic domain is commonly used as the main CAR endodomain component Other ITAM containing domains have also been tried but are not as effective 77 T cells also require co stimulatory molecules in addition to CD3 signaling in order to persist after activation For this reason the endodomains of CAR receptors typically also include one or more chimeric domains from co stimulatory proteins 3 Signaling domains from a wide variety of co stimulatory molecules have been successfully tested including CD28 CD27 CD134 OX40 and CD137 4 1BB 76 The intracellular signaling domain used defines the generation of a CAR T cell 4 First generation CARs include only a CD3 zeta cytoplasmic domain 4 Second generation CARs add a co stimulatory domain like CD28 or 4 1BB The involvement of these intracellular signaling domains improve T cell proliferation cytokine secretion resistance to apoptosis and in vivo persistence 4 Third generation CARs combine multiple co stimulatory domains such as CD28 41BB or CD28 OX40 to augment T cell activity Preclinical data show the third generation CARs exhibit improved effector functions and better in vivo persistence as compared to second generation CARs 4 Research directions editAntigen recognition edit Although the initial clinical remission rates after CAR T cell therapy in all patients are as high as 90 88 long term survival rates are much lower The cause is typically the emergence of leukemia cells that do not express CD19 and so evade recognition by the CD19 CAR T cells a phenomenon known as antigen escape 33 Preclinical studies developing CAR T cells with dual targeting of CD19 plus CD22 or CD19 plus CD20 have demonstrated promise and trials studying bispecific targeting to circumvent CD19 down regulation are ongoing 33 In 2018 a version of CAR was developed that is referred to as SUPRA CAR or split universal and programmable 89 Multiple mechanisms can be deployed to finely regulate the activity of SUPRA CAR which limits overactivation In contrast to the traditional CAR design SUPRA CAR allows targeting of multiple antigens without further genetic modification of a person s immune cells 90 SMDCs small molecule drug conjugates platform in immuno oncology is an experimental approach that makes possible the engineering of a single universal CAR T cell which binds with extraordinarily high affinity to a benign molecule designated as fluorescein isothiocyanate FITC These cells are then used to treat various cancer types when co administered with bispecific SMDC adaptor molecules These unique bispecific adaptors are constructed with a FITC molecule and a tumor homing molecule to precisely bridge the universal CAR T cell with the cancer cells which causes localized T cell activation Anti tumor activity in mice is induced only when both the universal CAR T cells plus the correct antigen specific adaptor molecules are present Anti tumor activity and toxicity can be controlled by adjusting the administered adaptor molecule dosing Treatment of antigenically heterogeneous tumors can be achieved by administration of a mixture of the desired antigen specific adaptors 91 92 CAR T function edit Fourth generation CARs also known as TRUCKs or armored CARs further add factors that enhance T cell expansion persistence and anti tumoral activity This can include cytokines such is IL 2 IL 5 IL 12 and co stimulatory ligands 93 94 Control mechanisms edit Adding a synthetic control mechanism to engineered T cells allows doctors to precisely control the persistence or activity of the T cells in the patient s body with the goal of reducing toxic side effects 95 The major control techniques trigger T cell death or limit T cell activation and often regulate the T cells via a separate drug that can be introduced or withheld as needed citation needed Suicide genes Genetically modified T cells are engineered to include one or more genes that can induce apoptosis when activated by an extracellular molecule Herpes simplex virus thymidine kinase HSV TK and inducible caspase 9 iCasp9 are two types of suicide genes that have been integrated into CAR T cells 95 96 97 In the iCasp9 system the suicide gene complex has two elements a mutated FK506 binding protein with high specificity to the small molecule rimiducid AP1903 and a gene encoding a pro domain deleted human caspase 9 Dosing the patient with rimiducid activates the suicide system leading to rapid apoptosis of the genetically modified T cells Although both the HSV TK and iCasp9 systems demonstrate a noticeable function as a safety switch in clinical trials some defects limit their application HSV TK is virus derived and may be immunogenic to humans 95 98 It is also currently unclear whether the suicide gene strategies will act quickly enough in all situations to halt dangerous off tumor cytotoxicity citation needed Dual antigen receptor CAR T cells are engineered to express two tumor associated antigen receptors at the same time reducing the likelihood that the T cells will attack non tumor cells Dual antigen receptor CAR T cells have been reported to have less intense side effects 99 An in vivo study in mice shows that dual receptor CAR T cells effectively eradicated prostate cancer and achieved complete long term survival 100 ON switch and OFF switch In this system CAR T cells can only function in the presence of both tumor antigen and a benign exogenous molecule To achieve this the CAR T cell s engineered chimeric antigen receptor is split into two separate proteins that must come together in order to function The first receptor protein typically contains the extracellular antigen binding domain while the second protein contains the downstream signaling elements and co stimulatory molecules such as CD3z and 4 1BB In the presence of an exogenous molecule such as a rapamycin analog the binding and signaling proteins dimerize together allowing the CAR T cells to attack the tumor 101 Human EGFR truncated form hEGFRt has been used as an OFF switch for CAR T cells using cetuximab 35 37 81 Bispecific molecules as switches Bispecific molecules target both a tumor associated antigen and the CD3 molecule on the surface of T cells This ensures that the T cells cannot become activated unless they are in close physical proximity to a tumor cell 102 The anti CD20 CD3 bispecific molecule shows high specificity to both malignant B cells and cancer cells in mice 103 FITC is another bifunctional molecule used in this strategy FITC can redirect and regulate the activity of the FITC specific CAR T cells toward tumor cells with folate receptors 104 Advances in CAR T cell manufacturing edit Due to the high costs of CAR T cell therapy 105 a number of alternative efforts are being investigated to improve CAR T cell manufacturing and reduce costs In vivo CAR T cell manufacturing strategies 106 107 are being tested In addition bioinstructive materials have been developed for CAR T cell generation 108 Rapid CAR T cell generation is also possible through shortening or eliminating the activation and expansion steps 109 In situ modification edit Another approach is to modify T cells and or B cells still in the body using viral vectors 110 Economics editThe cost of CAR T cell therapies has been criticized with the initial costs of tisagenlecleucel Kymriah and axicabtagene ciloleucel Yescarta being 375 000 and 475 000 respectively 105 The high cost of CAR T therapies is due to complex cellular manufacturing in specialized good manufacturing practice GMP facilities as well as the high level of hospital care necessary after CAR T cells are administered due to risks such as cytokine release syndrome 105 In the United States CAR T cell therapies are covered by Medicare and by many but not all private insurers 111 112 Manufacturers of CAR T cells have developed alternative payment programs due to the high cost of CAR T therapy such as by requiring payment only if the CAR T therapy induces a complete remission by a certain time point after treatment 113 Additionally CAR T cell therapies are not available worldwide yet CAR T cell therapies have been approved in China Australia Singapore the United Kingdom and some European countries 114 In February 2022 Brazil approved tisagenlecleucel Kymriah treatment 115 See also editCell therapy Checkpoint inhibitor Epcoritamab Gene therapy Immune checkpointReferences edit Fox M July 12 2017 New Gene Therapy for Cancer Offers Hope to Those With No Options Left NBC News Srivastava S Riddell SR August 2015 Engineering CAR T cells Design concepts Trends in Immunology 36 8 494 502 doi 10 1016 j it 2015 06 004 PMC 4746114 PMID 26169254 a b Sadelain M Brentjens R Riviere I April 2013 The basic principles of chimeric antigen receptor design Cancer Discovery 3 4 388 398 doi 10 1158 2159 8290 CD 12 0548 PMC 3667586 PMID 23550147 a b c d e f g Hartmann J Schussler Lenz M Bondanza A Buchholz CJ September 2017 Clinical development of CAR T cells challenges and opportunities in translating innovative treatment concepts EMBO Molecular Medicine 9 9 1183 1197 doi 10 15252 emmm 201607485 PMC 5582407 PMID 28765140 Tang XJ Sun XY Huang KM Zhang L Yang ZS Zou DD et al December 2015 Therapeutic potential of CAR T cell derived exosomes a cell free modality for targeted cancer therapy Oncotarget 6 42 44179 44190 doi 10 18632 oncotarget 6175 PMC 4792550 PMID 26496034 Zhang H Zhao P Huang H December 2020 Engineering better chimeric antigen receptor T cells Experimental Hematology amp Oncology 9 1 34 doi 10 1186 s40164 020 00190 2 PMC 7709221 PMID 33292660 Kuwana Y Asakura Y Utsunomiya N Nakanishi M Arata Y Itoh S et al December 1987 Expression of chimeric receptor composed of immunoglobulin derived V regions and T cell receptor derived C regions Biochemical and Biophysical Research Communications 149 3 960 968 doi 10 1016 0006 291x 87 90502 x PMID 3122749 Gross G Gorochov G Waks T Eshhar Z February 1989 Generation of effector T cells expressing chimeric T cell receptor with antibody type specificity Transplantation Proceedings 21 1 Pt 1 127 130 PMID 2784887 Rosenbaum L October 2017 Tragedy Perseverance and Chance The Story of CAR T Therapy The New England Journal of Medicine 377 14 1313 1315 doi 10 1056 NEJMp1711886 PMID 28902570 S2CID 205114161 Gross G Waks T Eshhar Z December 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 24 10024 10028 Bibcode 1989PNAS 8610024G doi 10 1073 pnas 86 24 10024 JSTOR 34790 PMC 298636 PMID 2513569 Eshhar Z Bach N Fitzer Attas CJ Gross G Lustgarten J Waks T Schindler DG 1996 The T body approach potential for cancer immunotherapy Springer Seminars in Immunopathology 18 2 199 209 doi 10 1007 BF00820666 PMID 8908700 S2CID 19872173 Irving BA Weiss A March 1991 The cytoplasmic domain of the T cell receptor zeta chain is sufficient to couple to receptor associated signal transduction pathways Cell 64 5 891 901 doi 10 1016 0092 8674 91 90314 o PMID 1705867 S2CID 23466990 a b Hege KM Roberts MR December 1996 T cell gene therapy Current Opinion in Biotechnology 7 6 629 634 doi 10 1016 s0958 1669 96 80074 7 PMID 8939644 June CH Sadelain M July 2018 Chimeric Antigen Receptor Therapy The New England Journal of Medicine 379 1 64 73 doi 10 1056 NEJMra1706169 PMC 7433347 PMID 29972754 a b c Braendstrup P Levine BL Ruella M February 2020 The long road to the first FDA approved gene therapy chimeric antigen receptor T cells targeting CD19 Cytotherapy 22 2 57 69 doi 10 1016 j jcyt 2019 12 004 PMC 7036015 PMID 32014447 Sadelain M Riviere I Brentjens R January 2003 Targeting tumours with genetically enhanced T lymphocytes Nature Reviews Cancer 3 1 35 45 doi 10 1038 nrc971 PMID 12509765 S2CID 33707802 Center for Biologics Evaluation and Research 2022 03 01 Approved Cellular and Gene Therapy Products FDA Jin C Yu D Hillerdal V Wallgren A Karlsson Parra A Essand M 2014 03 05 Allogeneic lymphocyte licensed DCs expand T cells with improved antitumor activity and resistance to oxidative stress and immunosuppressive factors Molecular Therapy Methods amp Clinical Development 1 14001 doi 10 1038 mtm 2014 1 PMC 4362340 PMID 26015949 a b c Li Nan Ho Mitchell 2022 Development of Glypican 2 Targeting Single Domain Antibody CAR T Cells for Neuroblastoma Single Domain Antibodies Methods in Molecular Biology Vol 2446 pp 451 468 doi 10 1007 978 1 0716 2075 5 23 ISBN 978 1 0716 2074 8 ISSN 1940 6029 PMID 35157288 S2CID 246813053 a b Makita S Yoshimura K Tobinai K June 2017 Clinical development of anti CD19 chimeric antigen receptor T cell therapy for B cell non Hodgkin lymphoma Cancer Science 108 6 1109 1118 doi 10 1111 cas 13239 PMC 5480083 PMID 28301076 Jin C Fotaki G Ramachandran M Nilsson B Essand M Yu D July 2016 Safe engineering of CAR T cells for adoptive cell therapy of cancer using long term episomal gene transfer EMBO Molecular Medicine 8 7 702 711 doi 10 15252 emmm 201505869 PMC 4931286 PMID 27189167 Jensen TI Axelgaard E Bak RO June 2019 Therapeutic gene editing in haematological disorders with CRISPR Cas9 British Journal of Haematology 185 5 821 835 doi 10 1111 bjh 15851 PMID 30864164 Muranski P Boni A Wrzesinski C Citrin DE Rosenberg SA Childs R Restifo NP December 2006 Increased intensity lymphodepletion and adoptive immunotherapy how far can we go Nature Clinical Practice Oncology 3 12 668 681 doi 10 1038 ncponc0666 PMC 1773008 PMID 17139318 a b c d Xin Yu J Hubbard Lucey VM Tang J October 2019 The global pipeline of cell therapies for cancer Nature Reviews Drug Discovery 18 11 821 822 doi 10 1038 d41573 019 00090 z PMID 31673124 S2CID 190862546 Brudno and Kochenderfer Chimeric antigen receptor T cell therapies for lymphoma Nature Reviews Clinical Oncology 2018 15 31 46 Mikkilineni and Kochenderfer Chimeric antigen receptor T cell therapies for multiple myeloma Blood 2017 130 2594 602 Almasbak H Aarvak T Vemuri MC 2016 CAR T Cell Therapy A Game Changer in Cancer Treatment Journal of Immunology Research 2016 5474602 doi 10 1155 2016 5474602 PMC 4889848 PMID 27298832 Jacobson CA Ritz J November 2011 Time to put the CAR T before the horse Blood 118 18 4761 4762 doi 10 1182 blood 2011 09 376137 PMID 22053170 Li Nan Spetz Madeline R Li Dan Ho Mitchell July 2021 Advances in immunotherapeutic targets for childhood cancers A focus on glypican 2 and B7 H3 Pharmacology amp Therapeutics 223 107892 doi 10 1016 j pharmthera 2021 107892 ISSN 1879 016X PMC 8202769 PMID 33992682 a b Li Dan Lin Shaoli Hong Jessica Ho Mitchell 2022 Immunotherapy for hepatobiliary cancers Emerging targets and translational advances Advances in Cancer Research 156 415 449 doi 10 1016 bs acr 2022 01 013 ISBN 9780323983921 ISSN 2162 5557 PMID 35961708 S2CID 246978004 Kochenderfer JN Wilson WH Janik JE Dudley ME Stetler Stevenson M Feldman SA et al November 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 20 4099 4102 doi 10 1182 blood 2010 04 281931 PMC 2993617 PMID 20668228 Kochenderfer JN Dudley ME Kassim SH Somerville RP Carpenter RO Stetler Stevenson M et al February 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 6 540 549 doi 10 1200 JCO 2014 56 2025 PMC 4322257 PMID 25154820 a b c d Schultz L Mackall C February 2019 Driving CAR T cell translation forward Science Translational Medicine 11 481 eaaw2127 doi 10 1126 scitranslmed aaw2127 PMID 30814337 Lim WA June CH February 2017 The Principles of Engineering Immune Cells to Treat Cancer Cell 168 4 724 740 doi 10 1016 j cell 2017 01 016 PMC 5553442 PMID 28187291 a b c d Li D Li N Zhang YF Fu H Feng M Schneider D et al June 2020 Persistent Polyfunctional Chimeric Antigen Receptor T Cells That Target Glypican 3 Eliminate Orthotopic Hepatocellular Carcinomas in Mice Gastroenterology 158 8 2250 2265 e20 doi 10 1053 j gastro 2020 02 011 PMC 7282931 PMID 32060001 Li Nan Fu Haiying Hewitt Stephen M Dimitrov Dimiter S Ho Mitchell 2017 08 08 Therapeutically targeting glypican 2 via single domain antibody based chimeric antigen receptors and immunotoxins in neuroblastoma Proceedings of the National Academy of Sciences of the United States of America 114 32 E6623 E6631 Bibcode 2017PNAS 114E6623L doi 10 1073 pnas 1706055114 ISSN 1091 6490 PMC 5559039 PMID 28739923 a b Li Nan Torres Madeline B Spetz Madeline R Wang Ruixue Peng Luyi Tian Meijie Dower Christopher M Nguyen Rosa Sun Ming Tai Chin Hsien de Val Natalia Cachau Raul Wu Xiaolin Hewitt Stephen M Kaplan Rosandra N 2021 06 15 CAR T cells targeting tumor associated exons of glypican 2 regress neuroblastoma in mice Cell Reports Medicine 2 6 100297 doi 10 1016 j xcrm 2021 100297 ISSN 2666 3791 PMC 8233664 PMID 34195677 Sakaguchi S Yamaguchi T Nomura T Ono M May 2008 Regulatory T cells and immune tolerance Cell 133 5 775 787 doi 10 1016 j cell 2008 05 009 PMID 18510923 S2CID 2315895 Zhang Q Lu W Liang CL Chen Y Liu H Qiu F Dai Z 2018 Chimeric Antigen Receptor CAR Treg A Promising Approach to Inducing Immunological Tolerance Frontiers in Immunology 9 2359 doi 10 3389 fimmu 2018 02359 PMC 6194362 PMID 30369931 Mougiakakos D Kronke G Volkl S Kretschmann S Aigner M Kharboutli S et al August 2021 CD19 Targeted CAR T Cells in Refractory Systemic Lupus Erythematosus The New England Journal of Medicine 385 6 567 569 doi 10 1056 NEJMc2107725 PMID 34347960 S2CID 236927691 a b Novartis receives first ever FDA approval for a CAR T cell therapy Kymriah TM CTL019 for children and young adults with B cell ALL that is refractory or has relapsed at least twice www novartis com 2017 08 30 a b c Novartis receives European Commission approval of its CAR T cell therapy Kymriah tisagenlecleucel www novartis com Retrieved 2023 11 18 a b c Novartis gets approval to sell Kymriah in Japan for 306 000 www reuters com 2019 05 15 KYMRIAH tisagenlecleucel US Food and Drug Administration 2019 04 05 FDA approves Novartis Kymriah CAR T cell therapy for adult patients with relapsed or refractory follicular lymphoma Novartis Retrieved 2022 06 05 Novartis Kymriah receives EC approval as first CAR T cell therapy for adults with relapsed or refractory follicular lymphoma www novartis com Retrieved 2023 11 18 Center for Biologics Evaluation and Research 2017 10 18 FDA approves CAR T cell therapy to treat adults with certain types of large B cell lymphoma FDA a b Yescarta Axicabtagene Ciloleucel Receives European Marketing Authorization for the Treatment of Relapsed or Refractory DLBCL and PMBCL After Two or More Lines of Systemic Therapy www gilead com Retrieved 2023 11 18 a b c d Kite Joint Venture Fosun Kite Gains the First CAR T cell Therapy Approval in China www gilead com Retrieved 2023 11 13 a b c d Yescarta Now Approved in Japan for Initial Treatment of Relapsed Refractory Large B Cell Lymphoma www gilead com 2022 12 22 Center for Biologics Evaluation and Research 2020 05 28 YESCARTA axicabtagene ciloleucel FDA Kite s CAR T cell Therapy Yescarta First in Europe to Receive Positive CHMP Opinion for Use in Second line Diffuse Large B cell Lymphoma and High grade B cell Lymphoma www gilead com Retrieved 2023 11 18 U S FDA Approves Yescarta for Relapsed or Refractory Follicular Lymphoma After Two or More Lines of Systemic Therapy www gilead com Retrieved 2021 03 15 Kite s CAR T cell Therapy Yescarta Granted European Marketing Authorization for the Treatment of Relapsed or Refractory Follicular Lymphoma www gilead com Retrieved 2023 11 18 Center for Biologics Evaluation and Research 2021 03 04 TECARTUS brexucabtagene autoleucel FDA a b c European Medicines Agency 2023 01 30 TECARTUS brexucabtagene autoleucel EMA a b Center for Biologics Evaluation and Research 2021 03 04 TECARTUS brexucabtagene autoleucel FDA U S Food and Drug Administration Approves Bristol Myers Squibb s Breyanzi lisocabtagene maraleucel a New CAR T Cell Therapy for Adults with Relapsed or Refractory Large B cell Lymphoma news bms com Retrieved 2023 11 21 a b European Medicines Agency 2023 11 10 Breyanzi EMA a b CAR T Cell Therapy Breyanzi Approved as Relapsed or Refractory Large B cell Lymphoma Second Line Therapy in Japan new bms com Center for Biologics Evaluation and Research 2022 06 24 FDA D I S C O Burst Edition FDA approval of Breyanzi lisocabtagene maraleucel for second line treatment of large B cell lymphoma FDA a b Center for Biologics Evaluation and Research 2021 03 27 ABECMA idecabtagene vicleucel FDA a b European Medicines Agency 2023 07 27 Abecma EMA a b Center for Biologics Evaluation and Research 2022 03 21 CARVYKTI FDA a b European Medicines Agency 2023 07 27 Carvykti EMA a b c d Bonifant CL Jackson HJ Brentjens RJ Curran KJ 2016 Toxicity and management in CAR T cell therapy Molecular Therapy Oncolytics 3 16011 doi 10 1038 mto 2016 11 PMC 5008265 PMID 27626062 Bupha Intr O Haeusler G Chee L Thursky K Slavin M Teh B June 2021 CAR T cell therapy and infection a review Expert Review of Anti Infective Therapy 19 6 749 758 doi 10 1080 14787210 2021 1855143 PMID 33249873 S2CID 227235627 Breslin S February 2007 Cytokine release syndrome overview and nursing implications Clinical Journal of Oncology Nursing 11 1 Suppl 37 42 doi 10 1188 07 CJON S1 37 42 PMID 17471824 S2CID 35773028 Lee DW Gardner R Porter DL Louis CU Ahmed N Jensen M et al July 2014 Current concepts in the diagnosis and management of cytokine release syndrome Blood 124 2 188 195 doi 10 1182 blood 2014 05 552729 PMC 4093680 PMID 24876563 Berg P Schonefeld S Ruppert Seipp G Funk MB 2022 11 29 Regulatory Measures to Improve the Safety of CAR T Cell Treatment Transfusion Medicine and Hemotherapy 50 3 218 225 doi 10 1159 000526786 ISSN 1660 3796 PMC 10331154 PMID 37435000 Gardner RA Ceppi F Rivers J Annesley C Summers C Taraseviciute A et al December 2019 Preemptive mitigation of CD19 CAR T cell cytokine release syndrome without attenuation of antileukemic efficacy Blood 134 24 2149 2158 doi 10 1182 blood 2019001463 PMC 6908832 PMID 31697826 Brudno JN Kochenderfer JN June 2016 Toxicities of chimeric antigen receptor T cells recognition and management Blood 127 26 3321 3330 doi 10 1182 blood 2016 04 703751 PMC 4929924 PMID 27207799 Study Evaluating the Efficacy and Safety of JCAR015 in Adult B cell Acute Lymphoblastic Leukemia B ALL ClinicalTrials gov Retrieved 2018 02 21 Turtle CJ Hanafi LA Berger C Gooley TA Cherian S Hudecek M et al June 2016 CD19 CAR T cells of defined CD4 CD8 composition in adult B cell ALL patients The Journal of Clinical Investigation 126 6 2123 2138 doi 10 1172 JCI85309 PMC 4887159 PMID 27111235 Gust Juliane October 5 2023 BCMA CAR T cell treatment associated parkinsonism ashpublications org doi 10 1182 blood 2023021860 Retrieved April 4 2024 a b c d e f g Chandran SS Klebanoff CA July 2019 T cell receptor based cancer immunotherapy Emerging efficacy and pathways of resistance Immunological Reviews 290 1 127 147 doi 10 1111 imr 12772 PMC 7027847 PMID 31355495 nbsp Material was copied from this source which is available under a Creative Commons Attribution 4 0 International License a b c Dotti G Gottschalk S Savoldo B Brenner MK January 2014 Design and development of therapies using chimeric antigen receptor expressing T cells Immunological Reviews 257 1 107 126 doi 10 1111 imr 12131 PMC 3874724 PMID 24329793 a b Zhang C Liu J Zhong JF Zhang X 2017 06 24 Engineering CAR T cells Biomarker Research 5 22 doi 10 1186 s40364 017 0102 y PMC 5482931 PMID 28652918 Baldo BA May 2015 Chimeric fusion proteins used for therapy indications mechanisms and safety Drug Safety 38 5 455 479 doi 10 1007 s40264 015 0285 9 PMID 25832756 S2CID 23852865 Li N Fu H Hewitt SM Dimitrov DS Ho M August 2017 Therapeutically targeting glypican 2 via single domain antibody based chimeric antigen receptors and immunotoxins in neuroblastoma Proceedings of the National Academy of Sciences of the United States of America 114 32 E6623 E6631 Bibcode 2017PNAS 114E6623L doi 10 1073 pnas 1706055114 PMC 5559039 PMID 28739923 a b c Li Nan Quan Alex Li Dan Pan Jiajia Ren Hua Hoeltzel Gerard de Val Natalia Ashworth Dana Ni Weiming Zhou Jing Mackay Sean Hewitt Stephen M Cachau Raul Ho Mitchell 2023 04 08 The IgG4 hinge with CD28 transmembrane domain improves VHH based CAR T cells targeting a membrane distal epitope of GPC1 in pancreatic cancer Nature Communications 14 1 1986 Bibcode 2023NatCo 14 1986L doi 10 1038 s41467 023 37616 4 ISSN 2041 1723 PMC 10082787 PMID 37031249 a b c Kolluri Aarti Li Dan Li Nan Duan Zhijian Roberts Lewis R Ho Mitchell 2023 02 01 Human VH based chimeric antigen receptor T cells targeting glypican 3 eliminate tumors in preclinical models of HCC Hepatology Communications 7 2 e0022 doi 10 1097 HC9 0000000000000022 ISSN 2471 254X PMC 9851680 PMID 36691969 Li Dan English Hejiao Hong Jessica Liang Tianyuzhou Merlino Glenn Day Chi Ping Ho Mitchell 2022 03 17 A novel PD L1 targeted shark VNAR single domain based CAR T cell strategy for treating breast cancer and liver cancer Molecular Therapy Oncolytics 24 849 863 doi 10 1016 j omto 2022 02 015 ISSN 2372 7705 PMC 8917269 PMID 35317524 Hudecek M Sommermeyer D Kosasih PL Silva Benedict A Liu L Rader C et al February 2015 The nonsignaling extracellular spacer domain of chimeric antigen receptors is decisive for in vivo antitumor activity Cancer Immunology Research 3 2 125 135 doi 10 1158 2326 6066 CIR 14 0127 PMC 4692801 PMID 25212991 Qin L Lai Y Zhao R Wei X Weng J Lai P et al March 2017 Incorporation of a hinge domain improves the expansion of chimeric antigen receptor T cells Journal of Hematology amp Oncology 10 1 68 doi 10 1186 s13045 017 0437 8 PMC 5347831 PMID 28288656 Bridgeman JS Hawkins RE Bagley S Blaylock M Holland M Gilham DE June 2010 The optimal antigen response of chimeric antigen receptors harboring the CD3zeta transmembrane domain is dependent upon incorporation of the receptor into the endogenous TCR CD3 complex Journal of Immunology 184 12 6938 6949 doi 10 4049 jimmunol 0901766 PMID 20483753 Casucci M Bondanza A 2011 Suicide gene therapy to increase the safety of chimeric antigen receptor redirected T lymphocytes Journal of Cancer 2 378 382 doi 10 7150 jca 2 378 PMC 3133962 PMID 21750689 A Cure for Cancer How CAR T Therapy is Revolutionizing Oncology Press release labiotech March 8 2018 Retrieved April 19 2018 Choe JH Williams JZ Lim WA 2020 Engineering T Cells to Treat Cancer The Convergence of Immuno Oncology and Synthetic Biology Annual Review of Cancer Biology 4 121 139 doi 10 1146 annurev cancerbio 030419 033657 Cho JH Collins JJ Wong WW May 2018 Universal Chimeric Antigen Receptors for Multiplexed and Logical Control of T Cell Responses Cell 173 6 1426 1438 e11 doi 10 1016 j cell 2018 03 038 PMC 5984158 PMID 29706540 SMDC technology Archived 2016 03 27 at the Wayback Machine ENDOCYTE Endocyte announces promising preclinical data for application of SMDC technology in CAR T cell therapy in late breaking abstract at American Association for Cancer Research AACR annual meeting 2016 Press release Endocyte April 19 2016 Archived from the original on July 30 2017 Retrieved December 20 2017 Kueberuwa G Kalaitsidou M Cheadle E Hawkins RE Gilham DE March 2018 CD19 CAR T Cells Expressing IL 12 Eradicate Lymphoma in Fully Lymphoreplete Mice through Induction of Host Immunity Molecular Therapy Oncolytics 8 41 51 doi 10 1016 j omto 2017 12 003 PMC 5772011 PMID 29367945 Chmielewski M Abken H 2015 TRUCKs the fourth generation of CARs Expert Opinion on Biological Therapy 15 8 1145 1154 doi 10 1517 14712598 2015 1046430 PMID 25985798 S2CID 42535203 a b c Zhang E Xu H January 2017 A new insight in chimeric antigen receptor engineered T cells for cancer immunotherapy Journal of Hematology amp Oncology 10 1 1 doi 10 1186 s13045 016 0379 6 PMC 5210295 PMID 28049484 Bonini C Ferrari G Verzeletti S Servida P Zappone E Ruggieri L et al June 1997 HSV TK gene transfer into donor lymphocytes for control of allogeneic graft versus leukemia Science 276 5319 1719 1724 doi 10 1126 science 276 5319 1719 PMID 9180086 Quintarelli C Vera JF Savoldo B Giordano Attianese GM Pule M Foster AE et al October 2007 Co expression of cytokine and suicide genes to enhance the activity and safety of tumor specific cytotoxic T lymphocytes Blood 110 8 2793 2802 doi 10 1182 blood 2007 02 072843 PMC 2018664 PMID 17638856 Riddell SR Elliott M Lewinsohn DA Gilbert MJ Wilson L Manley SA et al February 1996 T cell mediated rejection of gene modified HIV specific cytotoxic T lymphocytes in HIV infected patients Nature Medicine 2 2 216 223 doi 10 1038 nm0296 216 PMID 8574968 S2CID 35503876 Maher J Brentjens RJ Gunset G Riviere I Sadelain M January 2002 Human T lymphocyte cytotoxicity and proliferation directed by a single chimeric TCRzeta CD28 receptor Nature Biotechnology 20 1 70 75 doi 10 1038 nbt0102 70 PMID 11753365 S2CID 20302096 Wilkie S van Schalkwyk MC Hobbs S Davies DM van der Stegen SJ Pereira AC et al October 2012 Dual targeting of ErbB2 and MUC1 in breast cancer using chimeric antigen receptors engineered to provide complementary signaling Journal of Clinical Immunology 32 5 1059 1070 doi 10 1007 s10875 012 9689 9 PMID 22526592 S2CID 17660404 Wu CY Roybal KT Puchner EM Onuffer J Lim WA October 2015 Remote control of therapeutic T cells through a small molecule gated chimeric receptor Science 350 6258 aab4077 Bibcode 2015Sci 350 4077W doi 10 1126 science aab4077 PMC 4721629 PMID 26405231 Frankel SR Baeuerle PA June 2013 Targeting T cells to tumor cells using bispecific antibodies Current Opinion in Chemical Biology 17 3 385 392 doi 10 1016 j cbpa 2013 03 029 PMID 23623807 Sun LL Ellerman D Mathieu M Hristopoulos M Chen X Li Y et al May 2015 Anti CD20 CD3 T cell dependent bispecific antibody for the treatment of B cell malignancies Science Translational Medicine 7 287 287ra70 doi 10 1126 scitranslmed aaa4802 PMID 25972002 S2CID 24939667 Kim CH Axup JY Lawson BR Yun H Tardif V Choi SH et al October 2013 Bispecific small molecule antibody conjugate targeting prostate cancer Proceedings of the National Academy of Sciences of the United States of America 110 44 17796 17801 Bibcode 2013PNAS 11017796K doi 10 1073 pnas 1316026110 PMC 3816437 PMID 24127589 a b c Lyman GH Nguyen A Snyder S Gitlin M Chung KC April 2020 Economic Evaluation of Chimeric Antigen Receptor T Cell Therapy by Site of Care Among Patients With Relapsed or Refractory Large B Cell Lymphoma JAMA Network Open 3 4 e202072 doi 10 1001 jamanetworkopen 2020 2072 PMC 7136832 PMID 32250433 Smith Tyrel T Stephan Sirkka B Moffett Howell F McKnight Laura E Ji Weihang Reiman Diana Bonagofski Emmy Wohlfahrt Martin E Pillai Smitha P S Stephan Matthias T 2017 04 17 In situ programming of leukaemia specific T cells using synthetic DNA nanocarriers Nature Nanotechnology 12 8 813 820 doi 10 1038 nnano 2017 57 ISSN 1748 3387 PMC 5646367 PMID 28416815 Agarwal Shiwani Weidner Tatjana Thalheimer Frederic B Buchholz Christian J 2019 10 10 In vivo generated human CAR T cells eradicate tumor cells OncoImmunology 8 12 e1671761 doi 10 1080 2162402x 2019 1671761 ISSN 2162 402X PMC 6844313 PMID 31741773 Agarwalla Pritha Ogunnaike Edikan A Ahn Sarah Froehlich Kristen A Jansson Anton Ligler Frances S Dotti Gianpietro Brudno Yevgeny 2022 03 24 Bioinstructive implantable scaffolds for rapid in vivo manufacture and release of CAR T cells Nature Biotechnology 40 8 1250 1258 doi 10 1038 s41587 022 01245 x ISSN 1087 0156 PMC 9376243 PMID 35332339 Ghassemi Saba Durgin Joseph S Nunez Cruz Selene Patel Jai Leferovich John Pinzone Marilia Shen Feng Cummins Katherine D Plesa Gabriela Cantu Vito Adrian Reddy Shantan Bushman Frederic D Gill Saar I O Doherty Una O Connor Roddy S February 2022 Rapid manufacturing of non activated potent CAR T cells Nature Biomedical Engineering 6 2 118 128 doi 10 1038 s41551 021 00842 6 ISSN 2157 846X PMC 8860360 PMID 35190680 Ledford Heidi 2023 12 20 Cancer fighting CAR T cells could be made inside body with viral injection Nature 625 7994 225 226 doi 10 1038 d41586 023 03969 5 PMID 38129613 Decision Memo for Chimeric Antigen Receptor CAR T cell Therapy for Cancers CAG 00451N www cms gov Retrieved 2021 03 22 CAR T cell Therapy An Update on Coverage and Reimbursement Hematology org www hematology org Archived from the original on 2022 01 24 Retrieved 2021 03 22 Fiorenza S Ritchie DS Ramsey SD Turtle CJ Roth JA September 2020 Value and affordability of CAR T cell therapy in the United States Bone Marrow Transplantation 55 9 1706 1715 doi 10 1038 s41409 020 0956 8 PMID 32474570 S2CID 218987876 Eder M 25 November 2021 Which countries is CAR T cell therapy available in SingleUseSupport Retrieved 13 May 2022 Anvisa aprova produto de terapia avancada para tratamento de cancer Agencia Nacional de Vigilancia Sanitaria Anvisa in Brazilian Portuguese 2022 02 23 Retrieved 2022 06 07 External links editCAR T Cells Engineering Patients Immune Cells to Treat Their Cancers National Cancer Institute July 2019 Retrieved from https en wikipedia org w index php title CAR T cell amp oldid 1221385886, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.