Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
Mol Ther ; 31(1): 35-47, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36045585

ABSTRACT

CD19-targeting chimeric antigen receptors (CARs) with CD28 and CD3ζ signaling domains have been approved by the US FDA for treating B cell malignancies. Mutation of immunoreceptor tyrosine-based activation motifs (ITAMs) in CD3ζ generated a single-ITAM containing 1XX CAR, which displayed superior antitumor activity in a leukemia mouse model. Here, we investigated whether the 1XX design could enhance therapeutic potency against solid tumors. We constructed both CD19- and AXL-specific 1XX CARs and compared their in vitro and in vivo functions with their wild-type (WT) counterparts. 1XX CARs showed better antitumor efficacy in both pancreatic and melanoma mouse models. Detailed analysis revealed that 1XX CAR-T cells persisted longer in vivo and had a higher percentage of central memory cells. With fluorescence resonance energy transfer (FRET)-based biosensors, we found that decreased ITAM numbers in 1XX resulted in similar 70-kDa zeta chain-associated protein (ZAP70) activation, while 1XX induced higher Ca2+ elevation and faster extracellular signal-regulated kinase (Erk) activation than WT CAR. Thus, our results confirmed the superiority of 1XX against two targets in different solid tumor models and shed light on the underlying molecular mechanism of CAR signaling, paving the way for the clinical applications of 1XX CARs against solid tumors.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , T-Lymphocytes , Animals , Mice , CD28 Antigens/genetics , Cell Line, Tumor , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/antagonists & inhibitors , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays , Neoplasms/therapy
2.
Science ; 378(6625): 1194-1200, 2022 12 16.
Article in English | MEDLINE | ID: mdl-36480602

ABSTRACT

Chimeric antigen receptor (CAR) costimulatory domains derived from native immune receptors steer the phenotypic output of therapeutic T cells. We constructed a library of CARs containing ~2300 synthetic costimulatory domains, built from combinations of 13 signaling motifs. These CARs promoted diverse human T cell fates, which were sensitive to motif combinations and configurations. Neural networks trained to decode the combinatorial grammar of CAR signaling motifs allowed extraction of key design rules. For example, non-native combinations of motifs that bind tumor necrosis factor receptor-associated factors (TRAFs) and phospholipase C gamma 1 (PLCγ1) enhanced cytotoxicity and stemness associated with effective tumor killing. Thus, libraries built from minimal building blocks of signaling, combined with machine learning, can efficiently guide engineering of receptors with desired phenotypes.


Subject(s)
Machine Learning , Peptide Library , Receptors, Chimeric Antigen , T-Lymphocytes, Cytotoxic , Humans , Phenotype , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/immunology , Signal Transduction , Protein Domains , T-Lymphocytes, Cytotoxic/immunology
3.
Cells ; 11(21)2022 10 26.
Article in English | MEDLINE | ID: mdl-36359781

ABSTRACT

Chimeric antigen receptors (CARs) redirect T cells to recognize a specific target. CAR components play a pivotal role in antigen specificity, structure stability, expression on cell surface, and induction of cellular activation, which together determine the success of CAR T-cell therapy. CAR products targeting B-cell lymphoma encouraged the development of new CAR applications beyond cancer. For example, our group developed a CAR to specifically target glucuronoxylomannan (GXM) in the capsule of Cryptococcus species, called GXMR-CAR or GXMR-IgG4-28ζ. Cryptococcus are fungi that cause the life-threatening disease cryptococcosis, and GXMR-IgG4-28ζ redirected T cells to target yeast and titan cell forms of Cryptococcus spp. Here, we replaced the IgG4-hinge and CD28-transmembrane domains from GXMR-CAR with a CD8α molecule as the hinge/transmembrane and used CD28 or 4-1BB molecules as co-stimulatory domains, creating GXMR-8-28ζ and GXMR-8-BBζ, respectively. Jurkat cells expressing GXMR-CAR containing CD8α as the hinge/transmembrane improved the CAR expression and induced a tonic signaling. GXMR-8-28ζ and GXMR-8-BBζ induced high levels of IL-2 and up-regulation of CD69 expression in the presence of reference strains of C. neoformans and C. gattii. Moreover, GXMR-8-28ζ and GXMR-8-BBζ showed increased strength in response to incubation with clinical isolates of Cryptococcuss spp., and 4-1BB co-stimulatory domain triggered a more pronounced cellular activation. Dasatinib, a tyrosine kinase inhibitor, attenuated the GXMR-CAR signaling cascade's engagement in the presence or absence of its ligand. This study optimized novel second-generation GXMR-CARs containing the CD8-hinge/transmembrane domain that improved CAR expression, antigen recognition, and signal strength in T-cell activation.


Subject(s)
Cryptococcus , Receptors, Antigen, T-Cell , Receptors, Chimeric Antigen , Humans , CD28 Antigens/metabolism , Cryptococcus/immunology , Cryptococcus/metabolism , Immunoglobulin G , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/metabolism , Signal Transduction , Xenograft Model Antitumor Assays , Polysaccharides/chemistry , Polysaccharides/immunology , Cryptococcosis/immunology , Cryptococcosis/therapy
4.
Cancer Immunol Immunother ; 71(1): 165-176, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34046711

ABSTRACT

B7H6, a stress-induced ligand which binds to the NK cell receptor NKp30, has recently emerged as a promising candidate for immunotherapy due to its tumor-specific expression on a broad array of human tumors. NKp30 can function as a chimeric antigen receptor (CAR) extracellular domain but exhibits weak binding with a fast on and off rate to B7H6 compared to the TZ47 anti-B7H6 single-chain variable fragment (scFv). Here, directed evolution using yeast display was employed to isolate novel NKp30 variants that bind to B7H6 with higher affinity compared to the native receptor but retain its fast association and dissociation profile. Two variants, CC3 and CC5, were selected for further characterization and were expressed as soluble Fc-fusion proteins and CARs containing CD28 and CD3ς intracellular domains. We observed that Fc-fusion protein forms of NKp30 and its variants were better able to bind tumor cells expressing low levels of B7H6 than TZ47, and that the novel variants generally exhibited improved in vitro tumor cell killing relative to NKp30. Interestingly, CAR T cells expressing the engineered variants produced unique cytokine signatures in response to multiple tumor types expressing B7H6 compared to both NKp30 and TZ47. These findings suggest that natural CAR receptors can be fine-tuned to produce more desirable signaling outputs while maintaining evolutionary advantages in ligand recognition relative to scFvs.


Subject(s)
B7 Antigens/chemistry , Natural Cytotoxicity Triggering Receptor 3/chemistry , Receptors, Chimeric Antigen/chemistry , Animals , CD28 Antigens/chemistry , CD3 Complex/chemistry , Cell Line, Tumor , Cell Separation , Cytokines/metabolism , Flow Cytometry , Gene Expression Profiling , Gene Library , Genetic Variation , HEK293 Cells , Humans , Immunotherapy , Kinetics , Ligands , Mice , Mutation , Protein Conformation , Single-Chain Antibodies/chemistry
5.
Cell Mol Life Sci ; 79(1): 14, 2021 Dec 29.
Article in English | MEDLINE | ID: mdl-34966954

ABSTRACT

How single-chain variable fragments (scFvs) affect the functions of chimeric antigen receptors (CARs) has not been well studied. Here, the components of CAR with an emphasis on scFv were described, and then several methods to measure scFv affinity were discussed. Next, scFv optimization studies for CD19, CD38, HER2, GD2 or EGFR were overviewed, showing that tuning the affinity of scFv could alleviate the on-target/off-tumor toxicity. The affinities of scFvs for different antigens were also summarized to designate a relatively optimal working range for CAR design. Last, a synthetic biology approach utilizing a low-affinity synthetic Notch (synNotch) receptor to achieve ultrasensitivity of antigen-density discrimination and murine models to assay the on-target/off-tumor toxicity of CARs were highlighted. Thus, this review provides preliminary guidelines of choosing the right scFvs for CARs.


Subject(s)
Immunotherapy, Adoptive , Receptors, Chimeric Antigen/metabolism , Single-Chain Antibodies/metabolism , Animals , Disease Models, Animal , Humans , Neoplasms/immunology , Neoplasms/therapy , Receptors, Chimeric Antigen/chemistry , Single-Chain Antibodies/chemistry , Synthetic Biology
6.
Cells ; 10(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34943898

ABSTRACT

In recent years, cell-based immunotherapies have demonstrated promising results in the treatment of cancer. Chimeric antigen receptors (CARs) arm effector cells with a weapon for targeting tumor antigens, licensing engineered cells to recognize and kill cancer cells. The quality of the CAR-antigen interaction strongly depends on the selected tumor antigen and its expression density on cancer cells. CD19 CAR-engineered T cells approved by the Food and Drug Administration have been most frequently applied in the treatment of hematological malignancies. Clinical challenges in their application primarily include cytokine release syndrome, neurological symptoms, severe inflammatory responses, and/or other off-target effects most likely mediated by cytotoxic T cells. As a consequence, there remains a significant medical need for more potent technology platforms leveraging cell-based approaches with enhanced safety profiles. A promising population that has been advanced is the natural killer (NK) cell, which can also be engineered with CARs. NK cells which belong to the innate arm of the immune system recognize and kill virally infected cells as well as (stressed) cancer cells in a major histocompatibility complex I independent manner. NK cells play an important role in the host's immune defense against cancer due to their specialized lytic mechanisms which include death receptor (i.e., Fas)/death receptor ligand (i.e., Fas ligand) and granzyme B/perforin-mediated apoptosis, and antibody-dependent cellular cytotoxicity, as well as their immunoregulatory potential via cytokine/chemokine release. To develop and implement a highly effective CAR NK cell-based therapy with low side effects, the following three principles which are specifically addressed in this review have to be considered: unique target selection, well-designed CAR, and optimized gene delivery.


Subject(s)
Killer Cells, Natural/immunology , Receptors, Chimeric Antigen/metabolism , Animals , Electroporation , Humans , Microfluidics , Models, Biological , Protein Engineering , Receptors, Chimeric Antigen/chemistry
7.
Cells ; 10(11)2021 10 22.
Article in English | MEDLINE | ID: mdl-34831068

ABSTRACT

Chimeric antigen receptor (CAR) T-cell therapy was envisioned as a mechanism to re-direct effector T-cells to eliminate tumor cells. CARs are composed of the variable region of an antibody that binds a native cancer antigen coupled to the signaling domain of a TCR and co-stimulatory molecules. Its success and approval by the U.S. Food and Drug Administration for the treatment of B-cell malignancies revolutionized the immunotherapy field, leading to extensive research on its possible application for other cancer types. In this review, we will focus on the evolution of CAR-T cell therapy outlining current technologies as well as major obstacles for its wide application. We will highlight achievements, the efforts to increase efficacy and to evolve into an off-the-shelf treatment, and as a possible future treatment for non-cancer related diseases.


Subject(s)
Hematopoietic Stem Cell Transplantation , Receptors, Chimeric Antigen/therapeutic use , Animals , Clinical Trials as Topic , Humans , Immunotherapy, Adoptive , Protein Engineering , Receptors, Chimeric Antigen/chemistry , United States , United States Food and Drug Administration
8.
Nat Commun ; 12(1): 5271, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489470

ABSTRACT

Chimeric antigen receptor (CAR) T cells have emerged as a promising class of therapeutic agents, generating remarkable responses in the clinic for a subset of human cancers. One major challenge precluding the wider implementation of CAR therapy is the paucity of tumor-specific antigens. Here, we describe the development of a CAR targeting the tumor-specific isocitrate dehydrogenase 2 (IDH2) with R140Q mutation presented on the cell surface in complex with a common human leukocyte antigen allele, HLA-B*07:02. Engineering of the hinge domain of the CAR, as well as crystal structure-guided optimization of the IDH2R140Q-HLA-B*07:02-targeting moiety, enhances the sensitivity and specificity of CARs to enable targeting of this HLA-restricted neoantigen. This approach thus holds promise for the development and optimization of immunotherapies specific to other cancer driver mutations that are difficult to target by conventional means.


Subject(s)
HLA-B7 Antigen/chemistry , Isocitrate Dehydrogenase/metabolism , Protein Engineering/methods , Receptors, Chimeric Antigen/chemistry , Animals , Antigens, Neoplasm/metabolism , COS Cells , Cell Line , Chlorocebus aethiops , Epitopes , HLA-B7 Antigen/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/immunology , Mutation , Peptide Library , Protein Conformation , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/physiology
9.
Article in English | MEDLINE | ID: mdl-34592686

ABSTRACT

The proliferation of chimeric antigen receptor (CAR) T cells is closely related to their efficacy, but it is still a great challenge to monitor and quantify CAR T cells in vivo. Based on the high affinity (Kd ≈ 10-15 M) of streptavidin (SA) and biotin, radiolabeled biotin may be used to quantify SA-transduced CAR T cells (SA-CAR T cells). Radio-thin-layer chromatography (radio-TLC) and positron emission tomography (PET) are highly sensitive for trace analysis. Our aim was to develop radio-TLC and PET methods to quantify SA-CAR T cells in vitro and in vivo. First, we developed [68Ga]-DOTA-biotin. Commercially available SA was used as a standard, and quantitative standard curves were established in vitro and in vivo by radio-TLC and PET. Furthermore, the feasibility of the method was verified in Raji model mice. The linear range of radio-TLC was 0.02 âˆ¼ 0.15 pmol/µL with R2 = 0.9993 in vitro. The linear range of PET was 0.02 âˆ¼ 0.76 pmol/µL with R2 = 0.9986 in vivo. SA in CAR T cells can also be accurately quantified in a Raji leukemia model according to PET imaging. The radio-TLC/PET method established in this study is promising for using in the dynamic monitoring and analysis of SA-CAR T cells during therapy.


Subject(s)
Chromatography, Thin Layer/methods , Positron-Emission Tomography/methods , Receptors, Chimeric Antigen/chemistry , Streptavidin/pharmacology , T-Lymphocytes , Animals , Biotin/analogs & derivatives , Female , Mice , Organometallic Compounds , Reproducibility of Results , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
10.
Oncol Rep ; 46(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34296292

ABSTRACT

Melanoma is an aggressive type of cancer originating from the skin that arises from neoplastic changes in melanocytes. Transforming growth factor­ß (TGF­ß) is a pleiotropic cytokine and is known to contribute to melanoma progression by inducing the epithelial­mesenchymal transition (EMT) program and creating an environment that favors tumor progression. There are three TGF­ß isoforms, TGF­ß1, TGF­ß2 and TGF­ß3, all of which engage in pro­tumorigenic activities by activating SMAD signaling pathways. All TGF­ß isoforms activate signaling pathways by binding to their TGF­ß type I (TßRI) and type II (TßRII) receptors. Thus, effective targeting of all TGF­ß isoforms is of great importance. In the present study, chimeric proteins comprising the extracellular domains of TßRI and/or TßRII fused with the Fc portion of human immunoglobulin (IgG) were validated in the melanoma context. The Fc chimeric receptor comprising both TßRI and TßRII (TßRI­TßRII­Fc) effectively trapped all TGF­ß isoforms. Conversely, TßRII­Fc chimeric receptor, that comprises TßRII only, was able to interact with TGF­ß1 and TGF­ß3 isoforms, but not with TGF­ß2, which is a poor prognostic factor for melanoma patients. Accordingly, it was revealed that TßRI­TßRII­Fc chimeric receptor suppressed the EMT program in melanoma cells in vitro induced by any of the three TGF­ß isoforms, as revealed by decreased expression of mesenchymal markers. Conversely, TßRII­Fc chimeric receptor inhibited the EMT program induced by TGF­ß1 and TGF­ß3. In addition, it was established that tumor growth in subcutaneous mouse melanoma was inhibited by TßRI­TßRII­Fc chimeric receptor indicating that Fc chimeric receptor could be applied to modify the tumor microenvironment (TME) of melanoma. Therefore, designing of Fc chimeric receptors targeting TGF­ß signals that affect various components of the TME may result in the development of effective anti­melanoma agents.


Subject(s)
Melanoma/metabolism , Receptors, Fc/metabolism , Skin Neoplasms/metabolism , Transforming Growth Factor beta1/biosynthesis , Animals , Cell Proliferation , Cytokines/metabolism , Disease Progression , Epithelial-Mesenchymal Transition , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Melanoma/pathology , Melanoma, Experimental , Mice , Protein Binding , Protein Isoforms , Receptors, Chimeric Antigen/chemistry , Signal Transduction , Skin Neoplasms/pathology , Smad Proteins/metabolism , Tumor Microenvironment
11.
Front Immunol ; 12: 664329, 2021.
Article in English | MEDLINE | ID: mdl-33981310

ABSTRACT

Chimeric antigen receptors (CARs) or bispecific antibodies (bsAbs) redirected T cell against tumors is one of the most promising immunotherapy approaches. However, insufficient clinical outcomes are still observed in treatments of both solid and non-solid tumors. Limited efficacy and poor persistence are two major challenges in redirected T cell therapies. The immunological synapse (IS) is a vital component during the T cell response, which largely determines the clinical outcomes of T cell-based therapies. Here, we review the structural and signaling characteristics of IS formed by natural T cells and redirected T cells. Furthermore, inspired by the elaborate natural T cell receptor-mediated IS, we provide potential strategies for higher efficacy and longer persistence of redirected T cells.


Subject(s)
Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Antibodies, Bispecific/chemistry , Antibodies, Bispecific/immunology , Antibodies, Bispecific/metabolism , Antigens, Neoplasm/immunology , Humans , Immunological Synapses/immunology , Immunological Synapses/metabolism , Lymphocyte Activation/immunology , Neoplasms/immunology , Neoplasms/therapy , Protein Binding , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/metabolism , Signal Transduction , Structure-Activity Relationship , T-Cell Antigen Receptor Specificity
12.
Science ; 372(6537)2021 04 02.
Article in English | MEDLINE | ID: mdl-33795428

ABSTRACT

T cell exhaustion limits immune responses against cancer and is a major cause of resistance to chimeric antigen receptor (CAR)-T cell therapeutics. Using murine xenograft models and an in vitro model wherein tonic CAR signaling induces hallmark features of exhaustion, we tested the effect of transient cessation of receptor signaling, or rest, on the development and maintenance of exhaustion. Induction of rest through enforced down-regulation of the CAR protein using a drug-regulatable system or treatment with the multikinase inhibitor dasatinib resulted in the acquisition of a memory-like phenotype, global transcriptional and epigenetic reprogramming, and restored antitumor functionality in exhausted CAR-T cells. This work demonstrates that rest can enhance CAR-T cell efficacy by preventing or reversing exhaustion, and it challenges the notion that exhaustion is an epigenetically fixed state.


Subject(s)
Dasatinib/pharmacology , Epigenesis, Genetic , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenome , Female , Hepatocyte Nuclear Factor 1-alpha/metabolism , High Mobility Group Proteins/metabolism , Humans , Immunologic Memory , Lymphocyte Activation , Lymphoid Enhancer-Binding Factor 1/metabolism , Male , Mice , Neoplasms, Experimental/therapy , Protein Domains , Protein Stability , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/immunology , Signal Transduction , T-Lymphocytes/metabolism , Transcription, Genetic , Xenograft Model Antitumor Assays
13.
Int J Mol Sci ; 22(5)2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33804441

ABSTRACT

T cells that are genetically engineered to express chimeric antigen receptor (CAR) have a strong potential to eliminate tumor cells, yet the CAR-T cells may also induce severe side effects due to an excessive immune response. Although optimization of the CAR structure is expected to improve the efficacy and toxicity of CAR-T cells, the relationship between CAR structure and CAR-T cell functions remains unclear. Here, we constructed second-generation CARs incorporating a signal transduction domain (STD) derived from CD3ζ and a 2nd STD derived from CD28, CD278, CD27, CD134, or CD137, and investigated the impact of the STD structure and signaling on CAR-T cell functions. Cytokine secretion of CAR-T cells was enhanced by 2nd STD signaling. T cells expressing CAR with CD278-STD or CD137-STD proliferated in an antigen-independent manner by their STD tonic signaling. CAR-T cells incorporating CD28-STD or CD278-STD between TMD and CD3ζ-STD showed higher cytotoxicity than first-generation CAR or second-generation CARs with other 2nd STDs. The potent cytotoxicity of these CAR-T cells was not affected by inhibiting the 2nd STD signals, but was eliminated by placing the STDs after the CD3ζ-STD. Our data highlighted that CAR activity was affected by STD structure as well as by 2nd STD signaling.


Subject(s)
CD28 Antigens/immunology , Lymphoma, T-Cell/immunology , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , CD28 Antigens/metabolism , Cell Proliferation , Female , Humans , Immunotherapy, Adoptive , Lymphoma, T-Cell/pathology , Lymphoma, T-Cell/therapy , Mice , Mice, Inbred C57BL , Receptors, Chimeric Antigen/genetics , Sequence Homology, Amino Acid , Signal Transduction , Tumor Cells, Cultured
14.
Methods Mol Biol ; 2174: 219-244, 2021.
Article in English | MEDLINE | ID: mdl-32813253

ABSTRACT

The use of immunotherapy as an alternative treatment for cancer patients has become of great interest in the scientific community as it is required to overcome many of the currently unsolved problems such as tumor escape, immunosuppression and unwanted unspecific toxicity. The use of chimeric antigen receptor T cells has been a very successful strategy in some hematologic malignancies. However, the application of CAR T cells has been limited to solid tumors, and this has aimed the development of new generation of CARs with enhanced effectivity and specificity. Here, we review the state of the art of CAR T cell therapy with special emphasis on the current challenges and opportunities.


Subject(s)
Cell Transplantation/adverse effects , Cell Transplantation/methods , Neoplasms/therapy , Receptors, Chimeric Antigen , T-Lymphocytes/physiology , Genes, Transgenic, Suicide , Hematologic Neoplasms/therapy , Humans , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/cytology
15.
Immunotherapy ; 13(2): 177-184, 2021 02.
Article in English | MEDLINE | ID: mdl-33225803

ABSTRACT

Despite the success of antiretroviral therapy in suppressing HIV to an undetectable level in the blood and improving patients' quality of life, HIV persists in antiretroviral therapy-treated patients and threatens their lives. Anti-HIV chimeric antigen receptor (CAR) T cells could offer a cure by recognizing and killing virus-producing cells in an Env-specific manner. In this review, the authors summarize several important aspects of the development of anti-HIV CAR T cells, with a special focus on the evolution of CAR design for enhanced potency and targeting specificity, and also outline the challenges that still need to be addressed to take anti-HIV CAR T cells from a hopeful approach to a real HIV cure.


Subject(s)
Drug Development , HIV Infections/immunology , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , CD4 Antigens/immunology , Drug Development/trends , HIV Infections/therapy , HIV Infections/virology , HIV-1/immunology , Humans , Immune Evasion/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/therapeutic use , T-Lymphocytes/transplantation , T-Lymphocytes/virology , Virus Latency/immunology , env Gene Products, Human Immunodeficiency Virus/immunology
16.
J Biol Chem ; 296: 100116, 2021.
Article in English | MEDLINE | ID: mdl-33234592

ABSTRACT

CAR T cells targeting the B lymphocyte antigen CD19 have led to remarkable clinical results in B cell leukemia and lymphoma but eliminate all B lineage cells, leading to increased susceptibility to severe infections. As malignant B cells will express either immunoglobulin (Ig) light chain κ or λ, we designed a second-generation CAR targeting Igκ, IGK CAR. This construct demonstrated high target specificity but displayed reduced efficacy in the presence of serum IgG. Since CD19 CAR is insensitive to serum IgG, we designed various combinatorial CAR constructs in order to maintain the CD19 CAR T cell efficacy, but with IGK CAR target selectivity. The Kz-19BB design, combining CD19 CAR containing a 4-1BB costimulatory domain with an IGK CAR containing a CD3zeta stimulatory domain, maintained the target specificity of IgK CAR and was resistant to the presence of soluble IgG. Our results demonstrate that a combinatorial CAR approach can improve target selectivity and efficacy.


Subject(s)
Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Antigens, CD19/metabolism , B-Lymphocytes/metabolism , CD28 Antigens/metabolism , Cell Line, Tumor , Humans , Immunotherapy, Adoptive , Lymphoma/metabolism , Receptors, Chimeric Antigen/chemistry , T-Lymphocytes/metabolism
17.
Nat Rev Clin Oncol ; 18(2): 71-84, 2021 02.
Article in English | MEDLINE | ID: mdl-32978608

ABSTRACT

Despite several therapeutic advances over the past decade, multiple myeloma (MM) remains largely incurable, indicating a need for new treatment approaches. Chimeric antigen receptor (CAR) T cell therapy works by mechanisms distinct from those of other MM therapies and involves the modification of patient or donor T cells to target specific cell-surface antigens. B cell maturation antigen (BCMA) is expressed only on plasma cells, a small subset of B cells and MM cells, which makes it a suitable target antigen for such therapies. At the time of writing, data from >20 clinical trials involving anti-BCMA CAR T cells have demonstrated that patients with relapsed and/or refractory MM can achieve objective responses. These early investigations have been instrumental in demonstrating short-term safety and efficacy; however, most patients do not have disease remission lasting >18 months. Attempts to reduce or delay the onset of relapsed disease are underway and include identifying additional CAR T cell target antigens and methods of enhancing BCMA expression on MM cells. Engineering CAR T cells to enhance both the activity and safety of treatment continues to be a promising avenue for improvement. In this Review we summarize data from clinical trials that have been carried out to date, describe novel antigens that could be targeted in the future, and highlight potential future innovations that could enhance the efficacy and/or reduce the toxicities associated with CAR T cell therapies.


Subject(s)
Immunotherapy, Adoptive/methods , Multiple Myeloma/therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , B-Cell Maturation Antigen/immunology , Clinical Trials as Topic , Humans , Immunotherapy, Adoptive/adverse effects , Molecular Targeted Therapy/methods , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Neoplastic Stem Cells/pathology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology
18.
Bioorg Med Chem ; 28(24): 115834, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33166926

ABSTRACT

T-cell engaging bispecific antibodies (T-biAbs) mediate potent and selective cytotoxicity by combining specificities for target and effector cells in one molecule. Chemically programmed T-biAbs (cp-T-biAbs) are precisely assembled compositions of (i) small molecules that govern cancer cell surface targeting with high affinity and specificity and (ii) antibodies that recruit and activate T cells and equip the small molecule with confined biodistribution and longer circulatory half-life. Conceptually similar to cp-T-biAbs, switchable chimeric antigen receptor T cells (sCAR-Ts) can also be put under the control of small molecules by using a chemically programmed antibody as a bispecific adaptor molecule. As such, cp-T-biAbs and cp-sCAR-Ts can endow small molecules with the power of cancer immunotherapy. We here review the concept of chemically programmed antibodies for recruiting and activating T cells as a promising strategy for broadening the utility of small molecules in cancer therapy.


Subject(s)
Antibodies, Bispecific/chemistry , Receptors, Chimeric Antigen/chemistry , T-Lymphocytes, Cytotoxic/chemistry , Antibodies, Bispecific/immunology , Humans , Immunotherapy , Immunotherapy, Adoptive , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Peptides/chemistry , Receptors, Chimeric Antigen/immunology , Small Molecule Libraries/chemistry , T-Lymphocytes, Cytotoxic/immunology
19.
Med Oncol ; 37(11): 100, 2020 Oct 12.
Article in English | MEDLINE | ID: mdl-33047234

ABSTRACT

In the recent years, using genetically modified T cells has been known as a rapid developing therapeutic approach due to the heartwarming results of clinical trials with patients suffering from relapsed or refractory (R/R) hematologic malignancies such as R/R Acute Lymphoblastic Leukemia (R/R ALL). One of these renowned approaches is Chimeric antigen receptors (CARs). CARs are synthetic receptors with the ability to be expressed on the surface of T lymphocytes and are specifically designed to target a tumor-associated antigen (TAA) of interest. CAR-expressing T cells have the capability of proliferating and maintaining their immunological functionality in the recipient body but like any other therapeutic approach, the safety, effectiveness, and specificity enhancement of CAR T cells still lingers in the ambiguity arena. Genetic manipulation methods, expansion protocols, infusion dosage, and conditioning regimens are all among crucial factors which can affect the efficacy of CAR T cell-based cancer therapy. In this article, we discuss the studies that have focused on various aspects that affect the efficacy and persistence of CAR T-cell therapy for ALL treatment and provide a widespread overview regarding the practical approaches capable of elevating the effectiveness and lessening the relative toxicities attributed to it.


Subject(s)
Immunotherapy, Adoptive , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/transplantation , Antigens, CD19/immunology , Antigens, Neoplasm/immunology , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/chemistry , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
20.
Int J Mol Sci ; 21(19)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33050044

ABSTRACT

The impressive success of chimeric antigen receptor (CAR)-T cell therapies in treating advanced B-cell malignancies has spurred a frenzy of activity aimed at developing CAR-T therapies for other cancers, particularly solid tumors, and optimizing engineered T cells for maximum clinical benefit in many different disease contexts. A rapidly growing body of design work is examining every modular component of traditional single-chain CARs as well as expanding out into many new and innovative engineered immunoreceptor designs that depart from this template. New approaches to immune cell and receptor engineering are being reported with rapidly increasing frequency, and many recent high-quality reviews (including one in this special issue) provide comprehensive coverage of the history and current state of the art in CAR-T and related cellular immunotherapies. In this review, we step back to examine our current understanding of the structure-function relationships in natural and engineered lymphocyte-activating receptors, with an eye towards evaluating how well the current-generation CAR designs recapitulate the most desirable features of their natural counterparts. We identify key areas that we believe are under-studied and therefore represent opportunities to further improve our grasp of form and function in natural and engineered receptors and to rationally design better therapeutics.


Subject(s)
Cell Engineering/methods , Immunotherapy, Adoptive/methods , Lymphocyte Activation/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/immunology , Cell Communication/immunology , Humans , Neoplasms/therapy , Protein Domains , Receptors, Chimeric Antigen/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...