ABSTRACT
CD19-targeted chimeric antigen receptors (CAR) T cells are one of the most remarkable cellular therapies for managing B cell malignancies. However, long-term disease-free survival is still a challenge to overcome. Here, we evaluated the influence of different hinge, transmembrane (TM), and costimulatory CAR domains, as well as manufacturing conditions, cellular product type, doses, patient's age, and tumor types on the clinical outcomes of patients with B cell cancers treated with CD19 CAR T cells. The primary outcome was defined as the best complete response (BCR), and the secondary outcomes were the best objective response (BOR) and 12-month overall survival (OS). The covariates considered were the type of hinge, TM, and costimulatory domains in the CAR, CAR T cell manufacturing conditions, cell population transduced with the CAR, the number of CAR T cell infusions, amount of CAR T cells injected/Kg, CD19 CAR type (name), tumor type, and age. Fifty-six studies (3493 patients) were included in the systematic review and 46 (3421 patients) in the meta-analysis. The overall BCR rate was 56%, with 60% OS and 75% BOR. Younger patients displayed remarkably higher BCR prevalence without differences in OS. The presence of CD28 in the CAR's hinge, TM, and costimulatory domains improved all outcomes evaluated. Doses from one to 4.9 million cells/kg resulted in better clinical outcomes. Our data also suggest that regardless of whether patients have had high objective responses, they might have survival benefits from CD19 CAR T therapy. This meta-analysis is a critical hypothesis-generating instrument, capturing effects in the CD19 CAR T cells literature lacking randomized clinical trials and large observational studies.
Subject(s)
Antigens, CD19 , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Humans , Age Factors , Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/therapy , Leukemia, B-Cell/immunology , Leukemia, B-Cell/mortality , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/mortality , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Treatment OutcomeABSTRACT
Chimeric antigen receptor (CAR) engineering of natural killer (NK) cells has shown promising results in early-phase clinical studies. However, advancing CAR-NK cell therapeutic efficacy is imperative. In this study, we investigated the impact of a fourth-generation CD19-targeted CAR (CAR.19) coexpressing IL-27 on NK-92 cells. We observed a significant improvement in NK-92 cell proliferation and cytotoxicity activity against B-cell cancer cell lines, both in vitro and in a xenograft mouse B-cell lymphoma model. Our systematic transcriptome analysis of the activated NK-92 CAR variants further supports the potential of IL-27 in fourth-generation CARs to overcome limitations of NK cell-based targeted tumor therapies by providing essential growth and activation signals. Integrating IL-27 into CAR-NK cells emerges as a promising strategy to enhance their therapeutic potential and elicit robust responses against cancer cells. These findings contribute substantially to the mounting evidence supporting the potential of fourth-generation CAR engineering in advancing NK cell-based immunotherapies.
Subject(s)
Immunotherapy, Adoptive , Killer Cells, Natural , Receptors, Chimeric Antigen , Xenograft Model Antitumor Assays , Killer Cells, Natural/immunology , Humans , Animals , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Mice , Immunotherapy, Adoptive/methods , Cell Line, Tumor , Antigens, CD19/immunology , Cell Proliferation , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/immunology , Cytotoxicity, ImmunologicABSTRACT
In recent years, cancer has become one of the primary causes of mortality, approximately 10 million deaths worldwide each year. The most advanced, chimeric antigen receptor (CAR) T cell immunotherapy has turned out as a promising treatment for cancer. CAR-T cell therapy involves the genetic modification of T cells obtained from the patient's blood, and infusion back to the patients. CAR-T cell immunotherapy has led to a significant improvement in the remission rates of hematological cancers. CAR-T cell therapy presently limited to hematological cancers, there are ongoing efforts to develop additional CAR constructs such as bispecific CAR, tandem CAR, inhibitory CAR, combined antigens, CRISPR gene-editing, and nanoparticle delivery. With these advancements, CAR-T cell therapy holds promise concerning potential to improve upon traditional cancer treatments such as chemotherapy and radiation while reducing associated toxicities. This review covers recent advances and advantages of CAR-T cell immunotherapy.
Subject(s)
Immunotherapy, Adoptive , Neoplasms , Receptors, Chimeric Antigen , Humans , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/therapeutic use , Receptors, Chimeric Antigen/immunology , Neoplasms/therapy , Neoplasms/immunology , Hematologic Neoplasms/therapy , Gene Editing/methods , T-Lymphocytes/immunology , T-Lymphocytes/transplantationABSTRACT
The complete regression of clear cell renal cell carcinoma (ccRCC) obtained pre-clinically with anti-carbonic anhydrase IX (CAIX) G36 chimeric antigen receptor (CAR) T cells in doses equivalent to â 108 CAR T cells/kg renewed the potential of this target to treat ccRCC and other tumors in hypoxia. The immune checkpoint blockade (ICB) brought durable clinical responses in advanced ccRCC and other tumors. Here, we tested CD8α/4-1BB compared to CD28-based anti-CAIX CAR peripheral blood mononuclear cells (PBMCs) releasing anti-programmed cell death ligand-1 (PD-L1) IgG4 for human ccRCC treatment in vitro and in an orthotopic NSG mice model in vivo. Using a â 107 CAR PBMCs cells/kg dose, anti-CAIX CD28 CAR T cells releasing anti-PD-L1 IgG highly decrease both tumor volume and weight in vivo, avoiding the occurrence of metastasis. This antitumoral superiority of CD28-based CAR PBMCs cells compared to 4-1BB occurred under ICB via PD-L1. Furthermore, the T cell exhaustion status in peripheral CD4 T cells, additionally to CD8, was critical for CAR T cells efficiency. The lack of hepatotoxicity and nephrotoxicity upon the administration of a 107 CAR PMBCs cells/kg dose is the basis for carrying out clinical trials using anti-CAIX CD28 CAR PBMCs cells releasing anti-PD-L1 antibodies or anti-CAIX 4-1BB CAR T cells, offering exciting new prospects for the treatment of refractory ccRCC and hypoxic tumors.
Subject(s)
B7-H1 Antigen , Carbonic Anhydrase IX , Carcinoma, Renal Cell , Kidney Neoplasms , Receptors, Chimeric Antigen , Animals , Antibodies/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD28 Antigens , Carbonic Anhydrase IX/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/therapy , Immune Checkpoint Inhibitors , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Kidney Neoplasms/therapy , Leukocytes, Mononuclear/pathology , Mice , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunologyABSTRACT
Recently, many discoveries have elucidated the cellular and molecular diversity in the leukemic microenvironment and improved our knowledge regarding their complex nature. This has allowed the development of new therapeutic strategies against leukemia. Advances in biotechnology and the current understanding of T cell-engineering have led to new approaches in this fight, thus improving cell-mediated immune response against cancer. However, most of the investigations focus only on conventional cytotoxic cells, while ignoring the potential of unconventional T cells that until now have been little studied. γδ T cells are a unique lymphocyte subpopulation that has an extensive repertoire of tumor sensing and may have new immunotherapeutic applications in a wide range of tumors. The ability to respond regardless of human leukocyte antigen (HLA) expression, the secretion of antitumor mediators and high functional plasticity are hallmarks of γδ T cells, and are ones that make them a promising alternative in the field of cell therapy. Despite this situation, in particular cases, the leukemic microenvironment can adopt strategies to circumvent the antitumor response of these lymphocytes, causing their exhaustion or polarization to a tumor-promoting phenotype. Intervening in this crosstalk can improve their capabilities and clinical applications and can make them key components in new therapeutic antileukemic approaches. In this review, we highlight several characteristics of γδ T cells and their interactions in leukemia. Furthermore, we explore strategies for maximizing their antitumor functions, aiming to illustrate the findings destined for a better mobilization of γδ T cells against the tumor. Finally, we outline our perspectives on their therapeutic applicability and indicate outstanding issues for future basic and clinical leukemia research, in the hope of contributing to the advancement of studies on γδ T cells in cancer immunotherapy.
Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive , Intraepithelial Lymphocytes/drug effects , Intraepithelial Lymphocytes/transplantation , Leukemia/therapy , Animals , Antineoplastic Agents, Immunological/adverse effects , Cell Proliferation/drug effects , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy, Adoptive/adverse effects , Intraepithelial Lymphocytes/immunology , Intraepithelial Lymphocytes/metabolism , Leukemia/genetics , Leukemia/immunology , Leukemia/metabolism , Lymphocyte Activation/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Signal Transduction , Tumor MicroenvironmentABSTRACT
OBJECTIVE: CAR-T cell therapy has revolutionized the treatment of oncological diseases, and potential uses in autoimmune diseases have recently been described. The review aims to integrate the available data on treatment with CAR-T cells, emphasizing autoimmune diseases, to determine therapeutic advances and their possible future clinical applicability in autoimmunity. MATERIALS AND METHODS: A search was performed in PubMed with the keywords "Chimeric Antigen Receptor" and "CART cell". The documents of interest were selected, and a critical review of the information was carried out. RESULTS: In the treatment of autoimmune diseases, in preclinical models, three different cellular strategies have been used, which include Chimeric antigen receptor T cells, Chimeric autoantibody receptor T cells, and Chimeric antigen receptor in regulatory T lymphocytes. All three types of therapy have been effective. The potential adverse effects within them, cytokine release syndrome, cellular toxicity and neurotoxicity must always be kept in mind. CONCLUSIONS: Although information in humans is not yet available, preclinical models of CAR-T cells in the treatment of autoimmune diseases show promising results, so that in the future, they may become a useful and effective therapy in the treatment of these pathologies.
Subject(s)
Autoimmune Diseases/therapy , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/immunology , Animals , HumansABSTRACT
Recently, cell-mediated immune response in malignant neoplasms has become the focus in immunotherapy against cancer. However, in leukemia, most studies on the cytotoxic potential of T cells have concentrated only on T cells that recognize peptide antigens (Ag) presented by polymorphic molecules of the major histocompatibility complex (MHC). This ignores the great potential of unconventional T cell populations, which include gamma-delta T cells (γδ), natural killer T cells (NKT), and mucosal-associated invariant T cells (MAIT). Collectively, these T cell populations can recognize lipid antigens, specially modified peptides and small molecule metabolites, in addition to having several other advantages, which can provide more effective applications in cancer immunotherapy. In recent years, these cell populations have been associated with a repertoire of anti- or protumor responses and play important roles in the dynamics of solid tumors and hematological malignancies, thus, encouraging the development of new investigations in the area. This review focuses on the current knowledge regarding the role of unconventional T cell populations in the antitumor immune response in leukemia and discusses why further studies on the immunotherapeutic potential of these cells are needed.
Subject(s)
Immunotherapy, Adoptive/methods , Intraepithelial Lymphocytes/immunology , Leukemia/therapy , Mucosal-Associated Invariant T Cells/immunology , Natural Killer T-Cells/immunology , Clinical Trials, Phase I as Topic , Humans , Leukemia/immunology , Receptors, Chimeric Antigen/immunologyABSTRACT
PURPOSE: To describe long-term outcomes of anti-CD19 chimeric antigen receptor T (CART) cells in patients with relapsed or refractory chronic lymphocytic leukemia (CLL). METHODS: Between January 2013 and June 2016, 42 patients with relapsed or refractory CLL were enrolled in this study and 38 were infused with anti-CD19 CART cells (CART-19). Of these, 28 patients were initially randomly assigned to receive a low (5 × 107) or high (5 × 108) dose of CART-19, and 24 were evaluable for response assessment. After an interim analysis, 10 additional patients received the selected (high) dose and of these, eight were evaluable for response. Patients were followed for a median 31.5 months (range, 2 to 75 months). RESULTS: At 4 weeks, the complete and overall responses for the 32 evaluable patients were 28% (90% CI, 16% to 44%) and 44% (90% CI, 29% to 60%), respectively. The median overall survival (OS) for all patients was 64 months; there was no statistically significant difference between low- and high-dose groups (P = .84). Regardless of dose, prolonged survival was observed in patients who achieved a CR versus those who did not (P = .035), with median OS not reached in patients with CR versus 64 months in those without CR. The median progression-free survival was 40.2 months in patients with CR and 1 month in those without a CR (P < .0001). Toxicity was comparable in both dose groups. CONCLUSION: In patients with advanced CLL, a 5 × 108 dose of CART-19 may be more effective than 5 × 107 CART-19 at inducing CR without excessive toxicity. Attainment of a CR after CART-19 infusion, regardless of cell dose, is associated with longer OS and progression-free survival in patients with relapsed CLL.
Subject(s)
Immunotherapy, Adoptive/methods , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Aged , Antigens, CD19/immunology , Cytokine Release Syndrome/immunology , Dose-Response Relationship, Immunologic , Female , Humans , Immunotherapy, Adoptive/adverse effects , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Male , Middle Aged , Progression-Free Survival , Receptors, Chimeric Antigen/immunology , Recurrence , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/transplantationABSTRACT
CAR-T cell therapy emerged in the last years as a great promise to cancer treatment. Nowadays, there is a run to improve the breadth of its use, and thus, new chimeric antigen receptors (CAR) are being proposed. The antigen-binding counterpart of CAR is an antibody fragment, scFv (single chain variable fragment), that recognizes a membrane protein associated to a cancer cell. In this chapter, the use of human scFv phage display libraries as a source of new mAbs against surface antigen is discussed. Protocols focusing in the use of extracellular domains of surface protein in biotinylated format are proposed as selection antigen. Elution with unlabeled peptide and selection in solution is described. The analysis of enriched scFvs throughout the selection using NGS is also outlined. Taken together these protocols allow for the isolation of new scFvs able to be useful in the construction of new chimeric antigen receptors for application in cancer therapy.
Subject(s)
Cell Surface Display Techniques , Peptide Library , Receptors, Chimeric Antigen , Single-Chain Antibodies/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/immunology , Immunotherapy, Adoptive/methods , Protein Binding , Protein Interaction Mapping/methods , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/geneticsABSTRACT
Advances in the use of lentiviral vectors for gene therapy applications have created a need for large-scale manufacture of clinical-grade viral vectors for transfer of genetic materials. Lentiviral vectors can transduce a wide range of cell types and integrate into the host genome of dividing and nondividing cells, resulting in long-term expression of the transgene both in vitro and in vivo. In this chapter, we present a method to transfect human cells, creating an easy platform to produce lentiviral vectors for CAR-T cell application.
Subject(s)
Genetic Vectors/biosynthesis , Genetic Vectors/genetics , Immunotherapy, Adoptive , Lentivirus/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Cell Culture Techniques , Cell Line, Tumor , Genes, Reporter , Genetic Vectors/isolation & purification , Humans , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , TransgenesABSTRACT
Chimeric antigen receptor (CAR)-T cell therapy has revolutionized the immunotherapy field with high rate complete responses especially for hematological diseases. Despite the diversity of tumor specific-antigens, the manufacturing process is consistent and involves multiple steps, including selection of T cells, activation, genetic modification, and in vitro expansion. Among these complex manufacturing phases, the choice of culture system to generate a high number of functional cells needs to be evaluated and optimized. Flasks, bags, and rocking motion bioreactor are the most used platforms for CAR-T cell expansion in the current clinical trials but are far from being standardized. New processing options are available and a systematic effort seeking automation, standardization and the increase of production scale, would certainly help to bring the costs down and ultimately democratize this personalized therapy. In this review, we describe different cell expansion platforms available as well as the quality control requirements for clinical-grade production.
Subject(s)
Bioreactors , Cell Culture Techniques , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/metabolism , Cell Culture Techniques/standards , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/standards , Quality Control , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , WorkflowABSTRACT
Myelodysplastic syndrome (MDS) is a group of heterogeneous disorders caused by ineffective hematopoiesis and characterized by bone marrow dysplasia and cytopenia. Current treatment options for MDS are limited to supportive care, hypomethylating agents, and stem cell transplant. Most patients eventually succumb to the disease or progress to leukemia. Previously, we found that CD123 can be used to delineate MDS stem cells in patients at high risk for MDS and that the CD123-positive population is biologically distinct from normal hematopoietic stem cells. Furthermore, selective targeting of MDS stem cells can dramatically reduce tumor burden in preclinical models. On the basis of these findings, we propose CD123 as a candidate target for chimeric antigen receptor (CAR) T-cell therapy in high-risk MDS patients. To test this concept, we employed a CAR lentiviral vector containing a CD123-specific single-chain variable fragment in combination with the CD28 costimulatory domain, CD3ζ signaling domain, and truncated estimated glomerular filtration rate. Utilizing this system, we illustrate that CD123 CAR can be expressed on both healthy donor and MDS patient-derived T lymphocytes with high efficiency, leading to the successful elimination of MDS stem cells both in vitro and in patient-derived xenografts. These results provide the concept for the use of CD123-targeted CAR T cells as a therapeutic option for patients with MDS.
Subject(s)
Immunotherapy, Adoptive , Interleukin-3 Receptor alpha Subunit , Myelodysplastic Syndromes , Receptors, Chimeric Antigen , Animals , Female , Humans , Interleukin-3 Receptor alpha Subunit/genetics , Interleukin-3 Receptor alpha Subunit/immunology , Lentivirus , Male , Mice , Mice, Inbred NOD , Mice, SCID , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/immunology , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/immunologyABSTRACT
A brief review of tumor immunotherapies shows significant advancements in academic research and preclinical studies. Analysis of different immune cell pathways, including macrophage activation, natural killer cells, and dendritic cell presentation show promising clinical results when targeted with different nanoparticle polymer and gold materials. Following a brief discussion on immuno-oncology successes, detailed results are discussed in macrophage activation, dendritic cell presentation, and lysis of tumor cells with natural killer cells. Common targets include tumor-associated macrophages and induction of the proinflammatory phenotype, dual targeting of cell and humoral immunity with dendritic cells, and creating chimeric antigen receptors on natural killer cells. An analysis of the results shows a variety of nanoparticle synthesis methods are required depending on drug type and tissue type affected by tumors. Future research is discussed in conjunction with a brief analysis of completed clinical trial data on cancer therapies using nanoparticles to date. Although paclitaxel-loaded albumin nanoparticles are most frequently studied, academic research shows there may be additional mechanisms of action to elicit anti-tumor activity.
Subject(s)
Immunotherapy/methods , Nanoparticles/therapeutic use , Neoplasms/therapy , Antigens, Neoplasm , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Combined Modality Therapy/methods , Dendritic Cells/immunology , Docetaxel/therapeutic use , Doxorubicin/therapeutic use , Galectin 1/antagonists & inhibitors , Gold/therapeutic use , Humans , Immunity, Cellular , Immunity, Humoral , Killer Cells, Natural/immunology , Lymphocyte Activation , Macrophage Activation , Nanoparticles/administration & dosage , Neoplasms/immunology , Organ Specificity , Paclitaxel/therapeutic use , Receptors, Chimeric Antigen/immunologyABSTRACT
BACKGROUND: We have entered a new era of cancer therapy, with a number of immune-based therapies already used clinically as a standard of care. Adoptive cellular immunotherapy using T cells genetically modified with chimeric antigen receptors (CAR-T cells) represents a novel therapeutic approach. CAR-T cells have produced clinical responses in B-cell malignancies that are otherwise refractory to conventional therapies. Two CAR-T cell therapies obtained regulatory approval in 2017, with many more of these therapies under clinical development. CONTENT: This review focuses on the current state of adoptive cellular immunotherapy, specifically CAR-T cells, in the clinic and how this therapy differs from traditional small molecule and biologic therapies. Areas in which the clinical laboratory is affected by these novel therapies are discussed. Opportunities for the clinical laboratory to help guide these therapies are also highlighted. SUMMARY: The clinical laboratory will play an integral role in the care of patients undergoing adoptive cellular therapy with engineered T cells. There are many ways that this new therapeutic approach affects the clinical laboratory, and the clinical laboratory will likely play a critical role in managing patients that are treated with CAR-T cell therapy.
Subject(s)
Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Cell Transplantation/adverse effects , Cell Transplantation/methods , Humans , Immunotherapy, Adoptive/adverse effects , Neoplasms/immunology , Receptors, Chimeric Antigen/geneticsABSTRACT
Chimeric antigen receptor (CAR)-modified T cells are being investigated in many settings, including classical Hodgkin lymphoma (cHL). The unique biology of cHL, characterized by scant Hodgkin and Reed-Sternberg (HRS) cells within an immunosuppressive tumor microenvironment (TME), may pose challenges for cellular therapies directly targeting antigens expressed on HRS cells. We hypothesized that eradicating CD19+ B cells within the TME and the putative circulating CD19+ HRS clonotypic cells using anti-CD19-directed CAR-modified T cells (CART19) may indirectly affect HRS cells, which do not express CD19. Here we describe our pilot trial using CART19 in patients with relapsed or refractory cHL. To limit potential toxicities, we used nonviral RNA CART19 cells, which are expected to express CAR protein for only a few days, as opposed to CART19 generated by viral vector transduction, which expand in vivo and retain CAR expression. All 5 enrolled patients underwent successful manufacturing of nonviral RNA CART19, and 4 were infused with protocol-specified cell dose. There were no severe toxicities. Responses were seen, but these were transient. To our knowledge, this is the first CART19 clinical trial to use nonviral RNA gene delivery. This trial was registered at www.clinicaltrials.gov as #NCT02277522 (adult) and #NCT02624258 (pediatric).