Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nature ; 623(7989): 1034-1043, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37993715

ABSTRACT

Diet-derived nutrients are inextricably linked to human physiology by providing energy and biosynthetic building blocks and by functioning as regulatory molecules. However, the mechanisms by which circulating nutrients in the human body influence specific physiological processes remain largely unknown. Here we use a blood nutrient compound library-based screening approach to demonstrate that dietary trans-vaccenic acid (TVA) directly promotes effector CD8+ T cell function and anti-tumour immunity in vivo. TVA is the predominant form of trans-fatty acids enriched in human milk, but the human body cannot produce TVA endogenously1. Circulating TVA in humans is mainly from ruminant-derived foods including beef, lamb and dairy products such as milk and butter2,3, but only around 19% or 12% of dietary TVA is converted to rumenic acid by humans or mice, respectively4,5. Mechanistically, TVA inactivates the cell-surface receptor GPR43, an immunomodulatory G protein-coupled receptor activated by its short-chain fatty acid ligands6-8. TVA thus antagonizes the short-chain fatty acid agonists of GPR43, leading to activation of the cAMP-PKA-CREB axis for enhanced CD8+ T cell function. These findings reveal that diet-derived TVA represents a mechanism for host-extrinsic reprogramming of CD8+ T cells as opposed to the intrahost gut microbiota-derived short-chain fatty acids. TVA thus has translational potential for the treatment of tumours.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Oleic Acids , Animals , Cattle , Humans , Mice , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Dairy Products , Fatty Acids, Volatile/pharmacology , Fatty Acids, Volatile/therapeutic use , Milk/chemistry , Neoplasms/diet therapy , Neoplasms/immunology , Oleic Acids/pharmacology , Oleic Acids/therapeutic use , Red Meat , Sheep
2.
Mol Ther ; 30(4): 1381-1395, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35151842

ABSTRACT

T cells genetically engineered to recognize and eliminate tumor cells through synthetic chimeric antigen receptors (CARs) have demonstrated remarkable clinical efficacy against B cell leukemia over the past decade. This therapy is a form of highly personalized medicine that involves genetically modifying a patient's T cells to recognize and kill cancer cells. With the FDA approval of 5 CAR T cell products, this approach has been validated as a powerful new drug in the therapeutic armamentarium against cancer. Researchers are now studying how to expand this technology beyond its use in conventional polyclonal αß T cells to address limitations to the current therapy in cancer and applications beyond it. Considering the specific characteristics of immune cell from diverse lineages, several preclinical and clinical studies are under way to assess the advantages of CAR-redirected function in these cells and apply the lessons learned from CAR T cell therapy in cancer to other diseases.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Genetic Engineering , Humans , Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes
3.
Cancer Treat Res ; 183: 161-184, 2022.
Article in English | MEDLINE | ID: mdl-35551659

ABSTRACT

Genetic modification of T cells to express chimeric antigen receptors (CARs) has yielded remarkable clinical outcomes and initiated a novel era for cancer immunotherapy. The impressive clinical responses seen in hematologic malignancies have led to the investigation of CAR T cells in solid tumors but attaining similar results has been challenging to date. Glioblastoma (GBM) presents a particularly challenging malignancy for treatment and despite some progress in treatments over the past decade, prognosis remains poor for the vast majority of patients. However, recent data support the clinical efficacy and safety of CAR T cell therapy in GBM. In this review, common challenges associated with treating GBM will be discussed in addition to how CAR T cells can overcome such barriers. Additionally, emerging techniques of optimizing CAR T cell therapy for GBM will be emphasized, highlighting the prospective promise of cellular immunotherapy.


Subject(s)
Brain Neoplasms , Glioblastoma , Receptors, Chimeric Antigen , Brain Neoplasms/therapy , Cell- and Tissue-Based Therapy , Glioblastoma/drug therapy , Humans , Immunotherapy, Adoptive/methods , Prospective Studies , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/therapeutic use
4.
J Immunol ; 190(6): 2702-11, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23418626

ABSTRACT

CD20 is a widely validated, B cell-specific target for therapy in B cell malignancies. Rituximab is an anti-CD20 Ab that prolongs survival of chronic lymphocytic leukemia (CLL) patients when combined with chemotherapy. Ofatumumab and GA101 (obinutuzumab) are CD20-directed Abs currently being developed as alternative agents to rituximab in CLL based upon different properties of enhanced direct cell death, NK cell-mediated Ab-dependent cellular cytotoxicity, or complement-dependent cytotoxicity. Despite widespread study, ofatumumab and GA101 have not been compared with each other, nor studied for their interactions with monocytes and macrophages which are critical for the efficacy of anti-CD20 Abs in murine models. In CLL cells, we show that direct cell death and complement-dependent cytotoxicity are greatest with GA101 and ofatumumab, respectively. GA101 promotes enhanced NK cell activation and Ab-dependent cellular cytotoxicity at high Ab concentrations. Ofatumumab elicits superior Ab-dependent cellular phagocytosis with monocyte-derived macrophages. GA101 demonstrated reduced activation of monocytes with diminished pERK, TNF-α release, and FcγRIIa recruitment to lipid rafts. These data demonstrate that GA101 and ofatumumab are both superior to rituximab against CLL cells via different mechanisms of potential tumor elimination. These findings bear relevance to potential combination strategies with each of these anti-CD20 Abs in the treatment of CLL.


Subject(s)
Antibodies, Neoplasm/toxicity , Antigens, CD20/immunology , Drug Delivery Systems/methods , Killer Cells, Natural/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Macrophages/immunology , Monocytes/immunology , Antibodies, Neoplasm/therapeutic use , Antigens, CD20/metabolism , Cell Line, Tumor , Cytotoxicity Tests, Immunologic , Humans , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Macrophages/metabolism , Macrophages/pathology , Monocytes/metabolism , Monocytes/pathology , Tumor Cells, Cultured
5.
Adv Healthc Mater ; 13(14): e2302425, 2024 06.
Article in English | MEDLINE | ID: mdl-38245855

ABSTRACT

Despite the remarkable clinical efficacy of chimeric antigen receptor (CAR) T cells in hematological malignancies, only a subset of patients achieves a durable complete response (dCR). DCR has been correlated with CAR T cell products enriched with T cells memory phenotypes. Therefore, reagents that consistently promote memory phenotypes during the manufacturing of CAR T cells have the potential to significantly improve clinical outcomes. A novel modular multi-cytokine particle (MCP) platform is developed that combines the signals necessary for activation, costimulation, and cytokine support into a single "all-in-one" stimulation reagent for CAR T cell manufacturing. This platform allows for the assembly and screening of compositionally diverse MCP libraries to identify formulations tailored to promote specific phenotypes with a high degree of flexibility. The approach is leveraged to identify unique MCP formulations that manufacture CAR T cell products from diffuse large B cell patients   with increased proportions of memory-like phenotypes MCP-manufactured CAR T cells demonstrate superior anti-tumor efficacy in mouse models of lymphoma and ovarian cancer through enhanced persistence. These findings serve as a proof-of-principle of the powerful utility of the MCP platform to identify "all-in-one" stimulation reagents that can improve the effectiveness of cell therapy products through optimal manufacturing.


Subject(s)
Cytokines , Immunotherapy, Adoptive , Receptors, Chimeric Antigen , Animals , Humans , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Mice , Cytokines/metabolism , Immunotherapy, Adoptive/methods , Female , T-Lymphocytes/immunology , Hematologic Neoplasms/therapy , Hematologic Neoplasms/immunology , Cell Line, Tumor
6.
Res Sq ; 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38496632

ABSTRACT

Radiotherapy (RT) and anti-PD-L1 synergize to enhance local and distant (abscopal) tumor control. However, clinical results in humans have been variable. With the goal of improving clinical outcomes, we investigated the underlying synergistic mechanism focusing on a CD8+ PD-1+ Tcf-1+ stem-like T cell subset in the tumor-draining lymph node (TdLN). Using murine melanoma models, we found that RT + anti-PD-L1 induces a novel differentiation program in the TdLN stem-like population which leads to their expansion and differentiation into effector cells within the tumor. Our data indicate that optimal synergy between RT + anti-PD-L1 is dependent on the TdLN stem-like T cell population as either blockade of TdLN egress or specific stem-like T cell depletion reduced tumor control. Together, these data demonstrate a multistep stimulation of stem-like T cells following combination therapy which is initiated in the TdLN and completed in the tumor.

7.
Cancer Res Commun ; 3(7): 1248-1259, 2023 07.
Article in English | MEDLINE | ID: mdl-37465593

ABSTRACT

Siglec-15 (Sig15) has been implicated as an immune checkpoint expressed in solid tumor-infiltrating macrophages and is being targeted in clinical trials with mAbs to normalize the tumor immune microenvironment and stimulate antitumor immunity. However, the role of Sig15 in hematologic malignancies remains undefined. Sig15 mRNA and protein expression levels in hematologic malignancies were determined from publicly available databases, cell lines, and primary patient samples. Human B-cell acute lymphoblastic leukemia (B-ALL) cell lines were used to identify signaling pathways involved in the regulation of Sig15 expression. Secreted/soluble Sig15 and cytokine levels were measured from the plasma of children with leukemia and healthy controls. Knockdown and knockout of Siglec15 in a murine model of B-ALL was used to evaluate the effect of leukemia-derived Sig15 on the immune response to leukemia. We observed pathologic overexpression of Sig15 in a variety of hematologic malignancies, including primary B-ALL samples. This overexpression was driven by NFκB activation, which also increased the surface localization of Sig15. Secreted/soluble Sig15 was found to circulate at elevated levels in the plasma of children with B-ALL and correlated with an immune-suppressive cytokine milieu. Genetic inhibition of Sig15 in murine B-ALL promoted clearance of the leukemia by the immune system and a marked reversal of the immune-privileged leukemia bone marrow niche, including expanded early effector CD8+ T cells and reduction of immunosuppressive cytokines. Thus, Sig15 is a novel, potent immunosuppressive molecule active in leukemia that may be targeted therapeutically to activate T lymphocytes against leukemia cells. Significance: We demonstrate that Sig15 is overexpressed in hematologic malignancies driven by NFκB, is required for immune evasion in a mouse model of leukemia, and, for the first time, that it circulates at high levels in the plasma of children with leukemia.


Subject(s)
Burkitt Lymphoma , Hematologic Neoplasms , Leukemia , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Animals , Child , Humans , Mice , Adaptive Immunity , CD8-Positive T-Lymphocytes , Cytokines , Immunoglobulins , Membrane Proteins , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Sialic Acid Binding Immunoglobulin-like Lectins , Tumor Microenvironment/genetics
8.
Blood ; 116(7): 1025-34, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20427703

ABSTRACT

Mantle cell lymphoma (MCL) is a mostly incurable malignancy arising from naive B cells (NBCs) in the mantle zone of lymph nodes. We analyzed genomewide methylation in MCL patients with the HELP (HpaII tiny fragment Enrichment by Ligation-mediated PCR) assay and found significant aberrancy in promoter methylation patterns compared with normal NBCs. Using biologic and statistical criteria, we further identified 4 hypermethylated genes CDKN2B, MLF-1, PCDH8, and HOXD8 and 4 hypomethylated genes CD37, HDAC1, NOTCH1, and CDK5 when aberrant methylation was associated with inverse changes in mRNA levels. Immunohistochemical analysis of an independent cohort of MCL patient samples confirmed CD37 surface expression in 93% of patients, validating its selection as a target for MCL therapy. Treatment of MCL cell lines with a small modular immunopharmaceutical (CD37-SMIP) resulted in significant loss of viability in cell lines with intense surface CD37 expression. Treatment of MCL cell lines with the DNA methyltransferase inhibitor decitabine resulted in reversal of aberrant hypermethylation and synergized with the histone deacetylase inhibitor suberoylanilide hydroxamic acid in induction of the hypermethylated genes and anti-MCL cytotoxicity. Our data show prominent and aberrant promoter methylation in MCL and suggest that differentially methylated genes can be targeted for therapeutic benefit in MCL.


Subject(s)
Biomarkers, Tumor/genetics , DNA Methylation , Drug Design , Genome, Human , Lymphoma, Mantle-Cell/genetics , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Cells, Cultured , Drug Discovery , Fluorescent Antibody Technique , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Lymphoma, Mantle-Cell/pathology , Oligonucleotide Array Sequence Analysis , Tetraspanins
10.
Cancer J ; 27(2): 159-167, 2021.
Article in English | MEDLINE | ID: mdl-33750076

ABSTRACT

ABSTRACT: Redirection of T cell cytotoxicity by the chimeric antigen receptor (CAR) structure may not be sufficient for optimal antitumor function in the patient tumor microenvironment. Comodifying CAR T cells to secrete different classes of proteins can be used to optimize CAR T cell function, overcome suppressive signals, and/or alter the tumor microenvironment milieu. These modifications aim to improve initial responses to therapy and enhance the durability of response. Furthermore, CAR T cells can deliver these molecules locally to the tumor microenvironment, avoiding systemic distribution. This approach has been tested in preclinical models using a variety of different classes of agonistic and antagonistic proteins, and clinical trials are currently underway to assess efficacy in patients.


Subject(s)
Immunotherapy, Adoptive , Neoplasms/therapy , Receptors, Chimeric Antigen , Tumor Microenvironment , Humans , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes
11.
Adv Healthc Mater ; 10(15): e2002214, 2021 08.
Article in English | MEDLINE | ID: mdl-33690997

ABSTRACT

Cytokine signaling is critical to a range of biological processes including cell development, tissue repair, aging, and immunity. In addition to acting as key signal mediators of the immune system, cytokines can also serve as potent immunotherapies with more than 20 recombinant products currently Food and Drug Administration (FDA)-approved to treat conditions including hepatitis, multiple sclerosis, arthritis, and various cancers. Yet despite their biological importance and clinical utility, cytokine immunotherapies suffer from intrinsic challenges that limit their therapeutic potential including poor circulation, systemic toxicity, and low tissue- or cell-specificity. In the past decade in particular, methods have been devised to engineer cytokines in order to overcome such challenges and here, the myriad strategies are reviewed that may be employed in order to improve the therapeutic potential of cytokine and chemokine immunotherapies with applications in cancer and autoimmune disease therapy, as well as tissue engineering and regenerative medicine. For clarity, these strategies are collected and presented as they vary across size scales, ranging from single amino acid substitutions, to larger protein-polymer conjugates, nano/micrometer-scale particles, and macroscale implants. Together, this work aims to provide readers with a timely view of the field of cytokine engineering with an emphasis on early-stage therapeutic approaches.


Subject(s)
Autoimmune Diseases , Neoplasms , Autoimmune Diseases/therapy , Cytokines , Humans , Immunologic Factors , Immunotherapy , Neoplasms/therapy
12.
Cancer Immunol Res ; 9(11): 1245-1251, 2021 11.
Article in English | MEDLINE | ID: mdl-34544686

ABSTRACT

Recent success in the use of immunotherapy for a broad range of cancers has propelled the field of cancer immunology to the forefront of cancer research. As more and more young investigators join the community of cancer immunologists, the Arthur L. Irving Family Foundation Cancer Immunology Symposium provided a platform to bring this expanding and vibrant community together and support the development of the future leaders in the field. This commentary outlines the lessons that emerged from the inaugural symposium highlighting the areas of scientific and career development that are essential for professional growth in the field of cancer immunology and beyond. Leading scientists and clinicians in the field provided their experience on the topics of scientific trajectory, career trajectory, publishing, fundraising, leadership, mentoring, and collaboration. Herein, we provide a conceptual and practical framework for career development to the broader scientific community.


Subject(s)
Allergy and Immunology/education , Biomedical Research/methods , Neoplasms/epidemiology , Physicians/organization & administration , Humans , Leadership
13.
Nat Rev Clin Oncol ; 17(3): 147-167, 2020 03.
Article in English | MEDLINE | ID: mdl-31848460

ABSTRACT

T cells genetically engineered to express chimeric antigen receptors (CARs) have proven - and impressive - therapeutic activity in patients with certain subtypes of B cell leukaemia or lymphoma, with promising efficacy also demonstrated in patients with multiple myeloma. Nevertheless, various barriers restrict the efficacy and/or prevent the widespread use of CAR T cell therapies in these patients as well as in those with other cancers, particularly solid tumours. Key challenges relating to CAR T cells include severe toxicities, restricted trafficking to, infiltration into and activation within tumours, suboptimal persistence in vivo, antigen escape and heterogeneity, and manufacturing issues. The evolution of CAR designs beyond the conventional structures will be necessary to address these limitations and to expand the use of CAR T cells to a wider range of malignancies. Investigators are addressing the current obstacles with a wide range of engineering strategies in order to improve the safety, efficacy and applicability of this therapeutic modality. In this Review, we discuss the innovative designs of novel CAR T cell products that are being developed to increase and expand the clinical benefits of these treatments in patients with diverse cancers.


Subject(s)
Cell- and Tissue-Based Therapy , Neoplasms/drug therapy , Receptors, Antigen, T-Cell/therapeutic use , Receptors, Chimeric Antigen/therapeutic use , Cell Engineering , Humans , Lymphoma/drug therapy , Lymphoma/immunology , Neoplasms/genetics , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology
14.
Cancer Immunol Res ; 8(6): 732-742, 2020 06.
Article in English | MEDLINE | ID: mdl-32213625

ABSTRACT

Although clinical responses with CD19-targeting chimeric antigen receptor (CAR) T-cell treatment have been observed in patients with certain hematologic malignancies, high rates of disease relapse highlight the necessity to understand and improve mechanisms of CAR T-cell failure. Because T-cell dysfunction is thought to contribute to CAR T-cell treatment failure, understanding what mechanisms drive T cells into this dysfunctional state may aid optimal design of efficacious CAR T cells. Dysfunctional CAR T cells have been characterized as having upregulated inhibitory receptors and decreased cytolytic capabilities. Previous studies have identified a role for sustained CAR CD3ζ signaling in CAR T-cell dysfunction. Here, we demonstrate a mechanism that drives dysfunction in CAR T cells through excessive costimulation. Fully activated CD19-targeted CAR T cells were rendered dysfunctional upon stimulation with both endogenous CD28 stimulation and CAR-mediated CD28 costimulation. Costimulation-driven dysfunction of CAR T cells was demonstrated in a syngeneic immunocompetent mouse model, in which CAR T cells were activated with signals 1 (CD3ζ), 2 (CD28), and 3 (IL12). Thus, we show that CAR T-cell dysfunction can be driven through excessive CD28 and 4-1BB costimulation.See related article by Drakes et al., p. 743.


Subject(s)
CD28 Antigens/immunology , Immunotherapy, Adoptive/methods , Interleukin-12/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Thymoma/immunology , Thymus Neoplasms/immunology , Animals , Antigens, CD19/immunology , Antigens, CD19/metabolism , Apoptosis , CD28 Antigens/metabolism , Cell Proliferation , Cytokines , Humans , Mice , Mice, Inbred C57BL , Signal Transduction , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Thymoma/metabolism , Thymoma/pathology , Thymus Neoplasms/metabolism , Thymus Neoplasms/pathology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Cancer Immunol Res ; 8(6): 743-755, 2020 06.
Article in English | MEDLINE | ID: mdl-32209638

ABSTRACT

T-cell receptor (TCR)-modified T-cell gene therapy can target a variety of extracellular and intracellular tumor-associated antigens, yet has had little clinical success. A potential explanation for limited antitumor efficacy is a lack of T-cell activation in vivo We postulated that expression of proinflammatory cytokines in TCR-modified T cells would activate T cells and enhance antitumor efficacy. We demonstrate that expression of interleukin 18 (IL18) in tumor-directed TCR-modified T cells provides a superior proinflammatory signal than expression of interleukin 12 (IL12). Tumor-targeted T cells secreting IL18 promote persistent and functional effector T cells and a proinflammatory tumor microenvironment. Together, these effects augmented overall survival of mice in the pmel-1 syngeneic tumor model. When combined with sublethal irradiation, IL18-secreting pmel-1 T cells were able to eradicate tumors, whereas IL12-secreting pmel-1 T cells caused toxicity in mice through excessive cytokine secretion. In another xenograft tumor model, IL18 secretion enhanced the persistence and antitumor efficacy of NY-ESO-1-reactive TCR-modified human T cells as well as overall survival of tumor-bearing mice. These results demonstrate a rationale for optimizing the efficacy of TCR-modified T-cell cancer therapy through expression of IL18.See related commentary by Wijewarnasuriya et al., p. 732.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunotherapy, Adoptive/standards , Interleukin-12/metabolism , Interleukin-18/metabolism , Lymphocyte Activation/immunology , Melanoma/therapy , Receptors, Antigen, T-Cell/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation , Humans , Immunotherapy, Adoptive/methods , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Knockout , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Nat Commun ; 11(1): 6298, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293516

ABSTRACT

Immunosuppressive tumor microenvironment (TME) and ascites-derived spheroids in ovarian cancer (OC) facilitate tumor growth and progression, and also pose major obstacles for cancer therapy. The molecular pathways involved in the OC-TME interactions, how the crosstalk impinges on OC aggression and chemoresistance are not well-characterized. Here, we demonstrate that tumor-derived UBR5, an E3 ligase overexpressed in human OC associated with poor prognosis, is essential for OC progression principally by promoting tumor-associated macrophage recruitment and activation via key chemokines and cytokines. UBR5 is also required to sustain cell-intrinsic ß-catenin-mediated signaling to promote cellular adhesion/colonization and organoid formation by controlling the p53 protein level. OC-specific targeting of UBR5 strongly augments the survival benefit of conventional chemotherapy and immunotherapies. This work provides mechanistic insights into the novel oncogene-like functions of UBR5 in regulating the OC-TME crosstalk and suggests that UBR5 is a potential therapeutic target in OC treatment for modulating the TME and cancer stemness.


Subject(s)
Carcinoma, Ovarian Epithelial/immunology , Macrophages, Peritoneal/immunology , Ovarian Neoplasms/immunology , Peritoneal Neoplasms/immunology , Tumor Escape/immunology , Ubiquitin-Protein Ligases/metabolism , Adult , Aged , Animals , Ascites/genetics , Ascites/immunology , Ascites/pathology , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/secondary , Carcinoma, Ovarian Epithelial/therapy , Cell Line, Tumor/transplantation , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy, Adoptive/methods , Macrophages, Peritoneal/metabolism , Mice , Mice, Knockout , Middle Aged , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Paracrine Communication/immunology , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/secondary , Primary Cell Culture , Prognosis , Receptors, Chimeric Antigen/immunology , Spheroids, Cellular/immunology , Spheroids, Cellular/metabolism , Tumor Escape/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Ubiquitin-Protein Ligases/genetics
17.
Cell Physiol Biochem ; 24(5-6): 369-78, 2009.
Article in English | MEDLINE | ID: mdl-19910677

ABSTRACT

Rab proteins are small GTPases required for vesicle trafficking through the secretory and endocytic pathways. Rab GDP-dissociation inhibitor (rab-GDI) regulates Rab protein function and localization by maintaining Rab proteins in the GDP-bound conformation. Two isoforms of rab-GDI are present in most mammalian cells: GDI-1 and GDI-2. It has recently been demonstrated that a Heat shock protein 90 (Hsp90) chaperone complex regulates the interactions between Rab proteins and Rab-GDI-1. The AR42J cell line is derived from rat pancreatic exocrine tumor cells and develops an acinar-like phenotype when treated with dexamethasone (Dex). The aim of the present study was to examine the expression of rab-GDI isoforms and Hsp90 in AR42J cells in the presence or absence of Dex. Rab-GDI:Hsp90 interactions were also examined. Both rab-GDI isoforms were detected in AR42J cells by immunoblotting. In Dex-treated cells, quantitative immunoblotting revealed that rab-GDI-1 expression increased by 28%, although this change was not statistically significant. Rab-GDI-2 levels were unaltered by Dex treatment. Approximately 21% rab-GDI-1 was membrane associated, whereas rab-GDI-2 was exclusively cytosolic. Dex treatment did not affect the subcellular distribution of rab-GDI isoforms. Hsp90 was present in the cytosolic and membrane fractions of AR42J cells and co-immunoprecipitated with cytosolic rab-GDI-1. Moreover, density gradient centrifugation of AR42J cell membranes revealed that Hsp90 and rab-GDI-1 co-localize on low- and high-density membrane fractions, including amylase-containing secretory granules. The Hsp90 inhibitor, geldanamycin, inhibited CCK-8-induced amylase release from these cells in a dose-dependent manner. Our results indicate that as AR42J cells differentiate into acinar-like cells, rab-GDI isoform expression and localization is not significantly altered. Moreover, our findings suggest that Hsp90 regulates agonist-induced secretion in exocrine cells by interacting with rab-GDI-1.


Subject(s)
Amylases/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , HSP90 Heat-Shock Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Benzoquinones/pharmacology , Cell Line, Tumor , Centrifugation, Density Gradient , Dexamethasone/pharmacology , Guanine Nucleotide Dissociation Inhibitors/analysis , HSP90 Heat-Shock Proteins/analysis , Immunoprecipitation , Lactams, Macrocyclic/pharmacology , Protein Isoforms/analysis , Protein Isoforms/metabolism , Rats , Sincalide/metabolism , rab GTP-Binding Proteins/analysis , rho-Specific Guanine Nucleotide Dissociation Inhibitors
18.
EBioMedicine ; 39: 173-181, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30579863

ABSTRACT

BACKGROUND: Adoptive immunotherapy using T cells expressing chimeric antigen receptors (CARs) targeting CD19 has produced remarkable clinical outcomes. However, much of the mechanisms of action, such as the development of memory responses and sources of immune cytokines, remain elusive largely due to the challenge of characterizing human CAR T cell function in vivo. The lack of a suitable in vivo model also hinders the development of new CAR T cell therapies. METHODS: We established a humanized mouse (hu-mouse) model with a functional human immune system and genetically-matched (autologous) primary acute B-lymphoblastic leukemia (B-ALL) that permits modeling of CD19-targeted CAR T cell therapy in immunocompetent hosts without allogeneic or xenogeneic immune responses. FINDINGS: Anti-CD19 CAR T cells were detected in blood of leukemic hu-mice with kinetics and levels similar to those seen in patients receiving CAR T cell therapy. The levels of CAR T cells were correlated inversely with the burden of leukemia cells and positively with the survival times in anti-CD19 CAR T cell-treated leukemic hu-mice. Infusion of anti-CD19 CAR T cells also resulted in rapid production of T cell- and monocyte/macrophage-derived cytokines and an increase in frequency of regulatory T cells as reported in clinical studies. INTERPRETATION: These results provide a proof-of-principle that this novel preclinical model has the potential to be used to model human CAR T cell therapy and facilitate the design of new CARs with improved antitumor activity.


Subject(s)
Antigens, CD19/immunology , Leukemia, B-Cell/therapy , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/transplantation , Animals , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive , Leukemia, B-Cell/immunology , Mice , T-Lymphocytes/immunology , Treatment Outcome , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Nat Biotechnol ; 36(9): 847-856, 2018 10.
Article in English | MEDLINE | ID: mdl-30102295

ABSTRACT

The efficacy of chimeric antigen receptor (CAR) T cell therapy against poorly responding tumors can be enhanced by administering the cells in combination with immune checkpoint blockade inhibitors. Alternatively, the CAR construct has been engineered to coexpress factors that boost CAR-T cell function in the tumor microenvironment. We modified CAR-T cells to secrete PD-1-blocking single-chain variable fragments (scFv). These scFv-secreting CAR-T cells acted in both a paracrine and autocrine manner to improve the anti-tumor activity of CAR-T cells and bystander tumor-specific T cells in clinically relevant syngeneic and xenogeneic mouse models of PD-L1+ hematologic and solid tumors. The efficacy was similar to or better than that achieved by combination therapy with CAR-T cells and a checkpoint inhibitor. This approach may improve safety, as the secreted scFvs remained localized to the tumor, protecting CAR-T cells from PD-1 inhibition, which could potentially avoid toxicities associated with systemic checkpoint inhibition.


Subject(s)
Programmed Cell Death 1 Receptor/immunology , Receptors, Chimeric Antigen/immunology , Single-Chain Antibodies/immunology , T-Lymphocytes/immunology , Animals , Humans , Mice , Tumor Microenvironment , Xenograft Model Antitumor Assays
20.
Nat Rev Clin Oncol ; 13(6): 370-83, 2016 06.
Article in English | MEDLINE | ID: mdl-27000958

ABSTRACT

The engineered expression of chimeric antigen receptors (CARs) on the surface of T cells enables the redirection of T-cell specificity. Early clinical trials using CAR T cells for the treatment of patients with cancer showed modest results, but the impressive outcomes of several trials of CD19-targeted CAR T cells in the treatment of patients with B-cell malignancies have generated an increased enthusiasm for this approach. Important lessons have been derived from clinical trials of CD19-specific CAR T cells, and ongoing clinical trials are testing CAR designs directed at novel targets involved in haematological and solid malignancies. In this Review, we discuss these trials and present strategies that can increase the antitumour efficacy and safety of CAR T-cell therapy. Given the fast-moving nature of this field, we only discuss studies with direct translational application currently or soon-to-be tested in the clinical setting.


Subject(s)
Antigens, CD19/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , T-Lymphocytes/immunology , Antigens, Neoplasm/immunology , Cell- and Tissue-Based Therapy/methods , Chimera/immunology , Cytokines/metabolism , Humans , Lymphocyte Activation/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , RNA, Messenger/metabolism , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL