Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Cancer Res Commun ; 4(4): 1100-1110, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38551394

ABSTRACT

PURPOSE: TPST-1120 is a first-in-class oral inhibitor of peroxisome proliferator-activated receptor α (PPARα), a fatty acid ligand-activated transcription factor that regulates genes involved in fatty acid oxidation, angiogenesis, and inflammation, and is a novel target for cancer therapy. TPST-1120 displayed antitumor activity in xenograft models and synergistic tumor reduction in syngeneic tumor models when combined with anti-PD-1 agents. EXPERIMENTAL DESIGN: This phase I, open-label, dose-escalation study (NCT03829436) evaluated TPST-1120 as monotherapy in patients with advanced solid tumors and in combination with nivolumab in patients with renal cell carcinoma (RCC), cholangiocarcinoma (CCA), or hepatocellular carcinoma. Objectives included evaluation of safety, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity (RECIST v1.1). RESULTS: A total of 39 patients enrolled with 38 treated (20 monotherapy, 18 combination; median 3 prior lines of therapy). The most common treatment-related adverse events (TRAE) were grade 1-2 nausea, fatigue, and diarrhea. No grade 4-5 TRAEs or dose-limiting toxicities were reported. In the monotherapy group, 53% (10/19) of evaluable patients had a best objective response of stable disease. In the combination group, 3 patients had partial responses, for an objective response rate of 20% (3/15) across all doses and 30% (3/10) at TPST-1120 ≥400 mg twice daily. Responses occurred in 2 patients with RCC, both of whom had previously progressed on anti-PD-1 therapy, and 1 patient with late-line CCA. CONCLUSIONS: TPST-1120 was well tolerated as monotherapy and in combination with nivolumab and the combination showed preliminary evidence of clinical activity in PD-1 inhibitor refractory and immune compromised cancers. SIGNIFICANCE: TPST-1120 is a first-in-class oral inhibitor of PPARα, whose roles in metabolic and immune regulation are implicated in tumor proliferation/survival and inhibition of anticancer immunity. This first-in-human study of TPST-1120 alone and in combination with nivolumab supports proof-of-concept of PPARα inhibition as a target of therapeutic intervention in solid tumors.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Liver Neoplasms , PPAR alpha , Humans , Carcinoma, Renal Cell/drug therapy , Fatty Acids , Kidney Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Nivolumab/therapeutic use , PPAR alpha/antagonists & inhibitors
2.
Cancer Res Commun ; 3(8): 1486-1500, 2023 08.
Article in English | MEDLINE | ID: mdl-37559947

ABSTRACT

While the role of prostaglandin E2 (PGE2) in promoting malignant progression is well established, how to optimally block the activity of PGE2 signaling remains to be demonstrated. Clinical trials with prostaglandin pathway targeted agents have shown activity but without sufficient significance or dose-limiting toxicities that have prevented approval. PGE2 signals through four receptors (EP1-4) to modulate tumor progression. EP2 and EP4 signaling exacerbates tumor pathology and is immunosuppressive through potentiating cAMP production. EP1 and EP3 signaling has the opposite effect through increasing IP3 and decreasing cAMP. Using available small-molecule antagonists of single EP receptors, the cyclooxygenase-2 (COX-2) inhibitor celecoxib, or a novel dual EP2/EP4 antagonist generated in this investigation, we tested which approach to block PGE2 signaling optimally restored immunologic activity in mouse and human immune cells and antitumor activity in syngeneic, spontaneous, and xenograft tumor models. We found that dual antagonism of EP2 and EP4 together significantly enhanced the activation of PGE2-suppressed mouse and human monocytes and CD8+ T cells in vitro as compared with single EP antagonists. CD8+ T-cell activation was dampened by single EP1 and EP3 antagonists. Dual EP2/EP4 PGE2 receptor antagonists increased tumor microenvironment lymphocyte infiltration and significantly reduced disease burden in multiple tumor models, including in the adenomatous polyposis coli (APC)min+/- spontaneous colorectal tumor model, compared with celecoxib. These results support a hypothesis that redundancy of EP2 and EP4 receptor signaling necessitates a therapeutic strategy of dual blockade of EP2 and EP4. Here we describe TPST-1495, a first-in-class orally available small-molecule dual EP2/EP4 antagonist. Significance: Prostaglandin (PGE2) drives tumor progression but the pathway has not been effectively drugged. We demonstrate significantly enhanced immunologic potency and antitumor activity through blockade of EP2 and EP4 PGE2 receptor signaling together with a single molecule.


Subject(s)
Neoplasms , Prostaglandins , Humans , Animals , Mice , Dinoprostone/metabolism , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Celecoxib/pharmacology , CD8-Positive T-Lymphocytes/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism , Cyclooxygenase 2 Inhibitors , Tumor Microenvironment
3.
Clin Cancer Res ; 25(19): 5787-5798, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31263030

ABSTRACT

PURPOSE: Malignant pleural mesothelioma (MPM) is an aggressive cancer associated with poor prognosis. CRS-207 is a live-attenuated Listeria monocytogenes engineered to express mesothelin, a tumor-associated antigen highly expressed in MPM. CRS-207 induces antitumor immune responses and increases susceptibility of neoplastic cells to immune-mediated killing. PATIENTS AND METHODS: Patients with unresectable MPM, ECOG 0 or 1, and adequate organ and pulmonary function were enrolled in this multicenter, open-label phase Ib study. They received two priming infusions of 1 × 109 CFU CRS-207, followed by pemetrexed/cisplatin chemotherapy, and CRS-207 booster infusions. Primary objectives were safety and induction of immune response. Secondary/exploratory objectives included tumor response, progression-free survival (PFS), overall survival (OS), immune subset analysis, and gene-expression profiling of tumor. RESULTS: Of 35 evaluable patients, 89% (31/35) had disease control with one complete response (3%), 19 partial responses (54%), and 10 stable disease (29%). The estimated median duration of response was 5.0 months (95% CI, 3.9-11.5). The median PFS and OS were 7.5 (95% CI, 7.0-9.9) and 14.7 (95% CI, 11.2-21.9) months, respectively. Tumor size reduction was observed post-CRS-207 infusion prior to chemotherapy in 11 of 35 (31%) patients. No unexpected treatment-related serious adverse events or deaths were observed. IHC analysis of pre- and post-CRS-207 treatment tumor biopsies revealed possible reinvigoration and proliferation of T cells, increased infiltration of dendritic and natural killer cells, increased CD8:Treg ratio, and a shift from immunosuppressive M2-like to proinflammatory M1-like macrophages following CRS-207 administration. CONCLUSIONS: Combination of CRS-207 and chemotherapy induced significant changes in the local tumor microenvironment and objective tumor responses in a majority of treated patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , GPI-Linked Proteins/immunology , Listeria monocytogenes/immunology , Lung Neoplasms/therapy , Mesothelioma/therapy , Pleural Neoplasms/therapy , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/administration & dosage , Combined Modality Therapy , Female , GPI-Linked Proteins/metabolism , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Mesothelin , Mesothelioma/immunology , Mesothelioma/pathology , Mesothelioma, Malignant , Middle Aged , Pemetrexed/administration & dosage , Pleural Neoplasms/immunology , Pleural Neoplasms/pathology , Survival Rate , Tumor Microenvironment
4.
PLoS One ; 10(7): e0133627, 2015.
Article in English | MEDLINE | ID: mdl-26197454

ABSTRACT

While many age-associated immune changes have been reported, a comprehensive set of metrics of immune aging is lacking. Here we report data from 243 healthy adults aged 40-97, for whom we measured clinical and functional parameters, serum cytokines, cytokines and gene expression in stimulated and unstimulated PBMC, PBMC phenotypes, and cytokine-stimulated pSTAT signaling in whole blood. Although highly heterogeneous across individuals, many of these assays revealed trends by age, sex, and CMV status, to greater or lesser degrees. Age, then sex and CMV status, showed the greatest impact on the immune system, as measured by the percentage of assay readouts with significant differences. An elastic net regression model could optimally predict age with 14 analytes from different assays. This reinforces the importance of multivariate analysis for defining a healthy immune system. These data provide a reference for others measuring immune parameters in older people.


Subject(s)
Aging , Immune System/physiology , Leukocytes, Mononuclear/cytology , Adult , Aged , Aged, 80 and over , Area Under Curve , California , Chemokine CCL7/metabolism , Cytokines/metabolism , Female , Gene Expression Profiling , Humans , Immunophenotyping , Male , Middle Aged , Multivariate Analysis , Nitrogen/chemistry , Phenotype , Regression Analysis , Signal Transduction , Surveys and Questionnaires , Urea/chemistry
5.
Arthritis Res Ther ; 17: 127, 2015 May 18.
Article in English | MEDLINE | ID: mdl-25981462

ABSTRACT

The development of biomarkers for autoimmune diseases has been hampered by a lack of understanding of disease etiopathogenesis and of the mechanisms underlying the induction and maintenance of inflammation, which involves complex activation dynamics of diverse cell types. The heterogeneous nature and suboptimal clinical response to treatment observed in many autoimmune syndromes highlight the need to develop improved strategies to predict patient outcome to therapy and personalize patient care. Mass cytometry, using CyTOF®, is an advanced technology that facilitates multiparametric, phenotypic analysis of immune cells at single-cell resolution. In this review, we outline the capabilities of mass cytometry and illustrate the potential of this technology to enhance the discovery of cellular biomarkers for rheumatoid arthritis, a prototypical autoimmune disease.


Subject(s)
Arthritis, Rheumatoid/immunology , Mass Spectrometry/methods , Single-Cell Analysis/methods , Arthritis, Rheumatoid/blood , Biomarkers/analysis , Female , Humans , Male , Sensitivity and Specificity , Severity of Illness Index
6.
Diabetes ; 64(2): 604-17, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25187368

ABSTRACT

Peripheral tolerance is partially controlled by the expression of peripheral tissue antigens (PTAs) in lymph node stromal cells (LNSCs). We previously identified a transcriptional regulator, deformed epidermal autoregulatory factor 1 (Deaf1), that can regulate PTA expression in LNSCs of the pancreatic lymph nodes (PLNs). During the pathogenesis of type 1 diabetes (T1D), Deaf1 is spliced to form the dominant-negative isoform Deaf1-Var1. Here we show that Deaf1-Var1 expression correlates with the severity of disease in NOD mice and is reduced in the PLNs of mice that do not develop hyperglycemia. Inflammation and hyperglycemia independently drive Deaf1 splicing through activation of the splicing factors Srsf10 and Ptbp2, respectively. Inflammation induced by injection of activated splenocytes increased Deaf1-Var1 and Srsf10, but not Ptbp2, in the PLNs of NOD.SCID mice. Hyperglycemia induced by treatment with the insulin receptor agonist S961 increased Deaf1-Var1 and Ptbp2, but not Srsf10, in the PLNs of NOD.B10 and NOD mice. Overexpression of PTBP2 and/or SRSF10 also increased human DEAF1-VAR1 and reduced PTA expression in HEK293T cells. These data suggest that during the progression of T1D, inflammation and hyperglycemia mediate the splicing of DEAF1 and loss of PTA expression in LNSCs by regulating the expression of SRSF10 and PTBP2.


Subject(s)
Alternative Splicing , Diabetes Mellitus, Type 1/metabolism , Hyperglycemia/metabolism , Inflammation/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Aging , Animals , Blood Glucose , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA-Binding Proteins , Diabetes Mellitus, Type 1/genetics , Female , HEK293 Cells , Humans , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Lymph Nodes/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Pancreas/physiology , Pancreatic Elastase/genetics , Pancreatic Elastase/metabolism , Polypyrimidine Tract-Binding Protein/genetics , Polypyrimidine Tract-Binding Protein/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Serine-Arginine Splicing Factors , Spleen/cytology , Transcription Factors/genetics
7.
Diabetes ; 62(12): 4208-19, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24264405

ABSTRACT

Prediabetic NOD mice exhibit hyperglucagonemia, possibly due to an intrinsic α-cell defect. Here, we show that the expression of a potential glucagon inhibitor, the adenosine A1 receptor (Adora1), is gradually diminished in α-cells of NOD mice, autoantibody-positive (AA(+)) and overtly type 1 diabetic (T1D) patients during the progression of disease. We demonstrated that islet inflammation was associated with loss of Adora1 expression through the alternative splicing of Adora1. Expression of the spliced variant (Adora1-Var) was upregulated in the pancreas of 12-week-old NOD versus age-matched NOD.B10 (non-diabetes-susceptible) control mice and was detected in the pancreas of AA(+) patients but not in control subjects or overtly diabetic patients, suggesting that inflammation drives the splicing of Adora1. We subsequently demonstrated that Adora1-Var expression was upregulated in the islets of NOD.B10 mice after exposure to inflammatory cytokines and in the pancreas of NOD.SCID mice after adoptive transfer of activated autologous splenocytes. Adora1-Var encodes a dominant-negative N-terminal truncated isoform of Adora1. The splicing of Adora1 and loss of Adora1 expression on α-cells may explain the hyperglucagonemia observed in prediabetic NOD mice and may contribute to the pathogenesis of human T1D and NOD disease.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Glucagon-Secreting Cells/metabolism , Prediabetic State/metabolism , Receptor, Adenosine A1/metabolism , Adolescent , Adult , Animals , Child , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Female , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred NOD , Middle Aged , Prediabetic State/genetics , Prediabetic State/pathology , Receptor, Adenosine A1/genetics , Spleen/metabolism , Spleen/pathology
8.
FEBS J ; 278(1): 47-58, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21078124

ABSTRACT

GRAIL (gene related to anergy in lymphocytes, also known as RNF128), an ubiquitin-protein ligase (E3), utilizes a unique single transmembrane protein with a split-function motif, and is an important gatekeeper of T-cell unresponsiveness. Although it may play a role in other CD4 T-cell functions including activation, survival and differentiation, GRAIL is most well characterized as a negative regulator of T-cell receptor responsiveness and cytokine production. Here, we review the recent literature on this remarkable E3 in the regulation of human and mouse CD4 T-cell unresponsiveness.


Subject(s)
CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/metabolism , Animals , Autoimmune Diseases/enzymology , Autoimmune Diseases/physiopathology , Cell Cycle/physiology , Humans , Mice
9.
Blood ; 112(1): 120-30, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18323414

ABSTRACT

During thymocyte development, the T-cell receptor (TCR) can discriminate major histocompatibility complex (MHC)/peptide ligands over a narrow range of affinities and translate subtle differences into functional fate decisions. How small differences in TCR input are translated into absolute differences in functional output is unclear. We examined the effects of galectin-1 ablation in the context of class-I-restricted thymocyte development. Galectin-1 expression opposed TCR partial agonist-driven positive selection, but promoted TCR agonist-driven negative selection of conventional CD8(+) T cells. Galectin-1 expression also promoted TCR agonist-driven CD8alphaalpha intestinal intraepithelial lymphocytes (IEL) development. Recombinant galectin-1 enhanced TCR binding to agonist/MHC complexes and promoted a negative-selection-signaling signature, reflected in intensified rapid and transient extracellular signal-regulated kinase (ERK) activation. In contrast, galectin-1 expression antagonized ERK activity in thymocytes undergoing positive selection. We propose that galectin-1 aids in discriminating TCR-directed fate decisions by promoting TCR binding to agonist/MHC complexes and enforcing agonist-driven signals, while opposing partial-agonist signals. In this way, galectin-1 widens the distinction between TCR-directed functional fate cues.


Subject(s)
Galectin 1/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Base Sequence , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , DNA Primers/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Galectin 1/genetics , Galectin 1/metabolism , Galectin 1/pharmacology , Gene Expression , H-Y Antigen/genetics , H-Y Antigen/metabolism , Histocompatibility Antigens Class I/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Recombinant Proteins/pharmacology , T-Lymphocytes/cytology
10.
Ann N Y Acad Sci ; 1103: 45-62, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17376834

ABSTRACT

Type 1 diabetes is a complex, multifactorial disease characterized by T cell-mediated autoimmune destruction of insulin-secreting pancreatic beta cells. To facilitate research in type 1 diabetes, a large-scale dynamic mathematical model of the female non-obese diabetic (NOD) mouse was developed. In this model, termed the Entelos Type 1 Diabetes PhysioLab platform, virtual NOD mice are constructed by mathematically representing components of the immune system and islet beta cell physiology important for the pathogenesis of type 1 diabetes. This report describes the scope of the platform and illustrates some of its capabilities. Specifically, using two virtual NOD mice with either average or early diabetes-onset times, we demonstrate the reproducibility of experimentally observed dynamics involved in diabetes progression, therapeutic responses to exogenous IL-10, and heterogeneity in disease onset. Additionally, we use the Type 1 Diabetes PhysioLab platform to investigate the impact of disease heterogeneity on the effectiveness of exogenous IL-10 therapy to prevent diabetes onset. Results indicate that the inability of a previously published IL-10 therapy protocol to protect NOD mice who exhibit early diabetes onset is due to high levels of pancreatic lymph node (PLN) inflammation, islet infiltration, and beta cell destruction at the time of treatment initiation. Further, simulation indicates that earlier administration of the treatment protocol can prevent NOD mice from developing diabetes by initiating treatment during the period when the disease is still sensitive to IL-10's protective function.


Subject(s)
Diabetes Mellitus, Type 1 , Mice, Inbred NOD , Research Design , User-Computer Interface , Animals , Computer Simulation , Diabetes Mellitus, Type 1/physiopathology , Disease Progression , Humans , Mice , Models, Biological , Physiology/methods
11.
Ann N Y Acad Sci ; 1103: 63-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17376835

ABSTRACT

Several publications describing the use of anti-CD40L monoclonal antibodies (anti-CD40L) for the treatment of type 1 diabetes in non-obese diabetic (NOD) mice have reported different treatment responses to similar protocols. The Entelos Type 1 Diabetes PhysioLab platform, a dynamic large-scale mathematical model of the pathogenesis of type 1 diabetes, was used to study the effects of anti-CD40L therapy in silico. An examination of the impact of pharmacokinetic variability and the heterogeneity of disease progression rate on therapeutic outcome provided insights that could reconcile the apparently conflicting data. Optimal treatment protocols were identified by exploring the dynamics of key pathophysiological pathways.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD40 Ligand/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Animals , Computer Simulation , Drug Administration Schedule , Humans , Mice , Mice, Inbred NOD , Models, Biological
12.
Immunity ; 23(2): 115-26, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16111631

ABSTRACT

Type 1 diabetes (T1D) animal models such as the nonobese diabetic (NOD) mouse have improved our understanding of disease pathophysiology, but many candidate therapeutics identified therein have failed to prevent/cure human disease. We have performed a comprehensive evaluation of disease-modifying agents tested in the NOD mouse based on treatment timing, duration, study length, and efficacy. Interestingly, some popular tenets regarding NOD interventions were not confirmed: all treatments do not prevent disease, treatment dose and timing strongly influence efficacy, and several therapies have successfully treated overtly diabetic mice. The analysis provides a unique perspective on NOD interventions and suggests that the response of this model to therapeutic interventions can be a useful predictor of the human response as long as careful consideration is given to treatment dose, timing, and protocols; more thorough investigation of these parameters should improve clinical translation.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Disease Models, Animal , Animals , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/physiopathology , Humans , Mice , Mice, Inbred NOD
SELECTION OF CITATIONS
SEARCH DETAIL
...