Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Bioorg Med Chem ; 92: 117423, 2023 09 07.
Article in English | MEDLINE | ID: mdl-37531921

ABSTRACT

Hematopoietic progenitor kinase 1 (HPK1) is regarded as a highly validated target in pre-clinical immune oncology. HPK1 has been described as regulating multiple critical signaling pathway in both adaptive and innate cells. In support of this role, HPK1 KO T cells show enhanced sensitivity to TCR activation and HPK1 KO mice display enhanced anti-tumor activity. Taken together, inhibition of HPK1 has the potential to induce enhanced anti-tumor immune response. Herein, we described the discovery of highly potent HPK1 inhibitors starting form a weak HTS hit. Using a structure-based drug design, HPK1 inhibitors exhibiting excellent cellular single-digit nanomolar potency in both proximal (pSLP76) and distal (IL-2) biomarkers along with sustained elevation of IL-2 cytokine secretion were discovered.


Subject(s)
Interleukin-2 , Receptors, Antigen, T-Cell , Mice , Animals , Chlorocebus aethiops , Protein Serine-Threonine Kinases , COS Cells
2.
Front Public Health ; 7: 314, 2019.
Article in English | MEDLINE | ID: mdl-31737593

ABSTRACT

Introduction: The aim of this study was to explore the effectiveness in HbA1c lowering and self-efficacy of diabetes self-management of a 6 months coaching intervention. Methods: This paper was a two-armed coaching intervention study in which 116 participants who presented type 2 diabetes were recruited at a medical center. The intervention group had health coaching and usual care for 6 months, whereas the control had usual care only. The main outcome variables were HbA1c level and self-efficacy of diabetes self-management, in followed-up measure at 3 and 6 months. Results: We found that an approximate 0.68% (CI = 0.40 to 0.96) reduction in HbA1c was achieved after a 6-month health coaching. Both physical activity and self-efficacy of diabetes self-management were shown to benefit by health coaching. Conclusions: Health coaching might be an effective strategy to enhance self-management for diabetes patients in Taiwan where "Diabetes Shared Care Network" had been implemented for over 20 years. Consider limitations of this study, more studies with designs that yield higher quality evidence for the role of health coaching in diabetic patients are needed. Clinical Trial Registration: www.isrctn.com (ID number: ISRCTN52454940, date: 10 May, 2018, retrospectively registered).

3.
Mol Cancer Ther ; 18(2): 245-256, 2019 02.
Article in English | MEDLINE | ID: mdl-30401693

ABSTRACT

Pancreatic cancer has an abysmal 5-year survival rate of 8%, making it a deadly disease with a need for novel therapies. Here we describe a multitargeting heparin-based mimetic, necuparanib, and its antitumor activity in both in vitro and in vivo models of pancreatic cancer. Necuparanib reduced tumor cell proliferation and invasion in a three-dimensional (3D) culture model; in vivo, it extended survival and reduced metastasis. Furthermore, proteomic analysis demonstrated that necuparanib altered the expression levels of multiple proteins involved in cancer-driving pathways including organ development, angiogenesis, proliferation, genomic stability, cellular energetics, and invasion and metastasis. One protein family known to be involved in invasion and metastasis and altered by necuparanib treatment was the matrix metalloprotease (MMP) family. Necuparanib reduced metalloproteinase 1 (MMP1) and increased tissue inhibitor of metalloproteinase 3 (TIMP3) protein levels and was found to increase RNA expression of TIMP3. MMP enzymatic activity was also found to be reduced in the 3D model. Finally, we confirmed necuparanib's in vivo activity by analyzing plasma samples of patients enrolled in a phase I/II study in patients with metastatic pancreatic cancer; treatment with necuparanib plus standard of care significantly increased TIMP3 plasma protein levels. Together, these results demonstrate necuparanib acts as a broad multitargeting therapeutic with in vitro and in vivo anti-invasive and antimetastatic activity.


Subject(s)
Antineoplastic Agents/administration & dosage , Heparitin Sulfate/analogs & derivatives , Matrix Metalloproteinase 1/metabolism , Pancreatic Neoplasms/drug therapy , Tissue Inhibitor of Metalloproteinase-3/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Gene Expression Regulation, Neoplastic/drug effects , Heparitin Sulfate/administration & dosage , Heparitin Sulfate/pharmacology , Humans , Mice , Neoplasm Invasiveness , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Proteomics/methods , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Stromal Cells/drug effects , Tissue Inhibitor of Metalloproteinase-3/genetics , Xenograft Model Antitumor Assays
4.
J Cell Physiol ; 227(11): 3693-700, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22378222

ABSTRACT

Vascular endothelial growth factor A (VEGF-A) is a promoter of neovascularization and thus a popular therapeutic target for diseases involving excessive growth of blood vessels. In this study, we explored the potential of the disaccharide sucrose octasulfate (SOS) to alter VEGF165 diffusion through Descemet's membrane. Descemet's membranes were isolated from bovine eyes and used as a barrier between two chambers of a diffusion apparatus to measure VEGF transport. Diffusion studies revealed a dramatic increase in VEGF165 transport in the presence of SOS, with little diffusion of VEGF165 across the membrane over a 10-h time course in the absence of SOS. Diffusion studies with VEGF121, a non-heparin binding variant of VEGF, showed robust diffusion with or without SOS. To determine a possible mechanism, we measured the ability of SOS to inhibit VEGF interactions with extracellular matrix (ECM), using cell-free and cell surface binding assays. Binding studies showed SOS had no effect on VEGF165 binding to either heparin-coated plates or endothelial cell surfaces at less than mg/ml concentrations. In contrast, we show that SOS inhibited VEGF165 binding to fibronectin in a dose dependent manner and dramatically accelerated the rate of release of VEGF165 from fibronectin. SOS also inhibited the binding of VEGF165 to fibronectin-rich ECM deposited by vascular smooth muscle cells. These results suggest that fibronectin-rich extracellular matrices serve as barriers to VEGF165 diffusion by providing a network of binding sites that can trap and sequester the protein. Since the content of Descemet's membrane is typical of many basement membranes it is possible that they serve throughout the body as formidable barriers to VEGF165 diffusion and tightly regulate its bioavailability and distribution within tissues.


Subject(s)
Descemet Membrane , Facilitated Diffusion/drug effects , Sucrose/analogs & derivatives , Vascular Endothelial Growth Factor A/metabolism , Animals , Cattle , Cells, Cultured , Descemet Membrane/drug effects , Descemet Membrane/metabolism , Diffusion Chambers, Culture , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Fibronectins/chemistry , Fibronectins/metabolism , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Protein Binding/drug effects , Sucrose/chemistry , Sucrose/pharmacology , Vascular Endothelial Growth Factor A/chemistry
5.
PLoS One ; 6(6): e21106, 2011.
Article in English | MEDLINE | ID: mdl-21698156

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) play a key role in shaping the tumor microenvironment by presenting growth factors, cytokines, and other soluble factors that are critical for host cell recruitment and activation, as well as promoting tumor progression, metastasis, and survival. M402 is a rationally engineered, non-cytotoxic heparan sulfate (HS) mimetic, designed to inhibit multiple factors implicated in tumor-host cell interactions, including VEGF, FGF2, SDF-1α, P-selectin, and heparanase. A single s.c. dose of M402 effectively inhibited seeding of B16F10 murine melanoma cells to the lung in an experimental metastasis model. Fluorescent-labeled M402 demonstrated selective accumulation in the primary tumor. Immunohistological analyses of the primary tumor revealed a decrease in microvessel density in M402 treated animals, suggesting anti-angiogenesis to be one of the mechanisms involved in-vivo. M402 treatment also normalized circulating levels of myeloid derived suppressor cells in tumor bearing mice. Chronic administration of M402, alone or in combination with cisplatin or docetaxel, inhibited spontaneous metastasis and prolonged survival in an orthotopic 4T1 murine mammary carcinoma model. These data demonstrate that modulating HSPG biology represents a novel approach to target multiple factors involved in tumor progression and metastasis.


Subject(s)
Disease Progression , Heparitin Sulfate/analogs & derivatives , Heparitin Sulfate/pharmacology , Melanoma, Experimental/pathology , Molecular Mimicry , Neoplasm Metastasis , Animals , Cell Line, Tumor , Flow Cytometry , Melanoma, Experimental/blood supply , Mice , Surface Plasmon Resonance
6.
Ann Biomed Eng ; 36(12): 2134-48, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18839312

ABSTRACT

Growth factor binding to transmembrane protein receptors is generally understood to initiate cell signaling. Receptor binding of heparin-binding growth factors (HB-GFs), such as fibroblast growth factor-2 (FGF-2), is regulated by interactions with heparan sulfate proteoglycans. While there is some specificity for binding to heparan sulfate, overlap in sites for different growth factors may allow for cross regulation. Here we demonstrate, using experiments and computer simulations, that the HB-GFs FGF-2 and heparin-binding EGF-like growth factor (HB-EGF) can cross regulate receptor binding of the other despite having unique receptors. The ability of HSPG to stabilize HB-GF receptor binding is critical for competing growth factors to modulate receptor binding with both enhanced and reduced binding possible depending on this stabilization process. HSPG density and affinity for HB-GF are also critical factors for HB-GF cross regulation. Simulations further reveal that HB-GF can regulate receptor binding of non-HB-GFs such as EGF even when the two proteins share no binding sites when other HB-GF are present within the network. Proliferation studies demonstrate potentiation of HB-EGF-induced growth by FGF-2 indicating that competition networks can alter biological response. Exogenous manipulation of cellular responses to growth factors in complex living systems will require understanding the HSPG-controlled network.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Models, Biological , Myocytes, Smooth Muscle/metabolism , Acid Phosphatase/metabolism , Animals , Aorta/cytology , Cattle , Cell Count , Cells, Cultured , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fibroblast Growth Factor 2/genetics , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Iodine Radioisotopes/metabolism , Myocytes, Smooth Muscle/enzymology , Recombinant Proteins/metabolism
7.
J Cell Physiol ; 215(2): 434-41, 2008 May.
Article in English | MEDLINE | ID: mdl-18163458

ABSTRACT

The antithrombotic activity of heparin has largely been credited with the success found in some cancer treatment by heparin. There are, however, many potent growth factors involved in tumor and blood vessel growth that bind to heparin with high affinity and their regulation by heparin may play a role in heparin's efficacy. We therefore chose to study the activity of a heparin analog, sucrose octasulfate (SOS), which has been similarly shown to interact with heparin-binding growth factors. Using mouse melanoma and lung carcinoma models, we demonstrate in vivo inhibition of tumor growth by SOS. SOS, however, showed little effect in coagulation assays indicating that this activity was not a primary mechanism of action for this molecule. Studies were then performed to assess the effect of SOS on basic fibroblast growth factor (FGF-2) activity, a growth factor which promotes tumor and blood vessel growth and is produced by B16 melanoma cells. SOS potently inhibited FGF-2 binding to endothelial cells and stripped pre-bound FGF-2 from cells. SOS also regulated FGF-2 stimulated proliferation. Further, SOS facilitated FGF-2 diffusion through Descemet's membrane, a heparan sulfate-rich basement membrane from the cornea, suggesting a possible role in FGF-2 clearance. Our results suggest that molecules such as SOS have the potential to remove growth factors from tumor microenvironments and the approach offers an attractive area for further study.


Subject(s)
Carcinoma, Lewis Lung/pathology , Fibroblast Growth Factor 2/metabolism , Melanoma, Experimental/pathology , Sucrose/analogs & derivatives , Animals , Biological Transport/drug effects , Blood Coagulation/drug effects , Capillaries/metabolism , Carcinoma, Lewis Lung/blood supply , Cattle , Cell Division/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Descemet Membrane/metabolism , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/metabolism , Fibroblast Growth Factor 2/antagonists & inhibitors , Male , Melanoma, Experimental/blood supply , Mice , Mice, Inbred C57BL , Sucrose/administration & dosage , Sucrose/pharmacology
8.
Biochemistry ; 44(36): 12203-13, 2005 Sep 13.
Article in English | MEDLINE | ID: mdl-16142919

ABSTRACT

The structural complexity within heparan sulfate has suggested that it contains multiple protein-specific binding sites. To evaluate the selectivity of growth factor binding to heparan sulfate, we conducted a detailed study of the intercompetition of fibroblast growth factor-2 (FGF-2) and heparin-binding epidermal growth factor-like growth factor (HB-EGF) binding to heparan sulfate (HS) on bovine aortic smooth muscle cells. Radioligand binding assays were conducted, and an analytical method was developed for determining the apparent binding constants and numbers of specific and shared binding sites within HS. These studies revealed the presence of two general classes of HS-binding sites for FGF-2 and HB-EGF. The major class (approximately 10(6) sites per cell) was able to bind to either growth factor with relatively low affinity (K(d) = 12 and 44 nM for FGF-2 and HB-EGF, respectively) and was termed "common" binding sites. However, both FGF-2 and HB-EGF also showed specific high affinity (0.6 and 6.1 nM for FGF-2 and HB-EGF, respectively) binding to a minor subset (118,000 and 28,000 sites per cell for FGF-2 and HB-EGF, respectively) of "unique" binding sites, which were unable to bind the other growth factor. These studies indicate that growth factor binding to HS involves multiple binding sites of variable affinity, density, and selectivity. The approach outlined in this study could be applied to aid in the evaluation of the relative biological roles of these selective and nonselective growth factor binding domains within HS.


Subject(s)
Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/metabolism , Binding Sites , Fibroblast Growth Factor 2/genetics , Heparin-binding EGF-like Growth Factor , Humans , Intercellular Signaling Peptides and Proteins , Polysaccharide-Lyases/metabolism , Substrate Specificity
9.
Biochem J ; 379(Pt 2): 331-41, 2004 Apr 15.
Article in English | MEDLINE | ID: mdl-14717658

ABSTRACT

We investigated how lipid raft association of HSPG (heparan sulphate proteoglycans) modulates FGF-2 (fibroblast growth factor-2/basic fibroblast growth factor) interactions with vascular smooth-muscle cells. When lipid rafts were disrupted with sterol-binding agents, methyl-beta-cyclodextrin and filipin, FGF-2 binding to HSPG was reduced 2-5-fold, yet the amount and turnover of cell-surface HSPG were unaffected [corrected]. Approx. 50-65% of bound FGF-2 was in lipid raft-associated fractions based on insolubility in cold Triton X-100 and flotation in OptiPrep density gradients, and this level was increased with higher FGF-2 concentrations [corrected]. Less FGF-2 (50-90%) was associated in raft fractions when cholesterol was depleted or HSPG were degraded with heparinase III. To investigate how lipid raft-HSPG interactions altered binding, we compared the rates of FGF-2 dissociation with native, MbetaCD (methyl-beta-cyclodextrin)- and filipin-treated cells. We found that FGF-2 dissociation rates were increased when lipid rafts were disrupted. These results suggest that localization of HSPG within lipid rafts creates high local concentrations of binding sites such that dissociation of FGF-2 is hindered. The localization of FGF-2 and HSPG to lipid rafts also correlated with the activation of protein kinase Calpha. Thus raft association of HSPG might create growth factor traps resulting in increased binding and signal transduction to enhance cell sensitivity.


Subject(s)
Fibroblast Growth Factor 2/metabolism , Heparan Sulfate Proteoglycans/metabolism , Membrane Microdomains/metabolism , Muscle, Smooth, Vascular/metabolism , beta-Cyclodextrins , Animals , Binding Sites , Cattle , Cyclodextrins/pharmacology , Fibroblast Growth Factor 2/analysis , Fibroblast Growth Factor 2/pharmacology , Filipin/pharmacology , Heparan Sulfate Proteoglycans/analysis , Membrane Microdomains/chemistry , Muscle, Smooth, Vascular/cytology , Protein Kinase C/metabolism , Protein Kinase C-alpha , Proteoglycans/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...