Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
Add more filters

Publication year range
1.
Int J Mol Sci ; 25(3)2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38338842

ABSTRACT

Hepatocellular carcinoma (HCC) is a leading global cause of cancer-related mortality. Despite the widespread adoption of sorafenib as the standard HCC treatment, its efficacy is constrained, frequently encountering resistance. To augment the effectiveness of sorafenib, this study investigated the synergy of sorafenib and vinorelbine using 22 HCC patient-derived xenograft (PDX) models. In this study, mice bearing HCC tumors were treated with the vehicle, sorafenib (15 mg/kg), vinorelbine (3 mg/kg), and sorafenib-vinorelbine combination (Sora/Vino). Rigorous monitoring of the tumor growth and side effects coupled with comprehensive histological and molecular analyses was conducted. The overall survival (OS) of mice bearing HCC orthotopic tumors was also assessed. Our data showed a notable 86.4% response rate to Sora/Vino, surpassing rates of 31.8% for sorafenib and 9.1% for vinorelbine monotherapies. Sora/Vino significantly inhibited tumor growth, prolonged OS of mice bearing HCC orthotopic tumors (p < 0.01), attenuated tumor cell proliferation and angiogenesis, and enhanced necrosis and apoptosis. The combination therapy effectively suppressed the focal adhesion kinase (FAK) pathway, which is a pivotal player in cell proliferation, tumor angiogenesis, survival, and metastasis. The noteworthy antitumor activity in 22 HCC PDX models positions Sora/Vino as a promising candidate for early-phase clinical trials, leveraging the established use of sorafenib and vinorelbine in HCC and other cancers.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Sorafenib/pharmacology , Sorafenib/therapeutic use , Carcinoma, Hepatocellular/metabolism , Vinorelbine/pharmacology , Liver Neoplasms/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Cell Line, Tumor , Cell Proliferation , Xenograft Model Antitumor Assays , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Int J Mol Sci ; 25(1)2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38203186

ABSTRACT

Hepatocellular carcinoma (HCC) is a challenging cancer to treat, as traditional chemotherapies have shown limited effectiveness. The mammalian target of rapamycin/sirolimus (mTOR) and microtubules are prominent druggable targets for HCC. In this study, we demonstrated that co-targeting mTOR using mTOR inhibitors (everolimus and sirolimus) along with the microtubule inhibitor vinorelbine yielded results superior to those of the monotherapies in HCC PDX models. Our research showed that the vinorelbine arrests cells at the mitotic phase, induces apoptosis, and normalizes tumor blood vessels but upregulates survivin and activates the mTOR/p70S6K/4EBP1 pathway. The addition of the everolimus significantly improved the tumor response to the vinorelbine, leading to improved overall survival (OS) in most tested orthotopic HCC PDX models. The mechanistic investigation revealed that this marked antitumor effect was accompanied by the downregulations of mTOR targets (p-p70S6K, p-4EBP1, and p-S6K); several key cell-cycle regulators; and the antiapoptotic protein survivin. These effects did not compromise the normalization of the blood vessels observed in response to the vinorelbine in the vinorelbine-sensitive PDX models or to the everolimus in the everolimus-sensitive PDX models. The combination of the everolimus and vinorelbine (everolimus/vinorelbine) also promoted apoptosis with minimal toxicity. Given the cost-effectiveness and established effectiveness of everolimus, and especially sirolimus, this strategy warrants further investigation in early-phase clinical trials.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Everolimus/pharmacology , Carcinoma, Hepatocellular/drug therapy , Vinorelbine/pharmacology , Survivin , Ribosomal Protein S6 Kinases, 70-kDa , Liver Neoplasms/drug therapy , Sirolimus/pharmacology , TOR Serine-Threonine Kinases
3.
Liver Int ; 41(3): 608-620, 2021 03.
Article in English | MEDLINE | ID: mdl-33179425

ABSTRACT

BACKGROUND & AIMS: Infigratinib is a pan-FGFR (fibroblast growth factor receptor) inhibitor that has shown encouraging activity in FGFR-dependent hepatocellular carcinoma (HCC) models. However, long-term treatment results in the emergence of resistant colonies. We sought to understand the mechanisms behind infigratinib-induced tumour cell differentiation and resistance and to explore the potential of adding the CDK4/6 inhibitor ribociclib to prolong cell differentiation. METHODS: Nine high and three low FGFR1-3-expressing HCC patient-derived xenograft (PDX) tumours were subcutaneously implanted into SCID mice and subsequently treated with either infigratinib alone or in combination with ribociclib. Tumour tissues were then subjected to immunohistochemistry to assess cell differentiation, as indicated by the cytoplasmic-to-nuclear ratio and markers such as CYP3A4, HNF4α and albumin. Western blot analyses were performed to investigate the signalling pathways involved. RESULTS: Infigratinib induced cell differentiation in FGFR1-3-dependent HCC PDX models, as indicated by an increase in the cytoplasmic/nuclear ratio and an increase in CYP3A4, HNF4α and albumin. Resistant colonies emerged in long-term treatment, characterised by a reversal of differentiated cell morphology, a reduction in the cytoplasmic-to-nuclear ratio and a loss of differentiation markers. Western blot analyses identified an increase in the CDK4/Cdc2/Rb pathway. The addition of ribociclib effectively blocked this pathway and reversed resistance to infigratinib, resulting in prolonged cell differentiation and growth inhibition. CONCLUSIONS: Our findings demonstrate that the combined inhibition of FGFR/CDK4/6 pathways is highly effective in providing long-lasting tumour growth inhibition and cell differentiation and reducing drug resistance. Therefore, further clinical investigations in patients with FGFR1-3-dependant HCC are warranted.


Subject(s)
Aminopyridines , Carcinoma, Hepatocellular , Liver Neoplasms , Phenylurea Compounds , Purines , Pyrimidines , Aminopyridines/pharmacology , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Humans , Liver Neoplasms/drug therapy , Male , Mice , Mice, SCID , Phenylurea Compounds/pharmacology , Purines/pharmacology , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
4.
Hepatology ; 69(3): 943-958, 2019 03.
Article in English | MEDLINE | ID: mdl-30575985

ABSTRACT

The fibroblast growth factor (FGF) signaling cascade is a key signaling pathway in hepatocarcinogenesis. We report high FGF receptor (FGFR) expression in 17.7% (11 of 62) of hepatocellular carcinoma (HCC) models. Infigratinib, a pan-FGFR inhibitor, potently suppresses the growth of high-FGFR-expressing and sorafenib-resistant HCCs. Infigratinib inhibits FGFR signaling and its downstream targets, cell proliferation, the angiogenic rescue program, hypoxia, invasion, and metastasis. Infigratinib also induces apoptosis and vessel normalization and improves the overall survival of mice bearing FGFR-driven HCCs. Infigratinib acts in synergy with the microtubule-depolymerizing drug vinorelbine to promote apoptosis, suppress tumor growth, and improve the overall survival of mice. Increased expression levels of FGFR-2 and FGFR-3 through gene amplification correlate with treatment response and may serve as potential biomarkers for patient selection. Conclusion: Treatments with Infigratinib alone or in combination with vinorelbine may be effective in a subset of patients with HCC with FGFR-driven tumors.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phenylurea Compounds/therapeutic use , Pyrimidines/therapeutic use , Animals , Blood Vessels/drug effects , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology
5.
Int J Mol Sci ; 21(24)2020 Dec 10.
Article in English | MEDLINE | ID: mdl-33321903

ABSTRACT

The fibroblast growth factor (FGF) signaling cascade is one of the key signaling pathways in hepatocellular carcinoma (HCC). FGF has been shown to augment vascular endothelial growth factor (VEGF)-mediated HCC development and angiogenesis, as well as to potentially lead to resistance to VEGF/VEGF receptor (VEGFR)-targeted agents. Thus, novel agents targeting FGF/FGF receptor (FGFR) signaling may enhance and/or overcome de novo or acquired resistance to VEGF-targeted agents in HCC. Mice bearing high- and low-FGFR tumors were treated with Infigratinib (i.e., a pan-FGFR kinase inhibitor) and/or Bevacizumab (i.e., an angiogenesis inhibitor). The antitumor activity of both agents was assessed individually or in combination. Tumor vasculature, intratumoral hypoxia, and downstream targets of FGFR signaling pathways were also investigated. Infigratinib, when combined with Bevacizumab, exerted a synergistic inhibitory effect on tumor growth, invasion, and lung metastasis, and it significantly improved the overall survival of mice bearing FGFR-dependent HCC. Infigratinib/Bevacizumab promoted apoptosis, inhibited cell proliferation concomitant with upregulation of p27, and reduction in the expression of FGFR2-4, p-FRS-2, p-ERK1/2, p-p70S6K/4EBP1, Cdc25C, survivin, p-Cdc2, and p-Rb. Combining Infigratinib/Bevacizumab may provide therapeutic benefits for a subpopulation of HCC patients with FGFR-dependent tumors. A high level of FGFR-2/3 may serve as a potential biomarker for patient selection to Infigratinib/Bevacizumab.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Phenylurea Compounds/therapeutic use , Pyrimidines/therapeutic use , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Agents, Immunological/administration & dosage , Apoptosis , Bevacizumab/administration & dosage , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , Neoplasm Metastasis , Phenylurea Compounds/administration & dosage , Pyrimidines/administration & dosage , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Survivin/genetics , Survivin/metabolism , Tumor Hypoxia , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
6.
Appl Microbiol Biotechnol ; 102(5): 2191-2201, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29332217

ABSTRACT

Cellulose in plant cell walls is mainly covered by hemicellulose and lignin, and thus efficient removal of these components is thought to be a key step in the optimal utilization of lignocellulose. The recently discovered carbohydrate esterase (CE) 15 family of glucuronoyl esterases (GEs) which cleave the linkages between the free carboxyl group of D-glucuronic acid in hemicellulose and the benzyl groups in lignin residues could contribute to this process. Herein, we report the identification, functional expression, and enzymatic characterization of a GE, AfGE, from the filamentous fungus Aspergillus fumigatus. AfGE was heterologously expressed in Aspergillus oryzae, and the purified enzyme displayed the ability to degrade the synthetic substrates mimicking the ester linkage between hemicellulose and lignin. AfGE is a potentially industrially applicable enzyme due to its characteristic as a thermophilic enzyme with the favorable temperature of 40-50 °C at pH 5. Molecular modeling and site-directed mutagenesis studies of AfGE demonstrated that Lys209 plays an important role in the preference for the substrates containing 4-O-methyl group in the glucopyranose ring.


Subject(s)
Aspergillus fumigatus/enzymology , Esterases/metabolism , Esters/metabolism , Fungal Proteins/chemistry , Aspergillus fumigatus/chemistry , Aspergillus fumigatus/genetics , Aspergillus fumigatus/metabolism , Enzyme Stability , Esterases/chemistry , Esterases/genetics , Esterases/isolation & purification , Esters/chemistry , Fungal Proteins/genetics , Fungal Proteins/isolation & purification , Fungal Proteins/metabolism , Glucuronic Acid/metabolism , Molecular Structure , Polysaccharides/metabolism , Substrate Specificity
7.
Mol Ther ; 23(4): 746-56, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25619723

ABSTRACT

Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related death in the world. The multikinase inhibitor sorafenib only demonstrated marginal improvement in overall survival for advanced disease prompted the search for alternative treatment options. Human mesenchymal stem cells (MSCs) have the ability to home to tumor cells. However, its functional roles on the tumor microenvironment remain controversial. Herein, we showed that conditioned media derived from human fetal MSC (CM-hfMSCs) expressed high level of the insulin growth factor binding proteins IGFBPs and can sequester free insulin-like growth factors (IGFs) to inhibit HCC cell proliferation. The inhibitory effect of IGFBPs on IGF signaling was further evident from the reduction of activated IGF-1R and PI3K/Akt, leading eventually to the induction of cell cycle arrest. We also demonstrated that CM-hfMSCs could enhance the therapeutic efficacy of sorafenib and sunitinib. To the best of our knowledge, this is the first report to show that CM-hfMSCs has a tumor-specific, antiproliferative effect that is not observed with normal human hepatocyte cells and patient-derived matched normal tissues. Our results thus suggest that CM-hfMSCs can provide a useful tool to design alternative/adjuvant treatment strategies for HCC, especially in related function to potentiate the effects of chemotherapeutic drugs.


Subject(s)
Carcinoma, Hepatocellular/pathology , Fetus/cytology , Liver Neoplasms/pathology , Mesenchymal Stem Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction , Carcinoma, Hepatocellular/drug therapy , Cell Proliferation , Culture Media, Conditioned , Gene Knockdown Techniques , Humans , Indoles/therapeutic use , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Niacinamide/therapeutic use , Phenylurea Compounds/therapeutic use , Pyrroles/therapeutic use , Receptor, IGF Type 1/genetics , Sorafenib , Sunitinib
8.
BMC Cancer ; 15: 828, 2015 Oct 31.
Article in English | MEDLINE | ID: mdl-26520397

ABSTRACT

BACKGROUND: Despite advances in therapeutics, outcomes for hepatocellular carcinoma (HCC) remain poor and there is an urgent need for efficacious systemic therapy. Unfortunately, drugs that are successful in preclinical studies often fail in the clinical setting, and we hypothesize that this is due to functional differences between primary tumors and commonly used preclinical models. In this study, we attempt to answer this question by comparing tumor morphology and gene expression profiles between primary tumors, xenografts and HCC cell lines. METHODS: Hep G2 cell lines and tumor cells from patient tumor explants were subcutaneously (ectopically) injected into the flank and orthotopically into liver parenchyma of Mus Musculus SCID mice. The mice were euthanized after two weeks. RNA was extracted from the tumors, and gene expression profiling was performed using the Gene Chip Human Genome U133 Plus 2.0. Principal component analyses (PCA) and construction of dendrograms were conducted using Partek genomics suite. RESULTS: PCA showed that the commonly used HepG2 cell line model and its xenograft counterparts were vastly different from all fresh primary tumors. Expression profiles of primary tumors were also significantly divergent from their counterpart patient-derived xenograft (PDX) models, regardless of the site of implantation. Xenografts from the same primary tumors were more likely to cluster together regardless of site of implantation, although heat maps showed distinct differences in gene expression profiles between orthotopic and ectopic models. CONCLUSIONS: The data presented here challenges the utility of routinely used preclinical models. Models using HepG2 were vastly different from primary tumors and PDXs, suggesting that this is not clinically representative. Surprisingly, site of implantation (orthotopic versus ectopic) resulted in limited impact on gene expression profiles, and in both scenarios xenografts differed significantly from the original primary tumors, challenging the long-held notion that orthotopic PDX model is the gold standard preclinical model for HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Transcriptome , Animals , Cluster Analysis , Computational Biology/methods , Disease Models, Animal , Hep G2 Cells , Heterografts , Humans , Mice
9.
NMR Biomed ; 27(4): 486-94, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24535773

ABSTRACT

Contrary to the common notion that tumor necrotic regions are non-enhancing after contrast administration, recent evidence has shown that necrotic regions exhibit delayed and slow uptake of gadolinium tracer on dynamic contrast-enhanced MRI (DCE MRI). The purpose of this study is to explore whether the mapping of tumor voxels with delayed and slow enhancement on DCE MRI can be used to derive estimates of tumor necrotic fraction. Patient-derived tumor xenograft lines of seven human cancers were implanted in 26 mice which were subjected to DCE MRI performed using a spoiled gradient recalled sequence. Gadolinium tracer concentration was estimated using the variable flip angle technique. To identify tumor voxels exhibiting delayed and slow uptake of contrast medium, clustering analysis was performed using a k-means clustering algorithm that classified tumor voxels according to their contrast enhancement patterns. Comparison of the percentage of tumor voxels exhibiting delayed and slow enhancement with the tumor necrotic fraction estimated on histology showed a strong correlation (r = 0.962, p < 0.001). The mapping of tumor regions with delayed and slow contrast uptake on DCE MRI correlated strongly with tumor necrotic fraction, and can potentially serve as a non-invasive imaging surrogate for the in vivo assessment of necrotic fraction.


Subject(s)
Contrast Media , Magnetic Resonance Imaging , Neoplasms/diagnosis , Xenograft Model Antitumor Assays , Animals , Humans , Male , Mice , Mice, SCID , Necrosis , Neoplasms/pathology , Staining and Labeling
10.
Article in English | MEDLINE | ID: mdl-38604985

ABSTRACT

Challenges such as poor dispersion and insufficient polarization of BaTiO3 (BTO) nanoparticles (NPs) within poly(vinylidene fluoride-co-trifluoroethylene) (P(VDF-TrFE)) composites have hindered their piezoelectricity, limiting their uses in pressure sensors, nanogenerators, and artificial sensory synapses. Here, we introduce a high-performance piezoelectric nanocomposite material consisting of P(VDF-TrFE)/modified-BTO (mBTO) NPs for use as a self-activating component in a piezotronic artificial mechanoreceptor. To generate high-performance piezoelectric nanocomposite materials, the surface of BTO is hydroxylated, followed by the covalent attachment of (3-aminopropyl)triethoxysilane to improve the dispersibility of mBTO NPs within the P(VDF-TrFE) matrix. We also aim to enhance the crystallization degree of P(VDF-TrFE), the efficiency characteristics of mBTO, and the poling efficiency, even when incorporating small amounts of mBTO NPs. The piezoelectric potential mechanically induced from the P(VDF-TrFE)/mBTO NPs nanocomposite was three times greater than that from P(VDF-TrFE) and twice as high as that from the P(VDF-TrFE)/BTO NPs nanocomposite. The piezoelectric potential generated by mechanical stimuli on the piezoelectric nanocomposite was utilized to activate the synaptic ionogel-gated field-effect transistor for the development of self-powered piezotronics artificial mechanoreceptors on a polyimide substrate. The device successfully emulated fast-adapting (FA) functions found in biological FA mechanoreceptors. This approach has great potential for applications to future intelligent tactile perception technology.

11.
Langmuir ; 29(37): 11615-22, 2013 Sep 17.
Article in English | MEDLINE | ID: mdl-23924057

ABSTRACT

The rupture of a liquid bridge has many applications while the rupture of a gaseous bridge is gaining importance in the use of bubbles to affect the speed of liquid flow over surfaces. Here, comparative experiments were conducted for liquid and gaseous bridges dispensed at fixed volumes of 6 µL on silicone (hydrophobic) and silane coated glass (hydrophilic) surfaces and with the dispensing tip retracted at different speeds. With the liquid bridge, increasing the retracting speed left behind lower volumes on the substrate. The pinch off position and the contact line radius were factors that determined the volume. The bridge first entered into a receding state before being able to restore toward equilibrium in a relaxation process closer to rupture. On silicone the contact angle was able to undergo higher degrees of hysteresis with faster tip retraction speeds due to the lower free surface energy. With gaseous bridges, only a very small volume was left behind on the silane coated glass while the volume deposited on silicone could be tuned from almost none at low retraction speeds to virtually the entire gaseous volume bridge at high retraction speeds. The tip and neck distances from the substrate increased with tip speed until 0.5 mm/s on the silicone surface, but, beyond that, the position remained invariant until rupture. With the progress toward rupture for the gaseous bridge, the contact angle advanced rather than receded and there was no relaxation stage that brought the contact angle back toward equilibrium before rupture. Overall the gaseous bridges responded very differently to tip retraction than the liquid bridges.

12.
J Multidiscip Healthc ; 16: 101-117, 2023.
Article in English | MEDLINE | ID: mdl-36660039

ABSTRACT

Purpose: A total of 537 million suffered from diabetes mellitus in 2021, and the aging of the population will not abate this number in the future. Diabetes predisposes people to ailments and doubles the risk of COVID-19 mortality. mHealth has shown promise to help manage diabetes. The aim of this review is to objectively analyze research from the last 2.5 years to assess effectiveness where mHealth has been used as an intervention to help manage diabetes in older patients. We also analyzed patient satisfaction, quality, and barriers to adoption of mHealth to manage diabetes. Patients and Methods: No human subjects were involved in this review. We queried four research databases for mHealth to manage diabetes in older adults. We conducted the review based on the Kruse Protocol for writing as systematic review and we reported our findings in accordance with PRISMA (2020). Results: Thirty research articles from 11 countries were analyzed. Five interventions of mHealth were identified. Of these mHealth Short Message service (SMS) helped change behavior and encouraged self-care. mHealth SMS coupled with telemedicine for coaching showed positive effects on weight loss, BMI, diet, exercise, HbA1C, disease awareness, blood pressure, cholesterol, medication adherence, and foot care. Conclusion: mHealth SMS coupled with telemedicine for coaching shows the greatest promise for educating, changing behavior, and realizing positive outcomes across a broad spectrum of health factors. The largest drawback is the cost of acquiring equipment and training users.

13.
Angiogenesis ; 15(1): 59-70, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22187171

ABSTRACT

PURPOSE OF STUDY: Hepatocellular carcinoma (HCC) is the third leading cause of cancer death. Although sorafenib has been shown to improve survival of patients with advanced HCC, this improvement is modest and patients eventually have refractory disease. The purpose of this study is to assess the anti-tumor and anti-angiogenic activities of foretinib, a vascular endothelial growth factor receptor 2 (VEGFR-2) and c-Met inhibitor using mouse models of human HCC. EXPERIMENTAL TECHNIQUES: SK-HEP1 and 21-0208 HCC cells as well as patient-derived HCC models were employed to study the anti-tumor and antiangiogenic activities of foretinib. Changes of biomarkers relevant to hepatocyte growth factor (HGF) signaling pathways were determined by Western blotting. Microvessel density, apoptosis and cell proliferation were analyzed by immunohistochemistry. RESULTS: Treatment of SK-HEP1 cells with foretinib resulted in growth inhibition, G2/M cell cycle arrest, reduced colony formation and blockade of HGF-induced cell migration. In both orthotopic and ectopic models of HCC, foretinib potently inhibited tumor growth in a dose-dependent manner. Inhibition of angiogenesis correlated with inactivation of VEGFR-2/c-Met signaling pathways. Foretinib also caused elevation of p27 and Bim but reduced cyclin B1 expression and p-c-Myc, which resulted in a reduction in cellular proliferation and the induction of tumor cell apoptosis. In an orthotopic model, foretinib potently inhibited primary tumor growth and significantly prolonged mouse survival. DATA INTERPRETATIONS: Foretinib demonstrated significant antitumor activities in patient-derived HCC xenograft models. This study provides a compelling rationale for clinical investigation in patients with advanced HCC.


Subject(s)
Anilides/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Quinolines/therapeutic use , Xenograft Model Antitumor Assays , Anilides/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Quinolines/pharmacology , Survival Analysis , Time Factors
14.
J Hepatol ; 56(3): 595-601, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22027573

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is the third leading cause of cancer death. Although sorafenib has been shown to improve survival of patients with advanced HCC, this improvement is modest and patients eventually have refractory disease. This study aims at investigating the antitumor, antiangiogenesis and antimetastatic activities of dovitinib in preclinical models of HCC. METHODS: 21-0208 and SK-HEP1 cells as well as patient-derived HCC models were employed to study the antitumor effect of dovitinib. Changes of biomarkers relevant to FGFR/VEGFR/PDGFR pathways were determined by Western blotting. Microvessel density, apoptosis and cell proliferation were analyzed by immunohistochemistry. RESULTS: Treatment of SK-HEP1 cells with dovitinib resulted in G2/M cell cycle arrest, inhibition of colony formation in soft agar and blockade of bFGF-induced cell migration. Dovitinib inhibited basal expression and FGF-induced phosphorylation of FGFR-1, FRS2-α and ERK1/2. In vivo, dovitinib potently inhibited tumor growth of six HCC lines. Inhibition of angiogenesis correlated with inactivation of FGFR/PDGFR-ß/VEGFR-2 signaling pathways. Dovitinib also caused dephosphorylation of retinoblastoma, upregulation of p-histone H2A-X and p27, and downregulation of p-cdk-2 and cyclin B1, which resulted in a reduction in cellular proliferation and the induction of tumor cell apoptosis. In an orthotopic model, dovitinib potently inhibited primary tumor growth and lung metastasis and significantly prolonged mouse survival. CONCLUSIONS: Dovitinib demonstrated significant antitumor and antimetastatic activities in HCC xenograft models. This study provides a compelling rationale for clinical investigation in patients with advanced HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Quinolones/pharmacology , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Bevacizumab , Carcinoma, Hepatocellular/secondary , Cell Cycle Checkpoints/drug effects , Cell Division/drug effects , Cell Line, Tumor , Disease Models, Animal , G2 Phase/drug effects , Humans , Liver Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Mice , Mice, SCID , Survival Rate , Xenograft Model Antitumor Assays
15.
Healthcare (Basel) ; 10(2)2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35206935

ABSTRACT

Background and Objectives: Information regarding the COVID-19 pandemic has spread internationally through a variety of platforms, including social media. While efforts have been made to help reduce the spread of misinformation on social media, many platforms are still largely unregulated. The influence of social media use on vaccination promotion is not fully understood. This systematic review aims to identify facilitators and barriers associated with vaccine promotion through social media use. Materials and Methods: Reviewers analyzed 25 articles and identified common themes. Facilitators of vaccine promotion included an increase in the efforts of social media companies to reduce misinformation, the use of social media to spread information on public health and vaccine promotion, and the positive influence towards vaccinations of family and friends. Results and Conclusions: Identified barriers to vaccine promotion included the spread of misinformation, decreased vaccine acceptance among users of social media for COVID-19 related information due to polarization, and a lack of regulation on social media platforms. The results of this review provide insight for improving public health campaign promotion on social media and can help inform policy on social media regulation and misinformation prevention.

16.
J Cancer Res Clin Oncol ; 147(10): 2955-2968, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34156519

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC), the most common manifestation of liver cancer, is one of the leading causes of cancer-related mortality worldwide with limited treatment options. Infigratinib, a pan-FGFR inhibitor, has shown a potent antitumour effect in HCC. However, drug resistance is often observed in long-term treatment. In this study, we examined the potential feedback mechanism(s) leading to infigratinib and explored a combination therapy to overcome resistance in HCC. METHODS: Patient-derived xenograft (PDX) tumours were subcutaneously implanted into SCID mice and were subsequently treated with infigratinib. Tumour growth was monitored over time, and tumour samples were subjected to immunohistochemistry and Western blotting. For drug combination studies, mice were treated with infigratinib and/or varlitinib. Gene overexpression and knockdown studies were conducted to investigate the relationship between EZH2 and ErbB activity in infigratinib resistance. RESULTS: Infigratinib-resistant tumours exhibited higher levels of p-ErbB2 and p-ErbB3, concomitant with an increase in EZH2 expression. Gene overexpression and knockdown studies revealed that EZH2 directly regulates the levels of p-ErbB2 and p-ErbB3 in acquired resistance to infigratinib. The addition of varlitinib effectively overcame infigratinib resistance and prolonged the antitumour response, with minimal toxicity. CONCLUSION: The upregulation of the ErbB family by EZH2 appears to contribute to infigratinib resistance. The combination of infigratinib and varlitinib showed a potent antitumour effect and did not result in additional toxicity, warranting further clinical investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm/drug effects , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Receptor, ErbB-2/metabolism , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , Phenylurea Compounds/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrimidines/administration & dosage , Receptor, ErbB-2/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Hepatol Int ; 15(5): 1236-1246, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34333737

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is the most common liver cancer globally, claiming nearly 1 million lives each year. The overexpression of fibroblast growth factor (FGF) receptors (FGFRs) signaling cascade has been shown to contribute to tumorigenesis, metastasis, and poor prognosis in HCC. Therefore, targeted inhibition of the FGF/FGFR cascade may represent a new treatment strategy for HCC patients. METHODS: HCC patient-derived xenograft (PDX) models were implanted into either severe combined immunodeficient (SCID) or CD34+hu-NSG (humanized) mice and subsequently treated with vehicle, infigratinib (FGFR1-3 inhibitor), FGF401 (FGFR4 inhibitor), or the combination of infigratinib and FGF401. Tumor progressions, overall survival of mice, lung metastasis, and drug resistance were monitored, and samples collected at the end of the treatment cycle were subjected to Western blot analyses and immunohistochemistry. RESULTS: HCC PDX models expressing high levels of FGF19/FGFR4 or FGFR2/3 showed favorable initial treatment response to FGF401 and infigratinib, respectively. However, progressive disease due to acquired resistance was observed. Combination infigratinib/FGF401 augmented the antitumor activity, response rate, and overall survival of mice. This combination significantly increased the infiltration of B cells, macrophages, CD8+ T cells, and CD4+ T cells associated with granzyme-B-mediated apoptosis, delayed onset of resistance, and inhibited metastasis by potently inhibiting several critical signaling pathways involved in proliferation and metastasis. CONCLUSIONS: Our findings suggest that HCC patients with high FGFR2/3 or FGF19/FGFR4 expressing tumors might benefit from a combination infigratinib/FGF401; thus, supporting its evaluation in clinical trials.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Fibroblast Growth Factors , Humans , Liver Neoplasms/drug therapy , Mice , Mice, SCID , Phenylurea Compounds , Signal Transduction
18.
Mol Imaging Biol ; 23(1): 70-83, 2021 02.
Article in English | MEDLINE | ID: mdl-32909245

ABSTRACT

PURPOSE: Overexpression of fibroblast growth factor receptor (FGFR) contributes to tumorigenesis, metastasis, and poor prognosis of hepatocellular carcinoma (HCC). Infigratinib-a pan-FGFR inhibitor-potently suppresses the growth of high-FGFR-expressing HCCs in part via alteration of the tumor microenvironment and vessel normalization. In this study, we aim to assess the utility of dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) as a non-invasive imaging technique to detect microenvironment changes associated with infigratinib and sorafenib treatment in high-FGFR-expressing HCC xenografts. PROCEDURES: Serial DCE-MRIs were performed on 12 nude mice bearing high-FGFR-expressing patient-derived HCC xenografts to quantify tumor microenvironment pre- (day 0) and post-treatment (days 3, 6, 9, and 15) of vehicle, sorafenib, and infigratinib. DCE-MRI data were analyzed using extended generalized kinetic model and two-compartment distributed parameter model. After treatment, immunohistochemistry stains were performed on the harvested tumors to confirm DCE-MRI findings. RESULTS: By treatment day 15, infigratinib induced tumor regression (70 % volume reduction from baseline) while sorafenib induced relative growth arrest (185 % volume increase from baseline versus 694 % volume increase from baseline of control). DCE-MRI analysis revealed different changes in microcirculatory parameters upon exposure to sorafenib versus infigratinib. While sorafenib induced microenvironment changes similar to those of rapidly growing tumors, such as a decrease in blood flow (F), fractional intravascular volume (vp), and permeability surface area product (PS), infigratinib induced the exact opposite changes as early as day 3 after treatment: increase in F, vp, and PS. CONCLUSIONS: Our study demonstrated that DCE-MRI is a reliable non-invasive imaging technique to monitor tumor microcirculatory response to FGFR inhibition and VEGF inhibition in high-FGFR-expressing HCC xenografts. Furthermore, the microcirculatory changes from FGFR inhibition manifested early upon treatment initiation and were reliably detected by DCE-MRI, creating possibilities of combinatorial therapy for synergistic effect.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/drug therapy , Contrast Media/chemistry , Liver Neoplasms/drug therapy , Magnetic Resonance Imaging , Neovascularization, Pathologic/drug therapy , Phenylurea Compounds/therapeutic use , Pyrimidines/therapeutic use , Receptors, Fibroblast Growth Factor/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Hepatocellular/blood supply , Cell Proliferation/drug effects , Humans , Kinetics , Liver Neoplasms/blood supply , Mice, SCID , Perfusion , Sorafenib/pharmacology , Sorafenib/therapeutic use , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
19.
J Hepatol ; 52(1): 79-87, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19910069

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is a particularly vascularized solid tumor where the Raf/MEK/ERK pathway is activated; suggesting that inhibition of this pathway may have therapeutic potential. METHODS: We treated patient-derived HCC xenografts with (i) sorafenib, (ii) AZD6244 (ARRY-142886), and (iii) sorafenib plus AZD6244. Western blotting was employed to determine pharmacodynamic changes in biomarkers relevant to both angiogenesis and MEK signaling. Apoptosis, microvessel density, and cell proliferation were analyzed by immunohistochemistry. RESULTS: We report here that sorafenib treatment resulted in suppression of tumor growth, reduction in cell proliferation, induction of apoptosis and inhibition of mTOR targets. Sorafenib-induced elevation of the insulin-like growth factor receptor 1 (IGF-1R), phospho-c-Raf Ser338, phospho-MEK Ser217/221 and phospho-ERK Thr202/Tyr204 was attenuated by co-treating cells with anti-human IGF-1R antibody or over-expression of activated mutant p70S6K. Pharmacological inhibition of the MEK/ERK pathway by AZD6244 enhanced the anti-tumor effect of sorafenib in both orthotopic and ectopic models of HCC. Such inhibition led to a further increase in pro-apoptotic Bim, apoptosis and a profound inhibition of cell proliferation. CONCLUSION: Our findings underscore the potential of a combined therapeutic approach with sorafenib and MEK inhibitors in the treatment of HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Benzimidazoles/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Pyridines/therapeutic use , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzenesulfonates/pharmacology , Benzimidazoles/pharmacology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, SCID , Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Pathologic/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds , Pyridines/pharmacology , Signal Transduction/drug effects , Sorafenib , raf Kinases/metabolism
20.
J Gene Med ; 12(10): 848-58, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20963807

ABSTRACT

BACKGROUND: Herpes simplex virus type-1 (HSV-1) amplicon vectors are attractive tools for gene transfer because of their large DNA insert capacity, their broad host range of vector transduction and a minimal immune response as a result of the absence of helper viruses during viral packaging. However, the transient gene expression remains a challenge for the translation of HSV-1 amplicon based therapeutic strategies to a clinical setting. Although oriP/EBV nuclear antigen (EBNA)-1 elements of Epstein-Barr virus (EBV) have been successfully employed to achieve prolonged transgene expression, little is known about the stability of the EBNA-1 elements in the context of HSV-1 amplicon viral vectors. METHODS: We have generated HSV/EBV hybrid vectors expressing the mutant EBNA-1 gene with the luciferase reporter gene bicistronically to enable monitoring of EBNA-1 expression in real-time, both in vitro and in vivo. RESULTS: The results obtained showed that the HSV/EBV hybrid vectors could mediate high levels of transgene expression (ranging from approximately two-fold to nine-fold) in primary human tumor cells and human bone marrow-derived mesenchymal stem cells compared to the control vector. Prolonged transgene expression could also be observed in primary patient-derived human hepatocellular carcinoma xenografts and in the mouse brain parenchyma up to a period of 17 and 365 days, respectively. CONCLUSIONS: Taken together, we have demonstrated that these hybrid vectors could be promising tools as carriers of therapeutic genes in mesenchymal stem cells or even provide an alternative non-integrating platform for the generation of induced pluripotent stem cells.


Subject(s)
Genetic Vectors , Herpesvirus 4, Human/genetics , Mesenchymal Stem Cells/virology , Simplexvirus/genetics , Transgenes , Animals , Brain Neoplasms/pathology , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Chimera , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Feasibility Studies , Female , Gene Expression , Genes, Reporter , Glioma/pathology , HeLa Cells , Helper Viruses , Humans , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Mice, SCID , Transduction, Genetic , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL