Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Biochem Biophys Res Commun ; 443(2): 635-40, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24333418

ABSTRACT

The receptor tyrosine kinase Axl is overexpressed in and leads to patient morbidity and mortality in a variety of cancers. Axl-Gas6 interactions are critical for tumor growth, angiogenesis and metastasis. The goal of this study was to investigate the feasibility of imaging graded levels of Axl expression in tumors using a radiolabeled antibody. We radiolabeled anti-human Axl (Axl mAb) and control IgG1 antibodies with (125)I with high specific radioactivity and radiochemical purity, resulting in an immunoreactive fraction suitable for in vivo studies. Radiolabeled antibodies were investigated in severe combined immunodeficient mice harboring subcutaneous CFPAC (Axl(high)) and Panc1 (Axl(low)) pancreatic cancer xenografts by ex vivo biodistribution and imaging. Based on these results, the specificity of [(125)I]Axl mAb was also validated in mice harboring orthotopic Panc1 or CFPAC tumors and in mice harboring subcutaneous 22Rv1 (Axl(low)) or DU145 (Axl(high)) prostate tumors by ex vivo biodistribution and imaging studies at 72h post-injection of the antibody. Both imaging and biodistribution studies demonstrated specific and persistent accumulation of [(125)I]Axl mAb in Axl(high) (CFPAC and DU145) expression tumors compared to the Axl(low) (Panc1 and 22Rv1) expression tumors. Axl expression in these tumors was further confirmed by immunohistochemical studies. No difference in the uptake of radioactivity was observed between the control [(125)I]IgG1 antibody in the Axl(high) and Axl(low) expression tumors. These data demonstrate the feasibility of imaging Axl expression in pancreatic and prostate tumor xenografts.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Molecular Imaging/methods , Pancreatic Neoplasms/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Line, Tumor , Female , Male , Mice , Mice, Nude , Mice, SCID , Pancreatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/pathology , Radionuclide Imaging , Tissue Distribution , Axl Receptor Tyrosine Kinase
2.
Article in English | MEDLINE | ID: mdl-24521409

ABSTRACT

Cancer-related death is one of the most common causes of mortality in society. Small molecules have the capability to disrupt aberrant signaling pathways in tumors, leading to anticancer activities. Therefore the search for new molecules for cancer treatment continues to draw attention to the scientific research community. Synthesis and biological evaluation of hedgehog (Hh) pathway inhibitors SANT-1 and GANT-61 are disclosed. These molecules have been synthesized from common precursors using simple conversions, our synthesis features Vils-Meier-Haack reaction, imine formation reaction and N-arylation reaction. These drugs were evaluated using a Hh reporter assay to confirm pathway inhibitory activity, and tested for cell viability against pancreatic and prostate cancer cells. These methodologies can be applied to make potent analogs of both inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Piperazines/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Discovery , Enzyme Inhibitors/pharmacology , Hedgehog Proteins/metabolism , Humans , Male , Molecular Structure , Piperazines/chemical synthesis , Piperazines/chemistry , Pyrazoles/chemical synthesis , Pyrazoles/chemistry , Pyridines/chemical synthesis , Pyridines/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry
3.
Int J Cancer ; 132(4): 785-94, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-22821831

ABSTRACT

MIF is a proinflammatory cytokine and is implicated in cancer. A higher MIF level is found in many human cancer and cancer-prone inflammatory diseases, including chronic pancreatitis and pancreatic cancer. We tested the hypothesis that MIF contributes to pancreatic cancer aggressiveness and predicts disease outcome in resected cases. Consistent with our hypothesis we found that an elevated MIF mRNA expression in tumors was significantly associated with poor outcome in resected cases. Multivariate Cox-regression analysis further showed that MIF is independently associated with patients' survival (HR = 2.26, 95% CI = 1.17-4.37, p = 0.015). Mechanistic analyses revealed that MIF overexpression decreased E-cadherin and increased vimentin mRNA and protein levels in pancreatic cancer cell lines, consistent with the features of epithelial-to-mesenchymal transition (EMT). Furthermore, MIF-overexpression significantly increased ZEB1/2 and decreased miR-200b expression, while shRNA-mediated inhibition of MIF increased E-cadherin and miR-200b expression, and reduced the expression of ZEB1/2 in Panc1 cells. Re-expression of miR-200b in MIF overexpressing cells restored the epithelial characteristics, as indicated by an increase in E-cadherin and decrease in ZEB1/2 and vimentin expression. A reduced sensitivity to the chemotherapeutic drug, gemcitabine, occurred in MIF-overexpressing cells. Indicative of an increased malignant potential, MIF over-expressing cells showed significant increase in their invasion ability in vitro, and tumor growth and metastasis in an orthotopic xenograft mouse model. These results support a role of MIF in disease aggressiveness, indicating its potential usefulness as a candidate target for designing improved treatment in pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Epithelial-Mesenchymal Transition/genetics , Macrophage Migration-Inhibitory Factors/genetics , Pancreatic Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Animals , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Prognosis , RNA Interference , Transplantation, Heterologous , Gemcitabine
4.
Materials (Basel) ; 16(3)2023 Jan 20.
Article in English | MEDLINE | ID: mdl-36769971

ABSTRACT

In order to meet the great demand for green grain storage and low carbon emissions, paraffin, high-density polyethylene (HDPE), and expanded graphite (EG) were used to produce shape-stabilized phase change material (SSPCM) plates, which were then used to reconstruct building walls for existing granaries. A new type of SSPCM plate was then prefabricated with different thermal conductivities and a high latent heat. This plate could be directly adhered to the existing granary walls. In order to evaluate the thermal regulation performance of these phase change granary walls, experiments and numerical methods were established, specifically for the summer condition. The thermal behavior of the SSPCM granary wall was compared with that of the common concrete granary wall to obtain the optimal parameters. It was concluded that increasing the thickness of the SSPCM layer can reduce the temperature rise of the wall. However, the maximum latent heat utilization rate and energy storage effects were obtained when the SSPCM thickness was at an intermediate level of 30 mm. The thermal conductivity of the SSPCM had a controversial effect on the thermal resistance and latent heat utilization behaviors of the SSPCM. Considering the temperature level and energy saving rate, a 30 mm thick SSPCM plate with a thermal conductivity of 0.2 W/m·K provided a superior performance. When compared to the common wall, the optimized energy-saving rate was greatly enhanced by 35.83% for the SSPCM granary wall with a thickness of 30 mm and a thermal conductivity of 0.2 W/m·K.

5.
Clin Cancer Res ; 14(10): 3141-8, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483382

ABSTRACT

PURPOSE: The activity of imatinib in leukemia has recently been linked with expression of the organic cation transporter 1 (OCT1) gene SLC22A1. Here, we characterized the contribution of solute carriers to imatinib transport in an effort to further understand mechanisms involved in the intracellular uptake and retention (IUR) of the drug. EXPERIMENTAL DESIGN: IUR of [3H]imatinib was studied in Xenopus laevis oocytes and HEK293 cells expressing OATP1A2, OATP1B1, OATP1B3, OCT1-3, OCTN1-2, or OAT1-3. Gene expression was determined in nine leukemia cell lines using the Affymetrix U133 array. RESULTS: Imatinib was not found to be a substrate for OCT1 in oocytes (P = 0.21), whereas in HEK293 cells IUR was increased by only 1.20-fold relative to control cells (P = 0.002). Furthermore, in 74 cancer patients, the oral clearance of imatinib was not significantly altered in individuals carrying reduced-function variants in SLC22A1 (P = 0.99). Microarray analysis indicated that SLC22A1 was interrelated with gene expression of various transporters, including ABCB1, ABCC4, ABCG2 (negative), and OATP1A2 (positive). Imatinib was confirmed to be a substrate for the three efflux transporters (P < 0.05) as well as for OATP1A2 (P = 0.0001). CONCLUSIONS: This study suggests that SLC22A1 expression is a composite surrogate for expression of various transporters relevant to imatinib IUR. This observation provides a mechanistic explanation for previous studies that have linked SLC22A1 with the antitumor activity of imatinib. Because of its high expression in the intestine, ciliary body, gliomas, and leukemia cells, OATP1A2 may play a key role in imatinib pharmacokinetics-pharmacodynamics.


Subject(s)
Antineoplastic Agents/metabolism , Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/genetics , Organic Cation Transporter 1/genetics , Piperazines/metabolism , Pyrimidines/metabolism , Animals , Benzamides , Cell Line, Tumor , Gastrointestinal Stromal Tumors/drug therapy , Gene Expression , Humans , Imatinib Mesylate , Organic Cation Transporter 1/metabolism , Polymerase Chain Reaction , Xenopus laevis
6.
Mol Cancer Ther ; 6(9): 2429-40, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17876042

ABSTRACT

This study is intended to characterize the cellular target of gambogic acid (GA), a natural product isolated from the gamboge resin of Garcinia hurburyi tree, which possesses potent in vitro and in vivo antitumor activities. The antiproliferative activity of GA was further confirmed here in a panel of human tumor cells and multidrug-resistant cells. We found that GA significantly inhibited the catalytic activity of topoisomerase (Topo) II and, to a comparatively less extent, of Topo I, without trapping and stabilizing covalent topoisomerase-DNA cleavage complexes. Down-regulation of Topo IIalpha but not Topo I and Topo IIbeta, reduced GA-induced apoptosis and the phosphorylation of c-Jun, and restored cell proliferation upon GA treatment. Moreover, GA antagonized etoposide-induced DNA damage and abrogated the antiproliferative activity of etoposide, whereas it did not affect camptothecin-induced DNA damage. By dissecting the actions of GA on the individual steps of Topo IIalpha catalytic cycle, we found that GA inhibited DNA cleavage and ATP hydrolysis. Moreover, GA directly bound to the ATPase domain of Topo IIalpha, and may share common binding sites with ATP. The results reported here show that GA exerts its antiproliferative effect by inhibiting the catalytic activity Topo IIalpha. They also indicate that GA inhibits Topo IIalpha-mediated DNA cleavage and modulate the activity of Topo II poisons, which provide rationale for further clinical evaluation of GA.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA-Binding Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Topoisomerase II Inhibitors , Xanthones/pharmacology , Adenosine Triphosphatases/chemistry , Adenosine Triphosphate/metabolism , Antigens, Neoplasm/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Etoposide/pharmacology , Flow Cytometry , Genes, MDR , HeLa Cells , Humans , Hydrolysis , JNK Mitogen-Activated Protein Kinases/metabolism , Neoplasms/metabolism , Phosphorylation/drug effects , RNA Interference , Surface Plasmon Resonance , Tumor Stem Cell Assay
7.
Cancer Cell ; 29(1): 75-89, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26725216

ABSTRACT

Induction of compensatory mechanisms and ERK reactivation has limited the effectiveness of Raf and MEK inhibitors in RAS-mutant cancers. We determined that direct pharmacologic inhibition of ERK suppressed the growth of a subset of KRAS-mutant pancreatic cancer cell lines and that concurrent phosphatidylinositol 3-kinase (PI3K) inhibition caused synergistic cell death. Additional combinations that enhanced ERK inhibitor action were also identified. Unexpectedly, long-term treatment of sensitive cell lines caused senescence, mediated in part by MYC degradation and p16 reactivation. Enhanced basal PI3K-AKT-mTOR signaling was associated with de novo resistance to ERK inhibitor, as were other protein kinases identified by kinome-wide siRNA screening and a genetic gain-of-function screen. Our findings reveal distinct consequences of inhibiting this kinase cascade at the level of ERK.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/genetics , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Pancreatic Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Time
8.
Mol Cancer Ther ; 14(7): 1532-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25931518

ABSTRACT

KRAS is activated by mutation in the vast majority of cases of pancreatic cancer; unfortunately, therapeutic attempts to inhibit KRAS directly have been unsuccessful. Our previous studies showed that inhibition of cyclin-dependent kinase 5 (CDK5) reduces pancreatic cancer growth and progression, through blockage of the centrally important RAL effector pathway, downstream of KRAS. In the current study, the therapeutic effects of combining the CDK inhibitor dinaciclib (SCH727965; MK-7965) with the pan-AKT inhibitor MK-2206 were evaluated using orthotopic and subcutaneous patient-derived human pancreatic cancer xenograft models. The combination of dinaciclib (20 mg/kg, i.p., three times a week) and MK-2206 (60 mg/kg, orally, three times a week) dramatically blocked tumor growth and metastasis in all eight pancreatic cancer models examined. Remarkably, several complete responses were induced by the combination treatment of dinaciclib and MK-2206. The striking results obtained in these models demonstrate that the combination of dinaciclib with the pan-AKT inhibitor MK-2206 is promising for therapeutic evaluation in pancreatic cancer, and strongly suggest that blocking RAL in combination with other effector pathways downstream from KRAS may provide increased efficacy in pancreatic cancer. Based on these data, an NCI-CTEP-approved multicenter phase I clinical trial for pancreatic cancer of the combination of dinaciclib and MK-2206 (NCT01783171) has now been opened.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Pancreatic Neoplasms/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays/methods , Administration, Oral , Animals , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Proliferation/drug effects , Cyclic N-Oxides , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/metabolism , Drug Administration Schedule , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Immunohistochemistry , Indolizines , Injections, Intraperitoneal , Mice, Nude , Neoplasm Metastasis , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Pyridinium Compounds/administration & dosage , Pyridinium Compounds/pharmacology , Retinoblastoma Protein/metabolism , Treatment Outcome
9.
Oncotarget ; 5(9): 2575-87, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24810906

ABSTRACT

To identify potentially important genes dysregulated in pancreatic cancer, we analyzed genome-wide transcriptional analysis of pancreatic cancers and normal pancreatic duct samples and identified the transcriptional coactivator, EYA2 (Drosophila Eyes Absent Homologue-2) as silenced in the majority of pancreatic cancers. We investigated the role of epigenetic mechanisms of EYA2 gene silencing in pancreatic cancers, performed in vitro and in vivo proliferation and migration assays to assess the effect of EYA2 silencing on tumor cell growth and metastasis formation, and expression analysis to identify genes transcriptionally regulated by EYA2. We found loss of tumoral Eya2 expression in 63% of pancreatic cancers (120/189 cases). Silencing of EYA2 expression in pancreatic cancer cell lines correlated with promoter methylation and histone deacetylation and was reversible with DNA methyltransferase and HDAC inhibitors. EYA2 knockdown in pancreatic cancer cell lines increased cell proliferation. Compared to parental pancreatic cancer cells, pancreatic cancers stably-expressing EYA2 grew more slowly and had fewer metastases in orthotopic models. The transcriptional changes after stable expression of EYA2 in pancreatic cancer cells included induction of genes in the TGFbeta pathway. Epigenetic silencing of EYA2 is a common event in pancreatic cancers and stable expression EYA2 limits the growth and metastases of pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/secondary , Cell Proliferation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Silencing , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Pancreatic Neoplasms/pathology , Protein Tyrosine Phosphatases/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Base Sequence , Blotting, Western , Cell Cycle , Cell Movement , Chromatin Immunoprecipitation , DNA Methylation , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Nude , Molecular Sequence Data , Neoplasm Metastasis , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic/genetics , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
10.
Clin Cancer Res ; 19(15): 4067-78, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23757351

ABSTRACT

PURPOSE: Recent microarray and RNA-sequencing studies have uncovered aberrantly expressed microRNAs (miRNA) in Barrett's esophagus-associated esophageal adenocarcinoma. The functional significance of these miRNAs in esophageal adenocarcinoma initiation and progression is largely unknown. EXPERIMENTAL DESIGN: Expression levels of miR-199a/b-3p, -199a-5p, -199b-5p, -200b, -200c, -223, and -375 were determined in microdissected tissues from cardiac mucosa, Barrett's esophagus, dysplastic Barrett's esophagus, and esophageal adenocarcinoma using quantitative real-time PCR. miR-223 expression was validated in precursors and esophageal adenocarcinomas from 95 patients with esophageal adenocarcinoma by in situ hybridization (ISH). miR-223 was transfected into two esophageal adenocarcinoma cell lines, and in vitro assays were conducted. Target genes were identified using Illumina microarray, and results were validated in cell lines and human specimens. RESULTS: miR-199 family members and miR-223 were significantly overexpressed in esophageal adenocarcinoma, however, only miR-223 showed a stepwise increase during esophageal adenocarcinoma carcinogenesis. A similar trend was observed by ISH, which additionally showed that miR-223 is exclusively expressed by the epithelial compartment. miR-223-overexpressing cells had statistically significantly more migratory and invasive potential than scramble sequence-transfected cells. PARP1 was identified as a direct target gene of miR-223 in esophageal adenocarcinoma cells. Increased sensitivity to chemotherapy was observed in cells with enforced miR-223 expression and reduced PARP1. CONCLUSIONS: miR-223 is significantly upregulated during the Barrett's esophagus-dysplasia-esophageal adenocarcinoma sequence. Although high miR-223 levels might contribute to an aggressive phenotype, our results also suggest that patients with esophageal adenocarcinoma with high miR-223 levels might benefit from treatment with DNA-damaging agents.


Subject(s)
Adenocarcinoma/drug therapy , Barrett Esophagus/drug therapy , Esophageal Neoplasms/drug therapy , MicroRNAs/biosynthesis , Poly(ADP-ribose) Polymerases/biosynthesis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Barrett Esophagus/genetics , Barrett Esophagus/pathology , Disease Progression , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Humans , In Situ Hybridization , Poly (ADP-Ribose) Polymerase-1 , Up-Regulation
11.
Mol Cancer Ther ; 11(4): 921-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22389472

ABSTRACT

OCTN2 is a bifunctional transporter that reabsorbs filtered carnitine in a sodium-dependent manner and secretes organic cations into urine as a proton antiport mechanism. We hypothesized that inhibition of OCTN2 by anticancer drugs can influence carnitine resorption. OCTN2-mediated transport inhibition by anticancer drugs was assessed using cells transfected with human OCTN2 (hOCTN2) or mouse Octn2 (mOctn2). Excretion of carnitine and acetylcarnitine was measured in urine collected from mice and pediatric patients with cancer before and after administration of etoposide. Five of 27 tested drugs (50-100 µmol/L) inhibited hOCTN2-mediated carnitine uptake by 42% to 85% (P < 0.001). Of these inhibitors, etoposide was itself a transported substrate of hOCTN2 and mOctn2. Etoposide uptake by hOCTN2 was reversed in the presence of excess carnitine. This competitive inhibitory mechanism was confirmed in an in silico molecular docking analysis. In addition, etoposide inhibited the transcellular apical-to-basolateral flux of carnitine in kidney cells. Etoposide was also associated with a significant urinary loss of carnitine in mice (~1.5-fold) and in patients with cancer (~2.4-fold). Collectively, these findings indicate that etoposide can inhibit hOCTN2 function, potentially disturb carnitine homeostasis, and that this phenomenon can contribute to treatment-related toxicities.


Subject(s)
Carnitine/metabolism , Etoposide/pharmacology , Organic Cation Transport Proteins/antagonists & inhibitors , Acetylcarnitine/urine , Adolescent , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biological Transport , Carnitine/pharmacokinetics , Carnitine/urine , Cell Culture Techniques , Cell Line , Child , Etoposide/administration & dosage , Etoposide/pharmacokinetics , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/urine , Male , Mice , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Solute Carrier Family 22 Member 5 , Swine , Transfection
12.
Clin Cancer Res ; 18(5): 1291-302, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-21868763

ABSTRACT

PURPOSE: To illustrate the prognostic significance of hedgehog (Hh) signaling in patients with hepatocellular carcinoma (HCC) and to evaluate the efficacy of a novel nanoparticle-encapsulated inhibitor of the Hh transcription factor, Gli1 (NanoHHI) using in vitro and in vivo models of human HCCs. EXPERIMENTAL DESIGN: Patched1 (Ptch1) expression was detected in tumor tissue microarrays of 396 patients with HCC who underwent curative surgical resection during February 2000 to December 2002. Prognostic significance was assessed using Kaplan-Meier survival estimates and log-rank tests. The effects of NanoHHI alone and in combination with sorafenib were investigated on HCC cell lines. Primary HCC tumor growth and metastasis were examined in vivo using subcutaneous and orthotopic HCC xenografts in nude mice. RESULTS: Elevated expression of Ptch1 in HCC tissues was significantly related to disease recurrence, as well as a shorter time to recurrence in patients with HCC. In vitro, NanoHHI significantly inhibited the proliferation and invasion of HCC cell lines. NanoHHI potently suppressed in vivo tumor growth of HCC xenografts in both subcutaneous and orthotopic milieus, and in contrast to sorafenib, resulted in significant attenuation of systemic metastases in the orthotopic setting. Furthermore, NanoHHI significantly decreased the population of CD133-expressing HCC cells, which have been implicated in tumor initiation and metastases. CONCLUSION: Downstream Hh signaling has prognostic significance in patients with HCC as it predicts early recurrence. Gli inhibition through NanoHHI has profound tumor growth inhibition and antimetastatic effects in HCC models, which may provide a new strategy in the treatment of patients with HCC and prevention post-operative recurrence.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/metabolism , Hedgehog Proteins/antagonists & inhibitors , Liver Neoplasms/metabolism , Signal Transduction/drug effects , Adult , Aged , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzenesulfonates/administration & dosage , Benzenesulfonates/chemistry , Benzenesulfonates/pharmacology , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/mortality , Cell Line, Tumor , Cell Proliferation , Female , Humans , Immunophenotyping , Liver Neoplasms/diagnosis , Liver Neoplasms/drug therapy , Liver Neoplasms/mortality , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Nanocapsules/chemistry , Nanocapsules/therapeutic use , Neoplasm Invasiveness , Neoplasm Metastasis , Niacinamide/analogs & derivatives , Patched Receptors , Patched-1 Receptor , Phenylurea Compounds , Prognosis , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyridines/administration & dosage , Pyridines/chemistry , Pyridines/pharmacology , Receptors, Cell Surface/metabolism , Recurrence , Sorafenib , Transcription Factors/antagonists & inhibitors , Zinc Finger Protein GLI1
13.
Mol Cancer Ther ; 11(1): 165-73, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22027695

ABSTRACT

Aberrant activation of the hedgehog (Hh) signaling pathway is one of the most prevalent abnormalities in human cancer. Tumors with cell autonomous Hh activation (e.g., medulloblastomas) can acquire secondary mutations at the Smoothened (Smo) antagonist binding pocket, which render them refractory to conventional Hh inhibitors. A class of Hh pathway inhibitors (HPI) has been identified that block signaling downstream of Smo; one of these compounds, HPI-1, is a potent antagonist of the Hh transcription factor Gli1 and functions independent of upstream components in the pathway. Systemic administration of HPI-1 is challenging due to its minimal aqueous solubility and poor bioavailability. We engineered a polymeric nanoparticle from [poly(lactic-co-glycolic acid); (PLGA)] conjugated with polyethylene glycol (PEG), encapsulating HPI-1 (NanoHHI). NanoHHI particles have an average diameter of approximately 60 nm, forms uniform aqueous suspension, and improved systemic bioavailability compared with the parent compound. In contrast to the prototype targeted Smo antagonist, HhAntag (Genentech), NanoHHI markedly inhibits the growth of allografts derived from Ptch(-/+); Trp53(-/-) mouse medulloblastomas that harbor a Smo(D477G) binding site mutation (P < 0.001), which is accompanied by significant downregulation of mGli1 as well as bona fide Hh target genes (Akna, Cltb, and Olig2). Notably, NanoHHI combined with gemcitabine also significantly impedes the growth of orthotopic Pa03C pancreatic cancer xenografts that have a ligand-dependent, paracrine mechanism of Hh activation when compared with gemcitabine alone. No demonstrable hematologic or biochemical abnormalities were observed with NanoHHI administration. NanoHHI should be amenable to clinical translation in settings where tumors acquire mutational resistance to current Smo antagonists.


Subject(s)
Hedgehog Proteins/antagonists & inhibitors , Nanoparticles/chemistry , Pancreatic Neoplasms/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Hedgehog Proteins/metabolism , Humans , Male , Medulloblastoma/metabolism , Medulloblastoma/pathology , Mice , Mice, Nude , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Transcription Factors/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1 , Gemcitabine
14.
Curr Drug Targets ; 11(6): 708-15, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20298152

ABSTRACT

Apoptosis as a form of programmed cell death is a critical defense mechanism against the formation and progression of cancer and exhibits distinct morphological and biochemical traits. In an in vivo situation, apoptosis functions to eliminate potentially deleterious cells without causing such adverse effects as inflammatory response and ensuing scar formation. Therefore, targeting apoptotic pathways becomes an intriguing strategy for the development of chemotherapeutic agents. Marine natural products have become an important source in the discovery of antitumor drugs, especially when modern technology makes it more and more feasible to collect organisms from seas. Although lack of an analog of a long ethno-medical history for finding clues, as compared with terrestrial habitats, still hinders the progress, an increasing number of compounds have been isolated from marine organisms that have been found to possess apoptosis-inducing and anticancer activities. This primer summarizes several such compounds, based on their effects on apoptotic signaling pathways, although most of these products have not yet been studied in depth for their mechanisms of action.


Subject(s)
Apoptosis/drug effects , Biological Products/pharmacology , Neoplasms/drug therapy , Animals , Apoptosis/physiology , Biological Products/therapeutic use , Humans , Oceans and Seas
15.
Clin Cancer Res ; 16(19): 4789-99, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20858838

ABSTRACT

PURPOSE: Carnitine is an essential cofactor for mitochondrial fatty acid oxidation that is actively reabsorbed by the luminal transporter Octn2 (Slc22a5). Because the nephrotoxic agent cisplatin causes urinary loss of carnitine in humans, we hypothesized that cisplatin may affect Octn2 function. EXPERIMENTAL DESIGN: Excretion of carnitine and acetylcarnitine was measured in urine collected from mice with or without cisplatin administration. The transport of carnitine was assessed in cells that were transfected with OCT1 or OCT2. The effect of cisplatin treatment on gene expression was analyzed using a mouse GeneChip array and validated using quantitative reverse transcriptase-PCR. RESULTS: In wild-type mice, urinary carnitine excretion at baseline was ∼3-fold higher than in mice lacking the basolateral cisplatin transporters Oct1 and Oct2 [Oct1/2(-/-) mice], indicating that carnitine itself undergoes basolateral uptake into the kidney. Transport of carnitine by OCT2, but not OCT1, was confirmed in transfected cells. We also found that cisplatin caused an increase in the urinary excretion of carnitine and acetylcarnitine in wild-type mice but not in Oct1/2(-/-) mice, suggesting that tubular transport of cisplatin is a prerequisite for this phenomenon. Cisplatin did not directly inhibit the transport of carnitine by Octn2 but downregulated multiple target genes of the transcription factor peroxisome proliferator activated receptor α, including Slc22a5, in the kidney of wild-type mice that were absent in Oct1/2(-/-) mice. CONCLUSION: Our study shows a pivotal role of Oct1 and Oct2 in cisplatin-related disturbances in carnitine homeostasis. We postulate that this phenomenon is triggered by deactivation of peroxisome proliferator activated receptor α and leads to deregulation of carnitine-shuttle genes.


Subject(s)
Carnitine/urine , Cisplatin/pharmacology , Down-Regulation/drug effects , Organic Cation Transport Proteins/antagonists & inhibitors , Acetylcarnitine/urine , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Cell Line , Gene Expression Profiling , Humans , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Organic Cation Transport Proteins/deficiency , Organic Cation Transport Proteins/metabolism , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Solute Carrier Family 22 Member 5 , Symporters
16.
Clin Cancer Res ; 16(16): 4198-206, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20601443

ABSTRACT

PURPOSE: This study aimed to test the influence of functional renal organic cation transporters (OCT2 in humans, Oct1 and Oct2 in mice) on biomarkers of cisplatin nephrotoxicity, such as urinary activity of N-acetyl-beta-D-glucosaminidase (NAG). EXPERIMENTAL DESIGN: Temporal cisplatin-induced nephrotoxicity was assessed by histopathology and biomarkers. Cisplatin-mediated NAG changes and survival were determined in wild-type and Oct1/2(-/-) mice. Identification of OCT2 inhibitors was done in transfected 293Flp-In cells, and the NCI(60) cell line panel was used to assess contribution of OCT2 to cisplatin uptake in cancer cells. RESULTS: Classical biomarkers such as blood urea nitrogen and serum creatinine were not elevated until 72 hours after cisplatin administration and substantial kidney damage had occurred. Oct1/2(-/-) mice had 2.9-fold lower NAG by 4 hours (P < 0.0001) and 2.3-fold increased survival (P = 0.0097). Among 16 agents, cimetidine strongly inhibited uptake of tetraethylammonium bromide (P = 0.0006) and cisplatin (P < 0.0001), but did not have an influence on cisplatin uptake in SK-OV-3 cells, the cancer line with the highest OCT2 mRNA levels. In wild-type mice, cimetidine inhibited cisplatin-induced NAG changes (P = 0.016 versus cisplatin alone) to a degree similar to that seen in Oct1/2(-/-) mice receiving cisplatin (P = 0.91). Cumulative NAG activity of >0.4 absorbance units (AU) was associated with 21-fold increased odds for severe nephrotoxicity (P = 0.0017), which was linked with overall survival (hazard ratio, 8.1; 95% confidence interval, 2.1-31; P = 0.0078). CONCLUSIONS: Cimetidine is able to inhibit OCT2-mediated uptake of cisplatin in the kidney, and subsequently ameliorate nephrotoxicity likely with minimal effect on uptake in tumor cells.


Subject(s)
Acetylglucosaminidase/urine , Antineoplastic Agents/toxicity , Cisplatin/toxicity , Octamer Transcription Factor-1/deficiency , Organic Cation Transport Proteins/deficiency , Animals , Antineoplastic Agents/metabolism , Biomarkers/analysis , Cell Line, Tumor , Cimetidine/pharmacology , Cisplatin/metabolism , Enzyme Inhibitors/pharmacology , Humans , Kidney/drug effects , Kidney/metabolism , Male , Mice , Mice, Knockout , Organic Cation Transporter 2 , Polymerase Chain Reaction
17.
Chem Pharm Bull (Tokyo) ; 56(6): 827-30, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18520088

ABSTRACT

Three new coumarins containing a C(10) terpenoid side chain, clauslactones R - T (1 - 3), together with 14 known coumarins (4 - 17) and 11 known carbazole alkaloids (18 - 28), were isolated from the leaves and stems of Clausena excavata. Their structures were established by detailed spectroscopic analyses. Furthermore, the stereochemistry of 1 was confirmed by single-crystal X-ray diffraction analysis, which was the first example among coumarins with a C(10) terpenoid side chain. Additionally, compounds 22 and 27 were found to show moderate topoisomerase II inhibitory effects at 50 microM.


Subject(s)
Clausena/chemistry , Coumarins/chemistry , Coumarins/isolation & purification , Coumarins/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Magnetic Resonance Spectroscopy , Molecular Conformation , Plant Leaves/chemistry , Plant Stems/chemistry , Spectrometry, Mass, Electrospray Ionization , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Structure-Activity Relationship , Topoisomerase II Inhibitors , X-Ray Diffraction
18.
Cancer Biol Ther ; 6(5): 775-80, 2007 May.
Article in English | MEDLINE | ID: mdl-17426448

ABSTRACT

Using guanidine-HCl extraction, acetone precipitation, ultra-filtration and chromatography, a novel polypeptide with potent anti-angiogenic activity was purified from cartilage of the shark, Prionace glauca. N-terminal amino acid sequence analysis and SDS-PAGE revealed that the substance is a novel polypeptide with MW 15500 (PG155). The anti-angiogenic effects of PG155 were evaluated using zebrafish embryos model in vivo. Treatment of the embryos with 20 microg/ml PG155 resulted in a significant reduction in the growth of subintestinal vessels (SIVs). A higher dose resulted in almost complete inhibition of SIV growth, as observed by endogenous alkaline phosphatase (EAP) staining assay. An in vitro transwell experiment revealed that the polypeptide inhibited vascular endothelial growth factor (VEGF) induced migration and tubulogenesis of human umbilical vein endothelial cells (HUVECs). Exposure of HUVECs in 20 microg/ml PG155 significantly decreased the density of migrated cells. Almost complete inhibition of cell migration was found when HUVECs were treated with 40-80 microg/ml PG155. PG155 (20 microg/ml) markedly inhibited the tube formation of HUVECs and a dose-dependent effect was also found when treatment of HUVECs with PG155 at the concentration from 20-160 microg/ml.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cartilage/chemistry , Embryo, Nonmammalian/drug effects , Neovascularization, Physiologic/drug effects , Peptides/pharmacology , Sharks , Tissue Extracts/pharmacology , Alkaline Phosphatase/metabolism , Animals , Cell Movement/drug effects , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Microcirculation , Umbilical Veins/cytology , Umbilical Veins/drug effects , Umbilical Veins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Zebrafish/embryology , Zebrafish/metabolism
19.
Mol Pharmacol ; 70(5): 1593-601, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16914642

ABSTRACT

Salvicine, a structurally modified diterpenoid quinone derived from Salvia prionitis, is a nonintercalative topoisomerase II (topo II) poison. The compound possesses potent in vitro and in vivo antitumor activity with a broad spectrum of anti-multidrug resistance activity and is currently in phase II clinical trials. To elucidate the distinct antitumor properties of salvicine and obtain valuable structural information of salvicine-topo II interactions, we characterized the effects of salvicine on human topo IIalpha (htopo IIalpha), including possible binding sites and molecular interactions. The enzymatic assays disclosed that salvicine mainly inhibits the catalytic activity with weak DNA cleavage action, in contrast to the classic topo II poison etoposide (VP16). Molecular modeling studies predicted that salvicine binds to the ATP pocket in the ATPase domain and superimposes on the phosphate and ribose groups. In a surface plasmon resonance binding assay, salvicine exhibited higher affinity for the ATPase domain of htopo IIalpha than ATP and ADP. Competitive inhibition tests demonstrated that ATP competitively and dose-dependently blocked the interactions between salvicine and ATPase domain of htopo IIalpha. The data illustrate that salvicine shares a common binding site with ATP and functions as an ATP competitor. To our knowledge, this is the first report to identify an ATP-binding pocket as the structural binding motif for a nonintercalative eukaryotic topo II poison. These findings collectively support the potential value of an ATP competitor of htopo IIalpha in tumor chemotherapy.


Subject(s)
Adenosine Triphosphate/metabolism , Antigens, Neoplasm/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Naphthoquinones/metabolism , Naphthoquinones/pharmacology , Topoisomerase II Inhibitors , Adenosine Triphosphatases/isolation & purification , Adenosine Triphosphatases/metabolism , Antigens, Neoplasm/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Binding Sites , Catalysis , DNA/chemistry , DNA Topoisomerases, Type II/chemistry , DNA-Binding Proteins/chemistry , Dose-Response Relationship, Drug , Humans , Hydrolysis , Models, Molecular , Naphthoquinones/chemistry , Nucleic Acid Conformation , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL