Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 148
1.
Oncogene ; 39(23): 4619, 2020 Jun.
Article En | MEDLINE | ID: mdl-32366906

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

2.
Oncogene ; 37(6): 722-731, 2018 02 08.
Article En | MEDLINE | ID: mdl-29059175

Anti-vascular endothelial growth factor (VEGF) therapy has demonstrated efficacy in treating human metastatic cancers, but therapeutic resistance is a practical limitation and most tumors eventually become unresponsive. To identify microenvironmental factors underlying the resistance of cancer to antiangiogenesis therapy, we conducted genomic analyses of intraperitoneal ovarian tumors in which adaptive resistance to anti-VEGF therapy (B20 antibody) developed. We found that expression of the microseminoprotein, prostate-associated (MSMP) gene was substantially upregulated in resistant compared with control tumors. MSMP secretion from cancer cells was induced by hypoxia, triggering MAPK signaling in endothelial cells to promote tube formation in vitro. Recruitment of the transcriptional repressor CCCTC-binding factor (CTCF) to the MSMP enhancer region was decreased by histone acetylation under hypoxic conditions in cancer cells. MSMP siRNA, delivered in vivo using the DOPC nanoliposomes, restored tumor sensitivity to anti-VEGF therapy. In ovarian cancer patients treated with bevacizumab, serum MSMP concentration increased significantly only in non-responders. These findings imply that MSMP inhibition combined with the use of antiangiogenesis drugs may be a new strategy to overcome resistance to antiangiogenesis therapy.


Bevacizumab/pharmacology , Carcinoma, Ovarian Epithelial/pathology , Drug Resistance, Neoplasm , Fallopian Tube Neoplasms/pathology , Neoplasm Proteins/metabolism , Peritoneal Neoplasms/pathology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Apoptosis , Biomarkers, Tumor , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/metabolism , Cell Hypoxia , Cell Proliferation , Fallopian Tube Neoplasms/drug therapy , Fallopian Tube Neoplasms/metabolism , Female , Follow-Up Studies , Humans , Male , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neovascularization, Pathologic , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/metabolism , Prognosis , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Oncogene ; 36(7): 912-921, 2017 02 16.
Article En | MEDLINE | ID: mdl-27568980

Fatty acid binding protein 4 (FABP4) is a fatty acid chaperone, which is induced during adipocyte differentiation. Previously we have shown that FABP4 in endothelial cells is induced by the NOTCH1 signalling pathway, the latter of which is involved in mechanisms of resistance to antiangiogenic tumour therapy. Here, we investigated the role of FABP4 in endothelial fatty acid metabolism and tumour angiogenesis. We analysed the effect of transient FABP4 knockdown in human umbilical vein endothelial cells on fatty acid metabolism, viability and angiogenesis. Through therapeutic delivery of siRNA targeting mouse FABP4, we investigated the effect of endothelial FABP4 knockdown on tumour growth and blood vessel formation. In vitro, siRNA-mediated FABP4 knockdown in endothelial cells led to a marked increase of endothelial fatty acid oxidation, an increase of reactive oxygen species and decreased angiogenesis. In vivo, we found that increased NOTCH1 signalling in tumour xenografts led to increased expression of endothelial FABP4 that decreased when NOTCH1 and VEGFA inhibitors were used in combination. Angiogenesis, growth and metastasis in ovarian tumour xenografts were markedly inhibited by therapeutic siRNA delivery targeting mouse endothelial FABP4. Therapeutic targeting of endothelial FABP4 by siRNA in vivo has antiangiogenic and antitumour effects with minimal toxicity and should be investigated further.


Angiogenesis Inhibitors/metabolism , Cystadenocarcinoma, Serous/prevention & control , Fatty Acid-Binding Proteins/antagonists & inhibitors , Neovascularization, Pathologic/prevention & control , Ovarian Neoplasms/prevention & control , Animals , Apoptosis , Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Cystadenocarcinoma, Serous/blood supply , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Follow-Up Studies , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Mice, Nude , Neoplasm Grading , Neoplasm Invasiveness , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , Prospective Studies , Receptor, Notch1/metabolism , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
4.
Oncogene ; 36(10): 1339-1350, 2017 03.
Article En | MEDLINE | ID: mdl-27669434

KRas is mutated in a significant number of human cancers and so there is an urgent therapeutic need to target KRas signalling. To target KRas in lung cancers we used a systems approach of integrating a genome-wide miRNA screen with patient-derived phospho-proteomic signatures of the KRas downstream pathway, and identified miR-193a-3p, which directly targets KRas. Unique aspects of miR-193a-3p biology include two functionally independent target sites in the KRas 3'UTR and clinically significant correlation between miR-193a-3p and KRas expression in patients. Rescue experiments with mutated KRas 3'UTR showed very significantly that the anti-tumour effect of miR-193a-3p is via specific direct targeting of KRas and not due to other targets. Ex vivo and in vivo studies utilizing nanoliposome packaged miR-193a-3p demonstrated significant inhibition of tumour growth, circulating tumour cell viability and decreased metastasis. These studies show the broader applicability of using miR-193a-3p as a therapeutic agent to target KRas-mutant cancer.


Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Cells, Circulating/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RNA Interference , 3' Untranslated Regions , Cell Cycle , Cell Line, Tumor , Databases, Genetic , Genes, Reporter , Humans , Neoplasm Metastasis , Neoplastic Cells, Circulating/pathology , RNA, Messenger/genetics
6.
Oncogene ; 35(33): 4312-20, 2016 08 18.
Article En | MEDLINE | ID: mdl-26725326

MicroRNAs (miRNAs) are small RNA molecules that affect cellular processes by controlling gene expression. Recent studies have shown that hypoxia downregulates Drosha and Dicer, key enzymes in miRNA biogenesis, causing a decreased pool of miRNAs in cancer and resulting in increased tumor growth and metastasis. Here we demonstrate a previously unrecognized mechanism by which hypoxia downregulates Dicer. We found that miR-630, which is upregulated under hypoxic conditions, targets and downregulates Dicer expression. In an orthotopic mouse model of ovarian cancer, delivery of miR-630 using 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC) nanoliposomes resulted in increased tumor growth and metastasis, and decreased Dicer expression. Treatment with the combination of anti-miR-630 and anti-vascular endothelial growth factor antibody in mice resulted in rescue of Dicer expression and significantly decreased tumor growth and metastasis. These results indicate that targeting miR-630 is a promising approach to overcome Dicer deregulation in cancer. As demonstrated in the study, use of DOPC nanoliposomes for anti-miR delivery serves as a better alternative approach to cell line-based overexpression of sense or antisense miRNAs, while avoiding potential in vitro selection effects. Findings from this study provide a new understanding of miRNA biogenesis downregulation observed under hypoxia and suggest therapeutic avenues to target this dysregulation in cancer.


Cell Hypoxia , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/physiology , Neoplasms/etiology , Ribonuclease III/genetics , Animals , Cell Line, Tumor , Disease Progression , Female , Humans , Liposomes , Mice , MicroRNAs/antagonists & inhibitors , Neoplasms/therapy , Ovarian Neoplasms/metabolism , Phosphatidylcholines/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Oncogene ; 35(18): 2390-7, 2016 05 05.
Article En | MEDLINE | ID: mdl-26257064

Adrenergic stimulation adversely affects tumor growth and metastasis, but the underlying mechanisms are not well understood. Here, we uncovered a novel mechanism by which catecholamines induce inflammation by increasing prostaglandin E2 (PGE2) levels in ovarian cancer cells. Metabolic changes in tumors isolated from patients with depression and mice subjected to restraint stress showed elevated PGE2 levels. Increased metabolites, PTGS2 and PTGES protein levels were found in Skov3-ip1 and HeyA8 cells treated with norepinephrine (NE), and these changes were shown to be mediated by ADRB2 receptor signaling. Silencing PTGS2 resulted in significantly decreased migration and invasion in ovarian cancer cells in the presence of NE and decreased tumor burden and metastasis in restraint stress orthotopic models. In human ovarian cancer samples, concurrent increased ADRB2, PTGS2 and PTGES expression was associated with reduced overall and progression-free patient survival. In conclusion, increased adrenergic stimulation results in increased PGE2 synthesis via ADRB2-Nf-kB-PTGS2 axis, which drives tumor growth and metastasis.


Dinoprostone/biosynthesis , Norepinephrine/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Animals , Cell Line, Tumor , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/genetics , Female , Gene Silencing , Humans , Mice , Neoplasm Metastasis , Prostaglandin-E Synthases/metabolism
8.
Oncogene ; 35(6): 691-701, 2016 Feb 11.
Article En | MEDLINE | ID: mdl-25639871

The myc oncogene is overexpressed in almost half of all breast and ovarian cancers, but attempts at therapeutic interventions against myc have proven to be challenging. Myc regulates multiple biological processes, including the cell cycle, and as such is associated with cell proliferation and tumor progression. We identified a protein signature of high myc, low p27 and high phospho-Rb significantly correlated with poor patient survival in breast and ovarian cancers. Screening of a miRNA library by functional proteomics in multiple cell lines and integration of data from patient tumors revealed a panel of five microRNAs (miRNAs) (miR-124, miR-365, miR-34b*, miR-18a and miR-506) as potential tumor suppressors capable of reversing the p27/myc/phospho-Rb protein signature. Mechanistic studies revealed an RNA-activation function of miR-124 resulting in direct induction of p27 protein levels by binding to and inducing transcription on the p27 promoter region leading to a subsequent G1 arrest. Additionally, in vivo studies utilizing a xenograft model demonstrated that nanoparticle-mediated delivery of miR-124 could reduce tumor growth and sensitize cells to etoposide, suggesting a clinical application of miRNAs as therapeutics to target the functional effect of myc on tumor growth.


Breast Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression Regulation, Neoplastic , Genes, myc , MicroRNAs/physiology , Ovarian Neoplasms/genetics , Retinoblastoma Protein/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Phosphoproteins/metabolism , Proteomics/methods , Retinoblastoma Protein/metabolism , Transcriptome , Tumor Cells, Cultured
9.
Oncogene ; 35(17): 2186-96, 2016 04 28.
Article En | MEDLINE | ID: mdl-26212011

In the absence of extracellular stimulation the adaptor protein growth factor receptor-bound protein (Grb2) and the phospholipase Plcγ1 compete for the same binding site on fibroblast growth factor receptor 2 (FGFR2). Reducing cellular Grb2 results in upregulation of Plcγ1 and depletion of the phospholipid PI(4,5)P2. The functional consequences of this event on signaling pathways are unknown. We show that the decrease in PI(4,5)P2 level under non-stimulated conditions inhibits PTEN activity leading to the aberrant activation of the oncoprotein Akt. This results in excessive cell proliferation and tumor progression in a xenograft mouse model. As well as defining a novel mechanism of Akt phosphorylation with important therapeutic consequences, we also demonstrate that differential expression levels of FGFR2, Plcγ1 and Grb2 correlate with patient survival. Oncogenesis through fluctuation in the expression levels of these proteins negates extracellular stimulation or mutation and defines them as novel prognostic markers in ovarian cancer.


GRB2 Adaptor Protein/genetics , Oncogene Protein v-akt/genetics , Ovarian Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Phospholipase C gamma/genetics , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Female , GRB2 Adaptor Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Humans , Mice , Ovarian Neoplasms/pathology , PTEN Phosphohydrolase/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositols/metabolism , Phospholipase C gamma/biosynthesis , Prognosis , Receptor, Fibroblast Growth Factor, Type 2/biosynthesis , Signal Transduction
10.
J BUON ; 20(6): 1471-9, 2015.
Article En | MEDLINE | ID: mdl-26854443

PURPOSE: To evaluate the effectiveness of a neutral DOPC nanoliposome system for the delivery of siRNA to tumor cells in an obese murine cervical cancer model. METHODS: In vitro silencing of E6-E7 mRNA and E7 protein using siRNAE6 or siRNAE7 was analyzed in TC-1 cells by RT-PCR and Western blot. Silencing and antitumor capacities of siRNAE7-DOPC-nanoparticles (NP) were tested in vivo in both normal and obese mice using qPCR. These NPs were administered twice a week for 15 days and tumor volume and weight were recorded. RESULTS: Levels of in vitro E6-E7 silencing were 90% for mRNA and 60% for protein when siRNAE7 was used. On the other hand when siRNAE6 was used, the levels of silencing were 50% for E6-E7 mRNA and only 20% for protein. In vivo E7 mRNA silencing by siRNAE7-DOPC-NP was similar (60%) in both non-obese and obese mouse models. The therapeutic study showed a 65% decrease in tumor volume and a 57% reduction in tumor weight as compared to the control groups. CONCLUSION: There was no negative impact of obesity on the antitumor activity of siRNA-DOPC-NP in obese mice.


Obesity/complications , Papillomavirus E7 Proteins/genetics , Phosphatidylcholines/administration & dosage , RNA, Small Interfering/genetics , Uterine Cervical Neoplasms/therapy , Animals , Female , Gene Silencing , Liposomes , Mice , Mice, Inbred C57BL , Nanoparticles , Uterine Cervical Neoplasms/virology
11.
J Intern Med ; 267(1): 44-53, 2010 Jan.
Article En | MEDLINE | ID: mdl-20059643

Small interfering RNA (siRNA) technology holds great promise as a therapeutic intervention for targeted gene silencing in cancer and other diseases. However, in vivo systemic delivery of siRNA-based therapeutics to tumour tissues/cells remains a challenge. The major limitations against the use of siRNA as a therapeutic tool are its degradation by serum nucleases, poor cellular uptake and rapid renal clearance following systemic administration. Several siRNA-based loco-regional therapeutics are already in clinical trials. Further development of siRNAs for anti-cancer therapy depends on the development of safe and effective nanocarriers for systemic administration. To overcome these hurdles, nuclease-resistant chemically modified siRNAs and variety of synthetic and natural biodegradable lipids and polymers have been developed to systemically deliver siRNA with different efficacy and safety profiles. Cationic liposomes have emerged as one of the most attractive carriers because of their ability to form complexes with negatively charged siRNA and high in vitro transfection efficiency. However, their effectiveness as potential therapeutic carriers is limited by potential for pulmonary toxicity. Recently, our laboratories described the use of neutral 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine based nanoliposomes in murine tumour models. We found this approach to be safe and 10- and 30-fold more effective than cationic liposomes and naked siRNA, respectively, for systemic delivery of siRNA into tumour tissues. Here, we review potential approaches for systemic delivery of siRNA for cancer therapy.


Lipids/administration & dosage , Nanocapsules/administration & dosage , Neoplasms/drug therapy , RNA, Small Interfering/administration & dosage , Animals , Down-Regulation , Gene Silencing , Humans , Lipids/chemistry , Lipids/pharmacokinetics , Liposomes , Mice , Mice, Nude , Nanocapsules/chemistry , Nanomedicine/methods , Neoplasms/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/genetics
12.
Neuroscience ; 158(4): 1577-88, 2009 Feb 18.
Article En | MEDLINE | ID: mdl-19110040

Activation of muscarinic acetylcholine receptors (mAChRs) inhibits spinal nociceptive transmission by potentiation of GABAergic tone through M(2), M(3), and M(4) subtypes. To study the signaling mechanisms involved in this unique mAChR action, GABAergic spontaneous inhibitory postsynaptic currents (sIPSCs) of lamina II neurons were recorded using whole-cell patch clamp techniques in rat spinal cord slices. The mAChR agonist oxotremorine-M caused a profound increase in the frequency of GABAergic sIPSCs, which was abolished in the Ca(2+)-free solution. Inhibition of voltage-gated Ca(2+) channels with Cd(2+) and Ni(2+) largely reduced the effect of oxotremorine-M on sIPSCs. Blocking nonselective cation channels (NSCCs) with SKF96365 or 2-APB also largely attenuated the effect of oxotremorine-M. However, the KCNQ channel blocker XE991 and the adenylyl cyclase inhibitor MDL12330A had no significant effect on oxotremorine-M-induced increases in sIPSCs. Furthermore, the phosphoinositide-3-kinase (PI3K) inhibitor wortmannin or LY294002 significantly reduced the potentiating effect of oxotremorine-M on sIPSCs. In the spinal cord in which the M(3) subtype was specifically knocked down by intrathecal small interfering RNA (siRNA) treatment, SKF96365 and wortmannin still significantly attenuated the effect of oxotremorine-M. In contrast, SKF96365 and wortmannin both failed to alter the effect of oxotremorine-M on sIPSCs when the M(2)/M(4) mAChRs were blocked. Therefore, our study provides new evidence that activation of mAChRs increases synaptic GABA release through Ca(2+) influx and voltage-gated Ca(2+) channels. The PI3K-NSCC signaling cascade is primarily involved in the excitation of GABAergic interneurons by the M(2)/M(4) mAChRs in the spinal dorsal horn.


Inhibitory Postsynaptic Potentials/physiology , Neurons/physiology , Receptors, Muscarinic/physiology , Signal Transduction/physiology , Spinal Cord/cytology , gamma-Aminobutyric Acid/metabolism , Animals , Anthracenes/pharmacology , Biophysics , Cadmium/pharmacology , Calcium/metabolism , Drug Interactions , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Ion Channel Gating/drug effects , Ion Channel Gating/physiology , Male , Muscarinic Agonists/pharmacology , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons/drug effects , Nickel/pharmacology , Oxotremorine/analogs & derivatives , Oxotremorine/pharmacology , Patch-Clamp Techniques/methods , Potassium Channel Blockers/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/classification , Receptors, Muscarinic/genetics , Signal Transduction/drug effects
13.
Oncogene ; 27(57): 7192-200, 2008 Dec 04.
Article En | MEDLINE | ID: mdl-18806828

During development inhibitor of DNA-bind-2 (Id2) regulates proliferation and differentiation. Id2 expression has been detected in cancer cells, yet its cellular function and validity as a therapeutic target remains largely unknown. Immunohistochemical analysis of colorectal cancer (CRC) specimens revealed that Id2 was undetectable in normal colonic mucosa, but occurs in 40% of primary tumors and in most CRC liver metastases (P<0.0001). Additionally, Id2 was expressed in all CRC cell lines assayed. CRC cells with reduced Id2 expression demonstrated reduced proliferation. Analysis of CRC cell cycle regulatory proteins showed that reducing Id2 levels reduces cyclin D1 levels and increased p21 levels. Reduction of Id2 expression also enhanced tumor cell apoptosis, increasing levels of the pro-apoptotic protein Bim/Bod, and cleavage of caspase-7 and poly (ADP-ribose) polymerase. In vivo studies show tumors derived from cells with decreased Id2 levels formed smaller tumors with fewer metastases compared with tumors with normal levels (P<0.05). Furthermore, intraperitoneal administration of Id2 small interfering RNA (siRNA) conjugated with the neutral liposome 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine decreased tumor burden in mice compared with control treatment (P=0.006). We conclude that Id2 is upregulated in CRC, and is important in promoting cell survival. In vivo targeting of Id2 by siRNA establishes that it is a valid therapeutic target where its expression occurs.


Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Inhibitor of Differentiation Protein 2/metabolism , Liver Neoplasms/secondary , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Apoptosis/physiology , Autoradiography , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Flow Cytometry , Humans , Immunohistochemistry , Immunoprecipitation , Inhibitor of Differentiation Protein 2/genetics , Mice , RNA, Small Interfering , Signal Transduction/physiology , Up-Regulation , Xenograft Model Antitumor Assays
14.
Cancer Gene Ther ; 13(7): 706-19, 2006 Jul.
Article En | MEDLINE | ID: mdl-16514421

To ensure the success of systemic gene therapy, it is critical to enhance the tumor specificity and activity of the promoter. In the current study, we determined that topoisomerase IIalpha promoter is selectively activated in breast cancer cells. An element containing an inverted CCAAT box (ICB) was shown to be responsible for the breast cancer specificity. When the ICB-harboring topoisomerase IIalpha minimal promoter was linked with an enhancer sequence from the cytomegalovirus immediate early gene promoter (CMV promoter), this composite promoter, CT90, exhibited activity comparable to or higher than the CMV promoter in breast cancer cells in vitro and in vivo, yet expresses much lower activity in normal cell lines and normal organs than the CMV promoter. A CT90-driven construct expressing BikDD, a potent proapoptotic gene, was shown to selectively kill breast cancer cells in vitro, and to suppress mammary tumor development in an animal model of intravenously administrated, liposome-delivered gene therapy. Expression of BikDD was readily detectable in the tumors but not in the normal organs (such as heart) of CT90-BikDD-treated animals. The results indicate that liposomal CT90-BikDD is an effective systemic breast cancer-targeting gene therapy.


Antigens, Neoplasm/genetics , Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/therapy , DNA Topoisomerases, Type II/genetics , DNA-Binding Proteins/genetics , Genetic Therapy , Membrane Proteins/genetics , Mutation , Response Elements/genetics , Animals , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/genetics , Cell Line, Tumor , Cytomegalovirus/genetics , Enhancer Elements, Genetic/genetics , Female , Gene Expression , Genetic Vectors , Humans , Liposomes , Membrane Proteins/metabolism , Mice , Mice, Nude , Mitochondrial Proteins , Neoplasms, Experimental
15.
Leuk Lymphoma ; 43(5): 933-41, 2002 May.
Article En | MEDLINE | ID: mdl-12148903

Acute promyelocytic leukemia (APL) characterized by the translocation t(15;17) is uniquely sensitive to the differentiation-inducing effects of all-trans-retinoic acid (ATRA). All-trans-retinoic acid therapy induces complete clinical remissions (CRs) in most of patients with APL. However, chronic daily oral administration of ATRA results in accelerated metabolism of ATRA, leading to a progressive decline in plasma drug concentrations. These lower drug levels are associated with relapses and resistance to oral ATRA in patients with APL; thus the use of ATRA as a single agent is precluded. Liposomal ATRA (Lipo-ATRA) was designed to maintain high and stable plasma concentrations and to further improve the outcome of the APL disease by overcoming the development of ATRA resistance. Liposomal ATRA was shown to circumvent accelerated drug metabolism in the liver of rats in an animal model. In a phase I clinical study, intravenous (i.v.) administration of lipo-ATRA was shown to produce a significantly better pharmacokinetic profile than oral ATRA (non-liposomal) and to maintain higher and sustained plasma drug concentrations, with a similar side effects. More importantly, lipo-ATRA as a single agent induces PCR-negative molecular remissions in a high proportion of newly diagnosed patients with APL and maintain remissions up to 15-17 months or longer. In this review, we discuss the pharmacological features of lipo-ATRA and the molecular remissions induced by lipo-ATRA in newly diagnosed patients with APL or patients previously treated with ATRA or chemotherapy, and the possible impact of lipo-ATRA on the outcome of APL.


Antineoplastic Agents/administration & dosage , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/administration & dosage , Arsenic Trioxide , Arsenicals/therapeutic use , Cell Differentiation/drug effects , Clinical Trials as Topic , Drug Carriers , Histone Deacetylase Inhibitors , Humans , Liposomes , Oxides/therapeutic use , Tretinoin/pharmacology
16.
Semin Oncol ; 28(5 Suppl 16): 142-7, 2001 Oct.
Article En | MEDLINE | ID: mdl-11706405

The growth factor receptor-bound protein-2 (GRB2) is essential for multiple cellular functions. Inhibition of GRB2 function impairs developmental processes in various organisms and blocks transformation and proliferation of various cell types. GRB2 is most well known for its ability to link the epidermal growth factor receptor tyrosine kinase to the activation of RAS and its downstream kinases, ERK1,2. We recently reported that GRB2 does not link the HER2 tyrosine kinase to the activation of ERK1,2 but to another kinase, AKT. So, different tyrosine kinases may converge on GRB2 for signaling; however, they may not always use GRB2 to effect the same downstream kinases for cellular functions. It is likely that GRB2 will be found to regulate many more kinases because it plays such a pivotal role in signal transduction.


Adaptor Proteins, Signal Transducing , ErbB Receptors/metabolism , Neoplasms/genetics , Protein Serine-Threonine Kinases , Proteins/physiology , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology , Animals , GRB2 Adaptor Protein , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/pathology , Proteins/antagonists & inhibitors , Proteins/genetics , Proto-Oncogene Proteins c-akt , src Homology Domains
17.
J Biol Regul Homeost Agents ; 15(2): 107-22, 2001.
Article En | MEDLINE | ID: mdl-11501968

Acute promyelocytic leukemia (APL) is a unique disease that responds to differentiation-inducing effects of all-trans-retinoic acid (ATRA). ATRA induces complete clinical remissions (CRs) in most patients and now constitutes a standard therapy in patients with APL. However, CRs induced by ATRA are usually brief, and resistance to the therapy rapidly develops, leading to relapses in almost every patient; thus limiting the use of ATRA as a single agent. On the basis of clinical and in vitro studies, the following mechanisms have been proposed to explain ATRA resistance: 1) induction of accelerated metabolism of ATRA, 2) increased expression of cellular retinoic acid-binding proteins (CRABPs), 3) constitutive degradation of PML-RAR alpha, 4) point mutations in the ligand-binding domain of RAR alpha of PML-RAR alpha, 5) P-glycoprotein expression, 6) transcriptional repression by histone deacetylase activity, 7) isoforms of PML-RAR alpha, 8) persistent telomerase activity, and 9) expression of type II transglutaminase. In this review, we discuss the evidence provided in support of each mechanism, the mechanism's possible impact on the outcome of APL, and the newer approaches that are being employed to overcome ATRA resistance.


Antineoplastic Agents/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/therapeutic use , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/metabolism , Arsenic Trioxide , Arsenicals/therapeutic use , Drug Resistance, Neoplasm , GTP-Binding Proteins/genetics , Gene Expression/drug effects , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Humans , In Vitro Techniques , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Mutation , Oxides/therapeutic use , Protein Glutamine gamma Glutamyltransferase 2 , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Telomerase/metabolism , Transglutaminases/genetics , Tretinoin/administration & dosage , Tretinoin/metabolism
18.
J Clin Oncol ; 19(14): 3422-33, 2001 Jul 15.
Article En | MEDLINE | ID: mdl-11454891

PURPOSE: Preclinical studies have demonstrated that the adenovirus type 5 E1A gene is associated with antitumor activities by transcriptional repression of HER-2/neu and induction of apoptosis. Indeed, E1A gene therapy is known to induce regression of HER-2/neu-overexpressing breast and ovarian cancers in nude mice. Therefore, we evaluated the feasibility of intracavitary injection of E1A gene complexed with DC-Chol cationic liposome (DCC-E1A) in patients with both HER-2/neu-overexpressing and low HER-2/neu-expressing breast and ovarian cancers in a phase I clinical trial. PATIENTS AND METHODS: An E1A gene complexed with DCC-E1A cationic liposome was injected once a week into the thoracic or peritoneal cavity of 18 patients with advanced cancer of the breast (n = 6) or ovary (n = 12). RESULTS: E1A gene expression in tumor cells was detected by immunohistochemical staining and reverse transcriptase-polymerase chain reaction. This E1A gene expression was accompanied by HER-2/neu downregulation, increased apoptosis, and reduced proliferation. The most common treatment-related toxicities were fever, nausea, vomiting, and/or discomfort at the injection sites. CONCLUSION: These results argue for the feasibility of intracavitary DCC-E1A administration, provide a clear proof of preclinical concept, and warrant phase II trials to determine the antitumor activity of the E1A gene.


Adenovirus E1A Proteins/genetics , Breast Neoplasms/therapy , Gene Transfer, Horizontal , Genetic Therapy , Ovarian Neoplasms/therapy , Adult , Aged , Apoptosis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cholesterol/analogs & derivatives , Cytokines/metabolism , Female , Gene Expression , Genes, erbB-2 , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Injections , Ki-67 Antigen , Liposomes , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Peritoneal Cavity , Reverse Transcriptase Polymerase Chain Reaction , Thorax , Tumor Cells, Cultured
20.
Clin Infect Dis ; 33(1): 95-106, 2001 07 01.
Article En | MEDLINE | ID: mdl-11389501

Studies of invasive fungal infections have been and remain difficult to implement. Randomized clinical trials of fungal infections are especially slow and expensive to perform because it is difficult to identify eligible patients in a timely fashion, to prove the presence of the fungal infection in an unequivocal fashion, and to evaluate outcome in a convincing fashion. Because of these challenges, licensing decisions for antifungal agents have to date depended heavily on historical control comparisons and secondary advantages of the new agent. Although the availability of newer and potentially more effective agents makes these approaches less desirable, the fundamental difficulties of trials of invasive fungal infections have not changed. Therefore, there is a need for alternative trial designs and evaluation strategies for therapeutic studies of invasive mycoses, and this article summarizes the possible strategies in this area.


Antifungal Agents/therapeutic use , Controlled Clinical Trials as Topic/methods , Mycoses/drug therapy , Randomized Controlled Trials as Topic/methods , Research Design , Humans , Treatment Outcome
...