Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Breast Cancer Res Treat ; 153(2): 285-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26268696

ABSTRACT

Endoplasmic reticulum (ER) stress leads to activation of the unfolded protein response (UPR) signaling cascade and induction of an apoptotic cell death, autophagy, oncogenesis, metastasis, and/or resistance to cancer therapies. Mechanisms underlying regulation of ER transmembrane proteins PERK, IRE1α, and ATF6α/ß, and how the balance of these activities determines outcome of the activated UPR, remain largely unclear. Here, we report a novel molecule transmembrane protein 33 (TMEM33) and its actions in UPR signaling. Immunoblotting and northern blot hybridization assays were used to determine the effects of ER stress on TMEM33 expression levels in various cell lines. Transient transfections, immunofluorescence, subcellular fractionation, immunoprecipitation, and immunoblotting were used to study the subcellular localization of TMEM33, the binding partners of TMEM33, and the expression of downstream effectors of PERK and IRE1α. Our data demonstrate that TMEM33 is a unique ER stress-inducible and ER transmembrane molecule, and a new binding partner of PERK. Exogenous expression of TMEM33 led to increased expression of p-eIF2α and p-IRE1α and their known downstream effectors, ATF4-CHOP and XBP1-S, respectively, in breast cancer cells. TMEM33 overexpression also correlated with increased expression of apoptotic signals including cleaved caspase-7 and cleaved PARP, and an autophagosome protein LC3II, and reduced expression of the autophagy marker p62. TMEM33 is a novel regulator of the PERK-eIE2α-ATF4 and IRE1-XBP1 axes of the UPR signaling. Therefore, TMEM33 may function as a determinant of the ER stress-responsive events in cancer cells.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Signal Transduction , Unfolded Protein Response , Activating Transcription Factor 4/metabolism , Amino Acid Sequence , Animals , Autophagy/genetics , Base Sequence , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression , Humans , Membrane Proteins/chemistry , Molecular Sequence Data , Protein Serine-Threonine Kinases/metabolism , Protein Transport , Recombinant Fusion Proteins , Regulatory Factor X Transcription Factors , Transcription Factors/metabolism , X-Box Binding Protein 1 , eIF-2 Kinase/metabolism
2.
Int J Cancer ; 133(1): 31-42, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23280553

ABSTRACT

TNFAIP8 is a NF-κB-inducible, oncogenic molecule. Previous "promoter array" studies have identified differential methylation and regulation of TNFAIP8 in prostate epithelial and cancer cell lines. Here we demonstrate that TNFAIP8 expression is induced by androgen in hormone-responsive LNCaP prostate cancer cells. In athymic mice bearing hormone-refractory PC-3 prostate tumor xenografts, intravenous treatment with a liposomal formulation of TNFAIP8 antisense oligonucleotide (LE-AS5) caused reduced expression of TNFAIP8 in tumor tissues, and a combination of LE-AS5 and radiation or docetaxel treatment resulted in significant inhibition of PC-3 tumor growth as compared to single agents. The immunohistochemical evaluation of TNFAIP8 expression revealed correlation of both cytoplasmic and nuclear TNFAIP8 overexpression with high grade prostatic adenocarcinomas, while nuclear overexpression was found to be an independent predictor of disease recurrence controlling for tumor grade. Increased nuclear TNFAIP8 expression was statistically significantly associated with a 2.44 fold (95 % confidence interval: 1.01-5.91) higher risk of prostate cancer recurrence. Mechanistically, TNFAIP8 seems to function as a scaffold (or adaptor) protein. In the antibody microarray analysis of proteins associated with the TNFAIP8 immune-complex, we have identified Karyopherin alpha2 as a novel binding partner of nuclear TNFAIP8 in PC-3 cells. The Ingenuity Pathway Analysis of the TNFAIP8 interacting proteins suggested that TNFAIP8 influences cancer progression pathways and networks involving integrins and matrix metalloproteinases. Taken together, present studies demonstrate that TNFAIP8 is a novel therapeutic target in prostate cancer, and indicate a potential relationship of the nuclear trafficking of TNFAIP8 with adverse outcomes in a subset of prostate cancer patients.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/radiotherapy , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Biomarkers, Tumor/metabolism , Oligonucleotides, Antisense/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Taxoids/therapeutic use , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Chemotherapy, Adjuvant , Disease Progression , Docetaxel , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Immunohistochemistry , Liposomes , Male , Mice , Mice, Nude , Neoplasm Grading , Oligonucleotides, Antisense/chemical synthesis , Predictive Value of Tests , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Array Analysis , Radiotherapy, Adjuvant , Transplantation, Heterologous , Up-Regulation
3.
Methods Mol Biol ; 2660: 295-310, 2023.
Article in English | MEDLINE | ID: mdl-37191806

ABSTRACT

DEPDC1B (aliases BRCC3, XTP8, XTP1) is a DEP (Dishevelled, Egl-1, Pleckstrin) and Rho-GAP-like domains containing predominately membrane-associated protein. Earlier, we and others have reported that DEPDC1B is a downstream effector of Raf-1 and long noncoding RNA lncNB1, and an upstream positive effector of pERK. Consistently, DEPDC1B knockdown is associated with downregulation of ligand-stimulated pERK expression. We demonstrate here that DEPDC1B N-terminus binds to the p85 subunit of PI3K, and DEPDC1B overexpression results in decreased ligand-stimulated tyrosine phosphorylation of p85 and downregulation of pAKT1. Collectively, we propose that DEPDC1B is a novel cross-regulator of AKT1 and ERK, two of the prominent pathways of tumor progression. Our data showing high levels of DEPDC1B mRNA and protein during the G2/M phase have significant implications in cell entry into mitosis. Indeed, DEPDC1B accumulation during the G2/M phase has been associated with disassembly of focal adhesions and cell de-adhesion, referred to as a DEPDC1B-mediated de-adhesion mitotic checkpoint. DEPDC1B is a direct target of transcription factor SOX10, and SOX10-DEPDC1B-SCUBE3 axis has been associated with angiogenesis and metastasis. The Scansite analysis of the DEPDC1B amino acid sequence shows binding motifs for three well-established cancer therapeutic targets CDK1, DNA-PK, and aurora kinase A/B. These interactions and functionalities, if validated, may further implicate DEPDC1B in regulation of DNA damage-repair and cell cycle progression processes. Finally, a survey of the publicly available datasets indicates that high DEPDC1B expression is a viable biomarker in breast, lung, pancreatic and renal cell carcinomas, and melanoma. Currently, the systems and integrative biology of DEPDC1B is far from comprehensive. Future investigations are necessary in order to understand how DEPDC1B might impact AKT, ERK, and other networks, albeit in a context-dependent manner, and influence the actionable molecular, spatial, and temporal vulnerabilities within these networks in cancer cells.


Subject(s)
Cell Cycle Proteins , Mitosis , Cell Line, Tumor , Ligands , Cell Cycle , Cell Cycle Proteins/metabolism
4.
Methods Mol Biol ; 480: 65-83, 2009.
Article in English | MEDLINE | ID: mdl-19085118

ABSTRACT

By virtue of their potential to selectively silence oncogenic molecules in cancer cells, antisense oligonucleotides (ASO) and small interfering RNAs (siRNAs) are powerful tools for development of tailored anti-cancer drugs. The clinical benefit of ASO/siRNA therapeutic is, however, hampered due to poor pharmacokinetics and biodistribution, and suboptimal suppression of the target in tumor tissues. Raf-1 protein serine/threonine kinase is a druggable signaling molecule in cancer therapy. Our laboratory has developed cationic liposomes for systemic delivery of raf ASO (LErafAON) and raf siRNA (LErafsiRNA) to human tumor xenografts grown in athymic mice. LErafAON is also the first ASO containing liposomal drug tested in humans. In this article, we primarily focus on a modified formulation of systemically delivered cationic liposomes containing raf antisense oligonucleotide (md-LErafAON). The cationic liposomes were prepared using dimyristoyl 1,2-diacyl-3-trimethylammonium-propane (DMTAP), phosphatidylcholine (PC), and cholesterol (CHOL). The toxicology, pharmacokinetics, biodistribution, target selectivity, and anti-tumor efficacy studies of md-LErafAON were conducted in mice. We demonstrate that md-LErafAON is the next generation of systemically delivered and well-tolerated antisense therapeutic suitable for clinical evaluation.


Subject(s)
Drug Evaluation, Preclinical , Nanoparticles , Oligonucleotides, Antisense/administration & dosage , RNA, Small Interfering/administration & dosage , Animals , Base Sequence , Cations , Gene Silencing , Humans , Liposomes , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacokinetics , RNA, Small Interfering/pharmacokinetics , Tissue Distribution
5.
Mol Cancer Res ; 17(5): 1207-1219, 2019 05.
Article in English | MEDLINE | ID: mdl-30647104

ABSTRACT

Aberrant regulation of EGFR is common in non-small cell lung carcinomas (NSCLC), and tumor resistance to targeted therapies has been attributed to emergence of other co-occurring oncogenic events, parallel bypass receptor tyrosine kinase pathways including IGF1R, and TNFα-driven adaptive response via NF-κB. TNFAIP8, TNFα-inducible protein 8, is an NF-κB-activated prosurvival and oncogenic molecule. TNFAIP8 expression protects NF-κB-null cells from TNFα-induced cell death by inhibiting caspase-8 activity. Here, we demonstrate that knockdown of TNFAIP8 inhibited EGF and IGF-1-stimulated migration in NSCLC cells. TNFAIP8 knockdown cells showed decreased level of EGFR and increased expression of sorting nexin 1 (SNX1), a key regulator of the EGFR trafficking through the endosomal compartments, and treatment with SNX1 siRNA partially restored EGFR expression in these cells. TNFAIP8 knockdown cells also exhibited downregulation of IGF-1-induced pIGF1R and pAKT, and increased expression of IGF-1-binding protein 3 (IGFBP3), a negative regulator of the IGF-1/IGF1R signaling. Consistently, treatment of TNFAIP8 knockdown cells with IGFBP3 siRNA restored pIGF1R and pAKT levels. TNFAIP8 knockdown cells had enhanced sensitivities to inhibitors of EGFR, PI3K, and AKT. Furthermore, IHC expression of TNFAIP8 was associated with poor prognosis in NSCLC. These findings demonstrate TNFAIP8-mediated regulation of EGFR and IGF1R via SNX1 and IGFBP3, respectively. We posit that TNFAIP8 is a viable, multipronged target downstream of the TNFα/NF-κB axis, and silencing TNFAIP8 may overcome adaptive response in NSCLC. IMPLICATIONS: TNFAIP8 and its effectors SNX1 and IGFBP3 may be exploited to improve the efficacy of molecular-targeted therapies in NSCLC and other cancers.Visual Overview: http://mcr.aacrjournals.org/content/molcanres/17/5/1207/F1.large.jpg.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , RNA, Small Interfering/pharmacology , Receptor, IGF Type 1/metabolism , A549 Cells , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Insulin-Like Growth Factor Binding Protein 3/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Receptor, IGF Type 1/antagonists & inhibitors , Receptor, IGF Type 1/genetics , Signal Transduction , Sorting Nexins/genetics , Sorting Nexins/metabolism
6.
Methods Mol Biol ; 361: 163-85, 2007.
Article in English | MEDLINE | ID: mdl-17172711

ABSTRACT

Antisense oligonucleotides (ASO) against specific molecular targets (e.g., Bcl-2 and Raf-1) are important reagents in cancer biology and therapy. Phosphorothioate modification of the ASO backbone has resulted in an increased stability of ASO in vivo without compromising, in general, their target selectivity. Although the power of antisense technology remains unsurpassed, dose-limiting side effects of modified ASO and inadequate penetration into the tumor tissue have necessitated further improvements in ASO chemistry and delivery systems. Oligonucleotide delivery systems may increase stability of the unmodified or minimally modified ASO in plasma, enhance uptake of ASO by tumor tissue, and offer an improved therapy response. Here, we provide an overview of ASO design and in vivo delivery systems, and focus on preclinical validation of a liposomal nanoparticle containing minimally modified raf antisense oligodeoxynucleotide (LErafAON). Intact rafAON (15-mer) is present in plasma and in normal and tumor tissues of athymic mice systemically treated with LErafAON. Raf-1 expression is decreased in normal and tumor tissues of LErafAON-treated mice. Therapeutic benefit of a combination of LErafAON and radiation or an anticancer drug exceeds radiation or drug alone against human prostate, breast, and pancreatic tumors grown in athymic mice. Further improvements in ASO chemistry and nanoparticles are promising avenues in antisense therapy of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Nanoparticles , Neoplasms/metabolism , Oligodeoxyribonucleotides/pharmacology , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Combined Modality Therapy , Disease Models, Animal , Drug Carriers/chemistry , Drug Carriers/pharmacology , Drug Carriers/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Nude , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides/therapeutic use , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/therapeutic use , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-raf/biosynthesis , Proto-Oncogene Proteins c-raf/genetics , Thionucleotides/chemistry , Thionucleotides/genetics , Thionucleotides/pharmacology , Thionucleotides/therapeutic use
7.
Clin Cancer Res ; 12(4): 1251-9, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16489081

ABSTRACT

PURPOSE: Raf proteins are key elements of growth-related cellular signaling pathways and are a component of cancer cell resistance to radiation therapy. Antisense oligonucleotides to c-raf-1 permit highly selective inhibition of the gene product and offer a strategy for sensitizing cancer cells to radiation therapy. In this dose escalation study, we evaluated the safety of combined liposomal formulation of raf antisense oligonucleotide (LErafAON) and radiation therapy in patients with advanced malignancies. EXPERIMENTAL DESIGN: Patients with advanced solid tumors were treated with LErafAON in a phase I dose escalation study while receiving palliative radiation therapy. Drug-related and radiation-related toxicities were monitored. Pharmacokinetics and expression of c-raf-1 mRNA and Raf-1 protein were determined in peripheral blood mononuclear cells. RESULTS: Seventeen patients with palliative indications for radiation therapy were entered into this study. Thirteen patients received daily infusions of LErafAON and four received twice-weekly infusions. Radiation therapy was delivered in daily 300-cGy fractions over 2 weeks. Patients tolerated radiation, and no unexpected radiation-related side effects were observed. Drug-related reactions (grade > or =2), such as back pain, chills, dyspnea, fatigue, fever, flushing, and hypertension, were observed in most patients and were managed by premedication with corticosteroids and antihistamines. Serious adverse events occurred in five patients, including acute infusion-related symptoms, abnormal liver function tests, hypoxia, dehydration, diarrhea, esophagitis, fever, hypokalemia, pharyngitis, and tachypnea. Twelve of 17 patients were evaluable for tumor response at completion of treatment; four showed partial response, four showed stable disease, and four experienced progressive disease. The intact rafAON was detected in plasma for 30 minutes to several hours. Six patients with partial response or stable disease were evaluable for c-raf-1 mRNA and/or Raf-1 protein expression. Inhibition of c-raf-1 mRNA was observed in three of five patients. Raf-1 protein was inhibited in four of five patients. CONCLUSION: This is the first report of the combined modality treatment using antisense oligonucleotides with radiation therapy in patients with advanced cancer. A dose of 2.0 mg/kg of LErafAON administered twice weekly is tolerated with premedication and does not enhance radiation toxicity in patients. The observation of dose-dependent, infusion-related reactions has led to further modification of the liposomal composition for use in future clinical trials.


Subject(s)
Neoplasms/drug therapy , Neoplasms/radiotherapy , Oligodeoxyribonucleotides/administration & dosage , Abdominal Pain/etiology , Adult , Aged , Blotting, Western , Combined Modality Therapy/adverse effects , Diarrhea/etiology , Dose-Response Relationship, Drug , Female , Fever/etiology , Gene Expression/drug effects , Humans , Infusions, Intravenous , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Neoplasms/pathology , Oligodeoxyribonucleotides/adverse effects , Oligodeoxyribonucleotides/pharmacokinetics , Pharyngitis/etiology , Proto-Oncogene Proteins c-raf/blood , Proto-Oncogene Proteins c-raf/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
Methods Mol Biol ; 1513: 83-100, 2017.
Article in English | MEDLINE | ID: mdl-27807832

ABSTRACT

Tumor necrosis factor-α-inducible protein 8 (TNFAIP8) is the first discovered oncogenic and an anti-apoptotic member of a conserved TNFAIP8 or TIPE family of proteins. TNFAIP8 mRNA is induced by NF-kB, and overexpression of TNFAIP8 has been correlated with poor prognosis in many cancers. Downregulation of TNFAIP8 expression has been associated with decreased pulmonary colonization of human tumor cells, and enhanced sensitivities of tumor xenografts to radiation and docetaxel. Here we have investigated the effects of depletion of TNFAIP8 on the mRNA, microRNA and protein expression profiles in prostate and breast cancers and melanoma. Depending on the tumor cell type, knockdown of TNFAIP8 was found to be associated with increased mRNA expression of several antiproliferative and apoptotic genes (e.g., IL-24, FAT3, LPHN2, EPHA3) and fatty acid oxidation gene ACADL, and decreased mRNA levels of oncogenes (e.g., NFAT5, MALAT1, MET, FOXA1, KRAS, S100P, OSTF1) and glutamate transporter gene SLC1A1. TNFAIP8 knockdown cells also exhibited decreased expression of multiple onco-proteins (e.g., PIK3CA, SRC, EGFR, IL5, ABL1, GAP43), and increased expression of the orphan nuclear receptor NR4A1 and alpha 1 adaptin subunit of the adaptor-related protein complex 2 AP2 critical to clathrin-mediated endocytosis. TNFAIP8-centric molecules were found to be predominately implicated in the hypoxia-inducible factor-1α (HIF-1α) signaling pathway, and cancer and development signaling networks. Thus TNFAIP8 seems to regulate the cell survival and cancer progression processes in a multifaceted manner. Future validation of the molecules identified in this study is likely to lead to new subset of molecules and functional determinants of cancer cell survival and progression.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Prostatic Neoplasms/genetics , Proteomics/methods , Amino Acid Sequence , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Survival , Disease Progression , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Female , Humans , Male , Melanoma/diagnosis , Melanoma/metabolism , Melanoma/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Prognosis , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
9.
Int J Oncol ; 29(4): 861-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16964381

ABSTRACT

Raf-1 serine/threonine protein kinase plays an important role in cell growth, differentiation and cell survival. Recent reports using c-raf-1 gene-knockouts have observed MEK/ERK independent functions of Raf-1 in cell survival and protection from apoptosis. Raf-1 has also been shown to be involved in counteracting specific apoptotic pathways by restraining caspase activation, although the precise mechanism is unknown. XIAP is a potent inhibitor of apoptosis that blocks both the mitochondria and death receptor mediated pathways of apoptosis by directly binding to and inhibiting the initiator and effector caspases. In our efforts to understand the mechanism by which Raf-1 inhibits caspase activation, we discovered a novel interaction between Raf-1 and XIAP. In this study, we describe the physical interaction between Raf-1 and XIAP in vitro and in vivo in mammalian cells. We also demonstrate that Raf-1 phosphorylates XIAP in vitro and in vivo. Additionally, Raf-1 prevents XIAP degradation in response to different apoptotic triggers. Our studies identify XIAP as a new substrate of Raf-1 and provide potentially important insight into mechanisms underlying Raf-1 effects on cell survival.


Subject(s)
Proto-Oncogene Proteins c-raf/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Apoptosis , Humans , Immunoprecipitation , Proto-Oncogene Proteins c-raf/analysis , Tumor Cells, Cultured , X-Linked Inhibitor of Apoptosis Protein/analysis
10.
Int J Mol Med ; 17(3): 457-63, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16465392

ABSTRACT

Raf-1 protein serine-threonine kinase plays an important role in cell growth, proliferation, and cell survival. Previously, we and others have demonstrated that antisense raf oligonucleotide-mediated inhibition of Raf-1 expression leads to tumor growth arrest, radiosensitization and chemosensitization in vivo. Raf-1 inhibition is also associated with apoptotic cell death. In this study, we inhibited Raf-1 using an antisense raf oligonucleotide (AS-raf-ODN) to identify downstream targets of Raf-1 using microarray gene expression analysis. Treatment of MDA-MB-231 breast cancer cells with 250 nM AS-raf-ODN led to significant inhibition of Raf-1 protein (75.2 +/- 9.6%) and c-raf-1 mRNA levels (86.2 +/- 3.3%) as compared to untreated control cells. The lipofectin control or mismatch oligonucleotide had no effect on Raf-1 expression. To determine the changes in gene expression profiles that were due to inhibition of Raf-1, we simultaneously compared the gene expression patterns in AS-raf-ODN treated cells with untreated control cells and cells treated with lipofectin alone or MM-ODN. A total of 17 genes (4 upregulated and 13 down-regulated) including c-raf-1 were identified that were altered after AS-raf-ODN treatment. Functional clustering analysis revealed genes involved in apoptosis (Bcl-XL), cell adhesion (paxillin, plectin, Rho GDIalpha, CCL5), metabolism (GM2A, SLC16A3, PYGB), signal transduction (protein kinase C nu), and transcriptional regulation (HMGA1), and membrane-associated genes (GNAS, SLC16A3). Real-time PCR, Northern analysis and Western analysis confirmed the microarray findings. Our study provides insight into Raf-1 related signaling pathways and a model system to identify potential target genes.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Profiling , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Blotting, Northern , Blotting, Western , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Oligonucleotide Array Sequence Analysis , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-raf/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Int J Oncol ; 26(4): 1087-91, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15754006

ABSTRACT

Raf-1, a protein serine-threonine kinase, plays a critical role in mitogen-activated protein kinase kinase (MKK/MEK)- mitogen-activated protein kinase (extracellular signal-regulated kinase) (MAPK/ERK) pathways. We show here that systemically delivered novel cationic cardiolipin liposomes (NeoPhectin-AT) containing a small interfering RNA (siRNA) against Raf-1 silence the expression of Raf-1 in tumor tissues and inhibit tumor growth in xenograft model of human prostate cancer. The knockdown of Raf-1 expression by siRNA is also associated with down-regulation of cyclin D1 expression in vivo.


Subject(s)
Cardiolipins/pharmacology , Gene Silencing , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-raf/biosynthesis , RNA Interference , RNA, Small Interfering , Cations , Cyclin D1/biosynthesis , Down-Regulation , Humans , Liposomes , Male , Transplantation, Heterologous , Tumor Cells, Cultured
12.
Clin Cancer Res ; 8(11): 3611-21, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12429653

ABSTRACT

Raf-1 protein serine threonine kinase plays an important role in cell survival and proliferation. Antisense inhibition of Raf-1 expression has been shown to enhance the cytotoxic effects of radiation and anticancer drugs. Here we have evaluated the toxicity, pharmacokinetics, and antitumor efficacy of a novel formulation of liposome-entrapped raf antisense oligodeoxyribonucleotide (LErafAON). The LErafAON preparation showed high liposome entrapment efficiency of rafAON (>85%) and stability at room temperature. In CD2F1 mice, administration of LErafAON produced no morbidity/mortality (5-35 mg/kg/dose, i.v., x12). Dose-related elevations in liver enzymes (alanine aminotransferase and aspartate aminotransferase) and histopathological changes in liver were noted in LErafAON and blank liposome groups. No morbidity/mortality and changes in clinical chemistry or histopathology were observed in New Zealand white rabbits (3.75 mg/kg/dose, i.v., x8; 6.5 mg/kg/dose, i.v., x6) or in cynomolgous monkeys (3.75 or 6.25 mg/kg/dose, i.v., x9). Transient decrease in total hemolytic complement activity (approximately 62-74%) and increases in C3a (approximately 3-fold) and Bb levels (approximately 5-12-fold) were observed in LErafAON and blank liposome groups of monkeys. A 30 mg/kg i.v. dose of LErafAON in human prostate tumor (PC-3)-bearing BALB/c athymic mice gave a terminal plasma half-life of 27 h, and intact rafAON could be detected in plasma and in normal and tumor tissues for up to at least 48 h. In monkeys, the terminal plasma half-life of 30.36 +/- 23.87 h was observed at an i.v. dose of 6.25 mg/kg. LErafAON (25 mg/kg/dose, i.v., x10) or ionizing radiation (3.8 Gy/day, x5) treatment of PC-3 tumor-bearing athymic mice led to tumor growth arrest, whereas a combination of LErafAON and ionizing radiation treatments resulted in tumor regression. LErafAON treatment caused inhibition of Raf-1 protein expression in normal and tumor tissues in these mice (>50%, versus controls). These data have formed a basis of the clinical Phase I studies of LErafAON for cancer treatment.


Subject(s)
Liposomes/metabolism , Oligonucleotides, Antisense/pharmacokinetics , Proto-Oncogene Proteins c-raf/genetics , Animals , Cations , Cell Division , Cell Survival , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-raf/metabolism , Rabbits , Temperature , Time Factors , Tumor Cells, Cultured
13.
Clin Cancer Res ; 10(21): 7244-51, 2004 Nov 01.
Article in English | MEDLINE | ID: mdl-15534098

ABSTRACT

PURPOSE: Rapid cleavage in vivo and inefficient cellular uptake limit the clinical utility of antisense oligonucleotides (AON). Liposomal formulation may promote better intratumoral AON delivery and inhibit degradation in vivo. We conducted the first clinical evaluation of this concept using a liposomal AON complementary to the c-raf-1 proto-oncogene (LErafAON). EXPERIMENTAL DESIGN: A dose escalation study was done to determine the maximum tolerated dose and to characterize the toxicities of LErafAON given as weekly intravenous infusion for 8 weeks to adults with advanced solid tumors. Pharmacokinetic analysis and evaluation of c-raf-1 target suppression in peripheral blood mononuclear cells were included. RESULTS: Twenty-two patients received LErafAON (median 7 infusions; range 1-27) at doses of 1, 2, 4, and 6 mg/kg/week. Across all dose cohorts patients experienced infusion-related hypersensitivity reactions including flushing, dyspnea, hypoxia, rigors, back pain, and hypotension. Prolonged infusion duration and pretreatment with acetaminophen, H1- and H2-antagonists, and corticosteroids reduced the frequency and severity of these reactions. Progressive thrombocytopenia was dose-limiting at 6 mg/kg/week. No objective responses were observed. Two patients treated at the maximum tolerated dose of 4 mg/kg/week had evidence of stable disease, with dosing extended beyond 8 weeks. Pharmacokinetic analysis revealed persistence of detectable circulating rafAON at 24 hours in 7 of 10 patients in the highest 2 dose cohorts. Suppression of c-raf-1 mRNA was noted in two of five patients analyzed. CONCLUSIONS: Dose-independent hypersensitivity reactions and dose-dependent thrombocytopenia limited tolerance of LErafAON. Future clinical evaluation of this approach will depend on modification of the liposome composition.


Subject(s)
Genetic Therapy/adverse effects , Genetic Therapy/methods , Liposomes/metabolism , Neoplasms/therapy , Oligonucleotides, Antisense/pharmacology , Proto-Oncogene Proteins c-raf/genetics , Adult , Aged , Antineoplastic Agents/pharmacology , Cohort Studies , Dose-Response Relationship, Drug , Female , Gene Transfer Techniques , Humans , Hypoxia/metabolism , Leukocytes, Mononuclear/metabolism , Male , Maximum Tolerated Dose , Middle Aged , Neoplasm Transplantation , Proto-Oncogene Mas , RNA, Messenger/metabolism , Thrombocytopenia/etiology , Time Factors , Treatment Outcome
14.
Int J Oncol ; 24(5): 1181-8, 2004 May.
Article in English | MEDLINE | ID: mdl-15067340

ABSTRACT

Raf-1 protein kinase plays an important role in cell growth, proliferation and cell survival. We have previously described the use of liposome-entrapped antisense raf oligonucleotide (LErafAON) to inhibit Raf-1 expression resulting in tumor growth inhibition and radiosensitization. The present study was undertaken to evaluate the chemosensitization effects of LErafAON in combination with doxorubicin or paclitaxel on a panel of human tumor xenografts. LErafAON (25.0 mg/kg i.v. x 10) displayed significant antitumor activity (P<0.05) when administered as a single agent in prostate (PC-3), lung (A549) and breast (MDA-MB 231) carcinoma models. Doxorubicin (1.0-4.0 mg/kg i.v. per week x 3) and paclitaxel (1.0-4.0 mg/kg i.v. on alternate days x 3) were administered as single agents at non-toxic doses that led to only minimal to moderate antitumor activity. However, a combination of LErafAON with doxorubicin or paclitaxel led to significantly enhanced antitumor activity in all the tumor types tested (PC-3, P<0.03; A549, P<0.035; MDA-MB 231, P<0.045) as compared with LErafAON alone or chemotherapeutic agents alone treated groups. This effect of chemosensitization appeared to be sequence-specific because a mismatch control oligonucleotide continued to show significant tumor growth. Additionally, no inhibition in Raf-1 expression in MDA-MB 231 tumor tissue was observed with mismatch oligonucleotide treatment. On the other hand, LErafAON treatment led to >75% inhibition of Raf-1 expression in tumor tissue. These preclinical observations support the use of LErafAON in combination with chemotherapeutic agents to improve the treatment of human cancers.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-raf/genetics , Xenograft Model Antitumor Assays/methods , Animals , Breast Neoplasms/genetics , Doxorubicin/administration & dosage , Female , Humans , Liposomes , Lung Neoplasms/genetics , Male , Mice , Mice, Nude , Oligonucleotides, Antisense/administration & dosage , Paclitaxel/administration & dosage , Prostatic Neoplasms/genetics , Proto-Oncogene Proteins c-raf/metabolism , Tumor Cells, Cultured/transplantation
15.
Clin Cancer Res ; 16(11): 2939-48, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20400521

ABSTRACT

PURPOSE: BLID is a BH3-like motif containing apoptotic member of the Bcl-2 family of proteins. This study was designed to investigate the mechanism of BLID-induced apoptosis and to assess the significance of BLID expression in breast cancer. EXPERIMENTAL DESIGN: The interaction between BLID and Bcl-X(L) was examined using in vitro transcription/translation, coimmunoprecipitation, and immunoflourescence assays. The relationship between BLID mRNA expression and pathologic measures in breast cancer specimens (n = 55) was examined using the publicly available ONCOMINE microarray database. Immunohistochemistry was done using formalin-fixed, paraffin-embedded sections of 148 cases of invasive ductal breast carcinomas (IDC) and 58 cases of invasive lobular breast carcinomas, and breast tissue microarrays representing additional 437 cases (>85% IDC) with associated clinicopathologic database and long-term clinical follow-up (median 7 years). RESULTS: BLID was found to interact with Bcl-X(L), and the binding was enhanced in cancer cells exposed to doxorubicin or cisplatin. Exogenous expression of BLID correlated with activation of Bax and an increase in cytosolic cytochrome c. BLID mRNA expression was significantly reduced in grade 3 relative to grade 1 and 2 breast cancer (P = 0.023). Cytoplasmic BLID immunoreactivity was absent in IDC compared with invasive lobular breast carcinoma (P < 0.001). Lack of BLID expression was associated with younger age (median 40 years), African American ethnicity, tumor size, and triple-negative breast cancer (estrogen receptor negative, progesterone receptor negative, and human epidermal growth factor receptor 2 negative; all P < 0.005). Significant correlations were observed between BLID negativity and declines in overall, cause-specific, and local relapse-free survival (all P < 0.03). Multivariate analysis indicated that BLID is an independent prognostic factor of distant metastasis-free survival (hazard ratio, 0.302; 95% confidence interval, 0.160-0.570, P = 0.0002). CONCLUSION: BLID is a new binding partner of Bcl-X(L) and a significant prognostic factor in breast cancer.


Subject(s)
BRCA2 Protein/metabolism , Breast Neoplasms/metabolism , Adult , Apoptosis , Apoptosis Regulatory Proteins , Biomarkers, Tumor/analysis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Disease-Free Survival , Down-Regulation , Female , Humans , Middle Aged , Prognosis , RNA, Messenger/metabolism , Transfection , bcl-X Protein/metabolism
16.
Mol Ther ; 13(5): 947-55, 2006 May.
Article in English | MEDLINE | ID: mdl-16455304

ABSTRACT

SCC-S2/GG2-1/NDED (approved gene symbol TNFAIP8) is a transcription factor NF-kappaB-inducible, antiapoptotic, and oncogenic molecule. In this study, we examined the role of SCC-S2 in invasion and experimental metastasis. We demonstrate that expression of SCC-S2 cDNA in MDA-MB 435 human breast cancer cells is associated with enhanced invasion in vitro and increased frequency of pulmonary colonization of tumor cells in athymic mice. Systemic treatment of athymic mice with a cationic liposomal formulation of SCC-S2 antisense oligo led to decreased incidence of pulmonary metastasis and inhibition of SCC-S2 expression in vivo. Antisense inhibition of endogenous SCC-S2 expression correlated with decreased expression of VEGF receptor-2 in tumor cells and human lung microvascular endothelial cells and loss of endothelial cell viability. In addition, downregulation of SCC-S2 expression in tumor cells was associated with decreased expression of known metastasis-related molecules MMP-1 and MMP-9. These results demonstrate a novel role for SCC-S2 in tumor progression, involving multiple effectors, and provide a basis for SCC-S2-targeted cancer gene therapy.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Metalloproteases/metabolism , Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Line, Tumor , Female , Humans , Liposomes , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Metalloproteases/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Neoplasm Transplantation , Oligonucleotides, Antisense/therapeutic use , Proteins/genetics , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-2/genetics
17.
Anticancer Drugs ; 15(3): 243-53, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15014358

ABSTRACT

Raf-1 protein serine/threonine kinase plays an important role in cell proliferation and cell survival. We have previously described a novel cationic liposome-entrapped formulation of raf antisense oligodeoxyribonucleotide (LErafAON) and its use as a radiosensitizer. The aim of this study was to examine the effect of combination of LErafAON and a chemotherapeutic agent on growth of human prostate (PC-3) and pancreatic tumor xenografts in athymic mice (Aspc-1 and Colo 357). In PC-3 tumor-bearing mice, administration of a combination of LErafAON (i.v., 25 mg/kg/dose, x10/16) and cisplatin (i.v., 11.0 mg/kg/dose, x3), epirubicin (EPI) (i.v., 9.0 mg/kg/dose, x3) or mitoxantrone (MTO) (i.v., 2.5 mg/kg/dose, x3) led to enhanced tumor growth inhibition as compared with single agents (LErafAON+cisplatin versus cisplatin, p<0.0002, n=8; LErafAON+EPI versus EPI, p<0.0001, n=6; LErafAON+MTO versus MTO, p<0.05, n=5). In prostate or pancreatic tumor-bearing mice, combination of LErafAON (i.v., 25 mg/kg/dose, x10/13) with docetaxel (Taxotere) (i.v., 5, 7.5 or 10 mg/kg/dose, x2/4) led to tumor regression or enhanced growth inhibition as compared with single agents (PC-3: LErafAON+Taxotere versus Taxotere, p<0.02, n=7; Aspc-1: LErafAON+Taxotere versus Taxotere, p<0.03, n=5; Colo 357: LErafAON+Taxotere versus Taxotere, p<0.04, n=7). Combination of LErafAON (i.v., 25 mg/kg/dose, x10/13) with gemcitabine (i.v., 75 mg/kg/dose, x4/6) also caused a significant tumor growth inhibition in the two pancreatic carcinoma models studied (Aspc-1: LErafAON+gemcitabine versus gemcitabine, p<0.0001, n=7; Colo 357: LErafAON+gemcitabine versus gemcitabine, p<0.002, n =5). LErafAON treatment (i.v., 25 mg/kg/dose, x10) caused inhibition of Raf-1 protein expression in these tumor tissues (around 25-60%, n=4-7). Interestingly, Taxotere treatment per se also led to decreased steady state level of Raf-1 protein in PC-3 and Aspc-1 tumor tissues (i.v., 10 mg/kg/dose, x1 or 7.5 mg/kg/dose, x2; around 25-80%, n=2/6). Present studies demonstrate enhanced tumor growth inhibition or regression in response to a combination of a chemotherapeutic drug and LErafAON. These data provide a proof-of-principle for the clinical use of LErafAON in combination with chemotherapy for cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Deoxycytidine/analogs & derivatives , Oligonucleotides, Antisense/administration & dosage , Proto-Oncogene Proteins c-raf/administration & dosage , Xenograft Model Antitumor Assays/methods , Animals , Cell Line, Tumor , Cisplatin/administration & dosage , Deoxycytidine/administration & dosage , Docetaxel , Epirubicin/administration & dosage , Female , Humans , Liposomes , Male , Mice , Mice, Nude , Mitoxantrone , Proto-Oncogene Proteins c-raf/biosynthesis , Taxoids/administration & dosage , Gemcitabine
SELECTION OF CITATIONS
SEARCH DETAIL