Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters











Publication year range
1.
Sci Adv ; 8(3): eabj9815, 2022 Jan 21.
Article in English | MEDLINE | ID: mdl-35044832

ABSTRACT

Safe and effective vaccines are needed to end the COVID-19 pandemic. Here, we report the preclinical development of a lipid nanoparticle­formulated SARS-CoV-2 mRNA vaccine, PTX-COVID19-B. PTX-COVID19-B was chosen among three candidates after the initial mouse vaccination results showed that it elicited the strongest neutralizing antibody response against SARS-CoV-2. Further tests in mice and hamsters indicated that PTX-COVID19-B induced robust humoral and cellular immune responses and completely protected the vaccinated animals from SARS-CoV-2 infection in the lung. Studies in hamsters also showed that PTX-COVID19-B protected the upper respiratory tract from SARS-CoV-2 infection. Mouse immune sera elicited by PTX-COVID19-B vaccination were able to neutralize SARS-CoV-2 variants of concern, including the Alpha, Beta, Gamma, and Delta lineages. No adverse effects were induced by PTX-COVID19-B in either mice or hamsters. Based on these results, PTX-COVID19-B was authorized by Health Canada to enter clinical trials in December 2020 with a phase 2 clinical trial ongoing.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Synthetic/immunology , mRNA Vaccines/immunology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , CD4 Lymphocyte Count , CD8-Positive T-Lymphocytes/immunology , COVID-19/immunology , COVID-19 Vaccines/adverse effects , Canada , Cell Line , Cricetinae , Drug Evaluation, Preclinical , Female , HEK293 Cells , Humans , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Liposomes/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles , Spike Glycoprotein, Coronavirus/genetics , Th1 Cells/immunology
2.
Cancers (Basel) ; 14(2)2022 Jan 06.
Article in English | MEDLINE | ID: mdl-35053428

ABSTRACT

MicroRNA-21 (miR-21) is a small, non-coding RNA overexpressed in gastric cancer and many other solid malignancies, where it exhibits both pro-and anti-tumourigenic properties. However, the pathways regulating miR-21 and the consequences of its inhibition in gastric cancer remain incompletely understood. By exploiting the spontaneous Stat3-dependent formation of inflammation-associated gastric tumors in Gp130F/F mice, we functionally established miR-21 as a Stat3-controlled driver of tumor growth and progression. We reconciled our discoveries by identifying several conserved Stat3 binding motifs upstream of the miR-21 gene promoter, and showed that the systemic administration of a miR-21-specific antisense oligonucleotide antagomir reduced the established gastric tumor burden in Gp130F/F mice. We molecularly delineated the therapeutic benefits of miR-21 inhibition with the functional restoration of PTEN in vitro and in vivo, alongside an attenuated epithelial-to-mesenchymal transition and the extracellular matrix remodeling phenotype of tumors. We corroborated our preclinical findings by correlating high STAT3 and miR-21 expression with the reduced survival probability of gastric cancer patients. Collectively, our results provide a molecular framework by which miR-21 mediates inflammation-associated gastric cancer progression, and establish miR-21 as a robust therapeutic target for solid malignancies characterized by excessive Stat3 activity.

3.
Neuro Oncol ; 19(3): 372-382, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27765835

ABSTRACT

Background: Previously we showed therapeutic efficacy of unprotected miR-124 in preclinical murine models of glioblastoma, including in heterogeneous genetically engineered murine models by exploiting the immune system and thereby negating the need for direct tumor delivery. Although these data were promising, to implement clinical trials, we required a scalable formulation that afforded protection against circulatory RNases. Methods: We devised lipid nanoparticles that encapsulate and protect the miRs from degradation and provide enhanced delivery into the immune cell compartment and tested in vivo antitumor effects. Results: Treatment with nanoparticle-encapsulated miR-124, LUNAR-301, demonstrated a median survival exceeding 70 days, with an associated reversal of tumor-mediated immunosuppression and induction of immune memory. In both canine and murine models, the safety profile of LUNAR-301 was favorable. Conclusions: For the first time, we show that nanoparticles can direct a therapeutic response by targeting intracellular immune pathways. Although shown in the context of gliomas, this therapeutic approach would be applicable to other malignancies.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Immune Tolerance/genetics , Lipids/chemistry , MicroRNAs/genetics , Nanoparticles/administration & dosage , Animals , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Dogs , Glioma/genetics , Glioma/immunology , Humans , Immunologic Memory/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/administration & dosage , Nanoparticles/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Oncotarget ; 7(20): 29400-11, 2016 May 17.
Article in English | MEDLINE | ID: mdl-27102443

ABSTRACT

Small non-coding RNAs called miRNAs are key regulators in various biological processes, including tumor initiation, propagation, and metastasis in glioblastoma as well as other cancers. Recent studies have shown the potential for oncogenic miRNAs as therapeutic targets in glioblastoma. However, the application of antisense oligomers, or anti-miRs, to the brain is limited due to the blood-brain barrier (BBB), when administered in the traditional systemic manner. To induce a therapeutic effect in glioblastoma, anti-miR therapy requires a robust and effective delivery system to overcome this obstacle. To bypass the BBB, different delivery administration methods for anti-miRs were evaluated. Stereotaxic surgery was performed to administer anti-Let-7 through intratumoral (ITu), intrathecal (ITh), and intraventricular (ICV) routes, and each method's efficacy was determined by changes in the expression of anti-Let-7 target genes as well as by immunohistochemical analysis. ITu administration of anti-miRs led to a high rate of anti-miR delivery to tumors in the brain by both bolus and continuous administration. In addition, ICV administration, compared with ITu administration, showed a greater distribution of the miR across entire brain tissues. This study suggests that local administration methods are a promising strategy for anti-miR treatment and may overcome current limitations in the treatment of glioblastoma in preclinical animal models.


Subject(s)
Antagomirs/administration & dosage , Antineoplastic Agents/administration & dosage , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , MicroRNAs/antagonists & inhibitors , Animals , Blood-Brain Barrier , Humans , Injections, Intraventricular , Injections, Spinal , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays
5.
J Neurooncol ; 128(3): 395-404, 2016 07.
Article in English | MEDLINE | ID: mdl-27063952

ABSTRACT

Glioblastomas always recur despite surgery, radiotherapy and chemotherapy. A key player in the therapeutic resistance may be immature tumor cells with stem-like properties (TSCs) escaping conventional treatment. A group of promising molecular targets are microRNAs (miRs). miRs are small non-coding RNAs exerting post-transcriptional regulation of gene expression. In this study we aimed to identify over-expressed TSC-related miRs potentially amenable for therapeutic targeting. We used non-differentiated glioblastoma spheroid cultures (GSCs) containing TSCs and compared these to xenografts using a NanoString nCounter platform. This revealed 19 over-expressed miRs in the non-differentiated GSCs. Additionally, non-differentiated GSCs were compared to neural stem cells (NSCs) using a microarray platform. This revealed four significantly over-expressed miRs in the non-differentiated GSCs in comparison to the NSCs. The three most over-expressed miRs in the non-differentiated GSCs compared to xenografts were miR-126, -137 and -128. KEGG pathway analysis suggested the main biological function of these over-expressed miRs to be cell-cycle arrest and diminished proliferation. To functionally validate the profiling results suggesting association of these miRs with stem-like properties, experimental over-expression of miR-128 was performed. A consecutive limiting dilution assay confirmed a significantly elevated spheroid formation in the miR-128 over-expressing cells. This may provide potential therapeutic targets for anti-miRs to identify novel treatment options for GBM patients.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Heterografts , Humans , Male , Microarray Analysis , Neoplasm Transplantation , Neural Stem Cells/metabolism , Rats, Nude , Spheroids, Cellular/transplantation
6.
J Neurooncol ; 126(1): 47-55, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26428358

ABSTRACT

Over-expressed microRNAs (miRs) are promising new targets in glioblastoma (GBM) therapy. Inhibition of over-expressed miRs has been shown to diminish GBM proliferation, invasion and angiogenesis, indicating a significant therapeutic potential. However, the methods utilized for miR inhibition have had low translational potential. In clinical trials convection-enhanced delivery (CED) has been applied for local delivery of compounds in the brain. The aim of this study was to determine if safe and efficient miR inhibition was possible by CED of an anti-miR. We used a highly invasive GBM orthotopic xenograft model and targeted a well-validated miR, let-7a, with a 2'-O-methoxyethyl anti-miR with a combined phosphodiester/phosphorothioate backbone to establish an initial proof of concept. In vitro, anti-let-7a was delivered unassisted to the patient-derived T87 glioblastoma spheroid culture. In vivo, anti-let-7a or saline were administered by CED into orthotopic T87-derived tumors. After 1 month of infusion, tumors were removed and tumor mRNA levels of the target-gene High-mobility group AT-hook 2 (HMGA2) were determined. In vitro, 5 days inhibition was superior to 1 day at de-repressing the let-7a target HMGA2 and the inhibition was stable for 24 h. In vivo, anti-miR integrity was preserved in the pumps and no animals showed signs of severe adverse effects attributable to the anti-miR treatment. HMGA2 tumor level was significantly de-repressed in the anti-miR treated animals. The results showed-as an initial proof of concept-that miRs can be efficiently inhibited using CED delivery of anti-miR. The next step is to apply CED for anti-miR delivery focusing on key oncogenic miRs.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , MicroRNAs/metabolism , Animals , Antineoplastic Agents/administration & dosage , Brain Neoplasms/metabolism , Cell Line, Tumor , Convection , Drug Delivery Systems , Glioblastoma/metabolism , Glioma/pathology , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/therapeutic use , RNA, Messenger/metabolism , Statistics, Nonparametric , Transfection , Xenograft Model Antitumor Assays
7.
PLoS One ; 9(7): e100951, 2014.
Article in English | MEDLINE | ID: mdl-24992387

ABSTRACT

MicroRNAs (miRNAs), small RNA molecules that post-transcriptionally regulate mRNA expression, are crucial in diverse developmental and physiological programs and their misregulation can lead to disease. Chemically modified oligonucleotides have been developed to modulate miRNA activity for therapeutic intervention in disease settings, but their mechanism of action has not been fully elucidated. Here we show that the miRNA inhibitors (anti-miRs) physically associate with Argonaute proteins in the context of the cognate target miRNA in vitro and in vivo. The association is mediated by the seed region of the miRNA and is sensitive to the placement of chemical modifications. Furthermore, the targeted miRNAs are stable and continue to be associated with Argonaute. Our results suggest that anti-miRs specifically associate with Argonaute-bound miRNAs, preventing association with target mRNAs, which leads to subsequent stabilization and thus increased expression of the targeted mRNAs.


Subject(s)
Argonaute Proteins/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Oligonucleotides, Antisense/pharmacology , Animals , Female , Humans , Male , Mice, Inbred C57BL , Protein Binding/drug effects
8.
Cancer Cell ; 25(4): 501-15, 2014 Apr 14.
Article in English | MEDLINE | ID: mdl-24735924

ABSTRACT

Cancer-secreted microRNAs (miRNAs) are emerging mediators of cancer-host crosstalk. Here we show that miR-105, which is characteristically expressed and secreted by metastatic breast cancer cells, is a potent regulator of migration through targeting the tight junction protein ZO-1. In endothelial monolayers, exosome-mediated transfer of cancer-secreted miR-105 efficiently destroys tight junctions and the integrity of these natural barriers against metastasis. Overexpression of miR-105 in nonmetastatic cancer cells induces metastasis and vascular permeability in distant organs, whereas inhibition of miR-105 in highly metastatic tumors alleviates these effects. miR-105 can be detected in the circulation at the premetastatic stage, and its levels in the blood and tumor are associated with ZO-1 expression and metastatic progression in early-stage breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Endothelium, Vascular/pathology , MicroRNAs/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement , Endothelium, Vascular/metabolism , Female , Humans , MicroRNAs/genetics , Neoplasm Metastasis
9.
Cancer Res ; 73(9): 2884-96, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23338610

ABSTRACT

miRNAs (miR) are a critical class of small (21-25 nucleotides) noncoding endogenous RNAs implicated in gene expression regulation. We identified miR-23b and miR-27b as miRNAs that are highly upregulated in human breast cancer. We found that engineered knockdown of miR-23b and miR-27b substantially repressed breast cancer growth. Nischarin (NISCH) expression was augmented by knockdown of miR-23b as well as miR-27b. Notably, these miRNAs and Nischarin were inversely expressed in human breast cancers, underscoring their biologic relevance. We showed the clinical relevance of the expression of these miRNAs and showed that high expression of miR-23b and miR-27b correlates with poor outcome in breast cancer. Moreover, intraperitoneally delivered anti-miR-27b restored Nischarin expression and decreased tumor burden in a mouse xenograft model of human mammary tumor. Also, we report for the first time that HER2/neu (ERBB2), EGF, and TNF-α promote miR-23b/27b expression through the AKT/NF-κB signaling cascade. Nischarin was found to regulate miR-27b/23b expression through a feedback loop mechanism by suppressing NF-κB phosphorylation. Because anti-miR-27b compounds that suppress miR-27b inhibit tumor growth, the anti-miR-27b seems to be a good candidate for the development of new antitumor therapies.


Subject(s)
Breast Neoplasms/metabolism , Epidermal Growth Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/metabolism , MicroRNAs/metabolism , Receptor, ErbB-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement , Female , Humans , Mammary Neoplasms, Animal/genetics , Mice , Mice, Nude , Neoplasm Invasiveness , Treatment Outcome
10.
FEBS Lett ; 585(4): 671-6, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-21266177

ABSTRACT

RNA processing is vital for the high fidelity and diversity of eukaryotic transcriptomes and the encoded proteomes. However, control of RNA processing is not fully established. Σ RNA is a class of conserved large non-coding RNAs (murine Hepcarcin; human MALAT-1) up-regulated in carcinomas. Using antisense technology, we identified that RNA post-transcriptional modification is the most significant global function of Σ RNA. Specifically, processing of the pre-mRNAs of genes including Tissue Factor and Endoglin was altered by hydrolysis of Σ RNA/MALAT-1. These results support the hypothesis that Σ RNA/MALAT-1 is a regulatory molecule exerting roles in RNA post-transcriptional modification.


Subject(s)
Carcinoma/metabolism , RNA Processing, Post-Transcriptional , RNA, Neoplasm/metabolism , RNA, Untranslated/metabolism , Alternative Splicing , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cells, Cultured , Endoglin , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Oligonucleotides, Antisense , RNA Precursors/metabolism , RNA, Long Noncoding , RNA, Untranslated/antagonists & inhibitors , RNA, Untranslated/genetics , RNA-Binding Proteins/metabolism , Rats , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Serine-Arginine Splicing Factors , Thromboplastin/genetics , Thromboplastin/metabolism
11.
Cancer Res ; 70(20): 8077-87, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20924108

ABSTRACT

Hepatocellular carcinoma (HCC) is generally a fatal disease due to a paucity of effective treatment options. The identification of oncogenic microRNAs that exert pleiotropic effects in HCC cells may offer new therapeutic targets. In this study, we have identified the human microRNA miR-191 as a potential target for HCC therapy. Inhibition of miR-191 decreased cell proliferation and induced apoptosis in vitro and significantly reduced tumor masses in vivo in an orthotopic xenograft mouse model of HCC. Additionally, miR-191 was found to be upregulated by a dioxin, a known liver carcinogen, and was found to be a regulator of a variety of cancer-related pathways. Our findings offer a preclinical proof of concept for miR-191 targeting as a rational strategy to pursue for improving HCC treatment.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Asian People/genetics , Carcinogens/pharmacology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Division , Dioxins/pharmacology , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , MicroRNAs/drug effects , Models, Animal , Models, Genetic , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Transcription, Genetic/drug effects , Up-Regulation , White People/genetics
12.
Nat Med ; 16(10): 1134-40, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20871609

ABSTRACT

Inactivation of the p53 tumor suppressor pathway allows cell survival in times of stress and occurs in many human cancers; however, normal embryonic stem cells and some cancers such as neuroblastoma maintain wild-type human TP53 and mouse Trp53 (referred to collectively as p53 herein). Here we describe a miRNA, miR-380-5p, that represses p53 expression via a conserved sequence in the p53 3' untranslated region (UTR). miR-380-5p is highly expressed in mouse embryonic stem cells and neuroblastomas, and high expression correlates with poor outcome in neuroblastomas with neuroblastoma derived v-myc myelocytomatosis viral-related oncogene (MYCN) amplification. miR-380 overexpression cooperates with activated HRAS oncoprotein to transform primary cells, block oncogene-induced senescence and form tumors in mice. Conversely, inhibition of endogenous miR-380-5p in embryonic stem or neuroblastoma cells results in induction of p53, and extensive apoptotic cell death. In vivo delivery of a miR-380-5p antagonist decreases tumor size in an orthotopic mouse model of neuroblastoma. We demonstrate a new mechanism of p53 regulation in cancer and stem cells and uncover a potential therapeutic target for neuroblastoma.


Subject(s)
Gene Amplification , MicroRNAs/physiology , Neuroblastoma/pathology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Tumor Suppressor Protein p53/antagonists & inhibitors , 3' Untranslated Regions , Animals , Apoptosis , Binding Sites , DNA Damage , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Transgenic , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Oncogenes , Tumor Suppressor Protein p53/physiology
13.
Cancer Res ; 70(9): 3494-504, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20406980

ABSTRACT

The tropism of breast cancer cells for bone and their tendency to induce an osteolytic phenotype are a result of interactions between breast cancer cells and stromal cells and are of paramount importance for bone metastasis. However, the underlying molecular mechanisms remain poorly understood. We hypothesize that tumor-stromal interaction alters gene expression in malignant tumor cells and stromal cells creating a unique expression signature that promotes osteolytic breast cancer bone metastasis and that inhibition of such interactions can be developed as targeted therapeutics. Microarray analysis was performed to investigate gene expression profiling at the tumor-bone (TB) interface versus the tumor alone area from syngenic mice injected with three different syngenic mammary tumor cell lines that differ in their metastatic potential. We identified matrix metalloproteinase 13 (MMP13), receptor activator of NF-kappaB ligand (RANKL), and integrins binding sialoprotein to be genes upregulated at the TB interface and validated. To determine the functional role of MMP13 in tumor-induced osteolysis, mice with Cl66 mammary tumors were treated with MMP13 antisense oligonucleotides (MMP13-ASO) or control scrambled oligonucleotides (control-ASO). Knockdown of MMP13 expression at the TB interface leads to significant reduction in bone destruction and in the number of activated osteoclasts at the TB interface. Further analysis to evaluate the mechanism of MMP13-dependent osteolytic bone metastasis revealed that MMP13-ASO treatment decreased active MMP9, RANKL levels, and transforming growth factor-beta signaling at the TB interface. Together, our data indicate that upregulation of MMP13 at the TB interface is important in tumor-induced osteolysis and suggest that MMP13 is a potential therapeutic target for breast cancer bone metastasis.


Subject(s)
Bone Neoplasms/secondary , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/metabolism , Osteolysis/enzymology , Transforming Growth Factor beta/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/metabolism , Bone and Bones/enzymology , Bone and Bones/metabolism , Bone and Bones/pathology , Cell Line, Tumor , Enzyme Activation , Female , Gene Expression Profiling , Mammary Neoplasms, Experimental/metabolism , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase Inhibitors , Mice , Mice, Inbred BALB C , Osteoclasts/enzymology , Osteoclasts/pathology , Osteolysis/genetics , Osteolysis/metabolism , Osteolysis/pathology , Osteoprotegerin/biosynthesis , RANK Ligand/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction , Up-Regulation
14.
Nat Biotechnol ; 28(4): 341-7, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20351690

ABSTRACT

MicroRNAs (miRNAs) are increasingly implicated in the regulation of metastasis. Despite their potential as targets for anti-metastatic therapy, miRNAs have only been silenced in normal tissues of rodents and nonhuman primates. Therefore, the development of effective approaches for sequence-specific inhibition of miRNAs in tumors remains a scientific and clinical challenge. Here we show that systemic treatment of tumor-bearing mice with miR-10b antagomirs-a class of chemically modified anti-miRNA oligonucleotide-suppresses breast cancer metastasis. Both in vitro and in vivo, silencing of miR-10b with antagomirs significantly decreases miR-10b levels and increases the levels of a functionally important miR-10b target, Hoxd10. Administration of miR-10b antagomirs to mice bearing highly metastatic cells does not reduce primary mammary tumor growth but markedly suppresses formation of lung metastases in a sequence-specific manner. The miR-10b antagomir, which is well tolerated by normal animals, appears to be a promising candidate for the development of new anti-metastasis agents.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/therapy , MicroRNAs/administration & dosage , MicroRNAs/genetics , Neoplasm Metastasis/genetics , Neoplasm Metastasis/therapy , Animals , Female , Mice , Treatment Outcome
15.
Clin Exp Metastasis ; 26(7): 797-808, 2009.
Article in English | MEDLINE | ID: mdl-19590968

ABSTRACT

The bone microenvironment plays a critical role in tumor-induced osteolysis and osteolytic metastasis through tumor-bone (TB)-interaction. Receptor activator of nuclear factor-kappaB (RANK) ligand (RANKL) is one of the critical signaling molecules involved in osteolysis and bone metastasis. However, the regulation and functional significance of RANKL at the TB-interface in tumor-induced osteolysis remains unclear. In this report, we examined the role of tumor-stromal interaction in the regulation of RANKL expression and its functional significance in tumor-induced osteolysis. Using a novel mammary tumor model, we identified that RANKL expression was upregulated at the TB-interface as compared to the tumor alone area. We demonstrate increased generation of sRANKL at the TB-interface, which is associated with tumor-induced osteolysis. The ratio of RANKL to osteoprotegrin (OPG), a decoy receptor for RANKL, at the TB-interface was also increased. Targeting RANKL expression with antisense oligonucleotides (RANKL-ASO), significantly abrogated tumor-induced osteolysis, decreased RANKL expression and the RANKL:OPG ratio at the TB-interface. Together, these results demonstrate that upregulation of RANKL expression and sRANKL generation at the TB-interface potentiates tumor-induced osteolysis.


Subject(s)
Bone and Bones/metabolism , Mammary Neoplasms, Experimental/metabolism , RANK Ligand/metabolism , Animals , Base Sequence , Blotting, Western , Bone and Bones/pathology , DNA Primers , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Ligands , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Osteolysis , Reverse Transcriptase Polymerase Chain Reaction
16.
J Clin Invest ; 118(5): 1727-38, 2008 May.
Article in English | MEDLINE | ID: mdl-18431520

ABSTRACT

Deregulated activation of STAT3 is frequently associated with many human hematological and epithelial malignancies, including gastric cancer. While exaggerated STAT3 signaling facilitates an antiapoptotic, proangiogenic, and proproliferative environment for neoplastic cells, the molecular mechanisms leading to STAT3 hyperactivation remain poorly understood. Using the gp130(Y757F/Y757F) mouse model of gastric cancer, which carries a mutated gp130 cytokine receptor signaling subunit that cannot bind the negative regulator of cytokine signaling SOCS3 and is characterized by hyperactivation of the signaling molecules STAT1 and STAT3, we have provided genetic evidence that IL-11 promotes chronic gastric inflammation and associated tumorigenesis. Expression of IL-11 was increased in gastric tumors in gp130(Y757F/Y757F) mice, when compared with unaffected gastric tissue in wild-type mice, while gp130(Y757F/Y757F) mice lacking the IL-11 ligand-binding receptor subunit (IL-11Ralpha) showed normal gastric STAT3 activation and IL-11 expression and failed to develop gastric tumors. Furthermore, reducing STAT3 activity in gp130(Y757F/Y757F) mice, either genetically or by therapeutic administration of STAT3 antisense oligonucleotides, normalized gastric IL-11 expression and alleviated gastric tumor burden. Surprisingly, the genetic reduction of STAT1 expression also reduced gastric tumorigenesis in gp130(Y757F/Y757F) mice and coincided with reduced gastric inflammation and IL-11 expression. Collectively, our data have identified IL-11 as a crucial cytokine promoting chronic gastric inflammation and associated tumorigenesis mediated by excessive activation of STAT3 and STAT1.


Subject(s)
Cytokine Receptor gp130/immunology , Inflammation/metabolism , Interleukin-11/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Stomach Neoplasms/metabolism , Animals , Cytokine Receptor gp130/genetics , Gastric Mucosa/metabolism , Humans , Interleukin-11/genetics , Interleukin-6/immunology , Mice , Mice, Knockout , Mice, Transgenic , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Stomach/anatomy & histology , Stomach/pathology , Stomach Neoplasms/pathology
17.
Cancer Res ; 68(3): 631-4, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18245460

ABSTRACT

The eukaryotic translation initiation factor 4E (eIF4E) is frequently overexpressed in human cancers in relation to disease progression and drives cellular transformation, tumorigenesis, and metastatic progression in experimental models. Enhanced eIF4E function results from eIF4E overexpression and/or activation of the ras and phosphatidylinositol 3-kinase/AKT pathways and selectively increases the translation of key mRNAs involved in tumor growth, angiogenesis, and cell survival. Consequently, by simultaneously and selectively reducing the expression of numerous potent growth and survival factors critical for malignancy, targeting eIF4E for inhibition may provide an attractive therapy for many different tumor types. Recent work has now shown the plausibility of therapeutically targeting eIF4E and has resulted in the advance of the first eIF4E-specific therapy to clinical trials. These studies illustrate the increased susceptibility of tumor tissues to eIF4E inhibition and support the notion that the enhanced eIF4E function common to many tumor types may represent an Achilles' heel for cancer.


Subject(s)
Eukaryotic Initiation Factor-4E/antagonists & inhibitors , Neoplasms/therapy , Animals , Eukaryotic Initiation Factor-4E/biosynthesis , Eukaryotic Initiation Factor-4E/genetics , Humans , Neoplasms/genetics , Neoplasms/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics
18.
J Clin Invest ; 117(9): 2638-48, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17786246

ABSTRACT

Expression of eukaryotic translation initiation factor 4E (eIF4E) is commonly elevated in human and experimental cancers, promoting angiogenesis and tumor growth. Elevated eIF4E levels selectively increase translation of growth factors important in malignancy (e.g., VEGF, cyclin D1) and is thereby an attractive anticancer therapeutic target. Yet to date, no eIF4E-specific therapy has been developed. Herein we report development of eIF4E-specific antisense oligonucleotides (ASOs) designed to have the necessary tissue stability and nuclease resistance required for systemic anticancer therapy. In mammalian cultured cells, these ASOs specifically targeted the eIF4E mRNA for destruction, repressing expression of eIF4E-regulated proteins (e.g., VEGF, cyclin D1, survivin, c-myc, Bcl-2), inducing apoptosis, and preventing endothelial cells from forming vessel-like structures. Most importantly, intravenous ASO administration selectively and significantly reduced eIF4E expression in human tumor xenografts, significantly suppressing tumor growth. Because these ASOs also target murine eIF4E, we assessed the impact of eIF4E reduction in normal tissues. Despite reducing eIF4E levels by 80% in mouse liver, eIF4E-specific ASO administration did not affect body weight, organ weight, or liver transaminase levels, thereby providing the first in vivo evidence that cancers may be more susceptible to eIF4E inhibition than normal tissues. These data have prompted eIF4E-specific ASO clinical trials for the treatment of human cancers.


Subject(s)
Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Regulation, Neoplastic , Neoplasms/metabolism , Neoplasms/therapy , Protein Biosynthesis/genetics , Animals , Apoptosis , Base Sequence , Cells, Cultured , Endothelial Cells/metabolism , Eukaryotic Initiation Factor-4E/genetics , Humans , Mice , Neoplasms/blood supply , Neoplasms/pathology , Xenograft Model Antitumor Assays
19.
Cancer Res ; 67(8): 3583-93, 2007 Apr 15.
Article in English | MEDLINE | ID: mdl-17440069

ABSTRACT

The sonic hedgehog (Shh) pathway contributes to the initiation and progression of tumors with various origins when aberrantly activated. In this study, we investigated if the Shh pathway is important for the proliferation of hepatocellular carcinoma (HCC) cells and also began to identify which components of the pathway play a pivotal role in the biology of HCC. Expression levels of components in the pathway were measured, and glioma-associated oncogene (Gli) 2 levels were found to be considerably higher in human HCC lines compared with normal liver. Gli2 levels were also higher in tumor tissue from HCC patients compared with normal liver. Antisense oligonucleotides (ASO) were used to specifically down-regulate Gli2, and this led to decreased proliferation of various HCC cell lines. However, inhibition of Gli1 and Gli3 with ASOs did not decrease proliferation in most HCC cell lines and inhibitors targeting the upstream components of the pathway, including smoothened (Smo), displayed antiproliferative effects in only a subset of HCC cell lines. Moreover, in cancer cells harboring Smo mutations or unresponsive to the Smo inhibitor 3-keto-N-aminoethylaminoethylcaproyldihydrocinnamoyl cyclopamine, the Gli2 ASO was still able to inhibit proliferation. The importance of Gli2 in HCC proliferation was further confirmed by the changes in expression levels of genes, such as Bcl-2, c-Myc, and p27, following suppression of Gli2 expression. Taken together, these results suggest that, among the Gli transcription factors, Gli2 plays a predominant role in the proliferation of HCC cells and the suppression of Gli2 expression may provide a useful therapeutic option for the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , Liver Neoplasms/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Growth Processes/physiology , Down-Regulation , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nuclear Proteins/metabolism , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Veratrum Alkaloids/pharmacology , Zinc Finger Protein Gli2
20.
Nucleic Acids Res ; 34(16): 4467-76, 2006.
Article in English | MEDLINE | ID: mdl-16945958

ABSTRACT

Short interfering RNAs (siRNA) guide degradation of target RNA by the RNA-induced silencing complex (RISC). The use of siRNA in animals is limited partially due to the short half-life of siRNAs in tissues. Chemically modified siRNAs are necessary that maintain mRNA degradation activity, but are more stable to nucleases. In this study, we utilized alternating 2'-O-methyl and 2'-deoxy-2'-fluoro (OMe/F) chemically modified siRNA targeting PTEN and Eg5. OMe/F-modified siRNA consistently reduced mRNA and protein levels with equal or greater potency and efficacy than unmodified siRNA. We showed that modified siRNAs use the RISC mechanism and lead to cleavage of target mRNA at the same position as unmodified siRNA. We further demonstrated that siRNAs can compete with each other, where highly potent siRNAs can compete with less potent siRNAs, thus limiting the ability of siRNAs with lower potency to mediate mRNA degradation. In contrast, a siRNA with low potency cannot compete with a highly efficient siRNA. We established a correlation between siRNA potency and ability to compete with other siRNAs. Thus, siRNAs that are more potent inhibitors for mRNA destruction have the potential to out-compete less potent siRNAs indicating that the amount of a cellular component, perhaps RISC, limits siRNA activity.


Subject(s)
RNA Interference , RNA, Small Interfering/chemistry , RNA-Induced Silencing Complex/metabolism , Binding, Competitive , Cell Line, Tumor , Humans , Kinesins/genetics , PTEN Phosphohydrolase/genetics , RNA, Messenger/metabolism , Ribonucleases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL