Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Ther ; 17(2): 544-553, 2018 02.
Article in English | MEDLINE | ID: mdl-29282298

ABSTRACT

Colorectal carcinomas harbor well-defined genetic abnormalities, including aberrant activation of Wnt/ß-catenin and MAPK pathways, often simultaneously. Although the MAPK pathway can be targeted using potent small-molecule drugs, including BRAF and MEK inhibitors, ß-catenin inhibition has been historically challenging. RNAi approaches have advanced to the stage of clinical viability and are especially well suited for transcriptional modulators, such as ß-catenin. In this study, we report therapeutic effects of combined targeting of these pathways with pharmacologic agents. Using a recently described tumor-selective nanoparticle containing a ß-catenin-targeting RNAi trigger, in combination with the FDA-approved MEK inhibitor (MEKi) trametinib, we demonstrate synergistic tumor growth inhibition in in vivo models of colorectal cancer, melanoma, and hepatocellular carcinoma. At dose levels that were insufficient to significantly impact tumor growth as monotherapies, combination regimens resulted in synergistic efficacy and complete tumor growth inhibition. Importantly, dual MEKi/RNAi therapy dramatically improved survival of mice bearing colorectal cancer liver metastases. In addition, pharmacologic silencing of ß-catenin mRNA was effective against tumors that are inherently resistant or that acquire drug-induced resistance to trametinib. These results provide a strong rationale for clinical evaluation of this dual-targeting approach for cancers harboring Wnt/ß-catenin and MAPK pathway mutations. Mol Cancer Ther; 17(2); 544-53. ©2017 AACR.


Subject(s)
Colorectal Neoplasms/therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidinones/pharmacology , RNA, Messenger/genetics , RNA, Small Interfering/administration & dosage , beta Catenin/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Drug Synergism , Gene Silencing , Heterografts , Humans , Liver Neoplasms, Experimental/secondary , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Nude , Nanoparticles/administration & dosage , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
2.
Mol Cancer Ther ; 15(9): 2143-54, 2016 09.
Article in English | MEDLINE | ID: mdl-27390343

ABSTRACT

The Wnt/ß-catenin pathway is among the most frequently altered signaling networks in human cancers. Despite decades of preclinical and clinical research, efficient therapeutic targeting of Wnt/ß-catenin has been elusive. RNA interference (RNAi) technology silences genes at the mRNA level and therefore can be applied to previously undruggable targets. Lipid nanoparticles (LNP) represent an elegant solution for the delivery of RNAi-triggering oligonucleotides to disease-relevant tissues, but have been mostly restricted to applications in the liver. In this study, we systematically tuned the composition of a prototype LNP to enable tumor-selective delivery of a Dicer-substrate siRNA (DsiRNA) targeting CTNNB1, the gene encoding ß-catenin. This formulation, termed EnCore-R, demonstrated pharmacodynamic activity in subcutaneous human tumor xenografts, orthotopic patient-derived xenograft (PDX) tumors, disseminated hematopoietic tumors, genetically induced primary liver tumors, metastatic colorectal tumors, and murine metastatic melanoma. DsiRNA delivery was homogeneous in tumor sections, selective over normal liver and independent of apolipoprotein-E binding. Significant tumor growth inhibition was achieved in Wnt-dependent colorectal and hepatocellular carcinoma models, but not in Wnt-independent tumors. Finally, no evidence of accelerated blood clearance or sustained liver transaminase elevation was observed after repeated dosing in nonhuman primates. These data support further investigation to gain mechanistic insight, optimize dose regimens, and identify efficacious combinations with standard-of-care therapeutics. Mol Cancer Ther; 15(9); 2143-54. ©2016 AACR.


Subject(s)
Neoplasms/genetics , RNA Interference , RNA, Small Interfering/genetics , beta Catenin/genetics , Animals , Apolipoproteins E/chemistry , Apolipoproteins E/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Lipids/chemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Melanoma, Experimental , Mice , Nanoparticles/chemistry , Neoplasm Metastasis , Neoplasms/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/chemistry , Structure-Activity Relationship , Wnt Signaling Pathway , Xenograft Model Antitumor Assays , beta Catenin/metabolism , ras Proteins/genetics , ras Proteins/metabolism
3.
Mol Ther ; 24(4): 770-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26758691

ABSTRACT

Primary hyperoxaluria type 1 (PH1) is an autosomal recessive, metabolic disorder caused by mutations of alanine-glyoxylate aminotransferase (AGT), a key hepatic enzyme in the detoxification of glyoxylate arising from multiple normal metabolic pathways to glycine. Accumulation of glyoxylate, a precursor of oxalate, leads to the overproduction of oxalate in the liver, which accumulates to high levels in kidneys and urine. Crystalization of calcium oxalate (CaOx) in the kidney ultimately results in renal failure. Currently, the only treatment effective in reduction of oxalate production in patients who do not respond to high-dose vitamin B6 therapy is a combined liver/kidney transplant. We explored an alternative approach to prevent glyoxylate production using Dicer-substrate small interfering RNAs (DsiRNAs) targeting hydroxyacid oxidase 1 (HAO1) mRNA which encodes glycolate oxidase (GO), to reduce the hepatic conversion of glycolate to glyoxylate. This approach efficiently reduces GO mRNA and protein in the livers of mice and nonhuman primates. Reduction of hepatic GO leads to normalization of urine oxalate levels and reduces CaOx deposition in a preclinical mouse model of PH1. Our results support the use of DsiRNA to reduce liver GO levels as a potential therapeutic approach to treat PH1.


Subject(s)
Alcohol Oxidoreductases/genetics , Calcium Oxalate/metabolism , Hyperoxaluria, Primary/therapy , RNA, Small Interfering/administration & dosage , Animals , DEAD-box RNA Helicases/metabolism , Disease Models, Animal , Glyoxylates/urine , Humans , Hyperoxaluria, Primary/enzymology , Hyperoxaluria, Primary/urine , Liver/metabolism , Mice , Nanoparticles/chemistry , RNA, Small Interfering/pharmacology , Ribonuclease III/metabolism
4.
Mol Ther ; 22(1): 92-101, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24089139

ABSTRACT

Despite progress in identifying molecular drivers of cancer, it has been difficult to translate this knowledge into new therapies, because many of the causal proteins cannot be inhibited by conventional small molecule therapeutics. RNA interference (RNAi), which uses small RNAs to inhibit gene expression, provides a promising alternative to reach traditionally undruggable protein targets by shutting off their expression at the messenger RNA (mRNA) level. Challenges for realizing the potential of RNAi have included identifying the appropriate genes to target and achieving sufficient knockdown in tumors. We have developed high-potency Dicer-substrate short-interfering RNAs (DsiRNAs) targeting ß-catenin and delivered these in vivo using lipid nanoparticles, resulting in significant reduction of ß-catenin expression in liver cancer models. Reduction of ß-catenin strongly reduced tumor burden, alone or in combination with sorafenib and as effectively as DsiRNAs that target mitotic genes such as PLK1 and KIF11. ß-catenin knockdown also strongly reduced the expression of ß-catenin-regulated genes, including MYC, providing a potential mechanism for tumor inhibition. These results validate ß-catenin as a target for liver cancer therapy and demonstrate the promise of RNAi in general and DsiRNAs in particular for reaching traditionally undruggable cancer targets.


Subject(s)
Liver Neoplasms/genetics , Liver Neoplasms/pathology , RNA, Small Interfering/genetics , beta Catenin/genetics , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Gene Knockdown Techniques , Humans , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Male , Mice , Nanoparticles/administration & dosage , Nanoparticles/chemistry , RNA Interference , RNA, Small Interfering/chemistry , RNA, Small Interfering/metabolism , Ribonuclease III/metabolism , Tumor Burden/genetics , Xenograft Model Antitumor Assays , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...