226 results on '"Monia BP"'
Search Results
2. PND3 - Inotersen Improves Quality of Life and Neuropathy in Patients with Hereditary Transthyretin (HATTR) Amyloidosis with Polyneuropathy: Results of the Phase 3 Study Neuro-TTR
- Author
-
Berk, JL, Coelho, T, Wang, AK, Waddington-Cruz, M, Polydefkis, MJ, Dyck, PJ, Scheinberg, M, Plante-Bordeneuve, V, Barroso, F, Adams, D, Brannagan, TH, Whelan, C, Merlini, G, Drachman, BM, Heitner, SB, Conceicao, I, Schmidt, H, Vita, G, Campistol, JM, Gorevic, P, Monia, BP, Hughes, SG, Kwoh, J, Jung, B, Ackermann, EJ, Gertz, M, and Benson, MD
- Published
- 2018
- Full Text
- View/download PDF
3. Nicotinamide N-methyltransferase is a novel regulator of energy metabolism in adipose tissue
- Author
-
Kraus, D, primary, Yang, Q, additional, Kong, D, additional, Banks, AS, additional, Zhang, L, additional, Rodgers, JT, additional, Pirinen, E, additional, Pulinilkunnil, TC, additional, Gong, F, additional, Wang, YC, additional, Cen, Y, additional, Sauve, AA, additional, Asara, JM, additional, Peroni, OD, additional, Monia, BP, additional, Bhanot, S, additional, Alhonen, L, additional, Puigserver, P, additional, and Kahn, BB, additional
- Published
- 2014
- Full Text
- View/download PDF
4. A Study of Ras Gtpases and Prenylation in Human Mesangial Cell Proliferation in Vitro
- Author
-
Khwaja, S.A.I., primary, Sharpe, CC, additional, Monia, BP, additional, Sebti, SM, additional, and Hendry, BM, additional
- Published
- 2001
- Full Text
- View/download PDF
5. Polyethyleneimine grafted with pluronic P85 enhances Ku86 antisense delivery and the ionizing radiation treatment efficacy in vivo.
- Author
-
Belenkov, Al, Alakhov, VY, Kabanov, AV, Vinogradov, SV, Panasci, LC, Monia, BP, and Chow, TYK
- Subjects
ANTISENSE nucleic acids ,GENE therapy ,OLIGONUCLEOTIDES ,IONIZING radiation ,CANCER treatment - Abstract
In an effort to improve the efficacy of antisense delivery, we evaluated polyethyleneimine (PEI, 2 kDa) alone or grafted with nonionic amphiphilic block copolymer Pluronic® (P85) as a carrier for Ku86 antisense oligonucleotide (ASO) delivery. Ku86 is an abundant nuclear protein that plays an important role in nonhomologous DNA end joining and has implications in tumorigenesis and acquired drug resistance. Transfection of adherent and suspension cell lines with Ku86 ASOs complexed with P85-g-PEI (2 kDa) conjugates was associated with a specific decrease in Ku86 mRNA levels (EC
50 < 75 nM and EC50 < 250 nM, respectively, n = 3). More importantly, no requirement for reduced serum conditions was necessary during transfection. In contrast, whereas Ku86 ASOs complexed with PEI (2 kDa) alone were effective in decreasing Ku86 mRNA levels in adherent cell lines (EC5O < 75 nM, n = 3), the formulation did not produce any detectable decrease in Ku86 mRNA levels in suspension cell lines. Transfection of adherent cell lines with 500 nM Ku86 ASOs formulated with P85-g-PEI (2 kDa) was associated with a specific decrease (<10% remaining of control) in Ku86 protein expression and a two-fold increased cell death after treatment with ionizing radiation (IR). In athymic nude mice bearing subcutaneous human HT29 colon adenocarcinoma xenografts, Ku86 ASO-P85-g-PEI (2 kDa) administration (15 mg/kg, subcutaneously) with a Q1D × 7 treatment schedule, when combined with a single dose of IR (6 Gy), caused a significant inhibition of HT29 tumor growth compared with mismatch- and naked antisense-pretreated control groups (time from 200 to 1000 mm³, 126.9 versus 84.18 and 87.76 days, P < 0.005). A potentiation of the antitumor activity was observed in all mice treated with Ku86 ASO-P85-g-PEI (2 kDa) formulation; however, tumor growth inhibition was reversible upon treatment cessation. No morbidity/mortality or changes in histopathology were observed under this treatment regiment. Our results indicate that P85-g-PEI (2 kDa) conjugates may increase the efficacy of Ku86 ASO delivery in management of resistant malignancies, thus providing a rationale for their evaluation in cancer patients in combination with conventional anticancer therapies. [ABSTRACT FROM AUTHOR]- Published
- 2004
- Full Text
- View/download PDF
6. Targeting the contact system in a rabbit model of extracorporeal membrane oxygenation.
- Author
-
Tweddell JS, Kharnaf M, Zafar F, Riggs KW, Reagor JA, Monia BP, Revenko A, Leino DG, Owens AP, Martin JK, Gourley B, Rosenfeldt L, and Palumbo JS
- Subjects
- Animals, Rabbits, Factor XII, Thrombin metabolism, Factor XI metabolism, Extracorporeal Membrane Oxygenation adverse effects, Thrombosis etiology
- Abstract
Previous studies suggested that contact pathway factors drive thrombosis in mechanical circulation. We used a rabbit model of veno-arterial extracorporeal circulation (VA-ECMO) to evaluate the role of factors XI and XII in ECMO-associated thrombosis and organ damage. Factors XI and XII (FXI, FXII) were depleted using established antisense oligonucleotides before placement on a blood-primed VA-ECMO circuit. Decreasing FXII or FXI to < 5% of baseline activity significantly prolonged ECMO circuit lifespan, limited the development of coagulopathy, and prevented fibrinogen consumption. Histological analysis suggested that FXII depletion mitigated interstitial pulmonary edema and hemorrhage whereas heparin and FXI depletion did not. Neither FXI nor FXII depletion was associated with significant hemorrhage in other organs. In vitro analysis showed that membrane oxygenator fibers (MOFs) alone are capable of driving significant thrombin generation in a FXII- and FXI-dependent manner. MOFs also augment thrombin generation triggered by low (1 pM) or high (5 pM) tissue factor concentrations. However, only FXI elimination completely prevented the increase in thrombin generation driven by MOFs, suggesting MOFs augment thrombin-mediated FXI activation. Together, these results suggest that therapies targeting FXII or FXI limit thromboembolic complications associated with ECMO. Further studies are needed to determine the contexts wherein targeting FXI and FXII, either alone or in combination, would be most beneficial in ECMO. Moreover, studies are also needed to determine the potential mechanisms coupling FXII to end-organ damage in ECMO., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
7. TMPRSS6 as a Therapeutic Target for Disorders of Erythropoiesis and Iron Homeostasis.
- Author
-
Ganz T, Nemeth E, Rivella S, Goldberg P, Dibble AR, McCaleb ML, Guo S, Monia BP, and Barrett TD
- Subjects
- Mice, Animals, Humans, Iron metabolism, Liver metabolism, Homeostasis, Membrane Proteins genetics, Membrane Proteins metabolism, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Erythropoiesis genetics, Anemia, Iron-Deficiency drug therapy
- Abstract
TMPRSS6 is a serine protease highly expressed in the liver. Its role in iron regulation was first reported in 2008 when mutations in TMPRSS6 were shown to be the cause of iron-refractory iron deficiency anemia (IRIDA) in humans and in mouse models. TMPRSS6 functions as a negative regulator of the expression of the systemic iron-regulatory hormone hepcidin. Over the last decade and a half, growing understanding of TMPRSS6 biology and mechanism of action has enabled development of new therapeutic approaches for patients with diseases of erythropoiesis and iron homeostasis.ClinicalTrials.gov identifier NCT03165864., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. CFTR mRNAs with nonsense codons are degraded by the SMG6-mediated endonucleolytic decay pathway.
- Author
-
Sanderlin EJ, Keenan MM, Mense M, Revenko AS, Monia BP, Guo S, and Huang L
- Subjects
- Carrier Proteins metabolism, Codon, Nonsense, Humans, RNA, Messenger genetics, RNA, Messenger metabolism, Cystic Fibrosis genetics, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Endoribonucleases metabolism, Nonsense Mediated mRNA Decay
- Abstract
Approximately 10% of cystic fibrosis patients harbor nonsense mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene which can generate nonsense codons in the CFTR mRNA and subsequently activate the nonsense-mediated decay (NMD) pathway resulting in rapid mRNA degradation. However, it is not known which NMD branches govern the decay of CFTR mRNAs containing nonsense codons. Here we utilize antisense oligonucleotides targeting NMD factors to evaluate the regulation of nonsense codon-containing CFTR mRNAs by the NMD pathway. We observe that CFTR mRNAs with nonsense codons G542X, R1162X, and W1282X, but not Y122X, require UPF2 and UPF3 for NMD. Furthermore, we demonstrate that all evaluated CFTR mRNAs harboring nonsense codons are degraded by the SMG6-mediated endonucleolytic pathway rather than the SMG5-SMG7-mediated exonucleolytic pathway. Finally, we show that upregulation of all evaluated CFTR mRNAs with nonsense codons by NMD pathway inhibition improves outcomes of translational readthrough therapy., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
9. In Vitro and In Vivo Effects of SerpinA1 on the Modulation of Transthyretin Proteolysis.
- Author
-
Bezerra F, Niemietz C, Schmidt HHJ, Zibert A, Guo S, Monia BP, Gonçalves P, Saraiva MJ, and Almeida MR
- Subjects
- Age Factors, Amyloid metabolism, Amyloid Neuropathies, Familial genetics, Amyloid Neuropathies, Familial physiopathology, Amyloidosis genetics, Amyloidosis physiopathology, Animals, Disease Models, Animal, Female, Fibrinolysin, Hepatocytes metabolism, Humans, Male, Mice, Mice, Transgenic, Prealbumin genetics, Prealbumin physiology, Proteolysis, alpha 1-Antitrypsin physiology, Prealbumin metabolism, alpha 1-Antitrypsin metabolism
- Abstract
Transthyretin (TTR) proteolysis has been recognized as a complementary mechanism contributing to transthyretin-related amyloidosis (ATTR amyloidosis). Accordingly, amyloid deposits can be composed mainly of full-length TTR or contain a mixture of both cleaved and full-length TTR, particularly in the heart. The fragmentation pattern at Lys48 suggests the involvement of a serine protease, such as plasmin. The most common TTR variant, TTR V30M, is susceptible to plasmin-mediated proteolysis, and the presence of TTR fragments facilitates TTR amyloidogenesis. Recent studies revealed that the serine protease inhibitor, SerpinA1, was differentially expressed in hepatocyte-like cells (HLCs) from ATTR patients. In this work, we evaluated the effects of SerpinA1 on in vitro and in vivo modulation of TTR V30M proteolysis, aggregation, and deposition. We found that plasmin-mediated TTR proteolysis and aggregation are partially inhibited by SerpinA1. Furthermore, in vivo downregulation of SerpinA1 increased TTR levels in mice plasma and deposition in the cardiac tissue of older animals. The presence of TTR fragments was observed in the heart of young and old mice but not in other tissues following SerpinA1 knockdown. Increased proteolytic activity, particularly plasmin activity, was detected in mice plasmas. Overall, our results indicate that SerpinA1 modulates TTR proteolysis and aggregation in vitro and in vivo.
- Published
- 2021
- Full Text
- View/download PDF
10. Design and Rationale of the Global Phase 3 NEURO-TTRansform Study of Antisense Oligonucleotide AKCEA-TTR-L Rx (ION-682884-CS3) in Hereditary Transthyretin-Mediated Amyloid Polyneuropathy.
- Author
-
Coelho T, Ando Y, Benson MD, Berk JL, Waddington-Cruz M, Dyck PJ, Gillmore JD, Khella SL, Litchy WJ, Obici L, Monteiro C, Tai LJ, Viney NJ, Buchele G, Brambatti M, Jung SW, St L O'Dea L, Tsimikas S, Schneider E, Geary RS, Monia BP, and Gertz M
- Abstract
Introduction: AKCEA-TTR-L
Rx is a ligand-conjugated antisense (LICA) drug in development for the treatment of hereditary transthyretin amyloidosis (hATTR), a fatal disease caused by mutations in the transthyretin (TTR) gene. AKCEA-TTR-LRx shares the same nucleotide sequence as inotersen, an antisense medicine approved for use in hATTR polyneuropathy (hATTR-PN). Unlike inotersen, AKCEA-TTR-LRx is conjugated to a triantennary N-acetylgalactosamine moiety that supports receptor-mediated uptake by hepatocytes, the primary source of circulating TTR. This advanced design increases drug potency to allow for lower and less frequent dosing. The NEURO-TTRansform study will investigate whether AKCEA-TTR-LRx is safe and efficacious, with the aim of improving neurologic function and quality of life in hATTR-PN patients., Methods/design: Approximately 140 adults with stage 1 (independent ambulation) or 2 (requires ambulatory support) hATTR-PN are anticipated to enroll in this multicenter, open-label, randomized, phase 3 study. Patients will be assigned 6:1 to AKCEA-TTR-LRx 45 mg subcutaneously every 4 weeks or inotersen 300 mg once weekly until the prespecified week 35 interim efficacy analysis, after which patients receiving inotersen will receive AKCEA-TTR-LRx 45 mg subcutaneously every 4 weeks. All patients will then receive AKCEA-TTR-LRx through the remainder of the study treatment period. The final efficacy analysis at week 66 will compare the AKCEA-TTR-LRx arm with the historical placebo arm from the phase 3 trial of inotersen (NEURO-TTR). The primary outcome measures are between-group differences in the change from baseline in serum TTR, modified Neuropathy Impairment Score + 7, and Norfolk Quality of Life-Diabetic Neuropathy questionnaire., Conclusion: NEURO-TTRansform is designed to determine whether targeted delivery of AKCEA-TTR-LRx to hepatocytes with lower and less frequent doses will translate into clinical and quality-of-life benefits for patients with hATTR-PN., Trial Registration: The study is registered at ClinicalTrials.gov (NCT04136184) and EudraCT (2019-001698-10).- Published
- 2021
- Full Text
- View/download PDF
11. An oral antisense oligonucleotide for PCSK9 inhibition.
- Author
-
Gennemark P, Walter K, Clemmensen N, Rekić D, Nilsson CAM, Knöchel J, Hölttä M, Wernevik L, Rosengren B, Kakol-Palm D, Wang Y, Yu RZ, Geary RS, Riney SJ, Monia BP, Isaksson R, Jansson-Löfmark R, Rocha CSJ, Lindén D, Hurt-Camejo E, Crooke R, Tillman L, Rydén-Bergsten T, Carlsson B, Andersson U, Elebring M, Tivesten A, and Davies N
- Subjects
- Animals, Dogs, Macaca fascicularis, Rats, Serine Endopeptidases, Oligonucleotides, Antisense, PCSK9 Inhibitors
- Abstract
Inhibitors of proprotein convertase subtilisin/kexin type 9 (PCSK9) reduce low-density lipoprotein (LDL) cholesterol and are used for treatment of dyslipidemia. Current PCSK9 inhibitors are administered via subcutaneous injection. We present a highly potent, chemically modified PCSK9 antisense oligonucleotide (ASO) with potential for oral delivery. Past attempts at oral delivery using earlier-generation ASO chemistries and transient permeation enhancers provided encouraging data, suggesting that improving potency of the ASO could make oral delivery a reality. The constrained ethyl chemistry and liver targeting enabled by N -acetylgalactosamine conjugation make this ASO highly potent. A single subcutaneous dose of 90 mg reduced PCSK9 by >90% in humans with elevated LDL cholesterol and a monthly subcutaneous dose of around 25 mg is predicted to reduce PCSK9 by 80% at steady state. To investigate the feasibility of oral administration, the ASO was coformulated in a tablet with sodium caprate as permeation enhancer. Repeated oral daily dosing in dogs resulted in a bioavailability of 7% in the liver (target organ), about fivefold greater than the plasma bioavailability. Target engagement after oral administration was confirmed by intrajejunal administration of a rat-specific surrogate ASO in solution with the enhancer to rats and by plasma PCSK9 and LDL cholesterol lowering in cynomolgus monkey after tablet administration. On the basis of an assumption of 5% liver bioavailability after oral administration in humans, a daily dose of 15 mg is predicted to reduce circulating PCSK9 by 80% at steady state, supporting the development of the compound for oral administration to treat dyslipidemia., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2021
- Full Text
- View/download PDF
12. Antisense oligonucleotide-mediated knockdown of Mpzl3 attenuates the negative metabolic effects of diet-induced obesity in mice.
- Author
-
Worley BL, Auen T, Arnold AC, Monia BP, Hempel N, and Czyzyk TA
- Subjects
- Adipose Tissue metabolism, Animals, Diet, High-Fat adverse effects, Energy Metabolism, Gene Knockdown Techniques, Male, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Muscle, Skeletal metabolism, Obesity etiology, Obesity metabolism, Oligonucleotides, Antisense genetics, Lipogenesis, Membrane Proteins genetics, Obesity genetics
- Abstract
Previously, we demonstrated that global knockout (KO) of the gene encoding myelin protein zero-like 3 (Mpzl3) results in reduced body weight and adiposity, increased energy expenditure, and reduced hepatic lipid synthesis in mice. These mice also exhibit cyclic and progressive alopecia which may contribute to the observed hypermetabolic phenotype. The goal of the current study was to determine if acute and peripherally restricted knockdown of Mpzl3 could ameliorate the negative metabolic effects of exposure to a high-fat and sucrose, energy-dense (HED) diet similar to what was observed in global Mpzl3 KO mice in the absence of a skin phenotype. Mpzl3 antisense oligonucleotide (ASO) administration dose-dependently decreased fat mass and circulating lipids in HED-fed C57BL/6N mice. These changes were accompanied by a decrease in respiratory exchange ratio, a reduction in energy expenditure and food intake, a decrease in expression of genes regulating de novo lipogenesis in white adipose tissue, and an upregulation of genes associated with steroid hormone biosynthesis in liver, thermogenesis in brown adipose tissue and fatty acid transport in skeletal muscle. These data demonstrate that resistance to the negative metabolic effects of HED is a direct effect of Mpzl3 knockdown, rather than compensatory changes that could be associated with deletion of Mpzl3 during development in global KO mice. Inhibiting MPZL3 could be a potential therapeutic approach for the treatment of obesity and associated dyslipidemia., (© 2021 The Authors. Physiological Reports published by Wiley Periodicals LLC on behalf of The Physiological Society and the American Physiological Society.)
- Published
- 2021
- Full Text
- View/download PDF
13. Pharmacological targeting of coagulation factor XI mitigates the development of experimental atherosclerosis in low-density lipoprotein receptor-deficient mice.
- Author
-
Ngo ATP, Jordan KR, Mueller PA, Hagen MW, Reitsma SE, Puy C, Revenko AS, Lorentz CU, Tucker EI, Cheng Q, Hinds MT, Fazio S, Monia BP, Gailani D, Gruber A, Tavori H, and McCarty OJT
- Subjects
- Animals, Blood Coagulation, Factor XI genetics, Humans, Lipoproteins, LDL, Mice, Receptors, LDL genetics, Atherosclerosis drug therapy, Atherosclerosis genetics, Atherosclerosis prevention & control, Factor XI Deficiency
- Abstract
Background: Human coagulation factor (F) XI deficiency, a defect of the contact activation system, protects against venous thrombosis, stroke, and heart attack, whereas FXII, plasma prekallikrein, or kininogen deficiencies are asymptomatic. FXI deficiency, inhibition of FXI production, activated FXI (FXIa) inhibitors, and antibodies to FXI that interfere with FXI/FXII interactions reduce experimental thrombosis and inflammation. FXI inhibitors are antithrombotic in patients, and FXI and FXII deficiencies are atheroprotective in apolipoprotein E-deficient mice., Objectives: Investigate the effects of pharmacological targeting of FXI in experimental models of atherogenesis and established atherosclerosis., Methods and Results: Low-density lipoprotein receptor-knockout (Ldlr
-/- ) mice were administered high-fat diet (HFD) for 8 weeks; concomitantly, FXI was targeted with anti-FXI antibody (14E11) or FXI antisense oligonucleotide (ASO). 14E11 and FXI-ASO reduced atherosclerotic lesion area in proximal aortas when compared with controls, and 14E11 also reduced aortic sinus lesions. In an established disease model, in which therapy was given after atherosclerosis had developed, Ldlr-/- mice were fed HFD for 8 weeks and then administered 14E11 or FXI-ASO weekly until 16 weeks on HFD. In this established disease model, 14E11 and FXI-ASO reduced atherosclerotic lesion area in proximal aortas, but not in aortic sinus. In cultures of human endothelium, FXIa exposure disrupted VE-Cadherin expression and increased endothelial lipoprotein permeability. Strikingly, we found that 14E11 prevented the disruption of VE-Cadherin expression in aortic sinus lesions observed in the atherogenesis mouse model., Conclusion: Pharmacological targeting of FXI reduced atherogenesis in Ldlr-/- mice. Interference with the contact activation system may safely reduce development or progression of atherosclerosis., (© 2021 International Society on Thrombosis and Haemostasis.)- Published
- 2021
- Full Text
- View/download PDF
14. Ligand conjugated antisense oligonucleotide for the treatment of transthyretin amyloidosis: preclinical and phase 1 data.
- Author
-
Viney NJ, Guo S, Tai LJ, Baker BF, Aghajan M, Jung SW, Yu RZ, Booten S, Murray H, Machemer T, Burel S, Murray S, Buchele G, Tsimikas S, Schneider E, Geary RS, Benson MD, and Monia BP
- Subjects
- Humans, Healthy Volunteers, Ligands, Prealbumin genetics, Animals, Mice, Amyloid Neuropathies, Familial drug therapy, Amyloid Neuropathies, Familial genetics, Oligonucleotides, Antisense
- Abstract
Aims: Amyloidogenic transthyretin (ATTR) amyloidosis is a fatal disease characterized by progressive cardiomyopathy and/or polyneuropathy. AKCEA-TTR-L
Rx (ION-682884) is a ligand-conjugated antisense drug designed for receptor-mediated uptake by hepatocytes, the primary source of circulating transthyretin (TTR). Enhanced delivery of the antisense pharmacophore is expected to increase drug potency and support lower, less frequent dosing in treatment., Methods and Results: AKCEA-TTR-LRx demonstrated an approximate 50-fold and 30-fold increase in potency compared with the unconjugated antisense drug, inotersen, in human hepatocyte cell culture and mice expressing a mutated human genomic TTR sequence, respectively. This increase in potency was supported by a preferential distribution of AKCEA-TTR-LRx to liver hepatocytes in the transgenic hTTR mouse model. A randomized, placebo-controlled, phase 1 study was conducted to evaluate AKCEA-TTR-LRx in healthy volunteers (ClinicalTrials.gov: NCT03728634). Eligible participants were assigned to one of three multiple-dose cohorts (45, 60, and 90 mg) or a single-dose cohort (120 mg), and then randomized 10:2 (active : placebo) to receive a total of 4 SC doses (Day 1, 29, 57, and 85) in the multiple-dose cohorts or 1 SC dose in the single-dose cohort. The primary endpoint was safety and tolerability; pharmacokinetics and pharmacodynamics were secondary endpoints. All randomized participants completed treatment. No serious adverse events were reported. In the multiple-dose cohorts, AKCEA-TTR-LRx reduced TTR levels from baseline to 2 weeks after the last dose of 45, 60, or 90 mg by a mean (SD) of -85.7% (8.0), -90.5% (7.4), and -93.8% (3.4), compared with -5.9% (14.0) for pooled placebo (P < 0.001). A maximum mean (SD) reduction in TTR levels of -86.3% (6.5) from baseline was achieved after a single dose of 120 mg AKCEA-TTR-LRx ., Conclusions: These findings suggest an improved safety and tolerability profile with the increase in potency achieved by productive receptor-mediated uptake of AKCEA-TTR-LRx by hepatocytes and supports further development of AKCEA-TTR-LRx for the treatment of ATTR polyneuropathy and cardiomyopathy., (© 2020 Ionis Pharmaceuticals, INC. ESC Heart Failure published by John Wiley & Sons Ltd on behalf of European Society of Cardiology.)- Published
- 2021
- Full Text
- View/download PDF
15. SERPINA1 modulates expression of amyloidogenic transthyretin.
- Author
-
Niemietz C, Bezerra F, Almeida MR, Guo S, Monia BP, Saraiva MJ, Schütz P, Schmidt HH, and Zibert A
- Subjects
- Animals, Humans, Mice, RNA, Messenger genetics, alpha 1-Antitrypsin genetics, Amyloid Neuropathies, Familial metabolism, Prealbumin metabolism, alpha 1-Antitrypsin metabolism
- Abstract
Hereditary transthyretin amyloidosis (ATTR) is caused by amyloid deposition of misfolded transthyretin (TTR) in various tissues. Recently, reduction of circulating serum TTR, achieved via silencing oligonucleotides, was introduced as therapy of ATTR amyloidosis. We explored the impact of Serpin Family A Member 1 (SERPINA1) on TTR mRNA and protein expression. Oncostatin M (OSM) induced SERPINA1 in hepatoma cells and mice, while concomitantly TTR expression was significantly reduced. SERPINA1 knockdown resulted in specific elevated TTR expression in hepatoma cells; however other genes belonging to the group of acute phase proteins were unaffected. In mice, serum TTR was elevated after mSERPINA1 knockdown throughout antisense treatment. Following SERPINA1 knockdown, TTR deposition in several tissues, including dorsal root ganglia and intestine, was found to be increased, however numbers did not exceed significance levels. The data suggest that SERPINA1 is a co-factor of TTR expression. Our findings provide novel insight in the regulation of TTR and reveal a role of SERPINA1 in the pathogenesis of ATTR amyloidosis., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
16. Immunogenicity Assessment of Inotersen, a 2'- O -(2-Methoxyethyl) Antisense Oligonucleotide in Animals and Humans: Effect on Pharmacokinetics, Pharmacodynamics, and Safety.
- Author
-
Yu RZ, Wang Y, Norris DA, Kim TW, Narayanan P, Geary RS, Monia BP, and Henry SP
- Subjects
- Animals, Antibodies, Anti-Idiotypic blood, Antibodies, Anti-Idiotypic immunology, Blood Platelets immunology, Charcot-Marie-Tooth Disease blood, Drug-Related Side Effects and Adverse Reactions immunology, Drug-Related Side Effects and Adverse Reactions pathology, Female, Haplorhini, Humans, Immunogenicity, Vaccine genetics, Immunogenicity, Vaccine immunology, Kidney Function Tests, Male, Mice, Oligonucleotides adverse effects, Oligonucleotides, Antisense adverse effects, Oligonucleotides, Antisense blood, Oligonucleotides, Antisense pharmacokinetics, Oligoribonucleotides adverse effects, Oligoribonucleotides blood, Oligoribonucleotides pharmacokinetics, Prealbumin antagonists & inhibitors, Prealbumin immunology, Charcot-Marie-Tooth Disease drug therapy, Oligonucleotides administration & dosage, Oligonucleotides, Antisense administration & dosage, Oligoribonucleotides administration & dosage, Prealbumin genetics
- Abstract
Inotersen (TEGSEDI™) is a 2'- O -(2-methoxyethyl)-modified antisense oligonucleotide, intended for treating hereditary transthyretin (TTR) amyloidosis with polyneuropathy. The potential immunogenicity (IM) response to inotersen was evaluated in chronic nonclinical safety studies and the pivotal phase 2/3 clinical study. The evaluation was designed to assess the characteristics of antidrug antibodies (ADAs) and their effects on the pharmacokinetics, pharmacodynamics, clinical efficacy, and safety in animals and humans. No immunogenic response was observed after long-term treatment with inotersen in mice. In monkeys, the incidence rate of IM to inotersen appeared to be dose dependent, with 28.6%-50.0% of animals developing ADAs after 36 weeks of treatment. This was characterized as late onset (median onset of 185 days) with low titers (median titer of 8, or 400 if minimum required dilution of 50 is included). The overall incidence rate of patients who developed ADAs was 30% after 65 weeks of treatment with median onset of 203 days and median peak titer of 300. IM had minimal effect on plasma peak ( C
max ) and total exposure (i.e. area under curve, AUC) of inotersen, but showed elevated plasma trough levels in both IM-positive animals and humans. However, ADAs had no effect on tissue exposure, TTR messenger RNA, or plasma TTR levels in the long-term monkey study. Similarly, IM showed no effect on plasma TTR levels in clinical studies. Thus, ADAs antibodies were binding antibodies, but not neutralizing antibodies. Finally, no association was observed between IM and toxicity findings (eg, platelet, complement activation, and histopathology findings) in the inotersen 9-month monkey study. In humans, no difference was observed in hematology, including platelets, kidney function tests, or incidence of adverse events between IM-positive and -negative patients. Overall, IM showed no effect on toxicity or safety of inotersen evaluated in both monkeys and humans. ClinicalTrials.gov Identifier: NCT01737398.- Published
- 2020
- Full Text
- View/download PDF
17. Antisense Oligonucleotides Targeting Jagged 1 Reduce House Dust Mite-induced Goblet Cell Metaplasia in the Adult Murine Lung.
- Author
-
Carrer M, Crosby JR, Sun G, Zhao C, Damle SS, Kuntz SG, Monia BP, Hart CE, and Grossman TR
- Subjects
- Animals, Asthma metabolism, Biomarkers metabolism, Disease Models, Animal, Down-Regulation drug effects, Forkhead Transcription Factors metabolism, Lung metabolism, Male, Mice, Mice, Inbred BALB C, Pyroglyphidae, Receptors, Notch metabolism, Signal Transduction drug effects, Up-Regulation drug effects, Goblet Cells drug effects, Goblet Cells metabolism, Jagged-1 Protein metabolism, Lung drug effects, Metaplasia drug therapy, Metaplasia metabolism, Oligonucleotides, Antisense pharmacology
- Abstract
Goblet cell metaplasia, excessive mucus production, and inadequate mucus clearance accompany and exacerbate multiple chronic respiratory disorders, such as asthma and chronic obstructive pulmonary disease. Notch signaling plays a central role in controlling the fate of multiple cell types in the lung, including goblet cells. In the present study, we explored the therapeutic potential of modulating the Notch pathway in the adult murine lung using chemically modified antisense oligonucleotides (ASOs). To this end, we designed and characterized ASOs targeting the Notch receptors Notch1, Notch2, and Notch3 and the Notch ligands Jag1 (Jagged 1) and Jag2 (Jagged 2). Pulmonary delivery of ASOs in healthy mice or mice exposed to house dust mite, a commonly used mouse model of asthma, resulted in a significant reduction of the respective mRNAs in the lung. Furthermore, ASO-mediated knockdown of Jag1 or Notch2 in the lungs of healthy adult mice led to the downregulation of the club cell marker Scgb1a1 and the concomitant upregulation of the ciliated cell marker FoxJ1 (forkhead box J1). Similarly, ASO-mediated knockdown of Jag1 or Notch2 in the house dust mite disease model led to reduced goblet cell metaplasia and decreased mucus production. Because goblet cell metaplasia and excessive mucus secretion are a common basis for many lung pathologies, we propose that ASO-mediated inhibition of JAG1 could provide a novel therapeutic path for the treatment of multiple chronic respiratory diseases.
- Published
- 2020
- Full Text
- View/download PDF
18. Population Pharmacokinetic-Pharmacodynamic Modeling of Inotersen, an Antisense Oligonucleotide for Treatment of Patients with Hereditary Transthyretin Amyloidosis.
- Author
-
Yu RZ, Collins JW, Hall S, Ackermann EJ, Geary RS, Monia BP, Henry SP, and Wang Y
- Subjects
- Adult, Aged, Aged, 80 and over, Alanine Transaminase blood, Amyloid Neuropathies, Familial genetics, Amyloid Neuropathies, Familial pathology, Amyloid Neuropathies, Familial therapy, Bilirubin blood, Body Mass Index, Case-Control Studies, Drug Dosage Calculations, Female, Gene Expression, Glomerular Filtration Rate, Humans, Male, Middle Aged, Mutation, Neuroprotective Agents blood, Oligonucleotides blood, Prealbumin genetics, Prealbumin metabolism, RNA Interference, Serum Albumin metabolism, Amyloid Neuropathies, Familial blood, Models, Statistical, Neuroprotective Agents pharmacokinetics, Oligonucleotides pharmacokinetics, Prealbumin antagonists & inhibitors
- Abstract
A population pharmacokinetic (PK) and pharmacodynamic (PD) model was developed for inotersen to evaluate exposure-response relationships and to optimize therapeutic dosing regimen in patients with hereditary transthyretin (TTR) amyloidosis polyneuropathy (hATTR-PN). Inotersen PK and TTR level (PD) data were composed of one Phase 1 study in healthy subjects, one Phase 2/3 study in hATTR patients, and its one open-label extension study. Effects of intrinsic and extrinsic factors (covariates) on PK and PK/PD of inotersen were evaluated using a full model approach. Inotersen PK was characterized by a two-compartment model with elimination from the central compartment. The population PK analysis identified disease status and lean body mass (LBM) as significant covariates for inotersen PK. Nonetheless, the contribution of disease status and LBM on PK was small, as the difference in clearance (CL/ F ) was 11.1% between healthy subjects and patients with hATTR-PN and 38% between the lowest and highest LBM quartiles of the patient population. Age, race, sex, baseline renal function estimated glomerular filtration rate, and hepatic function markers (baseline albumin, bilirubin, and alanine aminotransferase values) were not statistically significant covariates affecting inotersen PK. An inhibitory effect indirect-response model (inhibition of TTR production) was used to describe the drug effect on TTR-time profiles, with baseline TTR included as a covariate. The overall population I
max and IC50 , together with 95% confidence interval, was estimated to be 0.913 (0.899-0.925) and 9.07 (8.08-10.1) ng/mL, respectively. V30M mutation showed no effect on the estimated IC50 value for hATTR patients. The final population PK and PK/PD model was used to simulate four different treatment regimens. The population PK/PD model developed well described the PK and PD of inotersen in patients with hATTR-PN and has been used for label recommendation and trial simulations.- Published
- 2020
- Full Text
- View/download PDF
19. Underlying Immune Disorder May Predispose Some Transthyretin Amyloidosis Subjects to Inotersen-Mediated Thrombocytopenia.
- Author
-
Narayanan P, Curtis BR, Shen L, Schneider E, Tami JA, Paz S, Burel SA, Tai LJ, Machemer T, Kwoh TJ, Xia S, Shattil SJ, Witztum JL, Engelhardt JA, Henry SP, Monia BP, and Hughes SG
- Subjects
- Adult, Aged, Amyloid Neuropathies, Familial genetics, Amyloid Neuropathies, Familial immunology, Amyloid Neuropathies, Familial pathology, Female, Genetic Predisposition to Disease, Humans, Immune System Diseases chemically induced, Immune System Diseases immunology, Immune System Diseases pathology, Immunoglobulin G, Intracranial Hemorrhages chemically induced, Intracranial Hemorrhages immunology, Intracranial Hemorrhages pathology, Male, Middle Aged, Oligodeoxyribonucleotides, Antisense administration & dosage, Oligonucleotides adverse effects, Oligonucleotides, Antisense adverse effects, Quality of Life, Thrombocytopenia chemically induced, Thrombocytopenia immunology, Thrombocytopenia pathology, Amyloid Neuropathies, Familial drug therapy, Oligonucleotides administration & dosage, Oligonucleotides, Antisense administration & dosage, Thrombocytopenia blood
- Abstract
Inotersen, a 2'-O-methoxyethyl (2'-MOE) phosphorothioate antisense oligonucleotide, reduced disease progression and improved quality of life in patients with hereditary transthyretin amyloidosis with polyneuropathy (hATTR-PN) in the NEURO-TTR and NEURO-TTR open-label extension (OLE) trials. However, 300 mg/week inotersen treatment was associated with platelet count reductions in several patients. Mean platelet counts in patients in the NEURO-TTR-inotersen group remained ≥140 × 10
9 /L in 50% and ≥100 × 109 /L in 80% of the subjects. However, grade 4 thrombocytopenia (<25 × 109 /L) occurred in three subjects in NEURO-TTR trial, and one of these suffered a fatal intracranial hemorrhage. The two others were treated successfully with corticosteroids and discontinuation of inotersen. Investigations in a subset of subjects in NEURO-TTR ( n = 17 placebo; n = 31 inotersen) and OLE ( n = 33) trials ruled out direct myelotoxicity, consumptive coagulopathy, and heparin-induced thrombocytopenia. Antiplatelet immunoglobulin G (IgG) antibodies were detected at baseline in 5 of 31 (16%) inotersen-treated subjects in NEURO-TTR, 4 of whom eventually developed grade 1 or 2 thrombocytopenia while on the drug. In addition, 24 subjects in the same group developed treatment-emergent antiplatelet IgG antibodies, of which 2 developed grade 2, and 3 developed grade 4 thrombocytopenia. Antiplatelet IgG antibodies in two of the three grade 4 thrombocytopenia subjects targeted GPIIb/IIIa. Plasma cytokines previously implicated in immune dysregulation, such as interleukin (IL)-23 and a proliferation-inducing ligand (APRIL) were often above the normal range at baseline. Collectively, these findings suggest an underlying immunologic dysregulation predisposing some individuals to immune-mediated thrombocytopenia during inotersen treatment.- Published
- 2020
- Full Text
- View/download PDF
20. Evaluation of the Antihemostatic and Antithrombotic Effects of Lowering Coagulation Factor VII Levels in a Non-human Primate.
- Author
-
Wallisch M, Olson SR, Crosby J, Johnson J, Murray SF, Shatzel JJ, Tucker EI, McCarty OJT, Hinds MT, Monia BP, and Gruber A
- Abstract
Introduction: Tissue factor (TF) and factor (F) VII, components of the extrinsic pathway of blood coagulation, are essential for hemostatic plug formation in response to injury; less clear are their roles in propagating thrombosis, as observational data in humans with congenital FVII deficiency suggests persistent thrombotic and bleeding risk even at significantly decreased FVII levels. We aimed to define the contribution of FVII to thrombus formation and hemostasis using a non-human primate model., Methods: We treated baboons with a FVII antisense oligonucleotide (ASO) and measured platelet and fibrin deposition inside and distal to collagen- or TF-coated vascular grafts. We assessed hemostasis by measuring bleeding time (BT) and prothrombin time (PT). Enoxaparin and vehicle treatments served as controls., Results: FVII-ASO treatment reduced FVII levels by 95% and significantly increased both the PT and BT. Lowering FVII levels did not decrease platelet deposition in collagen- or TF-coated grafts, in thrombi distal to the grafts, or fibrin content of either collagen- and TF-coated grafts. Lowering FVII levels were associated with a modest 25% reduction in platelet deposition at 60 min in the distal thrombus tail of TF-coated grafts only., Conclusions: FVII inhibition by way of ASO is feasible yet significantly impairs hemostasis while only exhibiting antithrombotic effects when thrombosis is initiated by vessel wall surface-associated TF exposure., (© Biomedical Engineering Society 2020.)
- Published
- 2020
- Full Text
- View/download PDF
21. Hepatic thrombopoietin gene silencing reduces platelet count and breast cancer progression in transgenic MMTV-PyMT mice.
- Author
-
Shirai T, Revenko AS, Tibbitts J, Ngo ATP, Mitrugno A, Healy LD, Johnson J, Tucker EI, Hinds MT, Coussens LM, McCarty OJT, Monia BP, and Gruber A
- Subjects
- Animals, Breast Neoplasms pathology, Cell Movement, Cell Transformation, Neoplastic, Disease Models, Animal, Disease Progression, Haplorhini, Mice, Mice, Transgenic, Neoplasm Staging, Tumor Microenvironment genetics, Breast Neoplasms blood, Breast Neoplasms etiology, Gene Silencing, Liver metabolism, Platelet Count, Thrombopoietin genetics
- Abstract
In humans, platelet count within the normal range is required for physiological hemostasis, but, adversely, platelets also support pathological thrombosis. Moreover, by releasing growth factors, they may enhance neoplastic proliferation. We hypothesize that platelet count correlates with platelet-dependent pathologies, even within the range of hemostatic competence. Because platelet production is promoted by thrombopoietin signaling through the myeloproliferative leukemia virus oncogene (cMPL), a receptor expressed on megakaryocytes, we evaluated the feasibility of selective targeting of hepatic thrombopoietin production to test this hypothesis. We synthesized murine- and primate-specific antisense oligonucleotides (THPO-ASO) that silence hepatic thrombopoietin gene (THPO) expression without blocking extrahepatic THPO. Repeated doses of THPO-ASO were administered to mice and a baboon, causing a sustained 50% decline in plasma thrombopoietin levels and platelet count within 4 weeks in both species. To investigate whether reducing platelet count within the translationally relevant hemostatic range could alter a neoplastic process, we administered THPO-ASO to 6-week-old transgenic MMTV-PyMT mice that develop early ductal atypia that progresses into cMPL-negative fatal metastatic breast cancer within 2 to 3 months. THPO-ASO treatment increased the average time to euthanasia (primary humane endpoint) at 2 cm3 combined palpable tumor volume. Our results show that THPO-ASO reduced blood platelet count, plasma platelet factor 4, vascular endothelial growth factor, thrombopoietin levels, bone marrow megakaryocyte density, tumor growth rate, proliferation index, vascularization, platelet and macrophage content, and pulmonary metastases vs untreated controls. These findings confirm that sustained and moderate pharmacological platelet count reduction is feasible with THPO-ASO administration and can delay progression of certain platelet-dependent pathological processes within a safe hemostatic platelet count range., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
22. Steric Inhibition of 5' UTR Regulatory Elements Results in Upregulation of Human CFTR.
- Author
-
Sasaki S, Sun R, Bui HH, Crosby JR, Monia BP, and Guo S
- Subjects
- 5' Untranslated Regions, Aminophenols pharmacology, Animals, Benzodioxoles pharmacology, Cell Line, Drug Combinations, Drug Synergism, Gene Expression Regulation drug effects, Genotype, HeLa Cells, Humans, Indoles pharmacology, Nucleic Acid Conformation, Oligonucleotides, Antisense chemistry, Quinolones pharmacology, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Oligonucleotides, Antisense pharmacology, Up-Regulation
- Abstract
Cystic fibrosis (CF) is an autosomal recessive monogenic disease caused by mutations in the CFTR gene. Therapeutic approaches that are focused on correcting CFTR protein face the challenge of the heterogeneity in CFTR mutations and resulting defects. Thus, while several small molecules directed at CFTR show benefit in the clinic for subsets of CF patients, these drugs cannot treat all CF patients. Additionally, the clinical benefit from treatment with these modulators could be enhanced with novel therapies. To address this unmet need, we utilized an approach to increase CFTR protein levels through antisense oligonucleotide (ASO)-mediated steric inhibition of 5' UTR regulatory elements. We identified ASOs to upregulate CFTR protein expression and confirmed the regulatory role of the sites amenable to ASO-mediated upregulation. Two ASOs were investigated further, and both increased CFTR protein expression and function in cell lines and primary human bronchial epithelial cells with distinct CF genotypes. ASO treatment further increased CFTR function in almost all CF genotypes tested on top of treatment with the FDA approved drug Symdeko (ivacaftor and tezacaftor). Thus, we present a novel approach to CFTR therapeutic intervention, through ASO-mediated modulation of translation., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
23. Nonsense-mediated RNA Decay Pathway Inhibition Restores Expression and Function of W1282X CFTR.
- Author
-
Keenan MM, Huang L, Jordan NJ, Wong E, Cheng Y, Valley HC, Mahiou J, Liang F, Bihler H, Mense M, Guo S, and Monia BP
- Subjects
- Aminophenols pharmacology, Cystic Fibrosis drug therapy, Cystic Fibrosis metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Homozygote, Humans, Mutation genetics, Quinolones pharmacology, RNA metabolism, Cystic Fibrosis Transmembrane Conductance Regulator metabolism, Nonsense Mediated mRNA Decay drug effects, RNA genetics
- Abstract
The recessive genetic disease cystic fibrosis (CF) is caused by loss-of-function mutations in the CFTR (CF transmembrane conductance regulator) gene. Approximately 10% of patients with CF have at least one allele with a nonsense mutation in CFTR. Nonsense mutations generate premature termination codons that can subject mRNA transcripts to rapid degradation through the nonsense-mediated mRNA decay (NMD) pathway. Currently, there are no approved therapies that specifically target nonsense mutations in CFTR. Here, we identified antisense oligonucleotides (ASOs) that target the NMD factor SMG1 to inhibit the NMD pathway, and determined their effects on the W1282X CFTR mutation. First, we developed and validated two in vitro models of the W1282X CFTR mutation. Next, we treated these cells with antisense oligonucleotides to inhibit NMD and measured the effects of these treatments on W1282X expression and function. SMG1-ASO-mediated NMD inhibition upregulated the RNA, protein, and surface-localized protein expression of the truncated W1282X gene product. Additionally, these ASOs increased the CFTR chloride channel function in cells homozygous for the W1282X mutation. Our approach suggests a new therapeutic strategy for patients harboring nonsense mutations and may be beneficial as a single agent in patients with CF and the W1282X mutation.
- Published
- 2019
- Full Text
- View/download PDF
24. Targeting Translation Termination Machinery with Antisense Oligonucleotides for Diseases Caused by Nonsense Mutations.
- Author
-
Huang L, Aghajan M, Quesenberry T, Low A, Murray SF, Monia BP, and Guo S
- Subjects
- Animals, Codon, Nonsense genetics, Disease Models, Animal, Gentamicins pharmacology, Hemophilia A genetics, Hemophilia A pathology, Humans, Liver drug effects, Liver metabolism, Mice, Molecular Targeted Therapy, Oligonucleotides, Antisense therapeutic use, Peptide Chain Termination, Translational drug effects, Peptide Termination Factors genetics, Protein Biosynthesis genetics, RNA, Messenger genetics, Factor IX genetics, Hemophilia A therapy, Oligonucleotides, Antisense genetics, Peptide Chain Termination, Translational genetics
- Abstract
Efforts to develop treatments for diseases caused by nonsense mutations have focused on identification of small molecules that promote translational read-through of messenger RNAs (mRNAs) harboring nonsense stop codons to produce full-length proteins. However, to date, no small molecule read-through drug has received FDA approval, probably because of a lack of balance between efficacy and safety. Depletion of translation termination factors eukaryotic release factor ( eRF ) 1 and eRF3a in cells was shown to promote translational read-through of a luciferase reporter gene harboring a nonsense mutation. In this study, we identified antisense oligonucleotides (ASOs) targeting translation termination factors and determined if ASO-mediated depletion of these factors could be a potentially effective and safe therapeutic approach for diseases caused by nonsense mutations. We found that ASO-mediated reduction of either eRF1 or eRF3a to 30%-40% of normal levels in the mouse liver is well tolerated. Hemophilia mice that express a mutant allele of human coagulation factor IX (FIX) containing nonsense mutation R338X were treated with eRF1 - or eRF3a -ASO. We found that although eRF1 - or eRF3a -ASO alone only elicited a moderate read-through effect on hFIX-R338X mRNA, both worked in synergy with geneticin, a small molecule read-through drug, demonstrating significantly increased production of functional full-length hFIX protein to levels that would rescue disease phenotypes in these mice. Overall our results indicate that modulating the translation termination pathway in the liver by ASOs may provide a novel approach to improving the efficacy of small molecule read-through drugs to treat human genetic diseases caused by nonsense mutations.
- Published
- 2019
- Full Text
- View/download PDF
25. Thrombin Signaling Promotes Pancreatic Adenocarcinoma through PAR-1-Dependent Immune Evasion.
- Author
-
Yang Y, Stang A, Schweickert PG, Lanman NA, Paul EN, Monia BP, Revenko AS, Palumbo JS, Mullins ES, Elzey BD, Janssen EM, Konieczny SF, and Flick MJ
- Subjects
- Adenocarcinoma immunology, Adenocarcinoma metabolism, Animals, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal metabolism, Mice, Mice, Inbred C57BL, Mice, Inbred NOD, Mice, SCID, Pancreatic Neoplasms immunology, Pancreatic Neoplasms metabolism, Signal Transduction, Thromboplastin metabolism, Tumor Cells, Cultured, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal pathology, Immune Evasion immunology, Pancreatic Neoplasms pathology, Receptor, PAR-1 physiology, Thrombin metabolism, Tumor Microenvironment immunology
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with robust activity of the coagulation system. To determine mechanisms by which clotting factors influence PDAC tumor progression, we generated and characterized C57Bl/6-derived KPC ( KRas
G12D , TRP53R172H ) cell lines. Tissue factor (TF) and protease-activated receptor-1 (PAR-1) were highly expressed in primary KPC pancreatic lesions and KPC cell lines similar to expression profiles observed in biopsies of patients with PDAC. In allograft studies, tumor growth and metastatic potential were significantly diminished by depletion of TF or Par-1 in cancer cells or by genetic or pharmacologic reduction of the coagulation zymogen prothrombin in mice. Notably, PAR-1-deleted KPC cells (KPC-Par-1KO ) failed to generate sizable tumors, a phenotype completely rescued by restoration of Par-1 expression. Expression profiling of KPC and KPC-Par-1KO cells indicated that thrombin-PAR-1 signaling significantly altered immune regulation pathways. Accordingly, KPC-Par-1KO cells failed to form tumors in immune-competent mice but displayed robust tumor growth comparable to that observed with control KPC cells in immune-compromised NSG mice. Immune cell depletion studies indicated that CD8 T cells, but not CD4 cells or natural killer cells, mediated elimination of KPC-Par-1KO tumor cells in C57Bl/6 mice. These results demonstrate that PDAC is driven by activation of the coagulation system through tumor cell-derived TF, circulating prothrombin, and tumor cell-derived PAR-1 and further indicate that one key mechanism of thrombin/PAR-1-mediated tumor growth is suppression of antitumor immunity in the tumor microenvironment. SIGNIFICANCE: The tissue factor-thrombin-PAR-1 signaling axis in tumor cells promotes PDAC growth and disease progression with one key mechanism being suppression of antitumor immunity in the microenvironment., (©2019 American Association for Cancer Research.)- Published
- 2019
- Full Text
- View/download PDF
26. Antisense oligonucleotide treatment ameliorates IFN-γ-induced proteinuria in APOL1-transgenic mice.
- Author
-
Aghajan M, Booten SL, Althage M, Hart CE, Ericsson A, Maxvall I, Ochaba J, Menschik-Lundin A, Hartleib J, Kuntz S, Gattis D, Ahlström C, Watt AT, Engelhardt JA, Monia BP, Magnone MC, and Guo S
- Subjects
- Animals, Cell Line, Female, Humans, Mice, Mice, Transgenic, Apolipoprotein L1 genetics, Interferon-gamma, Oligonucleotides, Antisense therapeutic use, Proteinuria drug therapy, Proteinuria etiology
- Abstract
African Americans develop end-stage renal disease at a higher rate compared with European Americans due to 2 polymorphisms (G1 and G2 risk variants) in the apolipoprotein L1 (APOL1) gene common in people of African ancestry. Although this compelling genetic evidence provides an exciting opportunity for personalized medicine in chronic kidney disease, drug discovery efforts have been greatly hindered by the fact that APOL1 expression is lacking in rodents. Here, we describe a potentially novel physiologically relevant genomic mouse model of APOL1-associated renal disease that expresses human APOL1 from the endogenous human promoter, resulting in expression in similar tissues and at similar relative levels as humans. While naive APOL1-transgenic mice did not exhibit a renal disease phenotype, administration of IFN-γ was sufficient to robustly induce proteinuria only in APOL1 G1 mice, despite inducing kidney APOL1 expression in both G0 and G1 mice, serving as a clinically relevant "second hit." Treatment of APOL1 G1 mice with IONIS-APOL1Rx, an antisense oligonucleotide (ASO) targeting APOL1 mRNA, prior to IFN-γ challenge robustly and dose-dependently inhibited kidney and liver APOL1 expression and protected against IFN-γ-induced proteinuria, indicating that the disease-relevant cell types are sensitive to ASO treatment. Therefore, IONIS-APOL1Rx may be an effective therapeutic for APOL1 nephropathies and warrants further development.
- Published
- 2019
- Full Text
- View/download PDF
27. Antisense oligonucleotide targeting of mRNAs encoding ENaC subunits α, β, and γ improves cystic fibrosis-like disease in mice.
- Author
-
Zhao C, Crosby J, Lv T, Bai D, Monia BP, and Guo S
- Subjects
- Animals, Disease Models, Animal, Gene Targeting methods, Ion Transport physiology, Lung metabolism, Lung physiopathology, Mice, Mucus metabolism, RNA, Messenger metabolism, Cystic Fibrosis genetics, Cystic Fibrosis metabolism, Cystic Fibrosis Transmembrane Conductance Regulator genetics, Epithelial Sodium Channels classification, Epithelial Sodium Channels genetics, Oligonucleotides, Antisense metabolism, Sodium metabolism
- Abstract
Background: The epithelial sodium channel ENaC consists of three subunits encoded by Scnn1a, Scnn1b, and Scnn1g and increased sodium absorption through this channel is hypothesized to lead to mucus dehydration and accumulation in cystic fibrosis (CF) patients., Methods: We identified potent and specific antisense oligonucleotides (ASOs) targeting mRNAs encoding the ENaC subunits and evaluated these ASOs in mouse models of CF-like lung disease., Results: ASOs designed to target mRNAs encoding each ENaC subunit or a control ASO were administered directly into the lungs of mice. The reductions in ENaC subunits correlated well with a reduction in amiloride sensitive channel conductance. In addition, levels of mucus markers Gob5, AGR2, Muc5ac, and Muc5b, periodic acid-Schiff's reagent (PAS) goblet cell staining, and neutrophil recruitment were reduced and lung function was improved when levels of any of the ENaC subunits were decreased., Conclusions: Delivery of ASOs targeting mRNAs encoding each of the three ENaC subunits directly into the lung improved disease phenotypes in a mouse model of CF-like lung disease. These findings suggest that targeting ENaC subunits could be an effective approach for the treatment of CF., (Copyright © 2018 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
28. IONIS-PKK Rx a Novel Antisense Inhibitor of Prekallikrein and Bradykinin Production.
- Author
-
Ferrone JD, Bhattacharjee G, Revenko AS, Zanardi TA, Warren MS, Derosier FJ, Viney NJ, Pham NC, Kaeser GE, Baker BF, Schneider E, Hughes SG, Monia BP, and MacLeod AR
- Subjects
- Angioedemas, Hereditary blood, Angioedemas, Hereditary genetics, Animals, Animals, Genetically Modified blood, Bradykinin blood, Complement C1 Inhibitor Protein pharmacology, Complement C1s antagonists & inhibitors, Dose-Response Relationship, Drug, Healthy Volunteers, Humans, Liver drug effects, Liver metabolism, Macaca fascicularis blood, Mice, Oligodeoxyribonucleotides, Antisense genetics, Oligodeoxyribonucleotides, Antisense pharmacology, Prekallikrein antagonists & inhibitors, Angioedemas, Hereditary therapy, Bradykinin genetics, Complement C1s genetics, Prekallikrein genetics
- Abstract
Kallikrein is the key contact system mediator responsible for the conversion of high-molecular-weight kininogen into the inflammatory vasodilator peptide bradykinin, a process regulated by C1-esterase inhibitor (C1-INH). In hereditary angioedema (HAE), genetic mutations result in deficient or dysfunctional C1-INH and dysregulation of the contact system leading to recurrent, sometimes fatal, angioedema attacks. IONIS-PKK
Rx is a second-generation 2'-O-(2-methoxyethyl)-modified chimeric antisense oligonucleotide, designed to bind and selectively reduce prekallikrein (PKK) mRNA in the liver. IONIS-PKKRx demonstrated dose-dependent reduction of human prekallikrein hepatic mRNA and plasma protein in transgenic mice and dose- and time-dependent reductions of plasma PKK in Cynomolgus monkeys. Similar dose-dependent reductions of plasma PKK levels were observed in healthy human volunteers accompanied by decreases in bradykinin generation capacity with an acceptable safety and tolerability profile. These results highlight a novel and specific approach to target PKK for the treatment of HAE and other diseases involving contact system activation and overproduction of bradykinin.- Published
- 2019
- Full Text
- View/download PDF
29. Inotersen (transthyretin-specific antisense oligonucleotide) for treatment of transthyretin amyloidosis.
- Author
-
Benson MD, Dasgupta NR, and Monia BP
- Subjects
- Clinical Trials, Phase III as Topic, Humans, Amyloid Neuropathies, Familial drug therapy, Oligodeoxyribonucleotides, Antisense pharmacology, Oligonucleotides pharmacology
- Abstract
Hereditary transthyretin amyloidosis (ATTR) is a fatal systemic disease that results from deposition of the misfolded protein transthyretin (TTR) in tissues. Common clinical manifestations of ATTR include peripheral neuropathy, cardiomyopathy, autonomic dysfunction, diarrhea and constipation. Historically there have not been effective therapies for this devastating disease. Inotersen/Tegsedi™ (Akcea Therapeutics, MA, USA) is a second-generation antisense oligonucleotide (ASO) specific for TTR that inhibits production of TTR by the liver. In the recently completed Phase III NEURO-TTR study, inotersen was shown to be effective in stabilizing or improving peripheral neuropathy as measured by the modified neurologic impairment score +7 (mNIS+7) and improving the quality of life assessed by the Norfolk Quality of Life-Diabetic Neuropathy (QOL-DN) questionnaire. Inotersen is a breakthrough therapy for treatment of ATTR.
- Published
- 2019
- Full Text
- View/download PDF
30. STAT3 antisense oligonucleotide AZD9150 in a subset of patients with heavily pretreated lymphoma: results of a phase 1b trial.
- Author
-
Reilley MJ, McCoon P, Cook C, Lyne P, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Curran M, Liu Q, Zhou T, Schmidt J, Jo M, Lee SJ, Yamashita M, Hughes SG, Fayad L, Piha-Paul S, Nadella MVP, Xiao X, Hsu J, Revenko A, Monia BP, MacLeod AR, and Hong DS
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Lymphoma pathology, Male, Middle Aged, Oligonucleotides pharmacology, Oligonucleotides, Antisense pharmacology, STAT3 Transcription Factor, Young Adult, Lymphoma drug therapy, Oligonucleotides therapeutic use, Oligonucleotides, Antisense therapeutic use
- Abstract
Background: The Janus kinase (JAK) and signal transduction and activation of transcription (STAT) signaling pathway is an attractive target in multiple cancers. Activation of the JAK-STAT pathway is important in both tumorigenesis and activation of immune responses. In diffuse large B-cell lymphoma (DLBCL), the transcription factor STAT3 has been associated with aggressive disease phenotype and worse overall survival. While multiple therapies inhibit upstream signaling, there has been limited success in selectively targeting STAT3 in patients. Antisense oligonucleotides (ASOs) represent a compelling therapeutic approach to target difficult to drug proteins such as STAT3 through of mRNA targeting. We report the evaluation of a next generation STAT3 ASO (AZD9150) in a non-Hodgkin's lymphoma population, primarily consisting of patients with DLBCL., Methods: Patients with relapsed or treatment refractory lymphoma were enrolled in this expansion cohort. AZD9150 was administered at 2 mg/kg and the 3 mg/kg (MTD determined by escalation cohort) dose levels with initial loading doses in the first week on days 1, 3, and 5 followed by weekly dosing. Patients were eligible to remain on therapy until unacceptable toxicity or progression. Blood was collected pre- and post-treatment for analysis of peripheral immune cells., Results: Thirty patients were enrolled, 10 at 2 mg/kg and 20 at 3 mg/kg dose levels. Twenty-seven patients had DLBCL. AZD9150 was safe and well tolerated at both doses. Common drug-related adverse events included transaminitis, fatigue, and thrombocytopenia. The 3 mg/kg dose level is the recommended phase 2 dose. All responses were seen among DLBCL patients, including 2 complete responses with median duration of response 10.7 months and 2 partial responses. Peripheral blood cell analysis of three patients without a clinical response to therapy revealed a relative increase in proportion of macrophages, CD4+, and CD8+ T cells; this trend did not reach statistical significance., Conclusions: AZD9150 was well tolerated and demonstrated efficacy in a subset of heavily pretreated patients with DLBCL. Studies in combination with checkpoint immunotherapies are ongoing., Trial Registration: Registered at ClinicalTrials.gov: NCT01563302 . First submitted 2/13/2012.
- Published
- 2018
- Full Text
- View/download PDF
31. Reducing dynamin 2 (DNM2) rescues DNM2 -related dominant centronuclear myopathy.
- Author
-
Buono S, Ross JA, Tasfaout H, Levy Y, Kretz C, Tayefeh L, Matson J, Guo S, Kessler P, Monia BP, Bitoun M, Ochala J, Laporte J, and Cowling BS
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Muscle, Skeletal metabolism, Mutation genetics, Protein Tyrosine Phosphatases, Non-Receptor genetics, Dynamin II genetics, Myopathies, Structural, Congenital genetics
- Abstract
Centronuclear myopathies (CNM) are a group of severe muscle diseases for which no effective therapy is currently available. We have previously shown that reduction of the large GTPase DNM2 in a mouse model of the X-linked form, due to loss of myotubularin phosphatase MTM1, prevents the development of the skeletal muscle pathophysiology. As DNM2 is mutated in autosomal dominant forms, here we tested whether DNM2 reduction can rescue DNM2 -related CNM in a knock-in mouse harboring the p.R465W mutation ( Dnm2
RW/+ ) and displaying a mild CNM phenotype similar to patients with the same mutation. A single intramuscular injection of adeno-associated virus-shRNA targeting Dnm2 resulted in reduction in protein levels 5 wk post injection, with a corresponding improvement in muscle mass and fiber size distribution, as well as an improvement in histopathological CNM features. To establish a systemic treatment, weekly i.p. injections of antisense oligonucleotides targeting Dnm2 were administered to Dnm2RW/+ mice for 5 wk. While muscle mass, histopathology, and muscle ultrastructure were perturbed in Dnm2RW/+ mice compared with wild-type mice, these features were indistinguishable from wild-type mice after reducing DNM2. Therefore, DNM2 knockdown via two different strategies can efficiently correct the myopathy due to DNM2 mutations, and it provides a common therapeutic strategy for several forms of centronuclear myopathy. Furthermore, we provide an example of treating a dominant disease by targeting both alleles, suggesting that this strategy may be applied to other dominant diseases., Competing Interests: Conflict of interest statement: H.T., J.L., and B.S.C. are inventors of a patent on targeting DNM2 for the treatment of centronuclear myopathies. J.L. and B.S.C. are scientific advisors for Dynacure.- Published
- 2018
- Full Text
- View/download PDF
32. Targeted delivery of antisense oligonucleotides to pancreatic β-cells.
- Author
-
Ämmälä C, Drury WJ 3rd, Knerr L, Ahlstedt I, Stillemark-Billton P, Wennberg-Huldt C, Andersson EM, Valeur E, Jansson-Löfmark R, Janzén D, Sundström L, Meuller J, Claesson J, Andersson P, Johansson C, Lee RG, Prakash TP, Seth PP, Monia BP, and Andersson S
- Subjects
- Animals, Forkhead Box Protein O1 genetics, Forkhead Box Protein O1 metabolism, Gene Silencing, Glucagon-Like Peptide-1 Receptor agonists, Glucagon-Like Peptide-1 Receptor genetics, HEK293 Cells, Humans, Mice, Inbred C57BL, Mice, Knockout, Oligonucleotides, Antisense chemistry, Oligonucleotides, Antisense pharmacokinetics, RNA, Long Noncoding genetics, Drug Delivery Systems methods, Glucagon-Like Peptide-1 Receptor metabolism, Insulin-Secreting Cells drug effects, Oligonucleotides, Antisense administration & dosage
- Abstract
Antisense oligonucleotide (ASO) silencing of the expression of disease-associated genes is an attractive novel therapeutic approach, but treatments are limited by the ability to deliver ASOs to cells and tissues. Following systemic administration, ASOs preferentially accumulate in liver and kidney. Among the cell types refractory to ASO uptake is the pancreatic insulin-secreting β-cell. Here, we show that conjugation of ASOs to a ligand of the glucagon-like peptide-1 receptor (GLP1R) can productively deliver ASO cargo to pancreatic β-cells both in vitro and in vivo. Ligand-conjugated ASOs silenced target genes in pancreatic islets at doses that did not affect target gene expression in liver or other tissues, indicating enhanced tissue and cell type specificity. This finding has potential to broaden the use of ASO technology, opening up novel therapeutic opportunities, and presents an innovative approach for targeted delivery of ASOs to additional cell types.
- Published
- 2018
- Full Text
- View/download PDF
33. Hereditary transthyretin amyloidosis: baseline characteristics of patients in the NEURO-TTR trial.
- Author
-
Waddington-Cruz M, Ackermann EJ, Polydefkis M, Heitner SB, Dyck PJ, Barroso FA, Wang AK, Berk JL, Dyck PJB, Monia BP, Hughes SG, Tai L, Jesse Kwoh T, Jung SW, Coelho T, Benson MD, and Gertz MA
- Subjects
- Adult, Aged, Aged, 80 and over, Amyloid Neuropathies, Familial drug therapy, Cardiomyopathies drug therapy, Cardiomyopathies genetics, Female, Humans, Male, Middle Aged, Oligonucleotides, Antisense therapeutic use, Polyneuropathies drug therapy, Polyneuropathies genetics, Quality of Life, Young Adult, Amyloid Neuropathies, Familial genetics, Mutation genetics, Prealbumin genetics
- Abstract
Background: Hereditary transthyretin (ATTRm) amyloidosis is a rare, progressive and fatal disease with a range of clinical manifestations., Objective: This study comprehensively evaluates disease characteristics in a large, diverse cohort of patients with ATTRm amyloidosis., Methods: Adult patients (N = 172) with Stage 1 or Stage 2 ATTRm amyloidosis who had polyneuropathy were screened and enrolled across 24 investigative sites and 10 countries in the NEURO-TTR trial ( www.clinicaltrials.gov , NCT01737398). Medical and disease history, quality of life, laboratory data, and clinical assessments were analyzed., Results: The NEURO-TTR patient population was diverse in age, disease severity, TTR mutation, and organ involvement. Twenty-seven different TTR mutations were present, with Val30Met being the most common (52%). One third of patients reported early onset disease (before age 50) and the average duration of neuropathy symptoms was 5.3 years. Symptoms affected multiple organs and systems, with nearly 70% of patients exhibiting broad involvement of weakness, sensory loss, and autonomic disturbance. Over 60% of patients had cardiomyopathy, with highest prevalence in the United States (72%) and lowest in South America/Australasia (33%). Cardiac biomarker NT-proBNP correlated with left ventricular wall thickness (p<.001). Quality of life, measured by Norfolk QoL-DN and SF-36 patient-reported questionnaires, was significantly impaired and correlated with disease severity., Conclusions: Baseline data from the NEURO-TTR trial demonstrates ATTRm amyloidosis as a systemic disease with deficits in multiple organs and body systems, leading to decreased quality of life. We report concomitant presentation of polyneuropathy and cardiomyopathy in most patients, and early involvement of multiple body systems.
- Published
- 2018
- Full Text
- View/download PDF
34. Inotersen Treatment for Patients with Hereditary Transthyretin Amyloidosis.
- Author
-
Benson MD, Waddington-Cruz M, Berk JL, Polydefkis M, Dyck PJ, Wang AK, Planté-Bordeneuve V, Barroso FA, Merlini G, Obici L, Scheinberg M, Brannagan TH 3rd, Litchy WJ, Whelan C, Drachman BM, Adams D, Heitner SB, Conceição I, Schmidt HH, Vita G, Campistol JM, Gamez J, Gorevic PD, Gane E, Shah AM, Solomon SD, Monia BP, Hughes SG, Kwoh TJ, McEvoy BW, Jung SW, Baker BF, Ackermann EJ, Gertz MA, and Coelho T
- Subjects
- Adult, Aged, Aged, 80 and over, Amyloid Neuropathies, Familial blood, Amyloid Neuropathies, Familial complications, Disease Progression, Double-Blind Method, Female, Glomerulonephritis chemically induced, Humans, Injections, Subcutaneous, Least-Squares Analysis, Male, Middle Aged, Oligonucleotides, Antisense adverse effects, Polyneuropathies etiology, Polyneuropathies therapy, Prealbumin analysis, Prealbumin genetics, Quality of Life, Severity of Illness Index, Thrombocytopenia chemically induced, Amyloid Neuropathies, Familial therapy, Oligonucleotides, Antisense therapeutic use, Prealbumin antagonists & inhibitors, RNAi Therapeutics
- Abstract
Background: Hereditary transthyretin amyloidosis is caused by pathogenic single-nucleotide variants in the gene encoding transthyretin ( TTR) that induce transthyretin misfolding and systemic deposition of amyloid. Progressive amyloid accumulation leads to multiorgan dysfunction and death. Inotersen, a 2'- O-methoxyethyl-modified antisense oligonucleotide, inhibits hepatic production of transthyretin., Methods: We conducted an international, randomized, double-blind, placebo-controlled, 15-month, phase 3 trial of inotersen in adults with stage 1 (patient is ambulatory) or stage 2 (patient is ambulatory with assistance) hereditary transthyretin amyloidosis with polyneuropathy. Patients were randomly assigned, in a 2:1 ratio, to receive weekly subcutaneous injections of inotersen (300 mg) or placebo. The primary end points were the change in the modified Neuropathy Impairment Score+7 (mNIS+7; range, -22.3 to 346.3, with higher scores indicating poorer function; minimal clinically meaningful change, 2 points) and the change in the score on the patient-reported Norfolk Quality of Life-Diabetic Neuropathy (QOL-DN) questionnaire (range, -4 to 136, with higher scores indicating poorer quality of life). A decrease in scores indicated improvement., Results: A total of 172 patients (112 in the inotersen group and 60 in the placebo group) received at least one dose of a trial regimen, and 139 (81%) completed the intervention period. Both primary efficacy assessments favored inotersen: the difference in the least-squares mean change from baseline to week 66 between the two groups (inotersen minus placebo) was -19.7 points (95% confidence interval [CI], -26.4 to -13.0; P<0.001) for the mNIS+7 and -11.7 points (95% CI, -18.3 to -5.1; P<0.001) for the Norfolk QOL-DN score. These improvements were independent of disease stage, mutation type, or the presence of cardiomyopathy. There were five deaths in the inotersen group and none in the placebo group. The most frequent serious adverse events in the inotersen group were glomerulonephritis (in 3 patients [3%]) and thrombocytopenia (in 3 patients [3%]), with one death associated with one of the cases of grade 4 thrombocytopenia. Thereafter, all patients received enhanced monitoring., Conclusions: Inotersen improved the course of neurologic disease and quality of life in patients with hereditary transthyretin amyloidosis. Thrombocytopenia and glomerulonephritis were managed with enhanced monitoring. (Funded by Ionis Pharmaceuticals; NEURO-TTR ClinicalTrials.gov number, NCT01737398 .).
- Published
- 2018
- Full Text
- View/download PDF
35. Antisense suppression of the nonsense mediated decay factor Upf3b as a potential treatment for diseases caused by nonsense mutations.
- Author
-
Huang L, Low A, Damle SS, Keenan MM, Kuntz S, Murray SF, Monia BP, and Guo S
- Subjects
- Animals, Cells, Cultured, Dystrophin genetics, Factor IX metabolism, Hemophilia B genetics, Hemophilia B metabolism, Hemophilia B therapy, Liver metabolism, Mice, RNA Stability, RNA, Messenger metabolism, RNA-Binding Proteins genetics, Transcriptome, Codon, Nonsense, Nonsense Mediated mRNA Decay, Oligonucleotides, Antisense, RNA-Binding Proteins antagonists & inhibitors
- Abstract
Background: About 11% of all human genetic diseases are caused by nonsense mutations that generate premature translation termination codons (PTCs) in messenger RNAs (mRNA). PTCs not only lead to the production of truncated proteins, but also often result in decreased mRNA abundance due to nonsense-mediated mRNA decay (NMD). Although pharmacological inhibition of NMD could be an attractive therapeutic approach for the treatment of diseases caused by nonsense mutations, NMD also regulates the expression of 10-20% of the normal transcriptome., Results: Here, we investigate whether NMD can be inhibited to stabilize mutant mRNAs, which may subsequently produce functional proteins, without having a major impact on the normal transcriptome. We develop antisense oligonucleotides (ASOs) to systematically deplete each component in the NMD pathway. We find that ASO-mediated depletion of each NMD factor elicits different magnitudes of NMD inhibition in vitro and are differentially tolerated in normal mice. Among all of the NMD factors, Upf3b depletion is well tolerated, consistent with previous reports that UPF3B is not essential for development and regulates only a subset of the endogenous NMD substrates. While minimally impacting the normal transcriptome, Upf3b-ASO treatment significantly stabilizes the PTC-containing dystrophin mRNA in mdx mice and coagulation factor IX mRNA in a hemophilia mouse model. Furthermore, when combined with reagents promoting translational read-through, Upf3b-ASO treatment leads to the production of functional factor IX protein in hemophilia mice., Conclusions: These data demonstrate that ASO-mediated reduction of the NMD factor Upf3b could be an effective and safe approach for the treatment of diseases caused by nonsense mutations.
- Published
- 2018
- Full Text
- View/download PDF
36. Inhaled ENaC antisense oligonucleotide ameliorates cystic fibrosis-like lung disease in mice.
- Author
-
Crosby JR, Zhao C, Jiang C, Bai D, Katz M, Greenlee S, Kawabe H, McCaleb M, Rotin D, Guo S, and Monia BP
- Subjects
- Administration, Inhalation, Animals, Disease Models, Animal, Epithelial Sodium Channel Blockers pharmacology, Mice, Respiratory System Agents pharmacology, Treatment Outcome, Cystic Fibrosis metabolism, Cystic Fibrosis physiopathology, Epithelial Sodium Channels metabolism, Oligonucleotides, Antisense pharmacology, Respiratory Mucosa metabolism, Respiratory Mucosa physiopathology
- Abstract
Background: Epithelial sodium channel (ENaC, Scnn1) hyperactivity in the lung leads to airway surface dehydration and mucus accumulation in cystic fibrosis (CF) patients and in mice with CF-like lung disease., Methods: We identified several potent ENaC specific antisense oligonucleotides (ASOs) and tested them by inhalation in mouse models of CF-like lung disease., Results: The inhaled ASOs distributed into lung airway epithelial cells and decreased ENaC expression by inducing RNase H1-dependent degradation of the targeted Scnn1a mRNA. Aerosol delivered ENaC ASO down-regulated mucus marker expression and ameliorated goblet cell metaplasia, inflammation, and airway hyper-responsiveness. Lack of systemic activity of ASOs delivered via the aerosol route ensures the safety of this approach., Conclusions: Our results demonstrate that antisense inhibition of ENaC in airway epithelial cells could be an effective and safe approach for the prevention and reversal of lung symptoms in CF and potentially other inflammatory diseases of the lung., (Copyright © 2017 European Cystic Fibrosis Society. Published by Elsevier B.V. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
37. Reduction in ocular complement factor B protein in mice and monkeys by systemic administration of factor B antisense oligonucleotide.
- Author
-
Grossman TR, Carrer M, Shen L, Johnson RB, Hettrick LA, Henry SP, Monia BP, and McCaleb ML
- Subjects
- Animals, Blotting, Western, Electrophoresis, Polyacrylamide Gel, Injections, Subcutaneous, Macaca fascicularis, Male, Mice, Mice, Inbred C57BL, Real-Time Polymerase Chain Reaction, Complement Factor B genetics, Complement Factor B metabolism, Eye metabolism, Liver metabolism, Oligonucleotides, Antisense administration & dosage, RNA, Messenger metabolism
- Abstract
Purpose: Age-related macular degeneration (AMD) is the leading cause of permanent vision loss among the elderly in many industrialized countries, and the complement system plays an important role in the pathogenesis of AMD. Inhibition of complement factor B, a key regulator of the alternative pathway, is implicated as a potential therapeutic intervention for AMD. Here we investigated the effect of liver factor B reduction on systemic and ocular factor B levels., Methods: Second-generation antisense oligonucleotides (ASOs) targeting mouse and monkey factor B mRNA were administered by subcutaneous injection to healthy mice or monkeys, and the level of factor B mRNA was assessed in the liver and the eye. In addition, the factor B protein level was determined in plasma and whole eyes from the treated animals., Results: Mice and monkeys treated with factor B ASOs demonstrated a robust reduction in liver factor B mRNA levels with no change in ocular factor B mRNA levels. Plasma factor B protein levels were significantly reduced in mice and monkeys treated with factor B ASOs, leading to a dramatic reduction in ocular factor B protein, below the assay detection levels., Conclusions: The results add to the increasing evidence that the liver is the main source of plasma and ocular factor B protein, and demonstrate that reduction of liver factor B mRNA by an ASO results in a significant reduction in plasma and ocular factor B protein levels. The results suggest that inhibition of liver factor B mRNA by factor B ASOs would reduce systemic alternative complement pathway activation and has potential to be used as a novel therapy for AMD.
- Published
- 2017
38. Navigating the Future of Cardiovascular Drug Development-Leveraging Novel Approaches to Drive Innovation and Drug Discovery: Summary of Findings from the Novel Cardiovascular Therapeutics Conference.
- Author
-
Povsic TJ, Scott R, Mahaffey KW, Blaustein R, Edelberg JM, Lefkowitz MP, Solomon SD, Fox JC, Healy KE, Khakoo AY, Losordo DW, Malik FI, Monia BP, Montgomery RL, Riesmeyer J, Schwartz GG, Zelenkofske SL, Wu JC, Wasserman SM, and Roe MT
- Subjects
- Animals, Cardiovascular Agents pharmacology, Cardiovascular Diseases physiopathology, Drug Discovery methods, Drug Evaluation, Preclinical methods, Drug Industry, Humans, Cardiovascular Agents therapeutic use, Cardiovascular Diseases drug therapy, Drug Design
- Abstract
Purpose: The need for novel approaches to cardiovascular drug development served as the impetus to convene an open meeting of experts from the pharmaceutical industry and academia to assess the challenges and develop solutions for drug discovery in cardiovascular disease., Methods: The Novel Cardiovascular Therapeutics Summit first reviewed recent examples of ongoing or recently completed programs translating basic science observations to targeted drug development, highlighting successes (protein convertase sutilisin/kexin type 9 [PCSK9] and neprilysin inhibition) and targets still under evaluation (cholesteryl ester transfer protein [CETP] inhibition), with the hope of gleaning key lessons to successful drug development in the current era. Participants then reviewed the use of innovative approaches being explored to facilitate rapid and more cost-efficient evaluations of drug candidates in a short timeframe., Results: We summarize observations gleaned from this summit and offer insight into future cardiovascular drug development., Conclusions: The rapid development in genetic and high-throughput drug evaluation technologies, coupled with new approaches to rapidly evaluate potential cardiovascular therapies with in vitro techniques, offer opportunities to identify new drug targets for cardiovascular disease, study new therapies with better efficiency and higher throughput in the preclinical setting, and more rapidly bring the most promising therapies to human testing. However, there must be a critical interface between industry and academia to guide the future of cardiovascular drug development. The shared interest among academic institutions and pharmaceutical companies in developing promising therapies to address unmet clinical needs for patients with cardiovascular disease underlies and guides innovation and discovery platforms that are significantly altering the landscape of cardiovascular drug development.
- Published
- 2017
- Full Text
- View/download PDF
39. Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic antisense oligonucleotide inhibitor of KRAS.
- Author
-
Ross SJ, Revenko AS, Hanson LL, Ellston R, Staniszewska A, Whalley N, Pandey SK, Revill M, Rooney C, Buckett LK, Klein SK, Hudson K, Monia BP, Zinda M, Blakey DC, Lyne PD, and Macleod AR
- Subjects
- Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cell Proliferation genetics, Disease Models, Animal, Humans, Mice, Mutation genetics, Oligonucleotides, Antisense therapeutic use, Signal Transduction drug effects, Signal Transduction genetics, Xenograft Model Antitumor Assays, ras Proteins antagonists & inhibitors, Carcinoma, Non-Small-Cell Lung drug therapy, Lung Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use, ras Proteins metabolism
- Abstract
Activating mutations in KRAS underlie the pathogenesis of up to 20% of human tumors, and KRAS is one of the most frequently mutated genes in cancer. Developing therapeutics to block KRAS activity has proven difficult, and no direct inhibitor of KRAS function has entered clinical trials. We describe the preclinical evaluation of AZD4785, a high-affinity constrained ethyl-containing therapeutic antisense oligonucleotide (ASO) targeting KRAS mRNA. AZD4785 potently and selectively depleted cellular KRAS mRNA and protein, resulting in inhibition of downstream effector pathways and antiproliferative effects selectively in KRAS mutant cells. AZD4785-mediated depletion of KRAS was not associated with feedback activation of the mitogen-activated protein kinase (MAPK) pathway, which is seen with RAS-MAPK pathway inhibitors. Systemic delivery of AZD4785 to mice bearing KRAS mutant non-small cell lung cancer cell line xenografts or patient-derived xenografts resulted in inhibition of KRAS expression in tumors and antitumor activity. The safety of this approach was demonstrated in mice and monkeys with KRAS ASOs that produced robust target knockdown in a broad set of tissues without any adverse effects. Together, these data suggest that AZD4785 is an attractive therapeutic for the treatment of KRAS -driven human cancers and warrants further development., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
40. Antisense oligonucleotide-mediated Dnm2 knockdown prevents and reverts myotubular myopathy in mice.
- Author
-
Tasfaout H, Buono S, Guo S, Kretz C, Messaddeq N, Booten S, Greenlee S, Monia BP, Cowling BS, and Laporte J
- Subjects
- Animals, Disease Models, Animal, Dynamin II metabolism, Female, Kidney metabolism, Liver metabolism, Male, Mice, Mice, Knockout, Muscle Contraction, Muscle, Skeletal metabolism, Mutation, Myopathies, Structural, Congenital metabolism, Myopathies, Structural, Congenital therapy, Phenotype, Protein Tyrosine Phosphatases, Non-Receptor metabolism, Recombination, Genetic, Dynamin II genetics, Myopathies, Structural, Congenital genetics, Oligonucleotides, Antisense genetics, Protein Tyrosine Phosphatases, Non-Receptor genetics
- Abstract
Centronuclear myopathies (CNM) are non-dystrophic muscle diseases for which no effective therapy is currently available. The most severe form, X-linked CNM, is caused by myotubularin 1 (MTM1) loss-of-function mutations, while the main autosomal dominant form is due to dynamin2 (DNM2) mutations. We previously showed that genetic reduction of DNM2 expression in Mtm1 knockout (Mtm1KO) mice prevents development of muscle pathology. Here we show that systemic delivery of Dnm2 antisense oligonucleotides (ASOs) into Mtm1KO mice efficiently reduces DNM2 protein level in muscle and prevents the myopathy from developing. Moreover, systemic ASO injection into severely affected mice leads to reversal of muscle pathology within 2 weeks. Thus, ASO-mediated DNM2 knockdown can efficiently correct muscle defects due to loss of MTM1, providing an attractive therapeutic strategy for this disease.
- Published
- 2017
- Full Text
- View/download PDF
41. Treatment of transthyretin cardiomyopathy with a TTR-specific antisense oligonucleotide (IONIS-TTR Rx ).
- Author
-
Benson MD, Ackermann EJ, and Monia BP
- Subjects
- Amyloid Neuropathies genetics, Humans, Prealbumin genetics, Amyloid Neuropathies, Familial genetics, Cardiomyopathies genetics, Oligonucleotides, Antisense genetics
- Published
- 2017
- Full Text
- View/download PDF
42. Knockdown of Z Mutant Alpha-1 Antitrypsin In Vivo Using Modified DNA Antisense Oligonucleotides.
- Author
-
Aghajan M, Guo S, and Monia BP
- Subjects
- Animals, DNA, Antisense administration & dosage, Disease Models, Animal, Humans, Liver metabolism, Mice, Oligonucleotides, Antisense administration & dosage, RNA Interference, RNA, Messenger genetics, RNA, Messenger metabolism, Solubility, alpha 1-Antitrypsin blood, alpha 1-Antitrypsin metabolism, Gene Knockdown Techniques methods, Mutation genetics, alpha 1-Antitrypsin genetics
- Abstract
Alpha-1 antitrypsin (AAT) is a serum protease inhibitor, mainly expressed in and secreted from hepatocytes, important for regulating neutrophil elastase activity among other proteases. Various mutations in AAT cause alpha-1 antitrypsin deficiency (AATD), a rare hereditary disorder that results in liver disease due to accumulation of AAT aggregates and lung disease from excessive neutrophil elastase activity. PiZ transgenic mice contain the human AAT genomic region harboring the most common AATD mutation, the Glu342Lys (Z) point mutation. These mice effectively recapitulate the liver disease exhibited in AATD patients, including AAT protein aggregates, hepatocyte death, and eventual liver fibrosis. Previously, we demonstrated that modified antisense oligonucleotides (ASOs) can dramatically reduce Z-AAT RNA and protein levels in PiZ mice enabling inhibition, prevention, and reversal of the associated liver disease. Here, we describe in detail usage of AAT-ASOs to knock down Z-AAT in PiZ mice with a focus on preparation and in vivo delivery of ASOs, as well as detailed workflows pertaining to the analysis of Z-AAT mRNA, plasma protein, and soluble/insoluble liver protein levels following ASO administration.
- Published
- 2017
- Full Text
- View/download PDF
43. Use of antisense oligonucleotides to correct the splicing error in ISCU myopathy patient cell lines.
- Author
-
Holmes-Hampton GP, Crooks DR, Haller RG, Guo S, Freier SM, Monia BP, and Rouault TA
- Subjects
- Acidosis, Lactic genetics, Acidosis, Lactic pathology, Acidosis, Lactic therapy, Female, Humans, Introns genetics, Male, Mitochondria drug effects, Mitochondria metabolism, Mitochondria pathology, Muscle, Skeletal drug effects, Muscle, Skeletal pathology, Muscular Diseases genetics, Muscular Diseases pathology, Muscular Diseases therapy, Oligonucleotides, Antisense therapeutic use, Phenotype, Point Mutation, RNA Splicing drug effects, RNA Splicing genetics, Succinate Dehydrogenase biosynthesis, Acidosis, Lactic congenital, Iron-Sulfur Proteins genetics, Muscular Diseases congenital, Oligonucleotides, Antisense genetics, Succinate Dehydrogenase genetics
- Abstract
ISCU myopathy is an inherited disease that primarily affects individuals of northern Swedish descent who share a single point mutation in the fourth intron of the ISCU gene. The current study shows correction of specific phenotypes associated with disease following treatment with an antisense oligonucleotide (ASO) targeted to the site of the mutation. We have shown that ASO treatment diminished aberrant splicing and increased ISCU protein levels in both patient fibroblasts and patient myotubes in a concentration dependent fashion. Upon ASO treatment, levels of SDHB in patient myotubular cell lines increased to levels observed in control myotubular cell lines. Additionally, we have shown that both patient fibroblast and myotubular cell lines displayed an increase in complex II activity with a concomitant decrease in succinate levels in patient myotubular cell lines after ASO treatment. Mitochondrial and cytosolic aconitase activities increased significantly following ASO treatment in patient myotubes. The current study suggests that ASO treatment may serve as a viable approach to correcting ISCU myopathy in patients., (Published by Oxford University Press 2016. This work is written by US Government employees and is in the public domain in the US.)
- Published
- 2016
- Full Text
- View/download PDF
44. Suppressing transthyretin production in mice, monkeys and humans using 2nd-Generation antisense oligonucleotides.
- Author
-
Ackermann EJ, Guo S, Benson MD, Booten S, Freier S, Hughes SG, Kim TW, Jesse Kwoh T, Matson J, Norris D, Yu R, Watt A, and Monia BP
- Subjects
- Adolescent, Adult, Amyloid Neuropathies, Familial therapy, Animals, Double-Blind Method, Female, Gene Expression, Healthy Volunteers, Hep G2 Cells, Hepatocytes cytology, Humans, Macaca fascicularis, Male, Mice, Mice, Transgenic, Middle Aged, Oligonucleotides, Antisense administration & dosage, Prealbumin biosynthesis, Prealbumin genetics, Primary Cell Culture, RNA Cleavage, RNA, Messenger biosynthesis, RNA, Messenger genetics, Hepatocytes metabolism, Liver metabolism, Oligonucleotides, Antisense pharmacokinetics, Prealbumin antagonists & inhibitors, RNA, Messenger antagonists & inhibitors, Ribonuclease H metabolism
- Abstract
Transthyretin amyloidosis (ATTR amyloidosis) is a rare disease that results from the deposition of misfolded transthyretin (TTR) protein from the plasma into tissues as amyloid fibrils, leading to polyneuropathy and cardiomyopathy. IONIS-TTR
Rx (ISIS 420915) is a 2nd-Generation 2'-O-(2-methoxyethyl) modified "2'-MOE" antisense oligonucleotide (ASO) that targets the TTR RNA transcript and reduces the levels of the TTR transcript through an RNaseH1 mechanism of action, leading to reductions in both mutant and wild-type TTR protein. The activity of IONIS-TTRRx to decrease TTR protein levels was studied in transgenic mice bearing the Ile84Ser human TTR mutant, in cynomolgus monkeys and in healthy human volunteers. Robust (>80%) reductions of plasma TTR protein were obtained in all three species treated with IONIS-TTRRx , which in mice and monkeys was associated with substantial reductions in hepatic TTR RNA levels. These effects were dose-dependent and lasted for weeks post-dosing. In a Phase 1 healthy volunteer study, treatment with IONIS-TTRRx for four weeks was well tolerated without any remarkable safety issues. TTR protein reductions up to 96% in plasma were observed. These nonclinical and clinical results support the ongoing Phase 3 development of IONIS-TTRRx in patients with ATTR amyloidosis.- Published
- 2016
- Full Text
- View/download PDF
45. Evaluation of Therapeutic Oligonucleotides for Familial Amyloid Polyneuropathy in Patient-Derived Hepatocyte-Like Cells.
- Author
-
Niemietz CJ, Sauer V, Stella J, Fleischhauer L, Chandhok G, Guttmann S, Avsar Y, Guo S, Ackermann EJ, Gollob J, Monia BP, Zibert A, and Schmidt HH
- Subjects
- Adult, Aged, Amyloid Neuropathies, Familial genetics, Amyloid Neuropathies, Familial urine, Cell Differentiation, Drug Evaluation, Preclinical, Female, Gene Knockdown Techniques, Humans, Induced Pluripotent Stem Cells cytology, Male, Middle Aged, Oligonucleotides, Antisense pharmacology, Prealbumin genetics, RNA, Messenger genetics, RNA, Small Interfering genetics, Amyloid Neuropathies, Familial drug therapy, Hepatocytes drug effects, Oligonucleotides, Antisense therapeutic use
- Abstract
Familial amyloid polyneuropathy (FAP) is caused by mutations of the transthyretin (TTR) gene, predominantly expressed in the liver. Two compounds that knockdown TTR, comprising a small interfering RNA (siRNA; ALN-TTR-02) and an antisense oligonucleotide (ASO; IONIS-TTRRx), are currently being evaluated in clinical trials. Since primary hepatocytes from FAP patients are rarely available for molecular analysis and commercial tissue culture cells or animal models lack the patient-specific genetic background, this study uses primary cells derived from urine of FAP patients. Urine-derived cells were reprogrammed to induced pluripotent stem cells (iPSCs) with high efficiency. Hepatocyte-like cells (HLCs) showing typical hepatic marker expression were obtained from iPSCs of the FAP patients. TTR mRNA expression of FAP HLCs almost reached levels measured in human hepatocytes. To assess TTR knockdown, siTTR1 and TTR-ASO were introduced to HLCs. A significant downregulation (>80%) of TTR mRNA was induced in the HLCs by both oligonucleotides. TTR protein present in the cell culture supernatant of HLCs was similarly downregulated. Gene expression of other hepatic markers was not affected by the therapeutic oligonucleotides. Our data indicate that urine cells (UCs) after reprogramming and hepatic differentiation represent excellent primary human target cells to assess the efficacy and specificity of novel compounds., Competing Interests: This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2016
- Full Text
- View/download PDF
46. Inhibition of the alternative complement pathway by antisense oligonucleotides targeting complement factor B improves lupus nephritis in mice.
- Author
-
Grossman TR, Hettrick LA, Johnson RB, Hung G, Peralta R, Watt A, Henry SP, Adamson P, Monia BP, and McCaleb ML
- Subjects
- Animals, Cells, Cultured, Complement C3 metabolism, Complement Factor B metabolism, Complement Pathway, Alternative genetics, Disease Models, Animal, Humans, Kidney pathology, Lupus Erythematosus, Systemic immunology, Lupus Nephritis immunology, Mice, Mice, Inbred MRL lpr, Mice, Inbred NZB, Proteinuria, Antigen-Antibody Complex metabolism, Complement Factor B genetics, Hepatocytes physiology, Kidney metabolism, Lupus Erythematosus, Systemic therapy, Lupus Nephritis therapy, Oligonucleotides, Antisense genetics
- Abstract
Systemic lupus erythematosus is an autoimmune disease that manifests in widespread complement activation and deposition of complement fragments in the kidney. The complement pathway is believed to play a significant role in the pathogenesis and in the development of lupus nephritis. Complement factor B is an important activator of the alternative complement pathway and increasing evidence supports reducing factor B as a potential novel therapy to lupus nephritis. Here we investigated whether pharmacological reduction of factor B expression using antisense oligonucleotides could be an effective approach for the treatment of lupus nephritis. We identified potent and well tolerated factor B antisense oligonucleotides that resulted in significant reductions in hepatic and plasma factor B levels when administered to normal mice. To test the effects of factor B antisense oligonucleotides on lupus nephritis, we used two different mouse models, NZB/W F1 and MRL/lpr mice, that exhibit lupus nephritis like renal pathology. Antisense oligonucleotides mediated reductions in circulating factor B levels were associated with significant improvements in renal pathology, reduced glomerular C3 deposition and proteinuria, and improved survival. These data support the strategy of using factor B antisense oligonucleotides for treatment of lupus nephritis in humans., (Copyright © 2015 The Authors. Published by Elsevier GmbH.. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
47. Combination of Tmprss6- ASO and the iron chelator deferiprone improves erythropoiesis and reduces iron overload in a mouse model of beta-thalassemia intermedia.
- Author
-
Casu C, Aghajan M, Oikonomidou PR, Guo S, Monia BP, and Rivella S
- Subjects
- Animals, Deferiprone, Disease Models, Animal, Erythropoiesis genetics, Membrane Proteins genetics, Membrane Proteins metabolism, Mice, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, beta-Thalassemia genetics, beta-Thalassemia metabolism, Erythropoiesis drug effects, Iron Chelating Agents pharmacology, Membrane Proteins antagonists & inhibitors, Pyridones pharmacology, beta-Thalassemia drug therapy
- Published
- 2016
- Full Text
- View/download PDF
48. Reversing Antisense Oligonucleotide Activity with a Sense Oligonucleotide Antidote: Proof of Concept Targeting Prothrombin.
- Author
-
Crosby JR, Zhao C, Zhang H, MacLeod AR, Guo S, and Monia BP
- Subjects
- Animals, Dose-Response Relationship, Drug, Male, Mice, Mice, Inbred BALB C, Oligonucleotides, Antisense pharmacology, Oligonucleotides pharmacology, Oligonucleotides, Antisense antagonists & inhibitors, Prothrombin drug effects
- Abstract
The tissue half-life of second-generation antisense oligonucleotide drugs (ASOs) is generally longer than traditional small molecule therapeutics. Thus, a strategy to reverse the activity of antisense drugs is warranted in certain settings. In this study, we describe a strategy employing the administration of a complementary sense oligonucleotide antidote (SOA). As a model system we have chosen to target the coagulation factor and antithrombotic drug target, prothrombin, to assess the feasibility of this approach. ASO targeting mouse prothrombin specifically suppressed >90% hepatic prothrombin mRNA levels and circulating prothrombin protein in mice. These effects were dose- and time-dependent, and as expected produced predictable increases in anticoagulation activity [prothrombin time/activated partial thromboplastin time (PT/aPTT)]. Treatment with prothrombin SOAs resulted in a dose-dependent reversal of ASO activity, as measured by a return in prothrombin mRNA levels and thrombin activity, and normalization of aPTT and PT. The antithrombotic activity of prothrombin ASOs was demonstrated in a FeCl3-induced thrombosis mouse model, and as predicted for this target, the doses required for antithrombotic activity were also associated with increased bleeding. Treatment with SOA was able to prevent prothrombin ASO-induced bleeding in a dose-dependent manner. These studies demonstrate for the first time the utility of SOAs to selectively and specifically reverse the intracellular effects of an antisense therapy.
- Published
- 2015
- Full Text
- View/download PDF
49. AZD9150, a next-generation antisense oligonucleotide inhibitor of STAT3 with early evidence of clinical activity in lymphoma and lung cancer.
- Author
-
Hong D, Kurzrock R, Kim Y, Woessner R, Younes A, Nemunaitis J, Fowler N, Zhou T, Schmidt J, Jo M, Lee SJ, Yamashita M, Hughes SG, Fayad L, Piha-Paul S, Nadella MV, Mohseni M, Lawson D, Reimer C, Blakey DC, Xiao X, Hsu J, Revenko A, Monia BP, and MacLeod AR
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Apoptosis, Carcinoma, Non-Small-Cell Lung genetics, Carcinoma, Non-Small-Cell Lung metabolism, Carcinoma, Non-Small-Cell Lung pathology, Cell Line, Tumor, Cell Proliferation, Down-Regulation, Female, Humans, Lung Neoplasms genetics, Lung Neoplasms metabolism, Lung Neoplasms pathology, Lymphoma genetics, Lymphoma metabolism, Lymphoma pathology, Male, Mice, Inbred BALB C, Mice, Inbred NOD, Mice, Nude, Mice, SCID, Middle Aged, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Time Factors, Treatment Outcome, Tumor Burden, Xenograft Model Antitumor Assays, Antineoplastic Agents therapeutic use, Carcinoma, Non-Small-Cell Lung therapy, Gene Expression Regulation, Neoplastic drug effects, Gene Knockdown Techniques, Lung Neoplasms therapy, Lymphoma therapy, Oligonucleotides therapeutic use, Oligonucleotides, Antisense therapeutic use, STAT3 Transcription Factor antagonists & inhibitors
- Abstract
Next-generation sequencing technologies have greatly expanded our understanding of cancer genetics. Antisense technology is an attractive platform with the potential to translate these advances into improved cancer therapeutics, because antisense oligonucleotide (ASO) inhibitors can be designed on the basis of gene sequence information alone. Recent human clinical data have demonstrated the potent activity of systemically administered ASOs targeted to genes expressed in the liver. We describe the preclinical activity and initial clinical evaluation of a class of ASOs containing constrained ethyl modifications for targeting the gene encoding the transcription factor STAT3, a notoriously difficult protein to inhibit therapeutically. Systemic delivery of the unformulated ASO, AZD9150, decreased STAT3 expression in a broad range of preclinical cancer models and showed antitumor activity in lymphoma and lung cancer models. AZD9150 preclinical activity translated into single-agent antitumor activity in patients with highly treatment-refractory lymphoma and non-small cell lung cancer in a phase 1 dose-escalation study., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
50. Genetic diminution of circulating prothrombin ameliorates multiorgan pathologies in sickle cell disease mice.
- Author
-
Arumugam PI, Mullins ES, Shanmukhappa SK, Monia BP, Loberg A, Shaw MA, Rizvi T, Wansapura J, Degen JL, and Malik P
- Subjects
- Anemia, Sickle Cell mortality, Anemia, Sickle Cell physiopathology, Animals, Blood Coagulation, Cells, Cultured, Hypertension, Pulmonary etiology, Immunoenzyme Techniques, Inflammation etiology, Magnetic Resonance Imaging, Male, Mice, Mice, Knockout, Oligoribonucleotides, Antisense pharmacology, Prothrombin antagonists & inhibitors, Survival Rate, Thrombin metabolism, Vascular Diseases etiology, Anemia, Sickle Cell complications, Genetic Therapy, Hypertension, Pulmonary prevention & control, Inflammation prevention & control, Prothrombin physiology, Vascular Diseases prevention & control
- Abstract
Sickle cell disease (SCD) results in vascular occlusions, chronic hemolytic anemia, and cumulative organ damage. A conspicuous feature of SCD is chronic inflammation and coagulation system activation. Thrombin (factor IIa [FIIa]) is both a central protease in hemostasis and a key modifier of inflammatory processes. To explore the hypothesis that reduced prothrombin (factor II [FII]) levels in SCD will limit vaso-occlusion, vasculopathy, and inflammation, we used 2 strategies to suppress FII in SCD mice. Weekly administration of FII antisense oligonucleotide "gapmer" to Berkeley SCD mice to selectively reduce circulating FII levels to ∼10% of normal for 15 weeks significantly diminished early mortality. More comprehensive, long-term comparative studies were done using mice with genetic diminution of circulating FII. Here, cohorts of FII(lox/-) mice (constitutively carrying ∼10% normal FII) and FII(WT) mice were tracked in parallel for a year following the imposition of SCD via hematopoietic stem cell transplantation. This genetically imposed suppression of FII levels resulted in an impressive reduction in inflammation (reduction in leukocytosis, thrombocytosis, and circulating interleukin-6 levels), reduced endothelial cell dysfunction (reduced endothelial activation and circulating soluble vascular cell adhesion molecule), and a significant improvement in SCD-associated end-organ damage (nephropathy, pulmonary hypertension, pulmonary inflammation, liver function, inflammatory infiltration, and microinfarctions). Notably, all of these benefits were achieved with a relatively modest 1.25-fold increase in prothrombin times, and in the absence of hemorrhagic complications. Taken together, these data establish that prothrombin is a powerful modifier of SCD-induced end-organ damage, and present a novel therapeutic target to ameliorate SCD pathologies., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.