Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
J Cardiovasc Pharmacol ; 78(1): e5-e11, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34232223

ABSTRACT

ABSTRACT: Atherosclerotic cardiovascular disease (ASCVD) continues to be the leading cause of preventable death in the United States. Elevated low-density lipoprotein cholesterol (LDL-C) is well known to result in cardiovascular disease. Mainstay therapy for reducing LDL-C and ASCVD risk is statin therapy. Despite achieving desired LDL-C levels with lipid-lowering therapy, cardiovascular residual risk often persists. Elevated lipoprotein(a) [Lp(a)] levels have been highlighted as an inherent independent predictor of ASCVD, and decreasing Lp(a) levels may result in a significant reduction in the residual risk in high-risk patients. To date, there are no approved medications to lower Lp(a) levels. Nicotinic acid, proprotein convertase subtilisin/kexin 9 inhibitors, and antisense oligonucleotide have demonstrated modest to potent Lp(a) reduction. Spotlight has been placed on antisense oligonucleotides and their role in Lp(a) lowering. APO(a)LRx is in the frontline for selectively decreasing Lp(a) concentrations and ongoing research may prove that this medication may lower Lp(a)-mediated residual risk, translating into cardiovascular benefit.


Subject(s)
Atherosclerosis/drug therapy , Dyslipidemias/drug therapy , Hypolipidemic Agents/therapeutic use , Lipoprotein(a)/blood , Oligodeoxyribonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Atherosclerosis/blood , Atherosclerosis/genetics , Biomarkers/blood , Clinical Trials as Topic , Down-Regulation , Dyslipidemias/blood , Dyslipidemias/genetics , Evidence-Based Medicine , Humans , Hypolipidemic Agents/adverse effects , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligonucleotides/adverse effects , Treatment Outcome
2.
Clin Cancer Res ; 27(7): 1912-1922, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33500356

ABSTRACT

PURPOSE: Despite standard of care (SOC) established by Stupp, glioblastoma remains a uniformly poor prognosis. We evaluated IGV-001, which combines autologous glioblastoma tumor cells and an antisense oligonucleotide against IGF type 1 receptor (IMV-001), in newly diagnosed glioblastoma. PATIENTS AND METHODS: This open-label protocol was approved by the Institutional Review Board at Thomas Jefferson University. Tumor cells collected during resection were treated ex vivo with IMV-001, encapsulated in biodiffusion chambers with additional IMV-001, irradiated, then implanted in abdominal acceptor sites. Patients were randomized to four exposure levels, and SOC was initiated 4-6 weeks later. On the basis of clinical improvements, randomization was halted after patient 23, and subsequent patients received only the highest exposure. Safety and tumor progression were primary and secondary objectives, respectively. Time-to-event outcomes were compared with the SOC arms of published studies. RESULTS: Thirty-three patients were enrolled, and median follow-up was 3.1 years. Six patients had adverse events (grade ≤3) possibly related to IGV-001. Median progression-free survival (PFS) was 9.8 months in the intent-to-treat population (vs. SOC, 6.5 months; P = 0.0003). In IGV-001-treated patients who met Stupp-eligible criteria, PFS was 11.6 months overall (n = 22; P = 0.001) and 17.1 months at the highest exposure (n = 10; P = 0.0025). The greatest overall survival was observed in Stupp-eligible patients receiving the highest exposure (median, 38.2 months; P = 0.044). Stupp-eligible patients with methylated O6-methylguanine-DNA methyltransferase promoter (n = 10) demonstrated median PFS of 38.4 months (P = 0.0008). Evidence of immune activation was noted. CONCLUSIONS: IGV-001 was well tolerated, PFS compared favorably with SOC, and evidence suggested an immune-mediated mechanism (ClinicalTrials.gov: NCT02507583).


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Receptor, IGF Type 1/antagonists & inhibitors , Adult , Aged , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Female , Glioblastoma/immunology , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Male , Middle Aged , Oligodeoxyribonucleotides, Antisense/adverse effects , Receptor, IGF Type 1/genetics
3.
Expert Rev Clin Pharmacol ; 12(8): 701-711, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31268366

ABSTRACT

Introduction: Hereditary transthyretin-mediated amyloidosis (ATTRv; v for variant) is an underdiagnosed, progressive, and fatal multisystemic disease with a heterogenous clinical phenotype that is caused by TTR gene mutations that destabilize the TTR protein, resulting in its misfolding, aggregation, and deposition in tissues throughout the body. Areas covered: Inotersen, an antisense oligonucleotide inhibitor, was recently approved in the United States and Europe for the treatment of the polyneuropathy of ATTRv based on the positive results obtained in the pivotal phase 3 trial, NEURO-TTR. This review will discuss the mechanism of action of inotersen and its pharmacology, clinical efficacy, and safety and tolerability. A PubMed search using the terms 'inotersen,' 'AG10,' 'antisense oligonucleotide,' 'hereditary transthyretin amyloidosis,' 'familial amyloid polyneuropathy,' and 'familial amyloid cardiomyopathy' was performed, and the results were screened for the most relevant English language publications. The bibliographies of all retrieved articles were manually searched to identify additional studies of relevance. Expert opinion: Inotersen targets the disease-forming protein, TTR, and has been shown to improve quality of life and neuropathy progression in patients with stage 1 or 2 ATTRv with polyneuropathy. Inotersen is well tolerated, with a manageable safety profile through regular monitoring for the development of glomerulonephritis or thrombocytopenia.


Subject(s)
Amyloid Neuropathies, Familial/drug therapy , Oligonucleotides/therapeutic use , Polyneuropathies/drug therapy , Adult , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/physiopathology , Animals , Humans , Mutation , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligodeoxyribonucleotides, Antisense/pharmacology , Oligodeoxyribonucleotides, Antisense/therapeutic use , Oligonucleotides/adverse effects , Oligonucleotides/pharmacology , Polyneuropathies/etiology , Prealbumin/genetics , Quality of Life
4.
J Cardiovasc Pharmacol ; 64(2): 164-71, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24691275

ABSTRACT

Mipomersen is a second-generation antisense oligonucleotide indicated as an adjunct therapy for homozygous familial hypercholesterolemia (HoFH). Warfarin is commonly prescribed for a variety of cardiac disorders in homozygous familial hypercholesterolemia population, and concurrent use of warfarin and mipomersen is likely. This open-label, single-sequence 2-period phase 1 study in healthy subjects evaluated the potential drug-drug interactions between mipomersen and warfarin. The subjects received a single oral 25 mg dose of warfarin alone on day 1, and after a 7-day washout period, received 200 mg mipomersen alone subcutaneously every other day on days 8-12, and received both concurrently on day 14. Coadministration of mipomersen did not change the pharmacodynamics (international normalized ratio, prothrombin time, and activated partial thromboplastin time) and pharmacokinetics (PK) of warfarin. There were no clinically significant changes in the PK of mipomersen with concurrent administration of warfarin. There were no events indicative of an increase in bleeding tendency when warfarin was coadministered with mipomersen, and the adverse event profile of mipomersen did not appear to be altered in combination with warfarin, as compared with that of the respective reference treatment. The combination of these 2 medications appeared to be safe and well tolerated. These results suggest that the dosage adjustment of warfarin or mipomersen is not expected to be necessary with coadministration.


Subject(s)
Oligodeoxyribonucleotides, Antisense/pharmacology , Oligodeoxyribonucleotides, Antisense/pharmacokinetics , Oligonucleotides/pharmacology , Oligonucleotides/pharmacokinetics , Warfarin/pharmacology , Warfarin/pharmacokinetics , Adolescent , Adult , Drug Interactions , Female , Healthy Volunteers , Humans , Hyperlipoproteinemia Type II/drug therapy , International Normalized Ratio , Male , Middle Aged , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligonucleotides/administration & dosage , Oligonucleotides/adverse effects , Partial Thromboplastin Time , Prothrombin Time , Warfarin/administration & dosage , Warfarin/adverse effects , Young Adult
5.
J Labelled Comp Radiopharm ; 57(2): 97-101, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24307558

ABSTRACT

Targeting and visualization of human telomerase reverse transcriptase (hTERT) represents a promising approach for providing diagnostic value. The uptake kinetics and imaging results of (99m) Tc-hTERT antisense oligonucleotides (ASON) in hTERT-expressing cells were examined in vitro and in vivo. The pharmacokinetics and acute toxicity studies of (99m) Tc-hTERT ASON were also performed. The labeling efficiencies of radiolabeled oligonucleotide reached 76 ± 5%, the specific activity was up to 1850 kBq/µg, and the radiochemical purity was above 96%. Radioactivity accumulated to a higher concentration in hTERT-expressing cells with antisense probe than with sense control (p < 0.05). Lipid carrier incorporation significantly increased the transmembrane delivery of radiolabeled probes (p < 0.05). hTERT-expressing xenografts in nude mice were clearly visualized at 6 h postinjection of the antisense probe but not the sense control probe. However, liposome did not increase the radioactivity accumulation of probes in tumors for either antisense or sense probe (p > 0.05). Radioactivity counts per minute versus time profiles for (99m) Tc-hTERT ASON were biphasic, indicative of a three-compartment model. The pharmacokinetics parameters of half-life of distribution (T1/2α ), half-life of elimination (T1/2ß ), total apparent volume of distribution (Vd), and total rate of clearance were 2.04 ± 0.48 min, 24 ± 4.8 min, 109.83 ± 17.20 mL, and 3.19 ± 0.17 mL/min, respectively. The acute toxicity study results showed the safe application of (99m) Tc-hTERT ASON in vivo. This study provides further evidences that (99m) Tc-hTERT ASON should be developed as a safe, potential molecular image-guided diagnostic agent.


Subject(s)
Oligodeoxyribonucleotides, Antisense/pharmacokinetics , Radiopharmaceuticals/pharmacokinetics , Technetium/pharmacokinetics , Telomerase/genetics , Animals , Hep G2 Cells , Humans , Mice , Mice, Nude , Neoplasms, Experimental/diagnostic imaging , Oligodeoxyribonucleotides, Antisense/adverse effects , Radiopharmaceuticals/adverse effects , Technetium/adverse effects , Tomography, Emission-Computed, Single-Photon , Xenograft Model Antitumor Assays
7.
Vasc Health Risk Manag ; 8: 651-9, 2012.
Article in English | MEDLINE | ID: mdl-23226021

ABSTRACT

Familial hypercholesterolemia (FH) is an autosomal dominant condition with a population prevalence of one in 300-500 (heterozygous) that is characterized by high levels of low-density lipoprotein (LDL) cholesterol, tendon xanthomata, and premature atherosclerosis and coronary heart disease (CHD). FH is caused mainly by mutations in the LDLR gene. However, mutations in other genes including APOB and PCSK9, can give rise to a similar phenotype. Homozygous FH with an estimated prevalence of one in a million is associated with severe hypercholesterolemia with accelerated atherosclerotic CHD in childhood and without treatment, death usually occurs before the age of 30 years. Current approaches for the treatment of homozygous FH include statin-based lipid-lowering therapies and LDL apheresis. Mipomersen is a second-generation antisense oligonucleotide (ASO) targeted to human apolipoprotein B (apoB)-100. This review provides an overview of the pathophysiology and current treatment options for familial hypercholesterolemia and describes novel therapeutic strategies focusing on mipomersen, an antisense apoB synthesis inhibitor. Mipomersen is distributed mainly to the liver where it silences apoB mRNA, thereby reducing hepatic apoB-100 and giving rise to reductions in plasma total cholesterol, LDL-cholesterol, and apoB concentrations in a dose-and time-dependent manner. Mipomersen has been shown to decrease apoB, LDL-cholesterol and lipoprotein(a) in patients with heterozygous and homozygous FH on maximally tolerated lipid-lowering therapy. The short-term efficacy and safety of mipomersen has been established, however, injection site reactions are common and concern exists regarding the long-term potential for hepatic steatosis with this ASO. In summary, mipomersen given alone or in combination with standard lipid-lowering medications shows promise as an adjunct therapy in patients with homozygous or refractory heterozygous FH at high risk of atherosclerotic CHD, who are not at target or are intolerant of statins.


Subject(s)
Anticholesteremic Agents/therapeutic use , Cholesterol/blood , Hyperlipoproteinemia Type II/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Anticholesteremic Agents/adverse effects , Apolipoproteins B/biosynthesis , Apolipoproteins B/genetics , Biomarkers/blood , Cholesterol, LDL/blood , Dose-Response Relationship, Drug , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/diagnosis , Hyperlipoproteinemia Type II/epidemiology , Hyperlipoproteinemia Type II/genetics , Liver/drug effects , Liver/metabolism , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligonucleotides/adverse effects , Prevalence , Time Factors , Treatment Outcome
8.
PLoS One ; 7(11): e49006, 2012.
Article in English | MEDLINE | ID: mdl-23152839

ABSTRACT

OBJECTIVES: Mipomersen, an antisense oligonucleotide targeting apolipoprotein B synthesis, significantly reduces LDL-C and other atherogenic lipoproteins in familial hypercholesterolemia when added to ongoing maximally tolerated lipid-lowering therapy. Safety and efficacy of mipomersen in patients with severe hypercholesterolemia was evaluated. METHODS AND RESULTS: Randomized, double-blind, placebo-controlled, multicenter trial. Patients (n  = 58) were ≥18 years with LDL-C ≥7.8 mmol/L or LDL-C ≥5.1 mmol/L plus CHD disease, on maximally tolerated lipid-lowering therapy that excluded apheresis. Weekly subcutaneous injections of mipomersen 200 mg (n  = 39) or placebo (n  = 19) were added to lipid-lowering therapy for 26 weeks. MAIN OUTCOME: percent reduction in LDL-C from baseline to 2 weeks after the last dose of treatment. Mipomersen (n = 27) reduced LDL-C by 36%, from a baseline of 7.2 mmol/L, for a mean absolute reduction of 2.6 mmol/L. Conversely, mean LDL-C increased 13% in placebo (n = 18) from a baseline of 6.5 mmol/L (mipomersen vs placebo p<0.001). Mipomersen produced statistically significant (p<0.001) reductions in apolipoprotein B and lipoprotein(a), with no change in high-density lipoprotein cholesterol. Mild-to-moderate injection site reactions were the most frequently reported adverse events with mipomersen. Mild-to-moderate flu-like symptoms were reported more often with mipomersen. Alanine transaminase increase, aspartate transaminase increase, and hepatic steatosis occurred in 21%, 13% and 13% of mipomersen treated patients, respectively. Adverse events by category for the placebo and mipomersen groups respectively were: total adverse events, 16(84.2%), 39(100%); serious adverse events, 0(0%), 6(15.4%); discontinuations due to adverse events, 1(5.3%), 8(20.5%) and cardiac adverse events, 1(5.3%), 5(12.8%). CONCLUSION: Mipomersen significantly reduced LDL-C, apolipoprotein B, total cholesterol and non-HDL-cholesterol, and lipoprotein(a). Mounting evidence suggests it may be a potential pharmacologic option for lowering LDL-C in patients with severe hypercholesterolemia not adequately controlled using existing therapies. Future studies will explore alternative dosing schedules aimed at minimizing side effects. TRIAL REGISTRATION: ClinicalTrials.gov NCT00794664.


Subject(s)
Anticholesteremic Agents/therapeutic use , Hypercholesterolemia/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/adverse effects , Cholesterol, LDL/blood , Female , Humans , Male , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligonucleotides/administration & dosage , Oligonucleotides/adverse effects , Treatment Outcome
10.
Immunol Invest ; 41(2): 171-82, 2012.
Article in English | MEDLINE | ID: mdl-21864114

ABSTRACT

Susceptible-strain animals immunized with P2 peptide could generate the disease of experimental autoimmune neuritis (EAN) with inflammation and demyelination of peripheral nerve. A myriad of transcription factors and inflammatory cytokines have been found to participate in this process; however, the roles of toll-like receptors (TLRs) are poorly understood in EAN. The aim of this study is to explore the role of TLR9 in the pathogenesis of EAN. The EAN was induced in Lewis rat by immunization with P2(53-78) and complete Freund's adjuvant. CpG oligodeoxynucleotides (ODN) (cODN), a suppressive ODN (sODN) and a control non-specific ODN (nODN) were respectively administered to explore the role of TLR9 in EAN both in vivo and vitro. Following immunization up to the peak phase of EAN, EAN rats inoculated with sODN had remarkably better clinical score of EAN and expressed a significantly inhibited TLR9 signaling pathway. Our study suggests that TLR9 may be involved in the pathogenesis of EAN.


Subject(s)
Guillain-Barre Syndrome/immunology , Neuritis, Autoimmune, Experimental/immunology , Oligodeoxyribonucleotides, Antisense/administration & dosage , Toll-Like Receptor 9/metabolism , Animals , Disease Models, Animal , Disease Progression , Disease Susceptibility , Freund's Adjuvant/administration & dosage , Guillain-Barre Syndrome/therapy , Humans , Immunization , Male , Myelin P2 Protein/administration & dosage , Neuritis, Autoimmune, Experimental/chemically induced , Neuritis, Autoimmune, Experimental/genetics , Neuritis, Autoimmune, Experimental/physiopathology , Neuritis, Autoimmune, Experimental/therapy , Oligodeoxyribonucleotides, Antisense/adverse effects , Peptide Fragments/administration & dosage , Rats , Rats, Inbred Lew , Toll-Like Receptor 9/genetics
11.
Mol Pharm ; 8(5): 1662-8, 2011 Oct 03.
Article in English | MEDLINE | ID: mdl-21740056

ABSTRACT

Lipoplexes and polyplexes represent the two major nanocarrier systems for nucleic acid delivery. Previous studies examining their uptake and intracellular unpacking rely on organic fluorophores fraught with low signal intensity and photobleaching. In this work quantum dot mediated Förster resonance energy transfer (QD-FRET) was first used to study and compare the cellular uptake and the intracellular fate of oligodeoxynucelotide (ODN)-based lipoplexes and polyplexes. QD605-amine and Cy5-labeled ODN (Cy5-GTI2040) were chosen as the FRET pair. By adjusting the lipid/ODN ratio of lipoplexes and the nitrogen/phosphate (N/P) ratio of polyplexes, lipoplexes and polyplexes with comparable physical properties were produced. The biological activities of dual-labeled lipoplexes and polyplexes remained unaltered compared to their unlabeled counterparts as evidenced by their comparable antisense activities against protein R2 in KB cells. Flow cytometry and confocal microscopy revealed similar pattern of uptake for these two types of nanoparticles, although polyplexes had a higher dissociation rate than lipoplexes in KB cells. We demonstrate that QD-FRET is a sensitive tool to study the uptake and intracellular unpacking of lipoplexes and polyplexes, which may help optimize their formulations for various theranostics applications.


Subject(s)
Antineoplastic Agents/metabolism , Drug Carriers/metabolism , Lipids/chemistry , Nanoparticles/chemistry , Oligodeoxyribonucleotides, Antisense/metabolism , Polyethyleneimine/chemistry , Quantum Dots , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/chemistry , Biological Transport , Cell Survival/drug effects , Chemical Phenomena , Drug Carriers/adverse effects , Drug Carriers/chemistry , Fluorescence Resonance Energy Transfer , Fluorescent Dyes/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Humans , KB Cells , Microscopy, Confocal , Nanoparticles/adverse effects , Nanoparticles/ultrastructure , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/ultrastructure , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligodeoxyribonucleotides, Antisense/chemistry , Particle Size , RNA, Messenger/metabolism , Ribonucleotide Reductases/antagonists & inhibitors , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/metabolism , Surface Properties
12.
Schizophr Res ; 130(1-3): 250-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21689907

ABSTRACT

Synapsin II is a synaptic vesicle-associated phosphoprotein that has been implicated in the pathophysiology of schizophrenia. Studies have demonstrated reductions in synapsin II mRNA and protein in medial prefrontal cortical post-mortem samples from patients with schizophrenia, genetic associations between synapsin II and schizophrenia, and synapsin II protein regulation by dopamine receptor activation. Collectively, this research indicates a relationship between synapsin II dysregulation and schizophrenia; however, it remains unknown whether perturbations in synapsin II play a role in the pathophysiology of this disease. The aim of this project was to evaluate animals with selective knock-down of synapsin II in the medial prefrontal cortex. After continuous infusion of synapsin II antisense sequences, animals were examined for the presence of schizophrenic-like behavioral phenotypes and assessed on the response to clinically relevant antipsychotic drugs. Our results indicate that rats with selective reductions in medial prefrontal cortical synapsin II demonstrate deficits in sensorimotor gating (prepulse inhibition), reduced social behavior, and hyperlocomotion, which are corrected by the atypical antipsychotic drug olanzapine. Additionally, synapsin II knock-down disrupts serial search efficiency. These behavioral changes are accompanied by reductions in vesicular neurotransmitter transporter protein concentrations for glutamate (VGLUT1 and VGLUT2) and GABA (VGAT), without affecting dopamine (VMAT2). These results implicate a causal role for decreased synapsin II in the medial prefrontal cortex in the pathophysiology of schizophrenia and the mechanisms of aberrant prefrontal cortical circuitry, and suggest that synapsin II may potentially serve as a novel therapeutic target for this disorder.


Subject(s)
Behavioral Symptoms/etiology , Prefrontal Cortex/metabolism , Schizophrenia/complications , Schizophrenia/pathology , Synapsins/metabolism , Animals , Antipsychotic Agents/therapeutic use , Disease Models, Animal , Drug Interactions , Inhibition, Psychological , Locomotion/drug effects , Locomotion/physiology , Male , Maze Learning/drug effects , Oligodeoxyribonucleotides, Antisense/adverse effects , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Reflex, Startle/physiology , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Sensory Gating/drug effects , Social Behavior , Synapsins/chemistry , Vesicular Neurotransmitter Transport Proteins/metabolism
13.
Neuromuscul Disord ; 21(1): 58-67, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20951042

ABSTRACT

The development of new animal models is a crucial step in determining the pathological mechanism underlying neurodegenerative diseases and is essential for the development of effective therapies. We have investigated the zebrafish (Danio rerio) as a new model to study CMT2A, a peripheral neuropathy characterized by the selective loss of motor neurons, caused by mutations of mitofusin 2 gene. Using a knock-down approach, we provide evidence that during embryonic development, mitofusin 2 loss of function is responsible of several morphological defects and motility impairment. Immunohistochemical investigations, revealing the presence of severe alterations in both motor neurons and muscles fibres, indicated the central role played by MFN2 in axonal and neuromuscular development. Finally, we demonstrated the ability of human MFN2 to balance the downregulation of endogenous mfn2 in zebrafish, further supporting the conserved function of the MFN2 gene. These results highlight the essential role of mitofusin 2 in the motor axon development and demonstrate the potential of zebrafish as a suitable and complementary platform for dissecting pathogenetic mechanisms of MFN2 mutations in vivo.


Subject(s)
Charcot-Marie-Tooth Disease , Developmental Disabilities/etiology , Down-Regulation/drug effects , Neuromuscular Diseases/etiology , Oligodeoxyribonucleotides, Antisense/adverse effects , Tolloid-Like Metalloproteinases/genetics , Zebrafish Proteins/genetics , Animals , Animals, Genetically Modified , Body Patterning/drug effects , Body Patterning/genetics , Charcot-Marie-Tooth Disease/chemically induced , Charcot-Marie-Tooth Disease/complications , Charcot-Marie-Tooth Disease/genetics , Computational Biology , Developmental Disabilities/genetics , Disease Models, Animal , Down-Regulation/genetics , Embryo, Nonmammalian , GTP Phosphohydrolases , Humans , Membrane Proteins/genetics , Mitochondrial Proteins/genetics , Motor Activity/drug effects , Motor Activity/genetics , Neuromuscular Diseases/genetics , RNA, Messenger/metabolism , Receptors, Cholinergic/metabolism , Tolloid-Like Metalloproteinases/metabolism , Tubulin/metabolism , Zebrafish , Zebrafish Proteins/metabolism
14.
Ann N Y Acad Sci ; 1132: 283-90, 2008.
Article in English | MEDLINE | ID: mdl-18567879

ABSTRACT

Acetylcholinesterase pre-mRNA is susceptible to alternative splicing. Myasthenia gravis has been shown to be associated with the expression of the readthrough transcript (AChE-R), which, unlike the normal "synaptic" transcript (AChE-S) is not tethered to the post-synaptic membrane, but is a soluble monomer in the synaptic cleft. In rats with experimental autoimmune myasthenia gravis (EAMG), inhibition of production of AChE-R using antisense is associated with a significant reduction in synaptic expression of AChE-R mRNA and protein, with improved muscle strength and stamina and increased survival. Synaptic AChE does not appear to be significantly affected by the induction of EAMG or treatment with antisense to AChE-R. Monarsen (EN101) is a synthetic 20-base antisense oligodeoxynucleotide directed against the human AChE gene. It is modified to achieve stability for oral administration. Sixteen patients with seropositive myasthenia gravis who were responsive to pyridostigmine were withdrawn from it and treated with Monarsen. Fourteen patients experienced a clinically significant response. In some, the improvement was dramatic. Although the dose of pyridostigmine was not optimized before the study, the majority of responders achieved better Quantitative Myasthenia Gravis scores than on pyridostigmine. The response of an individual muscle group to Monarsen was related to the degree of deterioration following the withdrawal of pyridostigmine. Cholinergic side effects were conspicuous by their absence. Monarsen is now being investigated in a phase II study.


Subject(s)
Myasthenia Gravis/genetics , Myasthenia Gravis/therapy , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/therapeutic use , Acetylcholinesterase/genetics , Acetylcholinesterase/metabolism , Adult , Aged , Animals , Combined Modality Therapy , Female , Genetic Therapy/adverse effects , Humans , Male , Middle Aged , Myasthenia Gravis/enzymology , Myasthenia Gravis/immunology , Oligodeoxyribonucleotides, Antisense/adverse effects , Transcription, Genetic/genetics , Treatment Outcome
15.
Bull Soc Pathol Exot ; 101(2): 90-7, 2008 Apr.
Article in French | MEDLINE | ID: mdl-18543699

ABSTRACT

The experimentation on human beings of one or several therapeutic molecules discovered in laboratory is necessary and important because it helps to find new treatments or new diagnostic methods. But, it presents serious ethical problems. In this article we are analysing the example of the HIV infection. We are succinctly describing the research methods in laboratory for therapeutic molecules, first the experimentation on animals and then on human being in clinical trials. We will then try to show, with several examples, how during these last 25 years of HIV infection, the research of new molecules has not always respected the ethical rules set out in Helsinki declaration, "Code de la santé publique" or "Guide de bonnes pratiques cliniques-ICH" etc. We are discussing here the way to avoid these irregularities.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Anti-HIV Agents/therapeutic use , Clinical Trials as Topic/ethics , Human Experimentation/ethics , Adenine/adverse effects , Adenine/analogs & derivatives , Adenine/therapeutic use , Animals , Anti-HIV Agents/adverse effects , Azathioprine/administration & dosage , Azathioprine/adverse effects , Azathioprine/therapeutic use , Clinical Trials as Topic/methods , Clinical Trials as Topic/standards , Congresses as Topic , Cyclosporine/adverse effects , Cyclosporine/therapeutic use , Developing Countries , Ditiocarb/adverse effects , Ditiocarb/therapeutic use , Double-Blind Method , Drug Evaluation, Preclinical , Drug Therapy, Combination , Ethics Committees, Research , Human Experimentation/standards , Humans , Informed Consent/ethics , Informed Consent/standards , Mass Media , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligodeoxyribonucleotides, Antisense/therapeutic use , Organophosphonates/adverse effects , Organophosphonates/therapeutic use , Practice Guidelines as Topic , Stavudine/adverse effects , Stavudine/therapeutic use , Tenofovir , Thionucleotides/adverse effects , Thionucleotides/therapeutic use
16.
Oligonucleotides ; 17(2): 201-12, 2007.
Article in English | MEDLINE | ID: mdl-17638524

ABSTRACT

Transforming growth factor-beta2 (TGF-beta2) is known to suppress the immune response to cancer cells and plays a pivotal role in tumor progression by regulating key mechanisms including proliferation, metastasis, and angiogenesis. For targeted protein suppression the TGF-beta2-specific antisense oligodeoxynucleotide AP 12009 was developed. In vitro experiments have been performed to prove specificity and efficacy of the TGF-beta2 inhibitor AP 12009 employing patient-derived malignant glioma cells as well as peripheral blood mononuclear cells (PBMCs) from patients. Clinically, the antisense compound AP 12009 was assessed in three Phase I/II-studies for the treatment of patients with recurrent or refractory malignant (high-grade) glioma WHO grade III or IV. Although the study was not primarily designed as an efficacy evaluation, prolonged survival compared to literature data and response data were observed, which are very rarely seen in this tumor indication. Two patients experienced long-lasting complete tumor remissions. These results implicate targeted TGF-beta2-suppression using AP 12009 as a promising novel approach for malignant gliomas and other highly aggressive, TGF-beta-2-overexpressing tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism , Adult , Antineoplastic Agents/adverse effects , Antineoplastic Agents/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioma/genetics , Glioma/metabolism , Humans , Male , Middle Aged , Oligodeoxyribonucleotides, Antisense/adverse effects , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/metabolism , Recurrence
17.
J Drug Target ; 14(10): 687-93, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17162738

ABSTRACT

OBJECTIVE: To compare transfection efficiency and safety for antisense oligodeoxynucleotides (AS-ODNs) between two type of phospholipids-based vectors. METHODS: An AS-ODNs sequence HA824 combined with luciferase reporter plasmid was used. Under low intensity ultrasound (US), a breast cancer cell line SK-BR-3 was exposed to different concentration of microbubbles and liposomes. Transfection efficiency was detected by fluorescence microscopy. Cell viability was verified by propidium iodide assay. Reverse transcription-polymerase chain reaction (RT-PCR) was used to detect the inhibitory effect of HA824 on HER-2 expression at mRNA level. Atomic force microscopy (AFM) scanning techniques was employed to observe the change of membrane pore size. RESULTS: AS-ODNs transfection efficiency showed an increasing tend with microbubble concentration, but not with liposome concentration. Maximum transfection efficiency with minimum cell viability was achieved under 2% microbubble concentration. Too strong sonoporation activity would enlarge membrane pores significantly and cause low cell viability. CONCLUSION: US-mediated AS-ODNs transfection enhanced by phospholipids-based microbubbles represents an effective and safe avenue.


Subject(s)
Oligodeoxyribonucleotides, Antisense/adverse effects , Oligodeoxyribonucleotides, Antisense/pharmacology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cell Survival/drug effects , Drug Carriers , Excipients , Female , Gene Transfer Techniques , Genes, Reporter/genetics , Genes, erbB-2/genetics , Humans , Liposomes , Luciferases/genetics , Microspheres , Oligodeoxyribonucleotides, Antisense/administration & dosage , Phospholipids , Porosity , Propidium , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spectrophotometry, Atomic , Transfection
18.
Neurosci Lett ; 410(1): 15-9, 2006 Dec 13.
Article in English | MEDLINE | ID: mdl-17055644

ABSTRACT

Selective and brain penetrating pharmacological antagonists for use in clarifying a role of alpha7 nicotinic acetylcholine receptors (nAChR) in behavioral paradigms are presently unavailable. Studies in alpha7 knock-out mice (KO) have not revealed convincing changes in behavioral phenotype, in particular measures of cognition that include contextual fear conditioning and spatial memory, which may be due to compensatory developmental changes. Therefore, an antisense oligonucleotide (aON) targeted toward the 3'- and 5'-UTR coding regions of the rat alpha7 nicotinic acetylcholine receptor was used. Following central injection of aON into the lateral ventricle of Long Evans rats for 6 days, treated rats exhibited a significant 42% and 25% decrease in alpha7 nAChR densities in hippocampus and cortex, respectively, as measured by [(3)H]-methyllycaconitine (MLA) binding. There was no change in alpha4beta2 densities measured by [(3)H]-cytisine binding. Acquisition of Morris Water Maze (MWM) performance, a measure of spatial memory, was impaired in aON-treated rats. In addition, a reduction in target platform crossings during a subsequent probe-trial was observed. These data demonstrate the ability of this aON to reduce hippocampal and cortical alpha7 nicotinic receptor densities associated with impaired MWM performance and support the specific involvement of the alpha7 nAChR in spatial learning and memory, a phenotype not affected in alpha7 KO mice.


Subject(s)
Memory Disorders/chemically induced , Oligodeoxyribonucleotides, Antisense/adverse effects , Receptors, Nicotinic/physiology , Space Perception/drug effects , Aconitine/analogs & derivatives , Aconitine/pharmacokinetics , Alkaloids/pharmacology , Animals , Azocines/pharmacology , Behavior, Animal/drug effects , Cerebral Cortex/drug effects , Dose-Response Relationship, Drug , Drug Interactions , Hippocampus/drug effects , Male , Maze Learning/drug effects , Memory Disorders/physiopathology , Nicotinic Antagonists/pharmacokinetics , Protein Binding/drug effects , Quinolizines/pharmacology , Rats , Rats, Long-Evans , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/genetics , Time Factors , Tritium/pharmacokinetics , alpha7 Nicotinic Acetylcholine Receptor
19.
Aliment Pharmacol Ther ; 23(10): 1403-13, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16669955

ABSTRACT

BACKGROUND: Alicaforsen is an antisense oligonucleotide inhibitor of intercellular adhesion molecule 1 protein expression with activity in subjects with ulcerative colitis and pouchitis. AIM: To compare the effects of alicaforsen enema to standard of care mesalazine (mesalamine) enema in subjects with mild to moderate active left-sided ulcerative colitis. METHOD: A randomized, double-blind, active-controlled multicentre clinical trial. Subjects received a nightly enema of 120 mg alicaforsen (n=55), 240 mg alicaforsen (n=50), or 4 g mesalazine (n=54) for 6 weeks, followed by a 24-week monitoring period. The primary end point was Disease Activity Index at week 6. Clinical improvement, remission and relapse were secondary end points. RESULTS: No significant difference was observed between treatment arms in the primary end point. However, the median duration of response to alicaforsen enema treatment was two- to threefold longer (128 and 146 days) in comparison with mesalazine (54 days). Complete mucosal healing occurred in 24% of the 240 mg alicaforsen group, when compared with 17% in the mesalazine. CONCLUSIONS: Alicaforsen enema demonstrated an acute response and safety profile similar to mesalazine enema, but was differentiated by a more durable response. The extended length of remission suggests that alicaforsen enema treatment may have a disease modifying effect.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Colitis, Ulcerative/drug therapy , Gastrointestinal Agents/administration & dosage , Mesalamine/administration & dosage , Oligodeoxyribonucleotides, Antisense/administration & dosage , Thionucleotides/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Enema , Female , Gastrointestinal Agents/adverse effects , Gastrointestinal Diseases/chemically induced , Humans , Male , Mesalamine/adverse effects , Middle Aged , Oligodeoxyribonucleotides, Antisense/adverse effects , Phosphorothioate Oligonucleotides , Thionucleotides/adverse effects , Treatment Outcome
20.
Aliment Pharmacol Ther ; 23(10): 1415-25, 2006 May 15.
Article in English | MEDLINE | ID: mdl-16669956

ABSTRACT

BACKGROUND: Alicaforsen is an antisense oligonucleotide designed to inhibit expression of human intercellular adhesion molecule 1. Previous clinical studies have demonstrated activity of alicaforsen enema in ulcerative colitis and pouchitis. AIM: To determine the minimally effective dosing regimen of alicaforsen enema in subjects with mild to moderate left-sided ulcerative colitis. METHODS: Randomized, placebo-controlled, double-blind, two-dose ranging multicentre study. One hundred and twelve subjects were equally randomized to receive one of four alicaforsen enema regimens or placebo daily for 6 weeks. Primary end point was Disease Activity Index at week 6. Secondary end points included evaluation of clinical improvement, relapse rates and durability of response. Analysis of data were performed on the intent-to-treat population. RESULTS: No significant difference was observed between treatment arms and placebo in the primary end point. A prolonged reduction in mean% Disease Activity Index relative to baseline was observed in the daily 240 mg alicaforsen enema treatment arm in comparison with placebo from week 18 (51% vs. 18%, P=0.04) to week 30 (50% vs. 11%, P=0.03). CONCLUSIONS: Alicaforsen enema was safe and well tolerated at all doses studied. The durability of the response to alicaforsen enema treatment may suggests a disease-modifying effect.


Subject(s)
Colitis, Ulcerative/drug therapy , Gastrointestinal Agents/administration & dosage , Oligodeoxyribonucleotides, Antisense/administration & dosage , Thionucleotides/administration & dosage , Adult , Aged , Double-Blind Method , Drug Administration Schedule , Enema , Female , Gastrointestinal Agents/adverse effects , Gastrointestinal Hemorrhage/etiology , Humans , Male , Middle Aged , Oligodeoxyribonucleotides, Antisense/adverse effects , Phosphorothioate Oligonucleotides , Rectum , Recurrence , Thionucleotides/adverse effects , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...