Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Conrad E. Raab is active.

Publication


Featured researches published by Conrad E. Raab.


Journal of Controlled Release | 2014

Development of a liver-targeted siRNA delivery platform with a broad therapeutic window utilizing biodegradable polypeptide-based polymer conjugates.

Stephanie E. Barrett; Rob Burke; Marc T. Abrams; Carol Bason; Marina Busuek; Edward Carlini; Brian A. Carr; Louis S. Crocker; Haihong Fan; Robert M. Garbaccio; Erin N. Guidry; Jun H. Heo; Bonnie J. Howell; Eric Kemp; Robert A. Kowtoniuk; Andrew H. Latham; Anthony Leone; Michael Lyman; Rubina G. Parmar; Mihir Patel; Sergey Pechenov; Tao Pei; Nicole T. Pudvah; Conrad E. Raab; Sean Riley; Laura Sepp-Lorenzino; Sheri Smith; Eric Soli; Steven J. Staskiewicz; Melissa Stern

The greatest challenge standing in the way of effective in vivo siRNA delivery is creating a delivery vehicle that mediates a high degree of efficacy with a broad therapeutic window. Key structure-activity relationships of a poly(amide) polymer conjugate siRNA delivery platform were explored to discover the optimized polymer parameters that yield the highest activity of mRNA knockdown in the liver. At the same time, the poly(amide) backbone of the polymers allowed for the metabolism and clearance of the polymer from the body very quickly, which was established using radiolabeled polymers to demonstrate the time course of biodistribution and excretion from the body. The fast degradation and clearance of the polymers provided for very low toxicity at efficacious doses, and the therapeutic window of this poly(amide)-based siRNA delivery platform was shown to be much broader than a comparable polymer platform. The results of this work illustrate that the poly(amide) platform has a promising future in the development of a siRNA-based drug approved for human use.


Molecular Pharmaceutics | 2014

Quantitation of Physiological and Biochemical Barriers to siRNA Liver Delivery via Lipid Nanoparticle Platform

Yan Xu; Mei Ou; Ed Keough; Jeff Roberts; Ken Koeplinger; Mike Lyman; Scott E. Fauty; Ed Carlini; Melissa Stern; Rena Zhang; Suzie Yeh; Elizabeth Mahan; Yi Wang; Don Slaughter; Marian Gindy; Conrad E. Raab; Charles D. Thompson; Jerome H. Hochman

Effective delivery of small interfering RNA (siRNA) requires efficient cellular uptake and release into cytosol where it forms an active complex with RNAi induced silencing complex (RISC). Despite rapid developments in RNAi therapeutics, improvements in delivery efficiency of siRNA are needed to realize the full potential of this modality in broad therapeutic applications. We evaluated potential physiological and biochemical barrier(s) to the effective liver delivery of siRNA formulated in lipid nanoparticle (LNP) delivery vehicles. The comparative siRNA delivery performance of three LNPs was investigated in rats. They were assembled with either C14- or C18-anchored PEG-lipid(s), cationic lipid(s), and various helper lipid(s) and contained the same siRNA duplex. These LNPs demonstrated differentiated potency with ED50s ranging from 0.02 to 0.25 mg/kg. The two C14-PEG-LNPs had comparable siRNA exposure in plasma and liver, while the C18-PEG-LNP demonstrated a higher plasma siRNA exposure and a slower but sustained liver uptake. RISC bound siRNA within the liver, a more proximal measure of the pharmacologically active siRNA species, displayed loading kinetics that paralleled the target mRNA knockdown profile, with greater RISC loading associated with more potent LNPs. Liver perfusion and hepatocyte isolation experiments were performed following treatment of rats with LNPs containing VivoTag-fluorescently labeled siRNA. One hour after dosing a majority of the siRNA within the liver was associated with hepatocytes and was internalized (within small subcellular vesicles) with no significant cell surface association, indicating good liver tissue penetration, hepatocellular distribution, and internalization. Comparison of siRNA amounts in hepatocytes and subcellular fractions of the three LNPs suggests that endosomal escape is a significant barrier to siRNA delivery where cationic lipid seems to have a great impact. Quantitation of Ago-2 associated siRNA revealed that after endosomal escape further loss of siRNA occurs prior to RISC loading. This quantitative assessment of LNP-mediated siRNA delivery has highlighted potential barriers with respect to endosomal escape and incomplete RISC loading for delivery optimization efforts.


Journal of Medicinal Chemistry | 2009

Discovery of N-{N-[(3-cyanophenyl)sulfonyl]-4(R)-cyclobutylamino-(L)-prolyl}-4-[(3',5'-dichloroisonicotinoyl) amino]-(L)-phenylalanine (MK-0668), an extremely potent and orally active antagonist of very late antigen-4.

Linus S. Lin; Thomas J. Lanza; James P. Jewell; Ping Liu; Carrie K. Jones; Gerard R. Kieczykowski; Kelly M. Treonze; Qian Si; Salony Manior; Gloria C. Koo; Xinchun Tong; Junying Wang; Anne Schuelke; James V. Pivnichny; Regina W. Wang; Conrad E. Raab; Stella H. Vincent; Philip Davies; Malcolm Maccoss; Richard A. Mumford; William K. Hagmann

Extremely potent very late antigen-4 (VLA-4) antagonists with picomolar, whole blood activity and slow dissociation rates were discovered by incorporating an amino substituent on the proline fragment of the initial lead structure. This level of potency against the unactivated form of VLA-4 was shown to be sufficient to overcome the poor pharmacokinetic profiles typical of this class of VLA-4 antagonists, and sustained activity as measured by receptor occupancy was achieved in preclinical species after oral dosing.


Journal of Organic Chemistry | 2009

Remote Electronic Control in the Regioselective Reduction of Succinimides: A Practical, Scalable Synthesis of EP4 Antagonist MF-310

Carmela Molinaro; Danny Gauvreau; Gregory Hughes; Stephen Lau; Sophie Lauzon; Remy Angelaud; Paul D. O’Shea; Jacob Janey; Michael Palucki; Scott R. Hoerrner; Conrad E. Raab; Rick R. Sidler; Michel Belley; Yongxin Han

A practical large-scale chromatography-free synthesis of EP4 antagonist MF-310, a potential new treatment for chronic inflammation, is presented. The synthetic route provided MF-310 as its sodium salt in 10 steps and 17% overall yield from commercially available pyridine dicarboxylate 7. The key features of this sequence include a unique regioselective reduction of succinimide 2 controlled by the electronic properties of a remote pyridine ring, preparation of cyclopropane carboxylic acid 3 via a Corey-Chaykovsky cyclopropanation, and a short synthesis of sulfonamide 5.


Journal of Pharmacology and Experimental Therapeutics | 2008

CYP2C75-Involved Autoinduction of Metabolism in Rhesus Monkeys of MK-0686, a Bradykinin B1 Receptor Antagonist

Cuyue Tang; Brian A. Carr; Frédéric Poignant; Bennett Ma; Stacey L. Polsky-Fisher; Yuhsin Kuo; Kristie Strong-Basalyga; Alisha Norcross; Karen Richards; Roy Eisenhandler; Edward Carlini; Christina Ng; Scott D. Kuduk; Nathan X. Yu; Conrad E. Raab; Thomas H. Rushmore; Clay B. Frederick; Mark G. Bock; Thomayant Prueksaritanont

After oral treatment (once daily) for 4 weeks with the potent bradykinin B1 receptor antagonist methyl 3-chloro-3′-fluoro-4′-{(1R)-1-[({1-[(trifluoroacetyl)amino]cyclopropyl}carbonyl)-amino]ethyl}-1,1′-biphenyl-2-carboxylate (MK-0686), rhesus monkeys (Macaca mulatta) exhibited significantly reduced systemic exposure of the compound in a dose-dependent manner, suggesting an occurrence of autoinduction of MK-0686 metabolism. This possibility is supported by two observations. 1) MK-0686 was primarily eliminated via biotransformation in rhesus monkeys, with oxidation on the chlorophenyl ring as one of the major metabolic pathways. This reaction led to appreciable formation of a dihydrodiol (M11) and a hydroxyl (M13) product in rhesus liver microsomes supplemented with NADPH. 2) The formation rate of these two metabolites determined in liver microsomes from MK-0686-treated groups was ≥2-fold greater than the value for a control group. Studies with recombinant rhesus P450s and monoclonal antibodies against human P450 enzymes suggested that CYP2C75 played an important role in the formation of M11 and M13. The induction of this enzyme by MK-0686 was further confirmed by a concentration-dependent increase of its mRNA in rhesus hepatocytes, and, more convincingly, the enhanced CYP2C proteins and catalytic activities toward CYP2C75 probe substrates in liver microsomes from MK-0686-treated animals. Furthermore, a good correlation was observed between the rates of M11 and M13 formation and hydroxylase activities toward probe substrates determined in a panel of liver microsomal preparations from control and MK-0686-treated animals. Therefore, MK-0686, both a substrate and inducer for CYP2C75, caused autoinduction of its own metabolism in rhesus monkeys by increasing the expression of this enzyme.


Journal of Pharmaceutical and Biomedical Analysis | 2002

LC/MS/MS plasma assay for the peptidomimetic VLA4 antagonist I and its major active metabolite II: for treatment of asthma by inhalation

Alison L Fisher; Elizabeth Depuy; Andrew Jayaraj; Conrad E. Raab; Matt Braun; Michel Ellis-Hutchings; Jin Zhang; J.D. Rogers; Donald G. Musson

In vitro and in animals, I is a potent and specific peptidomimetic for the potential treatment of airway inflammation in the pathogenesis of asthma. Preclinical studies indicated extensive conversion of I to an active metabolite II, and thus, a very sensitive assay for I and II was needed to support an inhalation ascending-dose study in man. The LC/MS/MS plasma/urine assay method (1.0 ml of sample) involves the following: liquid-liquid extraction of acidified plasma into pentane-ethyl acetate (90:10 v/v); evaporation of the organic extract, reconstitution into methanol; addition of water to the methanolic extract and freezing. After thawing, the extract is centrifuged and the clear supernatant injected for chromatography. Extract is chromatographed on a YMC ODS-AM column (50 x 2.0 mm). For detection, a Sciex 365 LC/MS/MS with an electrospray inlet and used in the positive ion, multiple reaction monitoring mode was used to monitor precursor-->fragment ions of m/z 709-->594 for I and m/z 513-->380 for II. The plasma assay was linear over the concentration range of 0.1-100 ng/ml in plasma for I and II. Accuracy and precision for I ranged from 97.9 to 102.1% of nominal with a 0.84-10.65% CV; similarly for II, 98.0-101.7% and 1.39-9.28% CV, respectively. Extraction recovery averaged 63.7% for I and 64.9% for II. This general assay methodology may be applied to assay small acidic peptides and peptidomimetics from biological fluids by LC/MS/MS.


Synthetic Communications | 1993

Enantioselective Approaches to Rhodium Catalysed Aldol-Type Reactions

Gregory Roos; Raymond J. Haines; Conrad E. Raab

Abstract Homochiral catalysts, [Rh (norbornadiene)-(homochiral diphosphine)]+ X−/H2, were tested to assess possible enantiocontrol in the aldol-type coupling of activated vinyl components with aldehydes. A related approach, via in situ rhodium silyl enolates, was also investigated.


Journal of Pharmacology and Experimental Therapeutics | 2008

CYP2C75-Involved Autoinduction of Metabolism in Rhesus Monkeys of Methyl 3-Chloro-3′-fluoro-4′-{(1R)-1-[({1-[(trifluoroacetyl)amino]cyclopropyl}carbonyl)amino]ethyl}-1,1′-biphenyl-2-carboxylate (MK-0686), a Bradykinin B1 Receptor Antagonist

Cuyue Tang; Brian A. Carr; Frédéric Poignant; Bennett Ma; Stacey L. Polsky-Fisher; Yuhsin Kuo; Kristie Strong-Basalyga; Alisha Norcross; Karen Richards; Roy Eisenhandler; Edward Carlini; Christina N. Di Marco; Scott D. Kuduk; Nathan X. Yu; Conrad E. Raab; Tom Rushmore; Clay B. Frederick; Mark G. Bock; Thomayant Prueksaritanont

After oral treatment (once daily) for 4 weeks with the potent bradykinin B1 receptor antagonist methyl 3-chloro-3′-fluoro-4′-{(1R)-1-[({1-[(trifluoroacetyl)amino]cyclopropyl}carbonyl)-amino]ethyl}-1,1′-biphenyl-2-carboxylate (MK-0686), rhesus monkeys (Macaca mulatta) exhibited significantly reduced systemic exposure of the compound in a dose-dependent manner, suggesting an occurrence of autoinduction of MK-0686 metabolism. This possibility is supported by two observations. 1) MK-0686 was primarily eliminated via biotransformation in rhesus monkeys, with oxidation on the chlorophenyl ring as one of the major metabolic pathways. This reaction led to appreciable formation of a dihydrodiol (M11) and a hydroxyl (M13) product in rhesus liver microsomes supplemented with NADPH. 2) The formation rate of these two metabolites determined in liver microsomes from MK-0686-treated groups was ≥2-fold greater than the value for a control group. Studies with recombinant rhesus P450s and monoclonal antibodies against human P450 enzymes suggested that CYP2C75 played an important role in the formation of M11 and M13. The induction of this enzyme by MK-0686 was further confirmed by a concentration-dependent increase of its mRNA in rhesus hepatocytes, and, more convincingly, the enhanced CYP2C proteins and catalytic activities toward CYP2C75 probe substrates in liver microsomes from MK-0686-treated animals. Furthermore, a good correlation was observed between the rates of M11 and M13 formation and hydroxylase activities toward probe substrates determined in a panel of liver microsomal preparations from control and MK-0686-treated animals. Therefore, MK-0686, both a substrate and inducer for CYP2C75, caused autoinduction of its own metabolism in rhesus monkeys by increasing the expression of this enzyme.


Xenobiotica | 2010

In vitro and in vivo metabolism of a novel cannabinoid-1 receptor inverse agonist, taranabant, in rats and monkeys

Vijay Bhasker G. Reddy; George A. Doss; Bindhu V. Karanam; Koppara Samuel; Thomas J. Lanza; Linus S. Lin; N.X. Yu; Andy Shiqiang Zhang; Conrad E. Raab; Ralph A. Stearns; Sanjeev Kumar

The metabolism and excretion of taranabant (MK-0364, N-[(1S,2S)-3-(4-chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl]-2-methyl-2{[5-(trifluoromethyl)pyridine-2-yl]oxy}propanamide), a potent cannabinoid-1 receptor inverse agonist, were evaluated in rats and rhesus monkeys. Following administration of [14C]taranabant, the majority of the radioactivity was excreted within 72 h. In both rats and rhesus monkeys, taranabant was eliminated primarily via oxidative metabolism, followed by excretion of metabolites into bile. Major pathways of metabolism that were common to rats and rhesus monkeys included hydroxylation at the benzylic carbon adjacent to the cyanophenyl ring to form a biologically active circulating metabolite M1, and oxidation of one of the two geminal methyl groups of taranabant or M1 to the corresponding diastereomeric carboxylic acids. Oxidation of the cyanophenyl ring, followed by conjugation with glutathione or glucuronic acid, was a major pathway of metabolism only in the rat and was not detected in the rhesus monkey. Metabolism profiles of taranabant in liver microsomes in vitro were qualitatively similar in rats, rhesus monkeys and humans and included formation of M1 and oxidation of taranabant or M1 to the corresponding carboxylic acids via oxidation of a geminal methyl group. In human liver microsomes, metabolism of taranabant was mediated primarily by CYP3A4.


Drug Metabolism and Disposition | 1999

Studies on Cytochrome P-450-Mediated Bioactivation of Diclofenac in Rats and in Human Hepatocytes: Identification of Glutathione Conjugated Metabolites

Wei Tang; Ralph A. Stearns; Stelvio M. Bandiera; Yong Zhang; Conrad E. Raab; Matthew P. Braun; Dennis C. Dean; Jianmei Pang; Kwan H. Leung; George A. Doss; John R. Strauss; Gloria Y. Kwei; Thomas H. Rushmore; Shuet Hing L Chiu; Thomas A. Baillie

Collaboration


Dive into the Conrad E. Raab's collaboration.

Researchain Logo
Decentralizing Knowledge