Network


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

Hotspot


Dive into the research topics where Mark A. Matulenko is active.

Publication


Featured researches published by Mark A. Matulenko.


Proceedings of the National Academy of Sciences of the United States of America | 2007

A-803467, a potent and selective Nav1.8 sodium channel blocker, attenuates neuropathic and inflammatory pain in the rat.

Michael F. Jarvis; Prisca Honore; Char Chang Shieh; Mark L. Chapman; Shailen K. Joshi; Xu Feng Zhang; Michael E. Kort; William L. Carroll; Brian Edward Marron; Robert Nelson Atkinson; James P. Thomas; Dong Liu; Michael J. Krambis; Yi Liu; Steve McGaraughty; Katharine L. Chu; Rosemarie Roeloffs; Chengmin Zhong; Joseph P. Mikusa; Gricelda Hernandez; Donna M. Gauvin; Carrie L. Wade; Chang Zhu; Madhavi Pai; Marc Scanio; Lei Shi; Irene Drizin; Robert J. Gregg; Mark A. Matulenko; Ahmed A. Hakeem

Activation of tetrodotoxin-resistant sodium channels contributes to action potential electrogenesis in neurons. Antisense oligonucleotide studies directed against Nav1.8 have shown that this channel contributes to experimental inflammatory and neuropathic pain. We report here the discovery of A-803467, a sodium channel blocker that potently blocks tetrodotoxin-resistant currents (IC50 = 140 nM) and the generation of spontaneous and electrically evoked action potentials in vitro in rat dorsal root ganglion neurons. In recombinant cell lines, A-803467 potently blocked human Nav1.8 (IC50 = 8 nM) and was >100-fold selective vs. human Nav1.2, Nav1.3, Nav1.5, and Nav1.7 (IC50 values ≥1 μM). A-803467 (20 mg/kg, i.v.) blocked mechanically evoked firing of wide dynamic range neurons in the rat spinal dorsal horn. A-803467 also dose-dependently reduced mechanical allodynia in a variety of rat pain models including: spinal nerve ligation (ED50 = 47 mg/kg, i.p.), sciatic nerve injury (ED50 = 85 mg/kg, i.p.), capsaicin-induced secondary mechanical allodynia (ED50 ≈ 100 mg/kg, i.p.), and thermal hyperalgesia after intraplantar complete Freunds adjuvant injection (ED50 = 41 mg/kg, i.p.). A-803467 was inactive against formalin-induced nociception and acute thermal and postoperative pain. These data demonstrate that acute and selective pharmacological blockade of Nav1.8 sodium channels in vivo produces significant antinociception in animal models of neuropathic and inflammatory pain.


Pain | 2005

Electrophysiological and in vivo characterization of A-317567, a novel blocker of acid sensing ion channels.

G.R. Dubé; Sonya G. Lehto; Nicole M. Breese; Scott J. Baker; Xueqing Wang; Mark A. Matulenko; Prisca Honore; Andrew O. Stewart; Robert B. Moreland; Jorge D. Brioni

&NA; Acid Sensing Ion Channels (ASICs) are a group of sodium‐selective ion channels that are activated by low extracellular pH. The role of ASIC in disease states remains unclear partly due to the lack of selective pharmacological agents. In this report, we describe the effects of A‐317567, a novel non‐amiloride blocker, on three distinct types of native ASIC currents evoked in acutely dissociated adult rat dorsal root ganglion (DRG) neurons. A‐317567 produced concentration‐dependent inhibition of all pH 4.5‐evoked ASIC currents with an IC50 ranging between 2 and 30 μM, depending upon the type of ASIC current activated. Unlike amiloride, A‐317567 equipotently blocked the sustained phase of ASIC3‐like current, a biphasic current akin to cloned ASIC3, which is predominant in DRG. When evaluated in the rat Complete Freuds Adjuvant (CFA)‐induced inflammatory thermal hyperalgesia model, A‐317567 was fully efficacious at a dose 10‐fold lower than amiloride. A‐317567 was also potent and fully efficacious when tested in the skin incision model of post‐operative pain. A‐317567 was entirely devoid of any diuresis or natriuresis activity and showed minimal brain penetration. In summary, A‐317567 is the first reported small molecule non‐amiloride blocker of ASIC that is peripherally active and is more potent than amiloride in vitro and in vivo pain models. The discovery of A‐317567 will greatly help to enhance our understanding of the physiological and pathophysiological role of ASICs.


Journal of Medicinal Chemistry | 2014

Discovery of ABT-267, a pan-genotypic inhibitor of HCV NS5A.

David A. Degoey; John T. Randolph; Dachun Liu; John K. Pratt; Charles W. Hutchins; Pamela Donner; A. Chris Krueger; Mark A. Matulenko; Sachin V. Patel; Christopher E. Motter; Lissa T. Nelson; Ryan G. Keddy; Michael D. Tufano; Daniel D. Caspi; Preethi Krishnan; Neeta Mistry; Gennadiy Koev; Thomas Reisch; Rubina Mondal; Tami Pilot-Matias; Yi Gao; David W. A. Beno; Clarence J. Maring; Akhter Molla; Emily O. Dumas; Andrew Campbell; Laura Williams; Christine Collins; Rolf Wagner; Warren M. Kati

We describe here N-phenylpyrrolidine-based inhibitors of HCV NS5A with excellent potency, metabolic stability, and pharmacokinetics. Compounds with 2S,5S stereochemistry at the pyrrolidine ring provided improved genotype 1 (GT1) potency compared to the 2R,5R analogues. Furthermore, the attachment of substituents at the 4-position of the central N-phenyl group resulted in compounds with improved potency. Substitution with tert-butyl, as in compound 38 (ABT-267), provided compounds with low-picomolar EC50 values and superior pharmacokinetics. It was discovered that compound 38 was a pan-genotypic HCV inhibitor, with an EC50 range of 1.7-19.3 pM against GT1a, -1b, -2a, -2b, -3a, -4a, and -5a and 366 pM against GT6a. Compound 38 decreased HCV RNA up to 3.10 log10 IU/mL during 3-day monotherapy in treatment-naive HCV GT1-infected subjects and is currently in phase 3 clinical trials in combination with an NS3 protease inhibitor with ritonavir (r) (ABT-450/r) and an NS5B non-nucleoside polymerase inhibitor (ABT-333), with and without ribavirin.


Neuropharmacology | 2010

A-887826 is a structurally novel, potent and voltage-dependent Nav1.8 sodium channel blocker that attenuates neuropathic tactile allodynia in rats

Xu-Feng Zhang; Char-Chang Shieh; Mark L. Chapman; Mark A. Matulenko; Ahmed H. Hakeem; Robert N. Atkinson; Michael E. Kort; Brian Edward Marron; Shailen K. Joshi; Prisca Honore; Connie R. Faltynek; Douglas S. Krafte; Michael F. Jarvis

Activation of sodium channels is essential to action potential generation and propagation. Recent genetic and pharmacological evidence indicates that activation of Na(v)1.8 channels contributes to chronic pain. Herein, we describe the identification of a novel series of structurally related pyridine derivatives as potent Na(v)1.8 channel blockers. A-887826 exemplifies this series and potently (IC(50)=11nM) blocked recombinant human Na(v)1.8 channels. A-887826 was approximately 3 fold less potent to block Na(v)1.2, approximately 10 fold less potent to block tetrodotoxin-sensitive sodium (TTX-S Na(+)) currents and was >30 fold less potent to block Na(V)1.5 channels. A-887826 potently blocked tetrodotoxin-resistant sodium (TTX-R Na(+)) currents (IC(50)=8nM) from small diameter rat dorsal root ganglion (DRG) neurons in a voltage-dependent fashion. A-887826 effectively suppressed evoked action potential firing when DRG neurons were held at depolarized potentials and reversibly suppressed spontaneous firing in small diameter DRG neurons from complete Freunds adjuvant inflamed rats. Following oral administration, A-887826 significantly attenuated tactile allodynia in a rat neuropathic pain model. Further characterization of TTX-R current block in rat DRG neurons demonstrated that A-887826 (100nM) shifted the mid-point of voltage-dependent inactivation of TTX-R currents by approximately 4mV without affecting voltage-dependent activation and did not exhibit frequency-dependent inhibition. The present data demonstrate that A-887826 is a structurally novel and potent Na(v)1.8 blocker that inhibits rat DRG TTX-R currents in a voltage-, but not frequency-dependent fashion. The ability of this structurally novel Na(v)1.8 blocker to effectively reduce tactile allodynia in neuropathic rats further supports the role of Na(v)1.8 sodium channels in pathological pain states.


Current Topics in Medicinal Chemistry | 2009

Voltage-gated sodium channel blockers for the treatment of chronic pain.

Mark A. Matulenko; Marc Scanio; Michael E. Kort

The voltage-gated sodium channels are a family of proteins that control the flow of sodium ions across cell membranes. Considerable data support the hypothesis that hyperexcitability and spontaneous action potential firing in peripheral sensory neurons mediated by voltage-gated sodium channels contribute to the pathophysiology of chronic pain. Sodium channel blockers are, therefore, appealing entities for therapeutic intervention in painful human neuropathies. This review will focus on the latest advances in the development of small molecule sodium channel blockers and their application to the treatment of chronic pain.


Bioorganic & Medicinal Chemistry Letters | 2010

Subtype-selective Nav1.8 sodium channel blockers: Identification of potent, orally active nicotinamide derivatives

Michael E. Kort; Robert N. Atkinson; James B. Thomas; Irene Drizin; Matthew Johnson; Matthew A. Secrest; Robert J. Gregg; Marc Scanio; Lei Shi; Ahmed H. Hakeem; Mark A. Matulenko; Mark L. Chapman; Michael J. Krambis; Dong Liu; Char-Chang Shieh; Xu-Feng Zhang; Gricelda Simler; Joseph P. Mikusa; Chengmin Zhong; Shailen K. Joshi; Prisca Honore; Rosemarie Roeloffs; Stephen Werness; Brett Antonio; Kennan C. Marsh; Connie R. Faltynek; Douglas S. Krafte; Michael F. Jarvis; Brian Edward Marron

A series of aryl-substituted nicotinamide derivatives with selective inhibitory activity against the Na(v)1.8 sodium channel is reported. Replacement of the furan nucleus and homologation of the anilide linker in subtype-selective blocker A-803467 (1) provided potent, selective derivatives with improved aqueous solubility and oral bioavailability. Representative compounds from this series displayed efficacy in rat models of inflammatory and neuropathic pain.


Tetrahedron Letters | 1991

Diastereoselectivity enhancement in vinylcuprate addition to β-alkoxyaldehydes via a vinylsilane.

Steven D. Burke; Anthony D. Piscopio; Brian E. Marron; Mark A. Matulenko; Gonghua Pan

Abstract The presence of a removable 1-(trimethylsilyl) residue greatly increases the chelation-controlled diastereoselectivity in alkenylcuprate additions to chiral β-alkoxyaldehydes. Several methods for protiodesilylation of the resultant allylic alcohols [5 » 2b (R=Me3SiCH2CH2OCH2-, PhCH2OCH2-, and PhCH2-)] are compared.


Journal of Medicinal Chemistry | 2016

Substituted Indazoles as Nav1.7 Blockers for the Treatment of Pain.

Jennifer M. Frost; David A. Degoey; Lei Shi; Rebecca J. Gum; Meagan M. Fricano; Greta L. Lundgaard; Odile F. El-Kouhen; Gin C. Hsieh; Torben R. Neelands; Mark A. Matulenko; Jerome F. Daanen; Madhavi Pai; Nayereh S. Ghoreishi-Haack; Cenchen Zhan; Xu-Feng Zhang; Michael E. Kort

The genetic validation for the role of the Nav1.7 voltage-gated ion channel in pain signaling pathways makes it an appealing target for the potential development of new pain drugs. The utility of nonselective Nav blockers is often limited due to adverse cardiovascular and CNS side effects. We sought more selective Nav1.7 blockers with oral activity, improved selectivity, and good druglike properties. The work described herein focused on a series of 3- and 4-substituted indazoles. SAR studies of 3-substituted indazoles yielded analog 7 which demonstrated good in vitro and in vivo activity but poor rat pharmacokinetics. Optimization of 4-substituted indazoles yielded two compounds, 27 and 48, that exhibited good in vitro and in vivo activity with improved rat pharmacokinetic profiles. Both 27 and 48 demonstrated robust activity in the acute rat monoiodoacetate-induced osteoarthritis model of pain, and subchronic dosing of 48 showed a shift to a lower EC50 over 7 days.


Tetrahedron Letters | 1999

Total synthesis of (+)-breynolide

Steven D. Burke; Jeffery J. Letourneau; Mark A. Matulenko

Abstract The total synthesis of (+)-breynolide ( 2 ) is described. The primary focus is the synthesis of the advanced aldehyde intermediate 12 , which comprises the cis -fused perhydrobenzothiophene ring system. Three stereoselective cyclohexene epoxidation/epoxide opening sequences and a glycolate ester Claisen rearrangement, by which all of the necessary oxygen functionality in 12 is introduced before installation of the thioether, are employed.


Journal of Medicinal Chemistry | 2018

Highlights of the Structure–Activity Relationships of Benzimidazole Linked Pyrrolidines Leading to the Discovery of the Hepatitis C Virus NS5A Inhibitor Pibrentasvir (ABT-530)

Rolf Wagner; John T. Randolph; Sachin V. Patel; Lissa T. Nelson; Mark A. Matulenko; Ryan G. Keddy; John K. Pratt; Dachun Liu; A. Chris Krueger; Pamela L. Donner; Douglas K. Hutchinson; Charles A. Flentge; David A. Betebenner; Todd W. Rockway; Clarence J. Maring; Teresa I. Ng; Preethi Krishnan; Tami Pilot-Matias; Christine A. Collins; Neeta S. Panchal; Thomas Reisch; Tatyana Dekhtyar; Rubina Mondal; DeAnne Stolarik; Yi Gao; Wenqing Gao; David W A Beno; Warren M. Kati

Curative interferon and ribavirin sparing treatments for hepatitis C virus (HCV)-infected patients require a combination of mechanistically orthogonal direct acting antivirals. A shared component of these treatments is usually an HCV NS5A inhibitor. First generation FDA approved treatments, including the component NS5A inhibitors, do not exhibit equivalent efficacy against HCV virus genotypes 1-6. In particular, these first generation NS5A inhibitors tend to select for viral drug resistance. Ombitasvir is a first generation HCV NS5A inhibitor included as a key component of Viekira Pak for the treatment of patients with HCV genotype 1 infection. Since the launch of next generation HCV treatments, functional cure for genotype 1-6 HCV infections has been achieved, as well as shortened treatment duration across a wider spectrum of genotypes. In this paper, we show how we have modified the anchor, linker, and end-cap architecture of our NS5A inhibitor design template to discover a next generation NS5A inhibitor pibrentasvir (ABT-530), which exhibits potent inhibition of the replication of wild-type genotype 1-6 HCV replicons, as well as improved activity against replicon variants demonstrating resistance against first generation NS5A inhibitors.

Collaboration


Dive into the Mark A. Matulenko's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Warren M. Kati

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

John T. Randolph

TAP Pharmaceutical Products

View shared research outputs
Top Co-Authors

Avatar

Michael E. Kort

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael D. Tufano

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge