1
|
Mokrov GV. Multitargeting in cardioprotection: An example of biaromatic compounds. Arch Pharm (Weinheim) 2023; 356:e2300196. [PMID: 37345968 DOI: 10.1002/ardp.202300196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023]
Abstract
A multitarget drug design approach is actively developing in modern medicinal chemistry and pharmacology, especially with regard to multifactorial diseases such as cardiovascular diseases, cancer, and neurodegenerative diseases. A detailed study of many well-known drugs developed within the single-target approach also often reveals additional mechanisms of their real pharmacological action. One of the multitarget drug design approaches can be the identification of the basic pharmacophore models corresponding to a wide range of the required target ligands. Among such models in the group of cardioprotectors is the linked biaromatic system. This review develops the concept of a "basic pharmacophore" using the biaromatic pharmacophore of cardioprotectors as an example. It presents an analysis of possible biological targets for compounds corresponding to the biaromatic pharmacophore and an analysis of the spectrum of biological targets for the five most known and most studied cardioprotective drugs corresponding to this model, and their involvement in the biological effects of these drugs.
Collapse
|
2
|
Alam KA, Svalastoga P, Martinez A, Glennon JC, Haavik J. Potassium channels in behavioral brain disorders. Molecular mechanisms and therapeutic potential: A narrative review. Neurosci Biobehav Rev 2023; 152:105301. [PMID: 37414376 DOI: 10.1016/j.neubiorev.2023.105301] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
Potassium channels (K+-channels) selectively control the passive flow of potassium ions across biological membranes and thereby also regulate membrane excitability. Genetic variants affecting many of the human K+-channels are well known causes of Mendelian disorders within cardiology, neurology, and endocrinology. K+-channels are also primary targets of many natural toxins from poisonous organisms and drugs used within cardiology and metabolism. As genetic tools are improving and larger clinical samples are being investigated, the spectrum of clinical phenotypes implicated in K+-channels dysfunction is rapidly expanding, notably within immunology, neurosciences, and metabolism. K+-channels that previously were considered to be expressed in only a few organs and to have discrete physiological functions, have recently been found in multiple tissues and with new, unexpected functions. The pleiotropic functions and patterns of expression of K+-channels may provide additional therapeutic opportunities, along with new emerging challenges from off-target effects. Here we review the functions and therapeutic potential of K+-channels, with an emphasis on the nervous system, roles in neuropsychiatric disorders and their involvement in other organ systems and diseases.
Collapse
Affiliation(s)
| | - Pernille Svalastoga
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway; Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | | | - Jeffrey Colm Glennon
- Conway Institute for Biomolecular and Biomedical Research, School of Medicine, University College Dublin, Dublin, Ireland.
| | - Jan Haavik
- Department of Biomedicine, University of Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Norway.
| |
Collapse
|
3
|
Hong E, Gu SM, Kim JM, Yoon KS, Lee JM, Kim YH, Suh SK, Lee D, Eom H, Yun J, Cha HJ. The designer benzodiazepine, flubromazepam, induces reward-enhancing and cardiotoxic effects in rodents. Toxicol Res (Camb) 2022; 11:644-653. [DOI: 10.1093/toxres/tfac039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/10/2022] [Accepted: 06/10/2022] [Indexed: 11/12/2022] Open
Abstract
Abstract
The use of many benzodiazepines is controlled worldwide due to their high likelihood of abuse and potential adverse effects. Flubromazepam—a designer benzodiazepine—is a long-acting gamma-aminobutyric acid subtype A receptor agonist. There is currently a lack of scientific evidence regarding the potential for flubromazepam dependence or other adverse effects. This study aimed to evaluate the dependence potential, and cardiotoxicity via confirmation of the QT and RR intervals which are the factors on the electrical properties of the heart of flubromazepam in rodents. Using a conditioned place preference test, we discovered that mice treated intraperitoneally with flubromazepam (0.1 mg/kg) exhibited a significant preference for the flubromazepam-paired compartment, suggesting a potential for flubromazepam dependence. In addition, we observed several cardiotoxic effects of flubromazepam; 100-μM flubromazepam reduced cell viability, increased RR intervals but not QT intervals in the electrocardiography measurements, and considerably inhibited potassium channels in a human ether-à-go-go-related gene assay. Collectively, these findings suggest that flubromazepam may have adverse effects on psychological and cardiovascular health, laying the foundation for further efforts to list flubromazepam as a controlled substance at both national and international levels.
Collapse
Affiliation(s)
- Eunchong Hong
- College of Pharmacy , Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Sun Mi Gu
- College of Pharmacy , Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Jin Mook Kim
- Pharmacological Research Division , National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Kyung Sik Yoon
- Pharmacological Research Division , National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Jin-Moo Lee
- Pharmacological Research Division , National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Young-Hoon Kim
- Pharmacological Research Division , National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Soo Kyung Suh
- Pharmacological Research Division , National Institute of Food and Drug Safety Evaluation (NIFDS), Ministry of Food and Drug Safety (MFDS), 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| | - Dohyun Lee
- Laboratory Animal Center , Osong Medical Innovation Foundation, 123 Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Heejong Eom
- Laboratory Animal Center , Osong Medical Innovation Foundation, 123 Osongsaengmyeong-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Jaesuk Yun
- College of Pharmacy , Chungbuk National University, 194-31 Osongsaengmyeong 1-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28160, Republic of Korea
| | - Hye Jin Cha
- Deputy Director General for Narcotics Safety Planning , Pharmaceutical Safety Bureau, Ministry of Food and Drug Safety (MFDS), 187 Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si, Chungcheongbuk-do 28159, Republic of Korea
| |
Collapse
|
4
|
Zeng H, Wang J, Clouse H, Lagrutta A, Sannajust F. Human-induced pluripotent stem cell-derived cardiomyocytes have limited I Ks for repolarization reserve as revealed by specific KCNQ1/KCNE1 blocker. JRSM Cardiovasc Dis 2019; 8:2048004019854919. [PMID: 31217965 PMCID: PMC6558757 DOI: 10.1177/2048004019854919] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/06/2019] [Accepted: 05/11/2019] [Indexed: 01/18/2023] Open
Abstract
Objective We investigated if there is IKs, and if there is repolarization
reserve by IKs in human-induced pluripotent stem cell-derived
cardiomyocytes (hiPSC-CMs). Design We used a specific KCNQ1/KCNE1 channel blocker, L-000768673, with an
IC50 of 9 nM, and four hERG-specific blockers, astemizole,
cisapride, dofetilide, and E-4031 to investigate the issue. Results L-000768673 concentration-dependently prolonged feature point duration
(FPD)―a surrogate signal of action potential duration―from 1 to 30 nM
without pacing or paced at 1.2 Hz, resulting from IKs blockade in
hiPSC-CMs. At higher concentrations, the effect of L-000768673 on
IKs was mitigated by its effect on ICa-L,
resulting in shortened FPD, reduced impedance amplitude, and increased
beating rate at 1 µM and above, recapitulating the self-limiting properties
of L-000768673 on action potentials. All four hERG-specific blockers
prolonged FPD as expected. Co-application of L-000768673 at sub-threshold
(0.1 and 0.3 nM) and threshold (1 nM) concentrations failed to
synergistically enhance the effects of hERG blockers on FPD prolongation,
rather it showed additive effects, inconsistent with the repolarization
reserve role of IKs in mature human myocytes that enhanced
IKr response, implying a difference between hiPSC-CMs used in
this study and mature human cardiomyocytes. Conclusion There was IKs current in hiPSC-CMs, and blockade of IKs
current caused prolongation of action potential of hiPSC-CMs. However, we
could not demonstrate any synergistic effects on action potential duration
prolongation of hiPSC-CMs by blocking hERG current and IKs
current simultaneously, implying little or no repolarization reserve by
IKs current in hiPSC-CMs used in this study.
Collapse
Affiliation(s)
- Haoyu Zeng
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | - Jixin Wang
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | - Holly Clouse
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | - Armando Lagrutta
- Merck & Co., Inc., Safety and Exploratory Pharmacology, West Point, PA, USA
| | | |
Collapse
|
5
|
Fanoe S, Kristensen D, Fink-Jensen A, Jensen HK, Toft E, Nielsen J, Videbech P, Pehrson S, Bundgaard H. Risk of arrhythmia induced by psychotropic medications: a proposal for clinical management. Eur Heart J 2014; 35:1306-15. [DOI: 10.1093/eurheartj/ehu100] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
6
|
Li GR, Dong MQ. Pharmacology of Cardiac Potassium Channels. CARDIOVASCULAR PHARMACOLOGY - HEART AND CIRCULATION 2010; 59:93-134. [DOI: 10.1016/s1054-3589(10)59004-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
7
|
Blockade of the IKs potassium channel: An overlooked cardiovascular liability in drug safety screening? J Pharmacol Toxicol Methods 2009; 60:1-10. [DOI: 10.1016/j.vascn.2009.04.197] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 04/25/2009] [Indexed: 12/25/2022]
|
8
|
Regan CP, Stump GL, Wallace AA, Anderson KD, McIntyre CJ, Liverton NJ, Lynch JJ. In Vivo Cardiac Electrophysiologic and Antiarrhythmic Effects of an Isoquinoline IKur Blocker, ISQ-1, in Rat, Dog, and Nonhuman Primate. J Cardiovasc Pharmacol 2007; 49:236-45. [PMID: 17438409 DOI: 10.1097/fjc.0b013e3180325b2a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The cardiac electrophysiologic effects of ISQ-1, an isoquinolinone I(Kur) blocker, were characterized in vivo. In rat, ISQ-1 elicited maximal 33% to 36% increases in atrial and ventricular refractoriness at a plasma concentration of 11.5 microM. In African green monkey, ISQ-1 increased atrial refractory period (maximal 17% at plasma concentration up to 20 microM) with no effect on ventricular refractory period or ECG QTc. Likewise in dog, ISQ-1 increased atrial refractory period (maximal 16% at plasma concentration up to 2 microM) with no effect on ventricular refractory period or QTc. In contrast, studies with ibutilide in nonhuman primate and dog demonstrated concomitant increases in atrial and ventricular refractoriness and QTc. Additionally, in a dog model of atrial flutter, ISQ-1 terminated ongoing flutter at doses (2.5 +/- 0.5 mg/kg IV) that selectively prolonged atrial refractoriness (13% increase), whereas flutter termination with ibutilide occurred at doses that increased both atrial and ventricular refractoriness as well as QTc. Of note, the cardiac electrophysiologic profiles displayed by ISQ-1 in these species were similar to those reported previously by our lab with a structurally distinct I(Kur) blocker. Taken together, these results further support the inhibition of I(Kur) as an approach to terminate atrial arrhythmia.
Collapse
Affiliation(s)
- Christopher P Regan
- Department of Stroke and Neurodegeneration, Merck Research Laboratories, West Point, Pennsylvania 19486, USA
| | | | | | | | | | | | | |
Collapse
|
9
|
Trotter BW, Nanda KK, Kett NR, Regan CP, Lynch JJ, Stump GL, Kiss L, Wang J, Spencer RH, Kane SA, White RB, Zhang R, Anderson KD, Liverton NJ, McIntyre CJ, Beshore DC, Hartman GD, Dinsmore CJ. Design and synthesis of novel isoquinoline-3-nitriles as orally bioavailable Kv1.5 antagonists for the treatment of atrial fibrillation. J Med Chem 2007; 49:6954-7. [PMID: 17125248 DOI: 10.1021/jm060927v] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Novel 3-cyanoisoquinoline Kv1.5 antagonists have been prepared and evaluated in in vitro and in vivo assays for inhibition of the Kv1.5 potassium channel and its associated cardiac potassium current, IKur. Structural modifications of isoquinolinone lead 1 afforded compounds with excellent potency, selectivity, and oral bioavailability.
Collapse
Affiliation(s)
- B Wesley Trotter
- Department of Medicinal Chemistry, Merck Research Laboratories, WP14-2, P.O. Box 4, Sumneytown Pike, West Point, Pennsylvania 19486, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Nanda KK, Nolt MB, Cato MJ, Kane SA, Kiss L, Spencer RH, Wang J, Lynch JJ, Regan CP, Stump GL, Li B, White R, Yeh S, Bogusky MJ, Bilodeau MT, Dinsmore CJ, Lindsley CW, Hartman GD, Wolkenberg SE, Trotter BW. Potent antagonists of the Kv1.5 potassium channel: Synthesis and evaluation of analogous N,N-diisopropyl-2-(pyridine-3-yl)acetamides. Bioorg Med Chem Lett 2006; 16:5897-901. [PMID: 16949818 DOI: 10.1016/j.bmcl.2006.08.054] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Revised: 08/08/2006] [Accepted: 08/10/2006] [Indexed: 11/29/2022]
Abstract
This letter describes the discovery of a novel series of potent Kv1.5 ion channel antagonists based on a diisopropyl amide scaffold. Structure-activity relationships of functionalized analogs are discussed. Key compound 1-(3-(diisopropylcarbamoyl)-2-phenyl-3-(pyridin-3-yl)propyl)-3-(2-fluorobenzyl)urea (10) exhibits significant atrial-selective effects in an in vivo model.
Collapse
Affiliation(s)
- Kausik K Nanda
- Department of Medicinal Chemistry, Merck Research Laboratories, Merck and Co., Inc., PO Box 4, West Point, PA 19486, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Brochu RM, Dick IE, Tarpley JW, McGowan E, Gunner D, Herrington J, Shao PP, Ok D, Li C, Parsons WH, Stump GL, Regan CP, Lynch JJ, Lyons KA, McManus OB, Clark S, Ali Z, Kaczorowski GJ, Martin WJ, Priest BT. Block of peripheral nerve sodium channels selectively inhibits features of neuropathic pain in rats. Mol Pharmacol 2006; 69:823-32. [PMID: 16301337 DOI: 10.1124/mol.105.018127] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Several sodium channel blockers are used clinically to treat neuropathic pain. However, many patients fail to achieve adequate pain relief from these highly brain-penetrant drugs because of dose-limiting central nervous system side effects. Here, we describe the functional properties of trans-N-{[2'-(aminosulfonyl)biphenyl-4-yl]methyl}-N-methyl-N'-[4-(trifluoromethoxy)benzyl]cyclopentane-1,2-dicarboxamide (CDA54), a peripherally acting sodium channel blocker. In whole-cell electrophysiological assays, CDA54 blocked the inactivated states of hNa(V)1.7 and hNa(V)1.8, two channels of the peripheral nervous system implicated in nociceptive transmission, with affinities of 0.25 and 0.18 microM, respectively. CDA54 displayed similar affinities for the tetrodotoxin-resistant Na+ current in small-diameter mouse dorsal root ganglion neurons. Peripheral nerve injury causes spontaneous electrical activity in normally silent sensory neurons. CDA54 inhibited these injury-induced spontaneous action potentials at concentrations 10-fold lower than those required to block normal A- and C-fiber conduction. Consistent with the selective inhibition of injury-induced firing, CDA54 (10 mg/kg p.o.) significantly reduced behavioral signs of neuropathic pain in two nerve injury models, whereas the same dose of CDA54 did not affect acute nociception or motor coordination. In anesthetized dogs, CDA54, at plasma concentrations of 6.7 microM, had no effect on cardiac electrophysiological parameters including conduction. Thus, the peripheral nerve sodium channel blocker CDA54 selectively inhibits sensory nerve signaling associated with neuropathic pain.
Collapse
Affiliation(s)
- Richard M Brochu
- Department of Ion Channels, Merck Research Laboratories, Rahway, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Du LP, Li MY, Tsai KC, You QD, Xia L. Characterization of binding site of closed-state KCNQ1 potassium channel by homology modeling, molecular docking, and pharmacophore identification. Biochem Biophys Res Commun 2005; 332:677-87. [PMID: 15904893 DOI: 10.1016/j.bbrc.2005.04.165] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2005] [Accepted: 04/29/2005] [Indexed: 10/25/2022]
Abstract
This investigation was performed to assess the importance of interaction in the binding of blockers to KCNQ1 potassium using molecular modeling. This work could be considered made up by three main steps: (1) the construction of closed-state structure of KCNQ1 through homology modeling; (2) the automated docking of three blockers: IKS-142, L-735821, and BMS-IKS, using DOCK program; (3) the generation and validation of pharmacophore for KCNQ1 ligands using Catalyst/HypoGen. The obtained results highlight the hydrophobic or aromatic residues involved in S6 transmembrane domain and the base of the pore helix of KCNQ1, confirming the mutagenesis data and pharmacophore model, and giving new suggestions for the rational design of novel KCNQ1 ligands.
Collapse
Affiliation(s)
- Lü-Pei Du
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, China
| | | | | | | | | |
Collapse
|
13
|
Lu HR, Vlaminckx E, Van De Water A, Gallacher DJ. Both β-adrenergic receptor stimulation and cardiac tissue type have important roles in elucidating the functional effects of IKs channel blockers in vitro. J Pharmacol Toxicol Methods 2005; 51:81-90. [PMID: 15767201 DOI: 10.1016/j.vascn.2004.10.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2004] [Accepted: 10/14/2004] [Indexed: 11/21/2022]
Abstract
INTRODUCTION Conflicting results associated with the use of I(Ks) blockers on the action potential duration (APD) have raised a question as to whether the variable results arise from the use of different cardiac tissues, beta-adrenergic stimulation, or by the "selectivity" of the chosen I(Ks) blockers. METHODS We used the highly selective I(Ks) blocker (-)-[3R, 4S] chromanol 293B [(-) chromanol] to mimic drug-induced long QT1 in isolated rabbit Purkinje fibers, papillary muscles, and ventricular trabeculae using the conventional microelectrode technique. RESULTS I(Ks) block with (-) chromanol at 1 x 10(-5) M did not significantly change the APD at different stimulation rates in all three cardiac tissues. Isoproterenol (Iso:1 x 10(-7) M) shortened APD(90), and (-) chromanol (1 x 10(-5) M) largely prevented this shortening in isolated papillary muscles at 1 Hz [-3% with Iso combined (-) chromanol group versus -16% with iso group; p<0.05] and also at 2 Hz (+7% versus -25% with Iso group; p<0.05), but did not significantly prevent this shortening in isolated Purkinje fibers. In isolated trabeculae, (-) chromanol combined with Iso significantly prolonged the APD(90) by 15% at 1 Hz (versus -10% with Iso group; p<0.05) and by 5% at 2 Hz (versus -11% with Iso group; p<0.05). DISCUSSION Our study shows that only during beta-adrenoceptor stimulation, pharmacological inhibition of the I(Ks) current plays an important role in the APD recorded from isolated ventricular trabeculae and papillary muscles, but not from Purkinje fibers. These results indicate that the APD prolonging effects of I(Ks)channel blockers during beta-adrenergic receptor stimulation can only be detected from isolated rabbit papillary muscles and ventricular trabeculae, but not Purkinje fibers.
Collapse
Affiliation(s)
- H R Lu
- Center of Excellence for Cardiovascular Safety Research, Johnson and Johnson Pharmaceutical Research and Development, A Division of Janssen Pharmaceutical N.V. B-2340, Beerse, Belgium
| | | | | | | |
Collapse
|
14
|
Ahmad S, Doweyko L, Ashfaq A, Ferrara FN, Bisaha SN, Schmidt JB, DiMarco J, Conder ML, Jenkins-West T, Normandin DE, Russell AD, Smith MA, Levesque PC, Lodge NJ, Lloyd J, Stein PD, Atwal KS. Tetrahydronaphthalene-derived amino alcohols and amino ketones as potent and selective inhibitors of the delayed rectifier potassium current IKs. Bioorg Med Chem Lett 2004; 14:99-102. [PMID: 14684307 DOI: 10.1016/j.bmcl.2003.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Class III anti-arrhythmic drugs (e.g., dofetilide) prolong cardiac action potential duration (APD) by blocking the fast component of the delayed rectifier potassium current (I(Kr)). The block of I(Kr) can result in life threatening ventricular arrhythmias (i.e., torsades de pointes). Unlike I(Kr), the role of the slow component of the delayed rectifier potassium current (I(Ks)) becomes significant only at faster heart rate. Therefore selective blockers of I(Ks) could prolong APD with a reduced propensity to cause pro-arrhythmic side effects. This report describes structure-activity relationships (SARs) of a series of I(Ks) inhibitors derived from 6-alkoxytetralones with good in vitro activity (IC(50) > or =30 nM) and up to 40-fold I(Ks)/I(Kr) selectivity.
Collapse
Affiliation(s)
- Saleem Ahmad
- Bristol-Myers Squibb Pharmaceutical Research Institute, PO Box 4000, Princeton, NJ 08543, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|