Network


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

Hotspot


Dive into the research topics where Elfatih Elzein is active.

Publication


Featured researches published by Elfatih Elzein.


Expert Opinion on Investigational Drugs | 2008

A1 adenosine receptor agonists and their potential therapeutic applications

Elfatih Elzein; Jeff Zablocki

Background: The challenges in developing any A1 adenosine receptor (A1-AdoR) agonist involve having the desired effect on target tissue while avoiding side effects due to activation of A1-AdoR on other tissues. A1-AdoR de-sensitization leading to tachyphylaxis is also another challenge. Objectives: The major goal of this review is twofold: to highlight the structure affinity relationships (SAR) of A1-AdoR agonists, starting with initial lead compounds that were the genesis for second-generation compounds with high selectivity, affinity, and partial agonism; and to give an overview of the A1-AdoR agonists under development for various indications. Results: Intense efforts by many pharmaceutical companies and academicians in the A1-AdoR agonist field have led to the discovery of clinical candidates for the following conditions: atrial arrhythmias – Tecadenoson, Selodenoson and PJ-875; type 2 diabetes (T2D) and insulin-sensitizing agents – GR79236, ARA, and CVT-3619; pain management – SDZ WAG 994, GW493838; and angina – BAY-68-4986. For the i.v. antiarrhythmic agents that act as ventricular rate control agents, a selective response can be accomplished by careful dosing paradigms. The treatment of T2D using A1-AdoR agonists has been met by limited success due to cardiovascular side effects and well-defined desensitization of full agonists in both animal models and human trials (GR79236 and ARA). However, new partial A1-AdoR agonists are in development, including CVT-3619 (hA1-AdoR Ki = 55 nm, selectivity A2A > 200; A2B > 1000; A3 > 20, CV Therapeutics), that have the potential to provide enhanced insulin sensitivity without cardiovascular side effects or tachyphylaxis. The A1-AdoR agonists GW493838 and GR792363 are under evaluation for pain management. The non-nucleosidic A1-AdoR agonist, BAY-68-4986 (Capadenoson), represents a unique approach to angina wherein both animal studies and early human studies are promising. Conclusion: The challenges associated with developing an A1-AdoR agonist for therapeutic intervention are now well defined in humans. Significant progress has been made in identifying agents for the treatment of atrial arrhythmias, T2D, and angina.


Handbook of experimental pharmacology | 2009

A1 Adenosine Receptor Antagonists, Agonists, and Allosteric Enhancers

William F. Kiesman; Elfatih Elzein; Jeff Zablocki

Intense efforts of many pharmaceutical companies and academicians in the A(1) adenosine receptor (AR) field have led to the discovery of clinical candidates that are antagonists, agonists, and allosteric enhancers. The A(1)AR antagonists currently in clinical development are KW3902, BG9928, and SLV320. All three have high affinity for the human (h) A(1)AR subtype (hA(1) K (i) < 10 nM), > 200-fold selectivity over the hA(2A) subtype, and demonstrate renal protective effects in multiple animal models of disease and pharmacologic effects in human subjects. In the A(1)AR agonist area, clinical candidates have been discovered for the following conditions: atrial arrhythmias (tecadenoson, selodenoson and PJ-875); Type II diabetes and insulin sensitizing agents (GR79236, ARA, RPR-749, and CVT-3619); and angina (BAY 68-4986). The challenges associated with the development of any A(1)AR agonist are to obtain tissue-specific effects but avoid off-target tissue side effects and A(1)AR desensitization leading to tachyphylaxis. For the IV antiarrhythmic agents that act as ventricular rate control agents, a selective response can be accomplished by careful IV dosing paradigms. The treatment of type II diabetes using A(1)AR agonists in the clinic has met with limited success due to cardiovascular side effects and a well-defined desensitization of full agonists in human trials (GR79236, ARA, and RPR 749). However, new partial A(1)AR agonists are in development, including CVT-3619 hA(1) AR K(i) = 55nM, hA(2A:hA2B:hA(3))1,000:20, CV Therapeutics), which have the potential to provide enhanced insulin sensitivity without cardiovascular side effects and tachyphylaxis. The nonnucleosidic A(1)AR agonist BAY 68-4986 (capadenoson) represents a novel approach to angina wherein both animal studies and early human studies are promising. T-62 is an A(1)AR allosteric enhancer that is currently being evaluated in clinical trials as a potential treatment for neuropathic pain. The challenges associated with developing A(1)AR antagonists, agonists, or allosteric enhancers for therapeutic intervention are now well defined in humans. Significant progress has been made in identifying A(1)AR antagonists for the treatment of edema associated with congestive heart failure (CHF), A(1)AR agonists for the treatment of atrial arrhythmias, type II diabetes and angina, and A(1)AR allosteric enhancers for the treatment of neuropathic pain.


Journal of Medicinal Chemistry | 2006

Novel 1,3-disubstituted 8-(1-benzyl-1H-pyrazol-4-yl) xanthines: high affinity and selective A2B adenosine receptor antagonists.

Rao Kalla; Elfatih Elzein; Thao Perry; Xiaofen Li; Venkata Palle; Vaibhav Varkhedkar; Arthur Gimbel; Tennig Maa; Dewan Zeng; Jeff Zablocki

Adenosine has been suggested to induce bronchial hyperresponsiveness in asthmatics, which is believed to be an A(2B) adenosine receptor (AdoR) mediated pathway. We hypothesize that a selective, high-affinity A(2B) AdoR antagonist may provide therapeutic benefit in the treatment of asthma. In an attempt to identify a high-affinity, selective antagonist for the A(2B) AdoR, we synthesized 8-(C-4-pyrazolyl) xanthines. Compound 22, 8-(1H-pyrazol-4-yl)-1,3-dipropyl xanthine, is a N-1 unsubstituted pyrazole derivative that has favorable binding affinity (K(i) = 9 nM) for the A(2B) AdoR, but it is only 2-fold selective versus the A(1) AdoR. Introduction of a benzyl group at the N-1-pyrazole position of 22 resulted in 19, which had moderate selectivity. The initial focus of the SAR study was on the preparation of substituted benzyl derivatives of 19 because the corresponding phenyl, phenethyl, and phenpropyl derivatives showed a decrease in A(2B) AdoR affinity and selectivity relative to 19. The preferred substitution on the phenyl ring of 19 contains an electron-withdrawing group, specifically F or CF(3) at the m-position, as in 33 and 36 respectively, increases the selectivity while retaining the affinity for the A(2B) AdoR. Exploring disubstitutions on the phenyl ring of derivatives 33 and36 led to the 2-chloro-5-trifluoromethylphenyl derivative 50, which retained the A(2B) AdoR affinity but enhanced the selectivity relative to 36. After optimization of the substitution on the 8-pyrazole xanthine, 1,3-disubstitution of the xanthine core was explored with methyl, ethyl, butyl, and isobutyl groups. In comparison to the corresponding dipropyl analogues, the smaller 1,3-dialkyl groups (methyl and ethyl) increased the A(2B) AdoR binding selectivity of the xanthine derivatives while retaining the affinity. However, the larger 1,3-dialkyl groups (isobutyl and butyl) resulted in a decrease in both A(2B) AdoR affinity and selectivity. This final SAR optimization led to the discovery of 1,3-dimethyl derivative 60, 8-(1-(3-(trifluoromethyl) benzyl)-1H-pyrazol-4-yl)-1,3-dimethyl xanthine, a high-affinity (K(i) = 1 nM) A(2B) AdoR antagonist with high selectivity (990-, 690-, and 1,000-) for the human A(1), A(2A,) and A(3) AdoRs.


Bioorganic & Medicinal Chemistry Letters | 2008

Selective, high affinity A2B adenosine receptor antagonists: N-1 monosubstituted 8-(pyrazol-4-yl)xanthines

Rao Kalla; Elfatih Elzein; Thao Perry; Xiaofen Li; Art Gimbel; Ming Yang; Dewan Zeng; Jeff Zablocki

A series of N-1 monosubstituted 8-pyrazolyl xanthines have been synthesized and evaluated for their affinity for the adenosine receptors (AdoRs). We have discovered two compounds 18 (CVT-7124) and 28 (CVT-6694) that display good affinity for the A(2B) AdoR (K(i)=6 nM and 7 nM, respectively) and greater selectivity for the human A(1), A(2A), and A(3) AdoRs (>1000-, >830-, and >1500-fold; >850-, >700-, and >1280-fold, respectively). CVT-6694 has been shown to block the release of interleukin-6 and monocyte chemotactic protein-1 from bronchial smooth muscle cells (BSMC), a process believed to be promoted by activation of A(2B) AdoR.


Expert Opinion on Therapeutic Patents | 2006

A2B adenosine receptor antagonists and their potential indications

Jeff Zablocki; Elfatih Elzein; Rao Kalla

The intense efforts by many pharmaceutical companies and academics in the A2B adenosine receptor (AdoR) antagonist field are driven by the plethora of disease states where the A2B AdoR has been implicated to play a role: asthma, in which it mediates inflammatory cytokine release; diabetes, in which it mediates gluconeogenesis; diabetic retinopathy and cancer, in which it mediates angiogenesis; and inflammatory pain, in which it mediates inflammatory cytokine release. Major advances have been made in the past 5 years in obtaining selective, high affinity A2B adenosine receptor (AdoR) antagonists containing different classes of core heterocycles, including xanthines, 7-deazadenines and pyrimidines. The high affinity A2B AdoR antagonists have a high degree of structural diversity that should aid in further drug design attempts and optimisation of drug properties (i.e., solubility, oral bioavailability and half-life) through combinations of structural features from different classes. The goal of obtaining a selective, high affinity A2B AdoR antagonist has been met by several research groups and this will help address the role of the A2B AdoR in asthma, diabetes, cancer and management of inflammatory pain.


British Journal of Pharmacology | 2016

The novel late Na+ current inhibitor, GS‐6615 (eleclazine) and its anti‐arrhythmic effects in rabbit isolated heart preparations

Sridharan Rajamani; Gongxin Liu; Nesrine El-Bizri; Donglin Guo; Cindy Hong Li; Xiao-Liang Chen; Kristopher M. Kahlig; Nevena Mollova; Elfatih Elzein; Jeff Zablocki; Luiz Belardinelli

Enhanced late Na+ current (late INa) in the myocardium is pro‐arrhythmic. Inhibition of this current is a promising strategy to stabilize ventricular repolarization and suppress arrhythmias. Here, we describe GS‐6615, a selective inhibitor of late INa, already in clinical development for the treatment of long QT syndrome 3 (LQT3).


Bioorganic & Medicinal Chemistry Letters | 2016

Discovery of triazolopyridine GS-458967, a late sodium current inhibitor (Late INai) of the cardiac NaV 1.5 channel with improved efficacy and potency relative to ranolazine.

Dmitry Koltun; Eric Parkhill; Elfatih Elzein; Tetsuya J. Kobayashi; Gregory T. Notte; Rao Kalla; Robert Jiang; Xiaofen Li; Thao Perry; Belem Avila; Wei-Qun Wang; Catherine Smith-Maxwell; Arvinder Dhalla; Sridharan Rajamani; Brian Stafford; Jennifer Tang; Nevena Mollova; Luiz Belardinelli; Jeff Zablocki

We started with a medium throughput screen of heterocyclic compounds without basic amine groups to avoid hERG and β-blocker activity and identified [1,2,4]triazolo[4,3-a]pyridine as an early lead. Optimization of substituents for Late INa current inhibition and lack of Peak INa inhibition led to the discovery of 4h (GS-458967) with improved anti-arrhythmic activity relative to ranolazine. Unfortunately, 4h demonstrated use dependent block across the sodium isoforms including the central and peripheral nervous system isoforms that is consistent with its low therapeutic index (approximately 5-fold in rat, 3-fold in dog). Compound 4h represents our initial foray into a 2nd generation Late INa inhibitor program and is an important proof-of-concept compound. We will provide additional reports on addressing the CNS challenge in a follow-up communication.


Journal of Medicinal Chemistry | 2016

Discovery of Dihydrobenzoxazepinone (GS-6615) Late Sodium Current Inhibitor (Late INai), a Phase II Agent with Demonstrated Preclinical Anti-Ischemic and Antiarrhythmic Properties.

Jeff Zablocki; Elfatih Elzein; Xiaofen Li; Dmitry Koltun; Eric Parkhill; Tetsuya J. Kobayashi; Ruben Martinez; Britton Kenneth Corkey; Haibo Jiang; Thao Perry; Rao Kalla; Gregory T. Notte; Oliver L. Saunders; Michael Graupe; Yafan Lu; Chandru Venkataramani; Juan Guerrero; Jason K. Perry; Mark Osier; Robert G. Strickley; Gongxin Liu; Wei-Qun Wang; Lufei Hu; Xiao-Jun Li; Nesrine El-Bizri; Ryoko Hirakawa; Kris M. Kahlig; Cheng Xie; Cindy Hong Li; Arvinder Dhalla

Late sodium current (late INa) is enhanced during ischemia by reactive oxygen species (ROS) modifying the Nav 1.5 channel, resulting in incomplete inactivation. Compound 4 (GS-6615, eleclazine) a novel, potent, and selective inhibitor of late INa, is currently in clinical development for treatment of long QT-3 syndrome (LQT-3), hypertrophic cardiomyopathy (HCM), and ventricular tachycardia-ventricular fibrillation (VT-VF). We will describe structure-activity relationship (SAR) leading to the discovery of 4 that is vastly improved from the first generation late INa inhibitor 1 (ranolazine). Compound 4 was 42 times more potent than 1 in reducing ischemic burden in vivo (S-T segment elevation, 15 min left anteriorior descending, LAD, occlusion in rabbits) with EC50 values of 190 and 8000 nM, respectively. Compound 4 represents a new class of potent late INa inhibitors that will be useful in delineating the role of inhibitors of this current in the treatment of patients.


Bioorganic & Medicinal Chemistry Letters | 2017

Discovery of potent and selective inhibitors of calmodulin-dependent kinase II (CaMKII)

Dmitry Koltun; Eric Parkhill; Rao Kalla; Thao Perry; Elfatih Elzein; Xiaofen Li; Scott P. Simonovich; Christopher Ziebenhaus; Timothy R. Hansen; Bruno Marchand; WaiLok K. Hung; Leanna Lagpacan; Magdeleine Hung; Ron G. Aoyama; Bernard P. Murray; Jason K. Perry; John R. Somoza; Armando G. Villaseñor; Nikos Pagratis; Jeff Zablocki

We hereby disclose the discovery of inhibitors of CaMKII (7h and 7i) that are highly potent in rat ventricular myocytes, selective against hERG and other off-target kinases, while possessing good CaMKII tissue isoform selectivity (cardiac γ/δ vs. neuronal α/β). In vitro and in vivo ADME/PK studies demonstrated the suitability of these CaMKII inhibitors for PO (7h rat F = 73%) and IV pharmacological studies.


Archive | 2004

A2B adenosine receptor antagonists

Rao Kalla; Thao Perry; Elfatih Elzein; Vaibhav Varkhedkar; Xiaofen Li; Prabha Ibrahim; Venkata D Oakwood Estates Palle; Dengming Xiao; Jeff Zablocki

Collaboration


Dive into the Elfatih Elzein's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dewan Zeng

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Brent Blackburn

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Suresh Kerwar

University of California

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge