Co-reporter:Erik Serrao ; Bikash Debnath ; Hiroyuki Otake ; Yuting Kuang ; Frauke Christ ; Zeger Debyser
Journal of Medicinal Chemistry 2013 Volume 56(Issue 6) pp:2311-2322
Publication Date(Web):February 28, 2013
DOI:10.1021/jm301632e
On the basis of an initial molecular modeling study suggesting the favorable binding of the “privileged” fragment 8-hydroxyquinoline with HIV-1 integrase (IN) at the IN–lens epithelium-derived growth factor/p75 (LEDGF/p75) interface , we developed a set of modified 8-hydroxyquinoline fragments demonstrating micromolar IC50 values for inhibition of the IN–LEDGF/p75 interaction, but significant cytotoxicity was associated with these initial compounds. Diverse modifications at the C5 and C7 carbons of the 8-hydroxyquinoline core improved potency, but reduction of diversity to only modifications at the C5 position ultimately yielded potent inhibitors with low cytotoxicity. Two of these particular compounds, 5-((p-tolylamino)methyl)quinolin-8-ol and 5-(((3,4-dimethylphenyl)amino)methyl)quinolin-8-ol, inhibited viral replication in MT-4 cells with low micromolar EC50. This is the first study providing evidence for 8-hydroxyquinolines as novel inhibitors of the IN–LEDGF/p75 interaction. Our lead compounds are druglike, have low molecular weights, and are amenable to various substitutions suitable for enhancing their potency and selectivity.
Co-reporter:Ya-Qiu Long ; Shao-Xu Huang ; Zahrah Zawahir ; Zhong-Liang Xu ; Huiyuan Li ; Tino W. Sanchez ; Ying Zhi ; Stephanie De Houwer ; Frauke Christ ; Zeger Debyser
Journal of Medicinal Chemistry 2013 Volume 56(Issue 13) pp:5601-5612
Publication Date(Web):June 12, 2013
DOI:10.1021/jm4006516
HIV-1 integrase (IN) catalyzes the integration of viral DNA into the host genome, involving several interactions with the viral and cellular proteins. We have previously identified peptide IN inhibitors derived from the α-helical regions along the dimeric interface of HIV-1 IN. Herein, we show that appropriate hydrocarbon stapling of these peptides to stabilize their helical structure remarkably improves the cell permeability, thus allowing inhibition of the HIV-1 replication in cell culture. Furthermore, the stabilized peptides inhibit the interaction of IN with the cellular cofactor LEDGF/p75. Cellular uptake of the stapled peptide was confirmed in four different cell lines using a fluorescein-labeled analogue. Given their enhanced potency and cell permeability, these stapled peptides can serve as not only lead IN inhibitors but also prototypical biochemical probes or “nanoneedles” for the elucidation of HIV-1 IN dimerization and host cofactor interactions within their native cellular environment.
Co-reporter:Supranee Sangthong, Helen Ha, Thapong Teerawattananon, Nattaya Ngamrojanavanich, Nouri Neamati, Nongnuj Muangsin
Bioorganic & Medicinal Chemistry Letters 2013 Volume 23(Issue 22) pp:6156-6160
Publication Date(Web):15 November 2013
DOI:10.1016/j.bmcl.2013.09.004
Overcoming drug resistance with remarkable cytotoxic activity by anthracene-9,10-dione derivatives would offer a potential therapeutic strategy. In this study, we report the synthesis and the cytotoxicity of a novel set of anthraquninones. (4-(4-Aminobenzylamino)-9,10-dioxo-9,10-dihydroanthracen-1-yl-4-methylbenzenesulfonate) (3) has excellent in vitro cytotoxicity against doxorubicin-resistant cancer cell line (IC50 = 0.8 μM), 20-fold higher than doxorubicin. The cytotoxic effect via G2/M arrest does not appear to be ROS.
Co-reporter:Shenghui Yu, Tino Wilson Sanchez, Yang Liu, Yanzhen Yin, Nouri Neamati, Guisen Zhao
Bioorganic & Medicinal Chemistry Letters 2013 Volume 23(Issue 22) pp:6134-6137
Publication Date(Web):15 November 2013
DOI:10.1016/j.bmcl.2013.09.018
A series of novel pyrimidone analogues have been designed and synthesized as HIV-1 integrase (IN) inhibitors. This study demonstrated that introducing a substituent in the N1-position of the pyrimidone scaffold does not significantly influence IN inhibitory activity. Molecular docking studies showed these compounds could occupy the IN active site and form pi–pi interactions with viral DNA nucleotides DC16 and DA17 to displace reactive viral DNA 3′OH and block intasome activity.A series of novel pyrimidone analogues were designed and synthesized as HIV-1 integrase inhibitors.
Co-reporter:Tapan Karmakar, Yuting Kuang, Nouri Neamati, Jubaraj B. Baruah
Polyhedron 2013 Volume 54() pp:285-293
Publication Date(Web):30 April 2013
DOI:10.1016/j.poly.2013.02.068
Cadmium and indium complexes [Cd(L1)(O2CCH3)2(H2O)] (1) [Cd(L2)2(O2CCH3)2(H2O)] (2), [L1In(H2O)Cl3]·L1 (3) and [L1In(MeOH)Cl3]·L1 (4) (where L1 = 2-(pyridin-2-yl)benzo[d]thiazole and L2 = 2-(pyridin-4-yl)benzo[d]thiazole) are synthesized and characterized. The cadmium complex 1 is hexa-coordinated in which L1 act as N,N-chelate. The two acetate ligands of the complex bind to cadmium in monodentate and bidentate fashion. In the complex 2, the ligand L2 binds to cadmium ion through the nitrogen atom of the pyridine ring. The indium complexes 3 and 4 are hexa-coordinated; each has one L1 as chelating ligand and another L1 as molecule of crystallization. Fluorescence study revealed that the ligand L1 can detect indium(III) ions in the presence of cadmium(II) ions. In cell-based cytotoxicity assays, the ligands are biologically inactive, while the cadmium complexes 1 and 2 are cytotoxic in pancreatic cancer cell lines Mia Paca-2, BxPC-3 and Panc-1, with IC50 values as low as 16.0 μM.Cadmium and indium complexes [Cd(L1)(O2CCH3)2(H2O)] (1) [Cd(L2)2(O2CCH3)2(H2O)] (2), [L1In(H2O)Cl3]·L1 (3) and [L1In(MeOH)Cl3]·L1.H2O (4) (where L1 = 2-(pyridin-2-yl)benzo[d]thiazole and L2 = 2-(pyridin-4-yl)benzo[d]thiazole) are synthesized and characterized. The cadmium complexes 1 and 2 are cytotoxic in pancreatic cancer cell lines Mia Paca-2, BxPC-3 and Panc-1, with IC50 values as low as 16.0 μM.
Co-reporter:Tino Wilson Sanchez, Bikash Debnath, Frauke Christ, Hiroyuki Otake, Zeger Debyser, Nouri Neamati
Bioorganic & Medicinal Chemistry 2013 21(4) pp: 957-963
Publication Date(Web):
DOI:10.1016/j.bmc.2012.12.012
Co-reporter:Zhe Wang, Peng Huang, Ashwinkumar Bhirde, Albert Jin, Ying Ma, Gang Niu, Nouri Neamati and Xiaoyuan Chen
Chemical Communications 2012 vol. 48(Issue 78) pp:9768-9770
Publication Date(Web):08 Aug 2012
DOI:10.1039/C2CC31974H
A nanoscale RGD–pyrene–graphene oxide (GO) biosensor was prepared for real-time in situ detection of a cancer cell surface marker, integrin αvβ3. This nanoscale GO-based biosensor is simple, robust, sensitive and of high selectivity. It can also be adapted to other cancer cell surface marker evaluation systems.
Co-reporter:Li-Fan Zeng ; Yong Wang ; Roza Kazemi ; Shili Xu ; Zhong-Liang Xu ; Tino W. Sanchez ; Liu-Meng Yang ; Bikash Debnath ; Srinivas Odde ; Hua Xie ; Yong-Tang Zheng ; Jian Ding ; Nouri Neamati ;Ya-Qiu Long
Journal of Medicinal Chemistry 2012 Volume 55(Issue 22) pp:9492-9509
Publication Date(Web):October 26, 2012
DOI:10.1021/jm300667v
Among a large number of HIV-1 integrase (IN) inhibitors, the 8-hydroxy-[1,6]naphthyridines (i.e., L-870,810) were one of the promising class of antiretroviral drugs developed by Merck Laboratories. In spite of its remarkable potency and efficacy, unfortunately upon completion of phase I clinical studies, development of L-870,810 was halted. Because of its desirable pharmacological and pharmaceutical properties we were intrigued to design novel analogues of L-870,810 with goals to (1) improve upon limitations of naphthyridine-7-carboxamides as antiviral agents and (2) to reposition their use as innovative cytotoxic agents for cancer therapeutics. Herein, we report on the design and synthesis of a series of 1,6-naphthyridine-7-carboxamides with various substitutions at the 5- and 8-positions. All the new 5-substituted-8-hydroxy-[1,6]naphthyridines were potent IN inhibitors and the 5-substituted-8-amino-[1,6]naphthyridines were significantly cytotoxic. Further optimization of the 5,8-disubstituted-[1,6]naphthyridines with structural variation on 7-carboxamide delivered novel compounds with significant cytotoxicity in a panel of cancer cell lines and effective inhibition against select oncogenic kinases.
Co-reporter:Bikash Debnath ; Shili Xu
Journal of Medicinal Chemistry 2012 Volume 55(Issue 15) pp:6645-6668
Publication Date(Web):May 31, 2012
DOI:10.1021/jm300207s
Co-reporter:Shili Xu;Roppei Yamada;Alexey N. Butkevich;Ebrahim Zandi;Roger Duncan;Bikash Debnath;Yu Zhou;Nicos A. Petasis
PNAS 2012 Volume 109 (Issue 40 ) pp:16348-16353
Publication Date(Web):2012-10-02
DOI:10.1073/pnas.1205226109
Protein disulfide isomerase (PDI), an endoplasmic reticulum chaperone protein, catalyzes disulfide bond breakage, formation,
and rearrangement. The effect of PDI inhibition on ovarian cancer progression is not yet clear, and there is a need for potent,
selective, and safe small-molecule inhibitors of PDI. Here, we report a class of propynoic acid carbamoyl methyl amides (PACMAs)
that are active against a panel of human ovarian cancer cell lines. Using fluorescent derivatives, 2D gel electrophoresis,
and MS, we established that PACMA 31, one of the most active analogs, acts as an irreversible small-molecule inhibitor of
PDI, forming a covalent bond with the active site cysteines of PDI. We also showed that PDI activity is essential for the
survival and proliferation of human ovarian cancer cells. In vivo, PACMA 31 showed tumor targeting ability and significantly
suppressed ovarian tumor growth without causing toxicity to normal tissues. These irreversible small-molecule PDI inhibitors
represent an important approach for the development of targeted anticancer agents for ovarian cancer therapy, and they can
also serve as useful probes for investigating the biology of PDI-implicated pathways.
Co-reporter:Stephanie M. Stanford ; Divya Krishnamurthy ; Matthew D. Falk ; Rossella Messina ; Bikash Debnath ; Sheng Li ; Tong Liu ; Roza Kazemi ; Russell Dahl ∞; Yantao He ×; Xiao Yu ×; Andrew C. Chan ●; Zhong-Yin Zhang ×; Amy M. Barrios ; Virgil L. Woods ; Jr.; Nouri Neamati ;Nunzio Bottini
Journal of Medicinal Chemistry 2011 Volume 54(Issue 6) pp:1640-1654
Publication Date(Web):February 22, 2011
DOI:10.1021/jm101202j
The lymphoid tyrosine phosphatase LYP, encoded by the PTPN22 gene, is a critical regulator of signaling in T cells and recently emerged as a candidate target for therapy of autoimmune diseases. Here, by library screening, we identified a series of noncompetitive inhibitors of LYP that showed activity in primary T cells. Kinetic analysis confirmed that binding of the compounds to the phosphatase is nonmutually exclusive with respect to a known bidentate competitive inhibitor. The mechanism of action of the lead inhibitor compound 4e was studied by a combination of hydrogen/deuterium-exchange mass spectrometry and molecular modeling. The results suggest that the inhibitor interacts critically with a hydrophobic patch located outside the active site of the phosphatase. Targeting of secondary allosteric sites is viewed as a promising yet unexplored approach to develop pharmacological inhibitors of protein tyrosine phosphatases. Our novel scaffold could be a starting point to attempt development of “nonactive site” anti-LYP pharmacological agents.
Co-reporter:Roppei Yamada ; Xuefei Cao ; Alexey N. Butkevich ; Melissa Millard ; Srinivas Odde ; Nick Mordwinkin ; Rambabu Gundla ; Ebrahim Zandi ; Stan G. Louie ; Nicos A. Petasis
Journal of Medicinal Chemistry 2011 Volume 54(Issue 8) pp:2902-2914
Publication Date(Web):March 28, 2011
DOI:10.1021/jm101655d
Herein, we discovered a series of propynoic acid carbamoyl methyl-amides (PACMAs) with potent cytotoxicity against a panel of cancer cell lines. These compounds interrupted cell cycle progression at low micromolar concentrations and induced early and late stage apoptosis. A representative compound suppressed tumor growth without apparent toxicity in an MDA-MB-435 mouse xenograft model. We used a Kinexus 628-antibody microarray and the Ingenuity Pathway Analysis (IPA) bioinformatics tools to better understand their mechanisms. The IPA analysis revealed the initiation of Nrf2-mediated oxidative stress through modulating the expression of SOD1 and STIP1 by compound 1. The involvement of the oxidative stress pathway was further validated by measuring the levels of the PACMA-induced mitochondrial superoxide species. To our knowledge, this is the first report on the discovery and biological evaluations of PACMAs as anticancer agents. Their broad-spectrum in vitro cytotoxicity, possibly through an oxidative stress-mediated pathway, and in vivo efficacy warrant further preclinical investigations.
Co-reporter:Xing Fan, Feng-Hua Zhang, Rasha I. Al-Safi, Li-Fan Zeng, Yumna Shabaik, Bikash Debnath, Tino W. Sanchez, Srinivas Odde, Nouri Neamati, Ya-Qiu Long
Bioorganic & Medicinal Chemistry 2011 Volume 19(Issue 16) pp:4935-4952
Publication Date(Web):15 August 2011
DOI:10.1016/j.bmc.2011.06.058
HIV-1 integrase (IN) is a validated therapeutic target for antiviral drug design. However, the emergence of viral strains resistant to clinically studied IN inhibitors demands the discovery of novel inhibitors that are structurally as well mechanistically different. Herein, we describe the design and discovery of novel IN inhibitors targeting the catalytic domain as well as its interaction with LEDGF/p75, which is essential for the HIV-1 integration as an IN cofactor. By merging the pharmacophores of salicylate and catechol, the 2,3-dihydroxybenzamide (5a) was identified as a new scaffold to inhibit the strand transfer reaction efficiently. Further structural modifications on the 2,3-dihydroxybenzamide scaffold revealed that the heteroaromatic functionality attached on the carboxamide portion and the piperidin-1-ylsulfonyl substituted at the phenyl ring are beneficial for the activity, resulting in a low micromolar IN inhibitor (5p, IC50 = 5 μM) with more than 40-fold selectivity for the strand transfer over the 3′-processing reaction. More significantly, this active scaffold remarkably inhibited the interaction between IN and LEDGF/p75 cofactor. The prototype example, N-(cyclohexylmethyl)-2,3-dihydroxy-5-(piperidin-1-ylsulfonyl) benzamide (5u) inhibited the IN-LEDGF/p75 interaction with an IC50 value of 8 μM. Using molecular modeling, the mechanism of action was hypothesized to involve the chelation of the divalent metal ions inside the IN active site. Furthermore, the inhibitor of IN-LEDGF/p75 interaction was properly bound to the LEDGF/p75 binding site on IN. This work provides a new and efficient approach to evolve novel HIV-1 IN inhibitors from rational integration and optimization of previously reported inhibitors.
Co-reporter:Jean-Philippe Monserrat, Rasha I. Al-Safi, Keshri Nath Tiwari, Lionel Quentin, Guy G. Chabot, Anne Vessières, Gérard Jaouen, Nouri Neamati, Elizabeth A. Hillard
Bioorganic & Medicinal Chemistry Letters 2011 Volume 21(Issue 20) pp:6195-6197
Publication Date(Web):15 October 2011
DOI:10.1016/j.bmcl.2011.07.078
We report here the discovery of a potent series of HIV-1 integrase (IN) inhibitors based on the ferrocenyl chalcone difluoridoborate structure. Ten new compounds have been synthesized and were generally found to have similar inhibitory activities against the IN 3′ processing and strand transfer (ST) processes. IC50 values were found to be in the low micromolar range, and significantly lower than those found for the non-coordinated ferrocenyl chalcones and other ferrocene molecules. The ferrocenyl chalcone difluoridoborates furthermore exhibited low cytotoxicity against cancer cells and low morphological activity against epithelial cells.
Co-reporter:Laith Q. Al-Mawsawi
ChemMedChem 2011 Volume 6( Issue 2) pp:228-241
Publication Date(Web):
DOI:10.1002/cmdc.201000443
Abstract
HIV-1 integrase (IN) is one of three essential enzymes for viral replication, and is a focus of ardent antiretroviral drug discovery and development efforts. Diligent research has led to the development of the strand-transfer-specific chemical class of IN inhibitors, with two compounds from this group, raltegravir and elvitegravir, advancing the farthest in the US Food and Drug Administration (FDA) approval process for any IN inhibitor discovered thus far. Raltegravir, developed by Merck & Co., has been approved by the FDA for HIV-1 therapy, whereas elvitegravir, developed by Gilead Sciences and Japan Tobacco, has reached phase III clinical trials. Although this is an undoubted success for the HIV-1 IN drug discovery field, the emergence of HIV-1 IN strand-transfer-specific drug-resistant viral strains upon clinical use of these compounds is expected. Furthermore, the problem of strand-transfer-specific IN drug resistance will be exacerbated by the development of cross-resistant viral strains due to an overlapping binding orientation at the IN active site and an equivalent inhibitory mechanism for the two compounds. This inevitability will result in no available IN-targeted therapeutic options for HIV-1 treatment-experienced patients. The development of allosterically targeted IN inhibitors presents an extremely advantageous approach for the discovery of compounds effective against IN strand-transfer drug-resistant viral strains, and would likely show synergy with all available FDA-approved antiretroviral HIV-1 therapeutics, including the IN strand-transfer-specific compounds. Herein we review the concept of allosteric IN inhibition, and the small molecules that have been investigated to bind non-active-site regions to inhibit IN function.
Co-reporter:Zahrah Zawahir ; Raveendra Dayam ; Jinxia Deng ; Cherelene Pereira
Journal of Medicinal Chemistry 2009 Volume 52(Issue 1) pp:20-32
Publication Date(Web):December 10, 2008
DOI:10.1021/jm800739m
Human apurinic/apyrimidinic endonuclease 1 (APE1) is an important enzyme in the base excision repair (BER) pathway that is essential for the repair of abasic sites in the genome. Evidence for APE1 as an attractive therapeutic target in anticancer drug development has been demonstrated by studies that link overexpression of APE1 in many cancers to resistance of tumor cells to radio- and chemotherapy. APE1 also shows a protective effect in several cancer cell models to a variety of DNA damaging agents. This study represents the first rational design of selective small-molecule APE1 inhibitors utilizing a three-dimensional interaction-based pharmacophore perception. All of our most potent molecules show inhibitory activity below 10 μM and are selective for APE1 inhibition.
Co-reporter:Kavya Ramkumar, Konstantin V. Tambov, Rambabu Gundla, Alexander V. Manaev, Vladimir Yarovenko, Valery F. Traven, Nouri Neamati
Bioorganic & Medicinal Chemistry 2009 Volume 17(Issue 2) pp:929
Publication Date(Web):15 January 2009
DOI:10.1016/j.bmc.2008.08.082
Co-reporter:Hongcai Li, Chao Wang, Tino Sanchez, Yanmei Tan, Chunying Jiang, Nouri Neamati, Guisen Zhao
Bioorganic & Medicinal Chemistry 2009 Volume 17(Issue 7) pp:2913-2919
Publication Date(Web):1 April 2009
DOI:10.1016/j.bmc.2009.01.077
HIV-1 integrase, which catalyzes the integration of the viral genome into the cellular chromosome, is an essential enzyme for retroviral replication, and represents an attractive and validated target in the development of therapeutics against AIDS. In this paper, 17 amide-containing novel diketoacids were designed and synthesized, and their ability to inhibit HIV-1 integrase was tested. The structure–activity relationships were also analyzed.A series of novel amide-containing diketoacids were designed and synthesized to develop potent HIV integrase inhibitors. Their inhibition of HIV integrase was tested and the structure–activity relationships were discussed.
Co-reporter:Mario Sechi, Giuseppe Rizzi, Alessia Bacchi, Mauro Carcelli, Dominga Rogolino, Nicolino Pala, Tino W. Sanchez, Laleh Taheri, Raveendra Dayam, Nouri Neamati
Bioorganic & Medicinal Chemistry 2009 Volume 17(Issue 7) pp:2925-2935
Publication Date(Web):1 April 2009
DOI:10.1016/j.bmc.2008.10.088
Previously, we discovered linomide analogues as novel HIV-1 integrase (IN) inhibitors. Here, to make possible structure–activity relationships, we report on the design and synthesis of a series of substituted dihydroquinoline-3-carboxylic acids. The crystal structure of the representative compound 2c has also been solved. Among the eight new analogues, 2e showed a potency in inhibiting IN strand transfer catalytic activity similar to the reference diketo acid inhibitor L-731,988 (IC50 = 0.9 μM vs. 0.54 μM, for 2e and L-731,988, respectively). Furthermore, none of the compounds showed significant cytotoxicity in two tested cancer cell lines. These compounds represent an interesting prototype of IN inhibitors, potentially involved in a metal chelating mechanism, and further optimization is warranted.
Co-reporter:Peng Wang, Chuan Liu, Tino Sanches, Yuan Zhong, Bo Liu, Junlong Xiong, Nouri Neamati, Guisen Zhao
Bioorganic & Medicinal Chemistry Letters 2009 Volume 19(Issue 16) pp:4574-4578
Publication Date(Web):15 August 2009
DOI:10.1016/j.bmcl.2009.06.100
A series of nitrogen-containing polyhydroxylated aromatics from caffeic acid phenethyl ester were designed and synthesized as HIV-1 integrase inhibitors. Most of these compounds exhibited potent inhibitory activities at micromolar concentrations against HIV-1 integrase in the 3′-end processing and the strand transfer. Their key structure–activity relationship was also discussed.A series of novel nitrogen-containing polyhydroxylated aromatics were evaluated for their ability to inhibit HIV-1 integrase at micromolar concentrations.
Co-reporter:Alessia Bacchi ; Mariano Biemmi ; Mauro Carcelli ; Fabrizio Carta ; Carlotta Compari ; Emilia Fisicaro ; Dominga Rogolino ; Mario Sechi ; Martin Sippel ; Christoph A. Sotriffer ; Tino W. Sanchez
Journal of Medicinal Chemistry 2008 Volume 51(Issue 22) pp:7253-7264
Publication Date(Web):November 4, 2008
DOI:10.1021/jm800893q
Previously, we synthesized a series of β-diketo acid metal complexes as novel HIV-1 integrase (IN) inhibitors (J. Med. Chem. 2006, 46, 4248−4260). Herein, a further extension of this study is reported. First, detailed docking studies were performed in order to investigate the mode of binding in the active site of the free ligands and of their metal complexes. Second, a series of potentiometric measurements were conducted for two diketo acids chosen as model ligands, with Mn2+ and Ca2+, in order to outline a speciation model. Third, we designed and synthesized a new set of complexes with different stoichiometries and tested them in an in vitro assay specific for IN. Finally, we obtained the first X-ray structure of a metal complex with HIV-1 IN inhibition activity. Analysis of these results supports the hypothesis that the diketo acids could act as complexes and form complexes with the metal ions on the active site of the enzyme.
Co-reporter:Raveendra Dayam ; Laith Q. Al-Mawsawi ; Zahrah Zawahir ; Myriam Witvrouw ; Zeger Debyser
Journal of Medicinal Chemistry 2008 Volume 51(Issue 5) pp:1136-1144
Publication Date(Web):February 19, 2008
DOI:10.1021/jm070609b
Two decades of intensive research efforts have led to the discovery of a large number of HIV-1 integrase (IN) inhibitors. Recently, the United States Food and Drug Administration (US FDA) approved MK-0518, or raltegravir (1), as the first IN inhibitor for HIV/AIDS treatment. Growing clinical evidence also demonstrates that the emergence of HIV-1 virus strains bearing IN amino acid substitutions that confer resistance to IN inhibitors is inevitable. The discovery of second generation inhibitors with potency against viral strains bearing drug resistant IN substitutions is necessary for ongoing effective treatment of viral infections. We generated common feature pharmacophore hypotheses using a training set of quinolone 3-carboxylic acid IN inhibitors, including the clinical candidate GS-9137 (2). A database search of small molecules using the quinolone 3-carboxylic acid pharmacophore model, followed by in vitro evaluation of selected hits in an assay specific to IN, resulted in the discovery of potential leads with diverse structural scaffolds useful for the design of second generation IN inhibitors.
Co-reporter:Li-Fan Zeng, Xiao-Hua Jiang, Tino Sanchez, Hu-Shan Zhang, Raveendra Dayam, Nouri Neamati, Ya-Qiu Long
Bioorganic & Medicinal Chemistry 2008 Volume 16(Issue 16) pp:7777-7787
Publication Date(Web):15 August 2008
DOI:10.1016/j.bmc.2008.07.008
Aryl diketoacids (ADK) and their bioisosteres are among the most promising HIV-1 integrase (IN) inhibitors. Previously, we designed a series of ADK dimers as a new class of IN inhibitors that were hypothesized to target two divalent metal ions on the active site of IN. Herein we present a further structure–activity relationship (SAR) study with respect to the substituent effect of the ADK and the dimerization with conformationally constrained linkers such as piperazine, 4-amino-piperidine, piperidin-4-ol, and trans-cyclohexan-1,4-diamine. The substituents on the phenyl ring as well as the spatial orientation of the two diketo units were observed to play important roles in the IN inhibitory potency. The hydrophobic group was an optimal substitution at the 3-position of the aryl ring. The piperazine and 4-amino-piperidine linkers brought about the most potent analogs among the hydrophobic group or halogen substituted ADK dimers. The docking studies suggested that the bulky hydrophobic substitution at 3-phenyl ring and the linker of 4-amino-piperidine were beneficial for adopting an active conformation to achieve strong interactions with the active site Mg2+ and the key residue E152 within the catalytic core domain. This study is a significant extension of our previous report on the dimeric ADK-containing IN inhibitors, providing a new promising template for further lead optimization.A new class of ADK dimer inhibitors of HIV-1 integrase was designed and synthesized by employing conformationally constrained diamine as linker. The substituent on the phenyl portion and the orientation of the linker were investigated with respect to the potency-enhancing effect.
Co-reporter:Kavya Ramkumar, Konstantin V. Tambov, Rambabu Gundla, Alexandr V. Manaev, Vladimir Yarovenko, Valery F. Traven, Nouri Neamati
Bioorganic & Medicinal Chemistry 2008 Volume 16(Issue 19) pp:8988-8998
Publication Date(Web):1 October 2008
DOI:10.1016/j.bmc.2008.08.067
HIV-1 integrase (IN) has emerged as an important therapeutic target for anti-HIV drug development. Its uniqueness to the virus and its critical role in the viral life cycle makes IN suitable for selective inhibition. The recent approval of Raltegravir (MK-0518) has created a surge in interest and great optimism in the field. In our ongoing IN drug design research, we herein report the discovery of substituted analogs of 3-acetyl-4-hydroxy-2-pyranones and their difluoridoborate complexes as novel IN inhibitors. In many of these compounds, complexation with boron difluoride increased the potency and selectivity of IN inhibition. Compound 9 was most active with an IC50 value of 9 μM and 3 μM for 3’-processing and strand transfer inhibition, respectively.
Co-reporter:Jinxia Deng;Laleh Taheri;Fedora Gre;Francesca Aiello;Antonio Garofalo
ChemMedChem 2008 Volume 3( Issue 11) pp:1677-1686
Publication Date(Web):
DOI:10.1002/cmdc.200800217
Abstract
Quinoxalinehydrazines represent a novel class of compounds with excellent potency in a panel of cancer cell lines. A prototype compound, SC144, showed significant in vivo efficacy in mice xenograft models of human breast cancer cells. The subsequent structure–activity relationship study resulted in the discovery of SC161 with better potency in cancer cell lines. Further exploring the possible conformational space by a 10 ns molecular dynamics simulation as presented herein, resulted in various pharmacophore orientations. The trajectory analysis indicated that in most of the simulation time, the molecule stays favorably in a compact planarlike orientation. We therefore built a pharmacophore model based on the cluster containing the highest number of frames to represent the most probable orientation. The model was used to screen a subset of our small molecule database containing 350,000 compounds. We selected 35 compounds for the initial cytotoxicity screen. Seventeen compounds belonging to oxadiazolopyrazine and quinoline class displayed cytotoxicity in various cancer cell lines. Five of them, compounds 2, 6, 15, 16, and 19, all bearing an oxadiazolopyrazine scaffold, showed IC50 values <3 μM in certain tumor cell lines. The most potent compound, 2, showed IC50 values <2 μM in HCT116 p53+/+, HCT116 p53−/−, and HEY cells, and 8 μM in NIH3T3 cells. This study shows that conformational sampling of a lead small molecule followed by representative pharmacophore model development is an efficient approach for the rational design of novel anticancer agents with similar or better potency than the original lead but with different physicochemical properties.
Co-reporter:Jinxia Deng, Tino Sanchez, Laith Q. Al-Mawsawi, Raveendra Dayam, Rosendo A. Yunes, Antonio Garofalo, Michael B. Bolger, Nouri Neamati
Bioorganic & Medicinal Chemistry 2007 Volume 15(Issue 14) pp:4985-5002
Publication Date(Web):15 July 2007
DOI:10.1016/j.bmc.2007.04.041
Recently, we reported small-molecule chalcones as a novel class of HIV-1 integrase (IN) inhibitors. The most potent compound showed an IC50 value of 2 μM for both IN-mediated 3′-processing and strand transfer reactions. To further utilize the chalcones, we developed pharmacophore models to identify chemical signatures important for biological activity. The derived models were validated with a collection of published inhibitors, and then were applied to screen a subset of our small molecule database. We tested 71 compounds in an in vitro assay specific for IN enzymatic activity. Forty-four compounds showed inhibitory potency <100 μM, and four of them exhibited IC50 values <10 μM. One compound, 62, with an IC50 value of 0.6 μM, displayed better potency than the original chalcone 2 against the strand transfer process. This study demonstrates the systematic use of pharmacophore technologies to discover novel structurally diverse inhibitors based on lead molecules that would exhibit poor characteristics in vivo. The identified compounds have the potential to exhibit favorable pharmacokinetic and pharmacodynamic profiles.
Co-reporter:Laith Q. Al-Mawsawi, Raveendra Dayam, Laleh Taheri, Myriam Witvrouw, Zeger Debyser, Nouri Neamati
Bioorganic & Medicinal Chemistry Letters 2007 Volume 17(Issue 23) pp:6472-6475
Publication Date(Web):1 December 2007
DOI:10.1016/j.bmcl.2007.09.102
The previously discovered salicylhydrazide class of compounds displayed potent HIV-1 integrase (IN) inhibitory activity. The development of this class of compounds as antiretroviral agents was halted due to cytotoxicity in the nanomolar to sub-micromolar range. We identified a novel class of non-cytotoxic hydrazide IN inhibitors utilizing the minimally required salicylhydrazide substructure as a template in a small-molecule database search. The novel hydrazides displayed low micromolar IN inhibitory activity and are several hundred-fold less cytotoxic than previously disclosed salicylhydrazide IN inhibitors.
Co-reporter:Raveendra Dayam;Laith Q. Al-Mawsawi;Dr Nouri Neamati
Drugs in R&D 2007 Volume 8( Issue 3) pp:155-168
Publication Date(Web):2007 May
DOI:10.2165/00126839-200708030-00003
From the discovery of HIV-1 integrase (IN) inhibitors using enzyme-based assays in 1992, it has taken 15 years to achieve success in human clinical trials. Currently available antiretroviral drugs set high clinical standards in efficacy and long-term safety for upcoming novel HIV/AIDS therapeutic agents. The results from advanced stages of human clinical trials with IN inhibitors indicate a promising future for these compounds as a novel class of antiretroviral drugs. Success and failure of previously discovered antiretroviral drugs have taught us that there are no magic bullets in eradicating HIV. However, approval of drugs selectively targeting IN has long been awaited. There is once again a surge of interest in the field focusing on clinical development of IN inhibitors. Here, we summarise the current status of IN inhibitors under clinical development. These agents include S-1360, GSK-364735, L-870,810, L-870,812, MK-0518, GS-9137, L-900564, GS-9224, and BMS-707035. Promising antiviral activity has already been achieved with MK-0518 and GS-9137 in late-stage clinical studies.
Co-reporter:Jinxia Deng, James A. Kelley, Joseph J. Barchi, Tino Sanchez, Raveendra Dayam, Yves Pommier, Nouri Neamati
Bioorganic & Medicinal Chemistry 2006 Volume 14(Issue 11) pp:3785-3792
Publication Date(Web):1 June 2006
DOI:10.1016/j.bmc.2006.01.040
HIV-1 integrase (IN) is an essential enzyme for effective viral replication and is a validated target for the development of antiretroviral drugs. Currently, there are no approved drugs targeting this enzyme. In this study, we have identified 11 structurally diverse small-molecule inhibitors of IN. These compounds have been selected by mining the moderately active antiviral molecules from a collection of 90,000 compounds screened by the National Cancer Institute (NCI) Antiviral Program. These compounds, which were screened at the NCI during the past 20 years, resulted in approximately 4000 compounds labeled as ‘moderately active.’ In our study, chalcone 11 shows the most potent activity with an IC50 of 2 ± 1 μM against purified IN in the presence of both Mn2+ and Mg2+ as cofactors. Docking simulations using the 11 identified inhibitors as a training set have elucidated two unique binding areas within the active site: The first encompasses the conserved D64-D116-E152 motif, while the other involves the flexible loop region formed by amino acid residues 140–149. The tested inhibitors exhibit favorable interactions with important amino acid residues through van der Waals and H-bonding contacts.
Co-reporter:Tino Sanchez;Raveendra Dayam
ChemMedChem 2006 Volume 1(Issue 2) pp:238-244
Publication Date(Web):20 DEC 2005
DOI:10.1002/cmdc.200500018
HIV-1 integrase (IN) is an essential enzyme for viral replication and a validated target for the development of drugs against AIDS. Currently there are no approved drugs that target IN. However, new IN inhibitors are under clinical investigation. As more IN inhibitors enter human drug trials, there is a growing need for the design of novel lead compounds with diverse structural scaffolds and promising pharmacokinetic properties to counteract the difficulties observed with first-generation IN inhibitors. We have identified a novel class of IN inhibitors through the systematic exploration of structure–activity relationships in a series of linomide analogues. The predicted bound conformation of the most active analogues inside the IN active site also supports the observed structure–activity correlation in this new compound class.
Co-reporter:Francesca Aiello, Antonella Brizzi, Antonio Garofalo, Fedora Grande, Gaetano Ragno, Raveendra Dayam, Nouri Neamati
Bioorganic & Medicinal Chemistry 2004 Volume 12(Issue 16) pp:4459-4466
Publication Date(Web):15 August 2004
DOI:10.1016/j.bmc.2004.05.037
Thiazolothiazepines are among the smallest and most constrained inhibitors of human immunodeficiency virus type-1 integrase (HIV-1 IN) inhibitors (J. Med. Chem.1999, 42, 3334). Previously, we identified two thiazolothiazepines lead IN inhibitors with antiviral activity in cell-based assays. Structural optimization of these molecules necessitated the design of easily synthesizable analogs. In order to design similar molecules with least number of substituent, herein we report the synthesis of 10 novel analogs. One of the new compounds (1) exhibited similar potency as the reference compounds, confirming that a thiazepinedione fused to a naphthalene ring system is the best combination for the molecule to accommodate into the IN active site. Thus, the replacement of sulfur in the thiazole ring with an oxygen does not seem considerably affect potency. On the other hand, the introduction of an extra methyl group at position 1 of the polycyclic system or the shift from a thiazepine to an oxazepine skeleton decreased potency. In order to understand their mode of interactions with IN active site, we docked all the compounds onto the previously reported X-ray crystal structure of IN. We observed that compounds 7–9 occupied an area close to D64 and Mg2+ and surrounded by amino acid residues K159, K156, N155, E152, D116, H67, and T66. The oxygen atom of the oxazolo ring of 7 and 8 could chelate Mg2+. These results indicate that the new analogs potentially interact with the highly conserved residues important for IN catalytic activities.Graphic
Co-reporter:Zdzislaw Brzozowski, Franciszek Saczewski, Tino Sanchez, Chih-Ling Kuo, Maria Gdaniec, Nouri Neamati
Bioorganic & Medicinal Chemistry 2004 Volume 12(Issue 13) pp:3663-3672
Publication Date(Web):1 July 2004
DOI:10.1016/j.bmc.2004.04.024
HIV-1 integrase (IN) is an essential enzyme for effective viral replication and is an attractive target for selective blockade of viral infection. Previously, we identified a series of sulfones, sulfonamides, and mercaptosalicylhydrazides (MBSAs) as IN inhibitors with antiviral activities in cell-based assays. In an effort to optimize a series of our active site directed lead compounds, we designed and synthesized novel benzodithiazines starting from MBSAs. In contrast to all reported IN inhibitors benzodithiazines are essentially nontoxic. Significant antiviral potency was only observed at concentration exceedingly higher than that required to inhibit purified IN.Graphic
Co-reporter:I-Jen Chen, Nouri Neamati , Marc C. Nicklaus, Ann Orr, Lynne Anderson, Joseph J. Barchi Jr., James A. Kelley, Yves Pommier, Alexander D. MacKerell Jr.
Bioorganic & Medicinal Chemistry 2000 Volume 8(Issue 10) pp:2385-2398
Publication Date(Web):October 2000
DOI:10.1016/S0968-0896(00)00180-2
Integration of viral DNA into the host cell genome is a critical step in the life cycle of HIV. This essential reaction is catalyzed by integrase (IN) through two steps, 3′-processing and DNA strand transfer. Integrase is an attractive target for drug design because there is no known cellular analogue and integration is essential for successful replication of HIV. A computational three-dimensional (3-D) database search was used to identify novel HIV-1 integrase inhibitors. Starting from the previously identified Y3 (4-acetylamino-5-hydroxynaphthalene-2,7-disulfonic acid) binding site on the avian sarcoma virus integrase (ASV IN), a preliminary search of all compounds in the nonproprietary, open part of the National Cancer Institute 3-D database yielded a collection of 3100 compounds. A more rigorous scoring method was used to rescreen the 3100 compounds against both ASV IN and HIV-1 IN. Twenty-two of those compounds were selected for inhibition assays against HIV-1 IN. Thirteen of the 22 showed inhibitory activity against HIV-1 IN at concentrations less than 200 μM and three of them showed antiviral activities in HIV-1 infected CEM cells with effective concentrations (EC50) ranging from 0.8 to 200 μM. Analysis of the computer-generated binding modes of the active compounds to HIV-1 IN showed that simultaneous interaction with the Y3 site and the catalytic site is possible. In addition, interactions between the active compounds and the flexible loop involved in the binding of DNA by IN are indicated to occur. The structural details and the unique binding motif between the HIV-1 IN and its inhibitors identified in the present work may contribute to the future development of IN inhibitors.
Co-reporter:Laith Q. Al-Mawsawi, Nouri Neamati
Trends in Pharmacological Sciences (October 2007) Volume 28(Issue 10) pp:526-535
Publication Date(Web):1 October 2007
DOI:10.1016/j.tips.2007.09.005
HIV-1 integrase (IN) executes the insertion of proviral DNA into the host cell genome, an essential step in the retroviral life cycle. This is a multi-step process that starts in the cytosol and culminates in the nucleus of the infected cell. It is becoming increasingly clear that IN interacts with a wide range of different host-cell proteins throughout the viral life cycle. These cellular cofactors are exploited for various functions, including nuclear import, DNA target-site selection and virion assembly. The disruption of key interactions between IN and direct cellular cofactors affords a novel therapeutic approach for the design and development of new classes of anti-retroviral agents. Here, we will discuss the rationale behind this emerging and promising therapeutic strategy for HIV drug discovery. Our discussion includes the identified IN cellular cofactors, key research developments in the field and the implications this approach will have on the current HIV treatment regimen.
Co-reporter:Mario Sechi, Fabrizio Carta, Luciano Sannia, Roberto Dallocchio, Alessandro Dessì, Rasha I. Al-Safi, Nouri Neamati
Antiviral Research (March 2009) Volume 81(Issue 3) pp:267-276
Publication Date(Web):March 2009
DOI:10.1016/j.antiviral.2008.12.010
Co-reporter:Divya Pathania, Melissa Millard, Nouri Neamati
Advanced Drug Delivery Reviews (30 November 2009) Volume 61(Issue 14) pp:1250-1275
Publication Date(Web):30 November 2009
DOI:10.1016/j.addr.2009.05.010
Cancer cells are characterized by self-sufficiency in the absence of growth signals, their ability to evade apoptosis, resistance to anti-growth signals, sustained angiogenesis, uncontrolled proliferation, and invasion and metastasis. Alterations in cellular bioenergetics are an emerging hallmark of cancer. The mitochondrion is the major organelle implicated in the cellular bioenergetic and biosynthetic changes accompanying cancer. These bioenergetic modifications contribute to the invasive, metastatic and adaptive properties typical in most tumors. Moreover, mitochondrial DNA mutations complement the bioenergetic changes in cancer. Several cancer management therapies have been proposed that target tumor cell metabolism and mitochondria. Glycolytic inhibitors serve as a classical example of cancer metabolism targeting agents. Several TCA cycle and OXPHOS inhibitors are being tested for their anticancer potential. Moreover, agents targeting the PDC/PDK (pyruvate dehydrogenase complex/pyruvate dehydrogenase kinase) interaction are being studied for reversal of Warburg effect. Targeting of the apoptotic regulatory machinery of mitochondria is another potential anticancer field in need of exploration. Additionally, oxidative phosphorylation uncouplers, potassium channel modulators, and mitochondrial redox are under investigation for their anticancer potential. To this end there is an increased demand for agents that specifically hit their target. Delocalized lipophilic cations have shown tremendous potential in delivering anticancer agents selectively to tumor cells. This review provides an overview of the potential anticancer agents that act by targeting cancer cell metabolism and mitochondria, and also brings us face to face with the emerging opportunities in cancer therapy.
Co-reporter:Laith Q. Al-Mawsawi, Frauke Christ, Raveendra Dayam, Zeger Debyser, Nouri Neamati
FEBS Letters (30 April 2008) Volume 582(Issue 10) pp:1425-1430
Publication Date(Web):30 April 2008
DOI:10.1016/j.febslet.2008.02.076
A lens epithelium-derived growth factor (LEDGF)/p75 peptide was evaluated for human immunodeficiency virus type 1 integrase (IN) inhibitory activity. The LEDGF/p75 peptide modestly inhibited IN catalysis and was dependent on IN–DNA assembly. The peptide was also effective at disrupting LEDGF/p75–IN complex formation. We next investigated the activity of the LEDGF/p75 peptide on IN mutant proteins that are unable to catalyze the DNA strand transfer reaction. The LEDGF/p75 peptide displayed an increased potency on these IN proteins, from 5-fold to greater than 10-fold, indicating the IN multimeric state greatly influences the peptide inhibitory effects. These results shed light on IN–DNA multimeric formation, and how this process influences the LEDGF/p75–IN interaction.
Co-reporter:Laith Q. Al-Mawsawi, Mario Sechi, Nouri Neamati
FEBS Letters (20 March 2007) Volume 581(Issue 6) pp:1151-1156
Publication Date(Web):20 March 2007
DOI:10.1016/j.febslet.2007.02.023
HIV-1 integrase (IN) mediates the insertion of viral cDNA into the cell genome, a vital process for replication. This step is catalyzed by two separate DNA reaction events, termed 3′-processing and strand transfer. Here, we show that six inhibitors from five structurally different classes of compounds display a selectivity shift towards preferential strand transfer inhibition over the 3′-processing activity of IN when a single serine is substituted at position C130. Even though IN utilizes the same active site for both reactions, this finding suggests a distinct conformational dissimilarity in the mechanistic details of each IN catalytic event.
Co-reporter:Rambabu Gundla ; Roza Kazemi ; Ramadevi Sanam ; Ravikumar Muttineni ; Jagarlapudi A. R. P. Sarma ; Raveendra Dayam
Journal of Medicinal Chemistry () pp:
Publication Date(Web):May 24, 2008
DOI:10.1021/jm7013875
Consensus virtual screening models were generated and validated utilizing a set of known human epidermal growth factor receptor-2 (HER2) inhibitors and modeled HER2 active and inactive state structures. The virtual screening models were successfully employed to discover a set of structurally diverse compounds with growth inhibitory activity against HER2-overexpressing SKBR3 breast cancer cell line. A search of a 3D database containing 350000 small-molecules using the consensus models retrieved 531 potential hits. Of the 531 hits, 57 were selected for testing in SKBR3 cells on the basis of structural novelty and desirable drug-like properties. Seven compounds inhibited growth of SKBR3 cells with IC50 values <10 µM. These lead compounds have desirable physicochemical properties and are excellent candidates for further optimization.
Co-reporter:Zhe Wang, Peng Huang, Ashwinkumar Bhirde, Albert Jin, Ying Ma, Gang Niu, Nouri Neamati and Xiaoyuan Chen
Chemical Communications 2012 - vol. 48(Issue 78) pp:NaN9770-9770
Publication Date(Web):2012/08/08
DOI:10.1039/C2CC31974H
A nanoscale RGD–pyrene–graphene oxide (GO) biosensor was prepared for real-time in situ detection of a cancer cell surface marker, integrin αvβ3. This nanoscale GO-based biosensor is simple, robust, sensitive and of high selectivity. It can also be adapted to other cancer cell surface marker evaluation systems.