1
|
Mohammed I, Sagurthi SR. Current Approaches and Strategies Applied in First-in-class Drug Discovery. ChemMedChem 2025; 20:e202400639. [PMID: 39648151 DOI: 10.1002/cmdc.202400639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/30/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
First-in-class drug discovery (FICDD) offers novel therapies, new biological targets and mechanisms of action (MOAs) toward targeting various diseases and provides opportunities to understand unexplored biology and to target unmet diseases. Current screening approaches followed in FICDD for discovery of hit and lead molecules can be broadly categorized and discussed under phenotypic drug discovery (PDD) and target-based drug discovery (TBDD). Each category has been further classified and described with suitable examples from the literature outlining the current trends in screening approaches applied in small molecule drug discovery (SMDD). Similarly, recent applications of functional genomics, structural biology, artificial intelligence (AI), machine learning (ML), and other such advanced approaches in FICDD have also been highlighted in the article. Further, some of the current medicinal chemistry strategies applied during discovery of hits and optimization studies such as hit-to-lead (HTL) and lead optimization (LO) have been simultaneously overviewed in this article.
Collapse
Affiliation(s)
- Idrees Mohammed
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
| | - Someswar Rao Sagurthi
- Drug Design & Molecular Medicine Laboratory, Department of Genetics & Biotechnology, Osmania University, Hyderabad, 500007, Telangana, India
- Special Center for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
2
|
Dunker C, Schlegel K, Junker A. Phenol (bio)isosteres in drug design and development. Arch Pharm (Weinheim) 2025; 358:e2400700. [PMID: 39580699 PMCID: PMC11726161 DOI: 10.1002/ardp.202400700] [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: 08/30/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/26/2024]
Abstract
Due to their versatile properties, phenolic compounds are integral to various biologically active molecules, including many pharmaceuticals. However, their application in drug design is often hindered by issues such as poor oral bioavailability, rapid metabolism, and potential toxicity. This review explores the use of phenol bioisosteres-structurally similar compounds that can mimic the biological activity of phenols while potentially offering improved drug-like properties. We provide an extensive analysis of various phenol bioisosteres, including benzimidazolones, benzoxazolones, indoles, quinolinones, and pyridones, highlighting their impact on the pharmacokinetic and pharmacodynamic profiles of drugs. Case studies illustrate the successful application of these bioisosteres in enhancing metabolic stability, receptor selectivity, and overall therapeutic efficacy. Additionally, the review addresses the challenges associated with phenol bioisosterism, such as maintaining potency and avoiding undesirable side effects. By offering a detailed examination of current strategies and potential future directions, this review serves as a valuable resource for medicinal chemists seeking to optimize phenolic scaffolds in drug development. The insights provided herein aim to facilitate the design of more effective and safer therapeutic agents through strategic bioisosteric modifications.
Collapse
Affiliation(s)
- Calvin Dunker
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and RadiopharmacyUniversity of TübingenTübingenGermany
| | - Katja Schlegel
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI)University of MuensterMuensterGermany
- Werner Siemens Imaging Center, Department of Preclinical Imaging and RadiopharmacyUniversity of TübingenTübingenGermany
| |
Collapse
|
3
|
Nada H, Choi Y, Kim S, Jeong KS, Meanwell NA, Lee K. New insights into protein-protein interaction modulators in drug discovery and therapeutic advance. Signal Transduct Target Ther 2024; 9:341. [PMID: 39638817 PMCID: PMC11621763 DOI: 10.1038/s41392-024-02036-3] [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: 04/11/2024] [Revised: 09/09/2024] [Accepted: 10/23/2024] [Indexed: 12/07/2024] Open
Abstract
Protein-protein interactions (PPIs) are fundamental to cellular signaling and transduction which marks them as attractive therapeutic drug development targets. What were once considered to be undruggable targets have become increasingly feasible due to the progress that has been made over the last two decades and the rapid technological advances. This work explores the influence of technological innovations on PPI research and development. Additionally, the diverse strategies for discovering, modulating, and characterizing PPIs and their corresponding modulators are examined with the aim of presenting a streamlined pipeline for advancing PPI-targeted therapeutics. By showcasing carefully selected case studies in PPI modulator discovery and development, we aim to illustrate the efficacy of various strategies for identifying, optimizing, and overcoming challenges associated with PPI modulator design. The valuable lessons and insights gained from the identification, optimization, and approval of PPI modulators are discussed with the aim of demonstrating that PPI modulators have transitioned beyond early-stage drug discovery and now represent a prime opportunity with significant potential. The selected examples of PPI modulators encompass those developed for cancer, inflammation and immunomodulation, as well as antiviral applications. This perspective aims to establish a foundation for the effective targeting and modulation of PPIs using PPI modulators and pave the way for future drug development.
Collapse
Affiliation(s)
- Hossam Nada
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, New York, USA
| | - Yongseok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Sungdo Kim
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| | - Kwon Su Jeong
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| | - Nicholas A Meanwell
- Baruch S. Blumberg Institute, Doylestown, PA, USA
- School of Pharmacy, University of Michigan, Ann Arbor, MI, USA
- Ernest Mario School of Pharmacy, Rutgers University New Brunswick, New Brunswick, NJ, USA
| | - Kyeong Lee
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea.
| |
Collapse
|
4
|
Shahab M, Ziyu P, Waqas M, Zheng G, Bin Jardan YA, Fentahun Wondmie G, Bouhrhia M. Targeting human progesterone receptor (PR), through pharmacophore-based screening and molecular simulation revealed potent inhibitors against breast cancer. Sci Rep 2024; 14:6768. [PMID: 38514638 PMCID: PMC10958019 DOI: 10.1038/s41598-024-55321-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/22/2024] [Indexed: 03/23/2024] Open
Abstract
Breast cancer, the prevailing malignant tumor among women, is linked to progesterone and its receptor (PR) in both tumorigenesis and treatment responsiveness. Despite thorough investigation, the precise molecular mechanisms of progesterone in breast cancer remain unclear. The human progesterone receptor (PR) serves as an essential therapeutic target for breast cancer treatment, warranting the rapid design of small molecule therapeutics that can effectively inhibit HPR. By employing cutting-edge computational techniques like molecular screening, simulation, and free energy calculation, the process of identifying potential lead molecules from natural products has been significantly expedited. In this study, we employed pharmacophore-based virtual screening and molecular simulations to identify natural product-based inhibitors of human progesterone receptor (PR) in breast cancer treatment. High-throughput molecular screening of traditional Chinese medicine (TCM) and zinc databases was performed, leading to the identification of potential lead compounds. The analysis of binding modes for the top five compounds from both database provides valuable structural insights into the inhibition of HPR for breast cancer treatment. The top five hits exhibited enhanced stability and compactness compared to the reference compound. In conclusion, our study provides valuable insights for identifying and refining lead compounds as HPR inhibitors.
Collapse
Affiliation(s)
- Muhammad Shahab
- State Key Laboratories of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China
| | - Peng Ziyu
- School of chemistry and chemical engineering, Wuhan University of Science and Technology, Wuhan, 430081, People's Republic of China
| | - Muhammad Waqas
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz, 616, Nizwa, Oman
| | - Guojun Zheng
- State Key Laboratories of Chemical Resources Engineering, Beijing University of Chemical Technology, Beijing, 100029, People's Republic of China.
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P. O. BOX 2455, 11451, Riyadh, Saudi Arabia
| | | | - Mohammed Bouhrhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco
| |
Collapse
|
5
|
Ghufran M, Rehman AU, Ayaz M, Ul-Haq Z, Uddin R, Azam SS, Wadood A. New lead compounds identification against KRas mediated cancers through pharmacophore-based virtual screening and in vitro assays. J Biomol Struct Dyn 2023; 41:8053-8067. [PMID: 36184737 DOI: 10.1080/07391102.2022.2128878] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 09/20/2022] [Indexed: 10/07/2022]
Abstract
Cancer remains the leading cause of mortality and morbidity in the world, with 19.3 million new diagnoses and 10.1 million deaths in 2020. Cancer is caused due to mutations in proto-oncogenes and tumor-suppressor genes. Genetic analyses found that Ras (Rat sarcoma) is one of the most deregulated oncogenes in human cancers. The Ras oncogene family members including NRas (Neuroblastoma ras viral oncogene homolog), HRas (Harvey rat sarcoma) and KRas are involved in different types of human cancers. The mutant KRas is considered as the most frequent oncogene implicated in the development of lung, pancreatic and colon cancers. However, there is no efficient clinical drug even though it has been identified as an oncogene for 30 years. Therefore there is an emerging need to develop potent, new anticancer drugs. In this study, computer-aided drug designing approaches as well as experimental methods were employed to find new and potential anti-cancer drugs. The pharmacophore model was developed from an already known FDA approved anti-cancer drug Bortezomib using the software MOE. The validated pharmacophore model was then used to screen the in-house and commercially available databases. The pharmacophore-based virtual screening resulted in 26 and 86 hits from in-house and commercial databases respectively. Finally, 6/13 (in-house database) and 24/64 hits (commercial databases) were selected with different scaffolds having good interactions with the significant active residues of KRasG12D protein that were predicted as potent lead compounds. Finally, the results of pharmacophore-based virtual screening were further validated by molecular dynamics simulation analysis. The 6 hits of the in-house database were further evaluated experimentally. The experimental results showed that these compounds have good anti-cancer activity which validate the protocol of our in silico studies. KRasG12D protein is a very important anti-cancer target and potent inhibitors for this target are still not available, so small lead compound inhibitors were identified to inhibit the activity of this protein by blocking the GTP-binding pocket.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Mehreen Ghufran
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
- Department of Pathology, Medical Teaching Institution Bacha Khan Medical College (BKMC) Mardan, Mardan, Pakistan
| | - Ashfaq Ur Rehman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Zaheer Ul-Haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, University of Karachi, Karachi, Pakistan
| | - Reaz Uddin
- Dr. Panjwani Center for Molecular Medicine and Drug Research, University of Karachi, Karachi, Pakistan
| | - Syed Sikander Azam
- Department of Bioinformatics, Quaid-e-Azam University, Islamabad, Pakistan
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| |
Collapse
|
6
|
Barresi E, Robello M, Baglini E, Poggetti V, Viviano M, Salerno S, Da Settimo F, Taliani S. Indol-3-ylglyoxylamide as Privileged Scaffold in Medicinal Chemistry. Pharmaceuticals (Basel) 2023; 16:997. [PMID: 37513909 PMCID: PMC10386336 DOI: 10.3390/ph16070997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/05/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
In recent years, indolylglyoxylamide-based derivatives have received much attention due to their application in drug design and discovery, leading to the development of a wide array of compounds that have shown a variety of pharmacological activities. Combining the indole nucleus, already validated as a "privileged structure," with the glyoxylamide function allowed for an excellent template to be obtained that is suitable to a great number of structural modifications aimed at permitting interaction with specific molecular targets and producing desirable therapeutic effects. The present review provides insight into how medicinal chemists have elegantly exploited the indolylglyoxylamide moiety to obtain potentially useful drugs, with a particular focus on compounds exhibiting activity in in vivo models or reaching clinical trials. All in all, this information provides exciting new perspectives on existing data that can be useful in further design of indolylglyoxylamide-based molecules with interesting pharmacological profiles. The aim of this report is to present an update of collection data dealing with the employment of this moiety in the rational design of compounds that are able to interact with a specific target, referring to the last 20 years.
Collapse
Affiliation(s)
- Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Marco Robello
- Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Monica Viviano
- Department of Pharmacy, University of Salerno, 84084 Fisciano, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
7
|
Rotella DP. Successes in antiviral drug discovery: a tribute to Nick Meanwell. Med Chem Res 2023; 32:1-10. [PMID: 37362321 PMCID: PMC10249547 DOI: 10.1007/s00044-023-03086-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023]
Abstract
Drug discovery is a difficult task, and is even more challenging when the target evolves during therapy. Antiviral drug therapy is an excellent example, exemplified by the evolution of therapeutic approaches for treatment of hepatitis C and HIV-1. Nick Meanwell and his colleagues made important contributions leading to molecules for treatment of hepatitis C and HIV-1, each with distinct mechanisms of action. This review summarizes the discovery and impact of these drugs, and will highlight, where applicable, the broader contributions of these discoveries to medicinal chemistry and drug discovery.
Collapse
Affiliation(s)
- David P. Rotella
- Department of Chemistry and Biochemistry, Sokol Institute of Pharmaceutical Life Sciences, Montclair State University, Montclair, NJ 07043 USA
| |
Collapse
|
8
|
Ballesteros-Garrido R. Recent developments in the synthesis of 4-, 5-, 6- and 7-azaindoles. ADVANCES IN HETEROCYCLIC CHEMISTRY 2023. [DOI: 10.1016/bs.aihch.2023.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
9
|
Shim KH, Sharma N, An SSA. Prion therapeutics: Lessons from the past. Prion 2022; 16:265-294. [PMID: 36515657 PMCID: PMC9754114 DOI: 10.1080/19336896.2022.2153551] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 12/15/2022] Open
Abstract
Prion diseases are a group of incurable zoonotic neurodegenerative diseases (NDDs) in humans and other animals caused by the prion proteins. The abnormal folding and aggregation of the soluble cellular prion proteins (PrPC) into scrapie isoform (PrPSc) in the Central nervous system (CNS) resulted in brain damage and other neurological symptoms. Different therapeutic approaches, including stalling PrPC to PrPSc conversion, increasing PrPSc removal, and PrPC stabilization, for which a spectrum of compounds, ranging from organic compounds to antibodies, have been explored. Additionally, a non-PrP targeted drug strategy using serpin inhibitors has been discussed. Despite numerous scaffolds being screened for anti-prion activity in vitro, only a few were effective in vivo and unfortunately, almost none of them proved effective in the clinical studies, most likely due to toxicity and lack of permeability. Recently, encouraging results from a prion-protein monoclonal antibody, PRN100, were presented in the first human trial on CJD patients, which gives a hope for better future for the discovery of other new molecules to treat prion diseases. In this comprehensive review, we have re-visited the history and discussed various classes of anti-prion agents, their structure, mode of action, and toxicity. Understanding pathogenesis would be vital for developing future treatments for prion diseases. Based on the outcomes of existing therapies, new anti-prion agents could be identified/synthesized/designed with reduced toxicity and increased bioavailability, which could probably be effective in treating prion diseases.
Collapse
Affiliation(s)
- Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Niti Sharma
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam, South Korea
| |
Collapse
|
10
|
A one pot diazo installation-photochemical oxidation (blue LED-O2)/ amidation of aryl/heteroaryl acetates with cyclic 2°-amines: An eco-friendly synthesis of aromatic α-ketoamides. Tetrahedron 2022. [DOI: 10.1016/j.tet.2022.133043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
11
|
Romanelli MN, Manetti D, Braconi L, Dei S, Gabellini A, Teodori E. The piperazine scaffold for novel drug discovery efforts: the evidence to date. Expert Opin Drug Discov 2022; 17:969-984. [PMID: 35848922 DOI: 10.1080/17460441.2022.2103535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION . Piperazine is a structural element present in drugs belonging to various chemical classes and used for numerous different therapeutic applications; it has been considered a privileged scaffold for drug design. AREAS COVERED The authors have searched examples of piperazine-containing compounds among drugs recently approved by the FDA, and in some research fields (nicotinic receptor modulators, compounds acting against cancer and bacterial multi-drug resistance), looking in particular to the design behind the insertion of this moiety. EXPERT OPINION Piperazine is widely used due to its peculiar characteristics, such as solubility, basicity, chemical reactivity, and conformational properties. This moiety has represented an important tool to modulate pharmacokinetic and pharmacodynamic properties of drugs.
Collapse
Affiliation(s)
- Maria Novella Romanelli
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), University of Florence, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Dina Manetti
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), University of Florence, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Laura Braconi
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), University of Florence, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Silvia Dei
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), University of Florence, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Alessio Gabellini
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), University of Florence, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| | - Elisabetta Teodori
- Department of Neuroscience, Psychology, Drug Research and Child's Health (NEUROFARBA), University of Florence, Section of Pharmaceutical and Nutraceutical Sciences, Via Ugo Schiff 6, 50019, Sesto Fiorentino, Italy
| |
Collapse
|
12
|
The Genesis and Future Prospects of Small Molecule HIV-1 Attachment Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:45-64. [DOI: 10.1007/978-981-16-8702-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
13
|
Recent research results have converted gp120 binders to a therapeutic option for the treatment of HIV-1 infection. A medicinal chemistry point of view. Eur J Med Chem 2021; 229:114078. [PMID: 34992041 DOI: 10.1016/j.ejmech.2021.114078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/16/2021] [Accepted: 12/22/2021] [Indexed: 11/21/2022]
Abstract
Current therapeutic armamentarium for treatment of HIV-1 infection is based on the use of highly active antiretroviral therapy that, unfortunately, does not act as a curative remedy. Moreover, duration of the therapy often results in lack of compliance with the consequent emergence of multidrug resistance. Finally, drug toxicity issues also arise during treatments. In the attempt to achieve a curative effect, in addition to invest substantial resources in finding new anti-HIV-1 agents and in optimizing antiviral lead compounds and drugs currently available, additional efforts should be done to deplete viral reservoir located within host CD4+ T cells. Gp120 binders represent a class of compounds able to affect the interactions between viral envelope proteins and host CD4, thus avoiding virus-to-cell attachment and fusion, and the consequent viral entry into host cells. This review summarizes the efforts done in the last five years to design new gp120 binders, that finally culminated in the approval of fostemsavir as an anti-HIV-1 drug.
Collapse
|
14
|
Schwärzer K, Rout SK, Bessinger D, Lima F, Brocklehurst CE, Karaghiosoff K, Bein T, Knochel P. Selective functionalization of the 1 H-imidazo[1,2- b]pyrazole scaffold. A new potential non-classical isostere of indole and a precursor of push-pull dyes. Chem Sci 2021; 12:12993-13000. [PMID: 34745530 PMCID: PMC8513920 DOI: 10.1039/d1sc04155j] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/28/2021] [Indexed: 12/28/2022] Open
Abstract
We report the selective functionalization of the 1H-imidazo[1,2-b]pyrazole scaffold using a Br/Mg-exchange, as well as regioselective magnesiations and zincations with TMP-bases (TMP = 2,2,6,6-tetramethylpiperidyl), followed by trapping reactions with various electrophiles. In addition, we report a fragmentation of the pyrazole ring, giving access to push-pull dyes with a proaromatic (1,3-dihydro-2H-imidazol-2-ylidene)malononitrile core. These functionalization methods were used in the synthesis of an isostere of the indolyl drug pruvanserin. Comparative assays between the original drug and the isostere showed that a substitution of the indole ring with a 1H-imidazo[1,2-b]pyrazole results in a significantly improved solubility in aqueous media.
Collapse
Affiliation(s)
- Kuno Schwärzer
- Department Chemie, Ludwig-Maximilians-Universität München Munich 81377 Germany
| | - Saroj K Rout
- Department Chemie, Ludwig-Maximilians-Universität München Munich 81377 Germany
| | - Derya Bessinger
- Department Chemie, Ludwig-Maximilians-Universität München Munich 81377 Germany
| | - Fabio Lima
- Global Discovery Chemistry, Novartis Institutes of BioMedical Research Basel 4057 Switzerland
| | - Cara E Brocklehurst
- Global Discovery Chemistry, Novartis Institutes of BioMedical Research Basel 4057 Switzerland
| | | | - Thomas Bein
- Department Chemie, Ludwig-Maximilians-Universität München Munich 81377 Germany
| | - Paul Knochel
- Department Chemie, Ludwig-Maximilians-Universität München Munich 81377 Germany
| |
Collapse
|
15
|
Wang T, Kadow JF, Meanwell NA. Innovation in the discovery of the HIV-1 attachment inhibitor temsavir and its phosphonooxymethyl prodrug fostemsavir. Med Chem Res 2021; 30:1955-1980. [PMID: 34602806 PMCID: PMC8476988 DOI: 10.1007/s00044-021-02787-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/19/2021] [Indexed: 11/25/2022]
Abstract
The discovery and development of fostemsavir (2), the tromethamine salt of the phosphonooxymethyl prodrug of temsavir (1), encountered significant challenges at many points in the preclinical and clinical development program that, in many cases, stimulated the implementation of innovative solutions in order to enable further progression. In the preclinical program, a range of novel chemistry methodologies were developed during the course of the discovery effort that enabled a thorough examination and definition of the HIV-1 attachment inhibitor (AI) pharmacophore. These discoveries helped to address the challenges associated with realizing a molecule with all of the properties necessary to successfully advance through development and this aspect of the program is the major focus of this retrospective. Although challenges and innovation are not unusual in drug discovery and development programs, the HIV-1 AI program is noteworthy not only because of the serial nature of the challenges encountered along the development path, but also because it resulted in a compound that remains the first and only example of a mechanistically novel class of HIV-1 inhibitor that is proving to be very beneficial for controlling virus levels in highly treatment-experienced HIV-1 infected patients. ![]()
Collapse
Affiliation(s)
- Tao Wang
- Beijing Kawin Technology Share-Holdiing Co., 6 Rongjing East Street, BDA, Beijing, PR China
| | - John F Kadow
- ViiV Healthcare, 36 East Industrial Road, Branford, CT 06405 USA
| | - Nicholas A Meanwell
- Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ 08543-4000 USA
| |
Collapse
|
16
|
Pyrroles as Privileged Scaffolds in the Search for New Potential HIV Inhibitors. Pharmaceuticals (Basel) 2021; 14:ph14090893. [PMID: 34577593 PMCID: PMC8468532 DOI: 10.3390/ph14090893] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 02/04/2023] Open
Abstract
Acquired immunodeficiency syndrome (AIDS) is caused by human immunodeficiency virus (HIV) and remains a global health problem four decades after the report of its first case. Despite success in viral load suppression and the increase in patient survival due to combined antiretroviral therapy (cART), the development of new drugs has become imperative due to strains that have become resistant to antiretrovirals. In this context, there has been a continuous search for new anti-HIV agents based on several chemical scaffolds, including nitrogenated heterocyclic pyrrole rings, which have been included in several compounds with antiretroviral activity. Thus, this review aims to describe pyrrole-based compounds with anti-HIV activity as a new potential treatment against AIDS, covering the period between 2015 and 2020. Our research allowed us to conclude that pyrrole derivatives are still worth exploring, as they may provide highly active compounds targeting different steps of the HIV-1 replication cycle and act with an innovative mechanism.
Collapse
|
17
|
Ding L, Pannecouque C, De Clercq E, Zhuang C, Chen FE. Improving Druggability of Novel Diarylpyrimidine NNRTIs by a Fragment-Based Replacement Strategy: From Biphenyl-DAPYs to Heteroaromatic-Biphenyl-DAPYs. J Med Chem 2021; 64:10297-10311. [PMID: 34197708 DOI: 10.1021/acs.jmedchem.1c00708] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A series of novel heteroaromatic-difluoro-biphenyl-diarylpyrimidines were designed as non-nucleoside anti-HIV inhibitors targeting reverse transcriptase by a fragment-based replacement strategy with the purpose of improving the druggability. Hopping five- or six-membered heterocycle groups on the biphenyl moiety as bioisosterism for intrinsically cyanophenyl gave 23 derivatives. All of these compounds possessed excellent HIV-1 inhibitory activity in the nanomolar range. Among them, 12g with a 4-pyridine group displayed excellent inhibitory activity toward WT and mutant HIV virus possessing significant selectivity. Moreover, this compound exhibited a decent improvement in druggability than etravirine and rilpivirine: (1) The hydrochloric acid salt of 12g exhibited significantly improved water solubility in different pH conditions. (2) 12g did not show apparent CYP enzymatic inhibitory activity or acute toxicity. (3) Excellent oral bioavailability was also revealed (F = 126%, rats) in 12g. Collectively, these novel heteroaromatic-biphenyl-DAPYs represent promising drug candidates for HIV clinical therapy.
Collapse
Affiliation(s)
- Li Ding
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai 200433, China.,Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, China
| | | | - Erik De Clercq
- Rega Institute for Medical Research, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Chunlin Zhuang
- Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai 200433, China.,Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, China
| | - Fen-Er Chen
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,Engineering Center of Catalysis and Synthesis for Chiral Molecules, Department of Chemistry, Fudan University, Shanghai 200433, China.,Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai 200433, China.,Institute of Pharmaceutical Science and Technology, Zhejiang University of Technology, 18 Chao Wang Road, Hangzhou 310014, China
| |
Collapse
|
18
|
Kumar D, Sharma P, Shabu, Kaur R, Lobe MMM, Gupta GK, Ntie-Kang F. In search of therapeutic candidates for HIV/AIDS: rational approaches, design strategies, structure-activity relationship and mechanistic insights. RSC Adv 2021; 11:17936-17964. [PMID: 35480193 PMCID: PMC9033207 DOI: 10.1039/d0ra10655k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 04/19/2021] [Indexed: 12/23/2022] Open
Abstract
The HIV/AIDS pandemic is a serious threat to the health and development of mankind, which has affected about 37.9 million people worldwide. The increasing negative health, economic and social impacts of this disease have led to the search for new therapeutic candidates for the mitigation of AIDS/HIV. However, to date, there is still no treatment that can cure this disease. Furthermore, the clinically available drugs have numerous severe side effects. Hence, the synthesis of novel agents from natural leads is one of the rational approaches to obtain new drugs in modern medicinal chemistry. This review article is an effort to summarize recent developments with regards to the discovery of novel analogs with promising biological potential against HIV/AIDS. Herein, we also aim to discuss prospective directions on the progress of more credible and specific analogues. Besides presenting design strategies, the present communication also highlights the structure-activity relationship together with the structural features of the most promising molecules, their IC50 values, mechanistic insights and some interesting key findings revealed during their biological evaluation. The interactions with the amino acid residues of the enzymes responsible for HIV-1 inhibition are also discussed. This collection will be of great interest for researchers working in this area.
Collapse
Affiliation(s)
- Dinesh Kumar
- Sri Sai College of Pharmacy Manawala Amritsar-143001 Punjab India +91-9988902489
| | - Pooja Sharma
- Sri Sai College of Pharmacy Manawala Amritsar-143001 Punjab India +91-9988902489
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala India
| | - Shabu
- Indian Institute of Integrative Medicine (CSIR-IIIM) Canal Road Jammu 180001 India
| | - Ramandeep Kaur
- Sri Sai College of Pharmacy Manawala Amritsar-143001 Punjab India +91-9988902489
| | - Maloba M M Lobe
- Department of Chemistry, Faculty of Science, University of Buea P. O. Box 63 Buea Cameroon +237 685625811
| | - Girish K Gupta
- Department of Pharmaceutical Chemistry, Sri Sai College of Pharmacy Badhani Pathankot-145001 Punjab India
| | - Fidele Ntie-Kang
- Department of Chemistry, Faculty of Science, University of Buea P. O. Box 63 Buea Cameroon +237 685625811
- Institute for Pharmacy, Martin-Luther-Universität Halle-Wittenberg Kurt-Mothes-Str. 3 06120 Halle (Saale) Germany +49 3455525043
- Institute of Botany, Technical University of Dresden Zellescher Weg 20b 01062 Dresden Germany
| |
Collapse
|
19
|
Petit E, Bosch L, Costa AM, Rodríguez-Izquierdo I, Sepúlveda-Crespo D, Muñoz-Fernández MA, Vilarrasa J. BMS Derivatives C7-Linked to β-Cyclodextrin and Hyperbranched Polyglycerol Retain Activity against R5-HIV-1 NLAD8 Isolates and Can Be Deemed Potential Microbicides. ChemMedChem 2021; 16:2217-2222. [PMID: 33843142 DOI: 10.1002/cmdc.202100080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 04/02/2021] [Indexed: 12/30/2022]
Abstract
Amides from indole-3-glyoxylic acid and 4-benzoyl-2-methylpiperazine, which are related to entry inhibitors developed by Bristol-Myers Squibb (BMS), have been synthesized with aliphatic chains located at the C7 position of the indole ring. These spacers contain an azido group suitable for the well-known Cu(I)-catalyzed (3+2)-cycloaddition or an activated triple bond for the nucleophilic addition of thiols under physiological conditions. Reaction with polyols (β-cyclodextrin and hyperbranched polyglycerol) decorated with complementary click partners has afforded polyol-BMS-like conjugates that are not cytotoxic (TZM.bl cells) and retain the activity against R5-HIV-1NLAD8 isolates. Thus, potential vaginal microbicides based on entry inhibitors, which can be called of 4th generation, are reported here for the first time.
Collapse
Affiliation(s)
- Elena Petit
- Organic Chemistry Section, Facultat de Química, Universitat de Barcelona, Diagonal 645, 08028, Barcelona, Catalonia, Spain
| | - Lluís Bosch
- Organic Chemistry Section, Facultat de Química, Universitat de Barcelona, Diagonal 645, 08028, Barcelona, Catalonia, Spain
| | - Anna M Costa
- Organic Chemistry Section, Facultat de Química, Universitat de Barcelona, Diagonal 645, 08028, Barcelona, Catalonia, Spain
| | - Ignacio Rodríguez-Izquierdo
- Laboratorio de Inmunobiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Dr. Esquerdo 46, 28007, Madrid, Spain
| | - Daniel Sepúlveda-Crespo
- Laboratorio de Inmunobiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Dr. Esquerdo 46, 28007, Madrid, Spain
| | - M Angeles Muñoz-Fernández
- Laboratorio de Inmunobiología Molecular, Hospital General Universitario Gregorio Marañón (HGUGM), Dr. Esquerdo 46, 28007, Madrid, Spain
| | - Jaume Vilarrasa
- Organic Chemistry Section, Facultat de Química, Universitat de Barcelona, Diagonal 645, 08028, Barcelona, Catalonia, Spain
| |
Collapse
|
20
|
Robello M, Barresi E, Baglini E, Salerno S, Taliani S, Settimo FD. The Alpha Keto Amide Moiety as a Privileged Motif in Medicinal Chemistry: Current Insights and Emerging Opportunities. J Med Chem 2021; 64:3508-3545. [PMID: 33764065 PMCID: PMC8154582 DOI: 10.1021/acs.jmedchem.0c01808] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the years, researchers in drug discovery have taken advantage of the use of privileged structures to design innovative hit/lead molecules. The α-ketoamide motif is found in many natural products, and it has been widely exploited by medicinal chemists to develop compounds tailored to a vast range of biological targets, thus presenting clinical potential for a plethora of pathological conditions. The purpose of this perspective is to provide insights into the versatility of this chemical moiety as a privileged structure in drug discovery. After a brief analysis of its physical-chemical features and synthetic procedures to obtain it, α-ketoamide-based classes of compounds are reported according to the application of this motif as either a nonreactive or reactive moiety. The goal is to highlight those aspects that may be useful to understanding the perspectives of employing the α-ketoamide moiety in the rational design of compounds able to interact with a specific target.
Collapse
Affiliation(s)
- Marco Robello
- Synthetic Bioactive Molecules Section, LBC, NIDDK, NIH, 8 Center Drive, Room 404, Bethesda, Maryland 20892, United States
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Emma Baglini
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| |
Collapse
|
21
|
Mahindra A, Janha O, Mapesa K, Sanchez-Azqueta A, Alam MM, Amambua-Ngwa A, Nwakanma DC, Tobin AB, Jamieson AG. Development of Potent PfCLK3 Inhibitors Based on TCMDC-135051 as a New Class of Antimalarials. J Med Chem 2020; 63:9300-9315. [PMID: 32787140 PMCID: PMC7497403 DOI: 10.1021/acs.jmedchem.0c00451] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Indexed: 12/20/2022]
Abstract
The protein kinase PfCLK3 plays a critical role in the regulation of malarial parasite RNA splicing and is essential for the survival of blood stage Plasmodium falciparum. We recently validated PfCLK3 as a drug target in malaria that offers prophylactic, transmission blocking, and curative potential. Herein, we describe the synthesis of our initial hit TCMDC-135051 (1) and efforts to establish a structure-activity relationship with a 7-azaindole-based series. A total of 14 analogues were assessed in a time-resolved fluorescence energy transfer assay against the full-length recombinant protein kinase PfCLK3, and 11 analogues were further assessed in asexual 3D7 (chloroquine-sensitive) strains of P. falciparum parasites. SAR relating to rings A and B was established. These data together with analysis of activity against parasites collected from patients in the field suggest that TCMDC-135051 (1) is a promising lead compound for the development of new antimalarials with a novel mechanism of action targeting PfCLK3.
Collapse
Affiliation(s)
- Amit Mahindra
- School
of Chemistry, University of Glasgow, Joseph Black Building, University
Avenue, Glasgow G12 8QQ, U.K.
| | - Omar Janha
- Centre
for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow G12 8QQ, U.K.
| | - Kopano Mapesa
- School
of Chemistry, University of Glasgow, Joseph Black Building, University
Avenue, Glasgow G12 8QQ, U.K.
| | - Ana Sanchez-Azqueta
- Centre
for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow G12 8QQ, U.K.
| | - Mahmood M. Alam
- Wellcome
Centre for Integrative Parasitology and Centre for Translational Pharmacology,
Institute of Infection Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, U.K.
| | - Alfred Amambua-Ngwa
- MRC
Unit The Gambia at LSHTM, Atlantic Boulevard,
Fajara, P. O. Box 273, Banjul, The Gambia
| | - Davis C. Nwakanma
- MRC
Unit The Gambia at LSHTM, Atlantic Boulevard,
Fajara, P. O. Box 273, Banjul, The Gambia
| | - Andrew B. Tobin
- Centre
for Translational Pharmacology, Institute of Molecular Cell and Systems Biology, University of Glasgow, Davidson Building, Glasgow G12 8QQ, U.K.
| | - Andrew G. Jamieson
- School
of Chemistry, University of Glasgow, Joseph Black Building, University
Avenue, Glasgow G12 8QQ, U.K.
| |
Collapse
|
22
|
Curreli F, Ahmed S, Benedict Victor SM, Iusupov IR, Belov DS, Markov PO, Kurkin AV, Altieri A, Debnath AK. Preclinical Optimization of gp120 Entry Antagonists as anti-HIV-1 Agents with Improved Cytotoxicity and ADME Properties through Rational Design, Synthesis, and Antiviral Evaluation. J Med Chem 2020; 63:1724-1749. [PMID: 32031803 DOI: 10.1021/acs.jmedchem.9b02149] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
We previously reported a milestone in the optimization of NBD-11021, an HIV-1 gp120 antagonist, by developing a new and novel analogue, NBD-14189 (Ref1), which showed antiviral activity against HIV-1HXB2, with a half maximal inhibitory concentration of 89 nM. However, cytotoxicity remained high, and the absorption, distribution, metabolism, and excretion (ADME) data showed relatively poor aqueous solubility. To optimize these properties, we replaced the phenyl ring in the compound with a pyridine ring and synthesized a set of 48 novel compounds. One of the new analogues, NBD-14270 (8), showed a marked improvement in cytotoxicity, with 3-fold and 58-fold improvements in selectivity index value compared with that of Ref1 and NBD-11021, respectively. Furthermore, the in vitro ADME data clearly showed improvements in aqueous solubility and other properties compared with those for Ref1. The data for 8 indicated that the pyridine scaffold is a good bioisostere for phenyl, allowing the further optimization of this molecule.
Collapse
Affiliation(s)
- Francesca Curreli
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute , New York Blood Center , 310 E 67th Street , New York 10065 , New York , United States
| | - Shahad Ahmed
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute , New York Blood Center , 310 E 67th Street , New York 10065 , New York , United States
| | - Sofia M Benedict Victor
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute , New York Blood Center , 310 E 67th Street , New York 10065 , New York , United States
| | - Ildar R Iusupov
- EDASA Scientific, Scientific Park , Moscow State University , Leninskie Gory Bld. 75, 77-101b , Moscow 119992 , Russia
| | - Dmitry S Belov
- EDASA Scientific, Scientific Park , Moscow State University , Leninskie Gory Bld. 75, 77-101b , Moscow 119992 , Russia
| | - Pavel O Markov
- EDASA Scientific, Scientific Park , Moscow State University , Leninskie Gory Bld. 75, 77-101b , Moscow 119992 , Russia
| | - Alexander V Kurkin
- EDASA Scientific, Scientific Park , Moscow State University , Leninskie Gory Bld. 75, 77-101b , Moscow 119992 , Russia
| | - Andrea Altieri
- EDASA Scientific, Scientific Park , Moscow State University , Leninskie Gory Bld. 75, 77-101b , Moscow 119992 , Russia
| | - Asim K Debnath
- Laboratory of Molecular Modeling & Drug Design, Lindsley F. Kimball Research Institute , New York Blood Center , 310 E 67th Street , New York 10065 , New York , United States
| |
Collapse
|
23
|
Lin L, Chen X, Zhao J, Lin S, Ma G, Liao X, Feng P. Facile One-pot Protocol of Derivatization Nitropyridines: Access to 3-Acetamidopyridin-2-yl 4-methylbenzenesulfonate Derivatives. HETEROCYCL COMMUN 2019. [DOI: 10.1515/hc-2019-0017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
AbstractThis paper discloses an efficient one-pot protocol to convert easily accessible 3-nitropyridines to 3-acetamidopyridin-2-yl 4-methylbenzenesulfonate derivatives which are core structures of many pharmaceutical molecules. The strategy successfully combined a three-step reaction in one pot via progressively adding different reactants at rt. The reaction displays good functional group tolerance and regioselectivity. Structurally diversified 3-nitropyridine could be time-efficiently (3.5 h) derivatized to various functional 2-O,3-N-pyridines which are apt for further elaborations. The transformation was amenable to gram-scale synthesis.
Collapse
Affiliation(s)
- Ling Lin
- Department of Chemistry, Jinan University, Guangzhou, 510632China
| | - Xiaoguang Chen
- Department of Chemistry, Jinan University, Guangzhou, 510632China
| | - Junhao Zhao
- Department of Chemistry, Jinan University, Guangzhou, 510632China
| | - Suitao Lin
- Department of Chemistry, Jinan University, Guangzhou, 510632China
| | - Guojian Ma
- Department of Chemistry, Jinan University, Guangzhou, 510632China
| | - Xiaojian Liao
- Department of Chemistry, Jinan University, Guangzhou, 510632China
| | - Pengju Feng
- Department of Chemistry, Jinan University, Guangzhou, 510632China
| |
Collapse
|
24
|
Vangala R, Sivan SK, Peddi SR, Manga V. Computational design, synthesis and evaluation of new sulphonamide derivatives targeting HIV-1 gp120. J Comput Aided Mol Des 2019; 34:39-54. [PMID: 31792886 DOI: 10.1007/s10822-019-00258-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/19/2019] [Indexed: 11/29/2022]
Abstract
Attachment of envelope glycoprotein gp120 to the host cell receptor CD4 is the first step during the human immunodeficiency virus-1 (HIV-1) entry into the host cells that makes it a promising target for drug design. To elucidate the crucial three dimensional (3D) structural features of reported HIV-1 gp120 CD4 binding inhibitors, 3D pharmacophores were generated and receptor based approach was employed to quantify these structural features. A four-partial least square factor model with good statistics and predictive ability was generated for the dataset of 100 molecules. To further ascertain the structural requirement for gp120-CD4 binding inhibition, molecular interaction studies of inhibitors with gp120 was carried out by performing molecular docking using Glide 5.6. Based on these studies, structural requirements were drawn and new molecules were designed accordingly to yield new sulphonamides derivatives. A water based green synthetic approach was adopted to obtain these compounds which were evaluated for their HIV-1 gp120 CD4 binding inhibition. The newly synthesized compounds exhibited remarkable activity (10-fold increase) when compared with the standard BMS 806. Further the stability of newly synthesized derivatives with HIV-1 gp120 was also investigated through molecular dynamics simulation studies. This provides a proof of concept for molecular modeling based design of new inhibitors for inhibition of HIV-1 gp120 CD4 interaction.
Collapse
Affiliation(s)
- Radhika Vangala
- Molecular Modeling and Medicinal Chemistry Group, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, 500 007, India
| | - Sree Kanth Sivan
- Department of Chemistry, Nizam College, Osmania University, Hyderabad, 500 001, India
| | - Saikiran Reddy Peddi
- Molecular Modeling and Medicinal Chemistry Group, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, 500 007, India
| | - Vijjulatha Manga
- Molecular Modeling and Medicinal Chemistry Group, Department of Chemistry, University College of Science, Osmania University, Hyderabad, Telangana, 500 007, India.
| |
Collapse
|
25
|
Huang YM, Alharbi NS, Sun B, Shantharam CS, Rakesh KP, Qin HL. Synthetic routes and structure-activity relationships (SAR) of anti-HIV agents: A key review. Eur J Med Chem 2019; 181:111566. [PMID: 31401538 DOI: 10.1016/j.ejmech.2019.111566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 01/05/2023]
Abstract
The worldwide increase of AIDS, an epidemic infection in constant development has an essential and still requires potent antiretroviral chemotherapeutic agents for reducing the integer of deaths caused by HIV. Thus, there is an urgent need for new anti-HIV drug candidates with increased strength, new targets, superior pharmacokinetic properties, and compact side effects. From this viewpoint, we first review present strategies of anti-HIV drug innovation and the synthesis of heterocyclic or natural compound as anti-HIV agents for facilitating the development of more influential and successful anti-HIV agents.
Collapse
Affiliation(s)
- Yu-Mei Huang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR China
| | - Njud S Alharbi
- Biotechnology Research Group, Deportment of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bing Sun
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR China.
| | - C S Shantharam
- Department of Chemistry, Pooja Bhagavath Memorial Mahajana Education Centre, Mysuru, 570016, Karnataka, India
| | - K P Rakesh
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR China.
| | - Hua-Li Qin
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR China.
| |
Collapse
|
26
|
A survey of core replacements in indole-based HIV-1 attachment inhibitors. Bioorg Med Chem Lett 2019; 29:1423-1429. [DOI: 10.1016/j.bmcl.2019.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 01/31/2019] [Accepted: 03/13/2019] [Indexed: 11/18/2022]
|
27
|
Meuser ME, Rashad AA, Ozorowski G, Dick A, Ward AB, Cocklin S. Field-Based Affinity Optimization of a Novel Azabicyclohexane Scaffold HIV-1 Entry Inhibitor. Molecules 2019; 24:molecules24081581. [PMID: 31013646 PMCID: PMC6514670 DOI: 10.3390/molecules24081581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 12/22/2022] Open
Abstract
Small-molecule HIV-1 entry inhibitors are an extremely attractive therapeutic modality. We have previously demonstrated that the entry inhibitor class can be optimized by using computational means to identify and extend the chemotypes available. Here we demonstrate unique and differential effects of previously published antiviral compounds on the gross structure of the HIV-1 Env complex, with an azabicyclohexane scaffolded inhibitor having a positive effect on glycoprotein thermostability. We demonstrate that modification of the methyltriazole-azaindole headgroup of these entry inhibitors directly effects the potency of the compounds, and substitution of the methyltriazole with an amine-oxadiazole increases the affinity of the compound 1000-fold over parental by improving the on-rate kinetic parameter. These findings support the continuing exploration of compounds that shift the conformational equilibrium of HIV-1 Env as a novel strategy to improve future inhibitor and vaccine design efforts.
Collapse
Affiliation(s)
- Megan E Meuser
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Adel A Rashad
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Gabriel Ozorowski
- Department of Integrative Structural and Computational Biology, Collaboration for AIDS Vaccine Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, PA 19102, USA.
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Collaboration for AIDS Vaccine Discovery, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Simon Cocklin
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, PA 19102, USA.
| |
Collapse
|
28
|
Zhou P, Chen G, Gao M, Wu J. Design, synthesis and evaluation of the osimertinib analogue (C-005) as potent EGFR inhibitor against NSCLC. Bioorg Med Chem 2018; 26:6135-6145. [PMID: 30442506 DOI: 10.1016/j.bmc.2018.10.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/08/2018] [Accepted: 10/19/2018] [Indexed: 01/26/2023]
Abstract
Osimertinib has been approved as a first-line treatment for non-small-cell lung cancer (NSCLC) patients whose tumor carries EGFR activation and / or resistant mutations. To mitigate Osimertinib's toxicity caused by AZ5104, the N-demethylation metabolite of Osimertinib, we designed and synthesized a series of Osimertinib analogs with different headpieces. In vitro and in vivo analysis rendered a potential clinical candidate C-005 which had pyrrolo-pyridine headpiece. Biochemically, C-005 and its main human hepatocyte metabolite showed over 30 fold selectivity of L858R/T790M mutant EGFR over WT EGFR. Such selectivity profile was retained at cellular level. In general, C-005 is 2-14 fold more selective than Osimertinib in a panel of WT EGFR cancer cell lines. Furthermore, C-005 demonstrated robust antitumor efficacy and good tolerability in NCI-H1975, PC-9 and HCC827 xenograft mouse models, making it a potential candidate for human test in clinical.
Collapse
Affiliation(s)
- Ping Zhou
- Wuxi Shuangliang Biotechnology Co., Ltd., Jiangyin, Jiangsu Province 214437, People's Republic of China
| | - Gang Chen
- Nanjing Galaxy Biological Technology Co., Ltd., Nanjing, Jiangsu 210032, People's Republic of China
| | - Minqi Gao
- Wuxi Biortus Biosciences Co., Ltd., Jiangyin, Jiangsu Province 214437, People's Republic of China
| | - Jiaquan Wu
- Wuxi Shuangliang Biotechnology Co., Ltd., Jiangyin, Jiangsu Province 214437, People's Republic of China; Wuxi Biortus Biosciences Co., Ltd., Jiangyin, Jiangsu Province 214437, People's Republic of China.
| |
Collapse
|
29
|
Wang T, Ueda Y, Zhang Z, Yin Z, Matiskella J, Pearce BC, Yang Z, Zheng M, Parker DD, Yamanaka GA, Gong YF, Ho HT, Colonno RJ, Langley DR, Lin PF, Meanwell NA, Kadow JF. Discovery of the Human Immunodeficiency Virus Type 1 (HIV-1) Attachment Inhibitor Temsavir and Its Phosphonooxymethyl Prodrug Fostemsavir. J Med Chem 2018; 61:6308-6327. [PMID: 29920093 DOI: 10.1021/acs.jmedchem.8b00759] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The optimization of the 4-methoxy-6-azaindole series of HIV-1 attachment inhibitors (AIs) that originated with 1 to deliver temsavir (3, BMS-626529) is described. The most beneficial increases in potency and pharmacokinetic (PK) properties were attained by incorporating N-linked, sp2-hybridized heteroaryl rings at the 7-position of the heterocyclic nucleus. Compounds that adhered to a coplanarity model afforded targeted antiviral potency, leading to the identification of 3 with characteristics that provided for targeted exposure and PK properties in three preclinical species. However, the physical properties of 3 limited plasma exposure at higher doses, both in preclinical studies and in clinical trials as the result of dissolution- and/or solubility-limited absorption, a deficiency addressed by the preparation of the phosphonooxymethyl prodrug 4 (BMS-663068, fostemsavir). An extended-release formulation of 4 is currently in phase III clinical trials where it has shown promise as part of a drug combination therapy in highly treatment-experienced HIV-1 infected patients.
Collapse
|
30
|
Murafuji T, F. M. Hafizur Rahman A, Yamashita K, Narita M, Ishiguro K, Kamijo S, Miyakawa I, Mikata Y. Synthesis and Antifungal Activities of Pyridine Bioisosteres of a Bismuth Heterocycle Derived from Diphenyl Sulfone. HETEROCYCLES 2018. [DOI: 10.3987/com-18-13876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Meanwell NA, Krystal MR, Nowicka-Sans B, Langley DR, Conlon DA, Eastgate MD, Grasela DM, Timmins P, Wang T, Kadow JF. Inhibitors of HIV-1 Attachment: The Discovery and Development of Temsavir and its Prodrug Fostemsavir. J Med Chem 2017; 61:62-80. [PMID: 29271653 DOI: 10.1021/acs.jmedchem.7b01337] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human immunodeficiency virus-1 (HIV-1) infection currently requires lifelong therapy with drugs that are used in combination to control viremia. The indole-3-glyoxamide 6 was discovered as an inhibitor of HIV-1 infectivity using a phenotypic screen and derivatives of this compound were found to interfere with the HIV-1 entry process by stabilizing a conformation of the virus gp120 protein not recognized by the host cell CD4 receptor. An extensive optimization program led to the identification of temsavir (31), which exhibited an improved antiviral and pharmacokinetic profile compared to 6 and was explored in phase 3 clinical trials as the phosphonooxymethyl derivative fostemsavir (35), a prodrug designed to address dissolution- and solubility-limited absorption issues. In this drug annotation, we summarize the structure-activity and structure-liability studies leading to the discovery of 31 and the clinical studies conducted with 35 that entailed the development of an extended release formulation suitable for phase 3 clinical trials.
Collapse
Affiliation(s)
| | | | | | | | - David A Conlon
- Chemical and Synthetic Development, Bristol-Myers Squibb Research and Development , 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Martin D Eastgate
- Chemical and Synthetic Development, Bristol-Myers Squibb Research and Development , 1 Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Dennis M Grasela
- Innovative Medicines Development, Bristol-Myers Squibb Research and Development , PO Box 4000, Princeton, New Jersey 08543-4000, United States
| | - Peter Timmins
- Drug Product Science and Technology, Bristol-Myers Squibb , Reeds Lane, Moreton, Merseyside CH46 1QW, United Kingdom
| | | | | |
Collapse
|
32
|
Wang D, Feng H, Li L, Liu Z, Yan Z, Yu P. Access to 8-Azachromones via Activation of C–H in N-Oxides. J Org Chem 2017; 82:11275-11287. [DOI: 10.1021/acs.joc.7b02063] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Dong Wang
- China
International Science and Technology Cooperation Base of Food Nutrition/Safety
and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hairong Feng
- China
International Science and Technology Cooperation Base of Food Nutrition/Safety
and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Linna Li
- China
International Science and Technology Cooperation Base of Food Nutrition/Safety
and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhenlin Liu
- China
International Science and Technology Cooperation Base of Food Nutrition/Safety
and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhongli Yan
- The
Research Centre of Modern Analysis Technology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Peng Yu
- China
International Science and Technology Cooperation Base of Food Nutrition/Safety
and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
33
|
Pancera M, Lai YT, Bylund T, Druz A, Narpala S, O’Dell S, Schön A, Bailer RT, Chuang GY, Geng H, Louder MK, Rawi R, Soumana DI, Finzi A, Herschhorn A, Madani N, Sodroski J, Freire E, Langley DR, Mascola JR, McDermott AB, Kwong PD. Crystal structures of trimeric HIV envelope with entry inhibitors BMS-378806 and BMS-626529. Nat Chem Biol 2017; 13:1115-1122. [PMID: 28825711 PMCID: PMC5676566 DOI: 10.1038/nchembio.2460] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 07/19/2017] [Indexed: 01/27/2023]
Abstract
The HIV-1 envelope (Env) spike is a conformational machine that transitions between prefusion (closed, CD4- and CCR5-bound) and postfusion states to facilitate HIV-1 entry into cells. Although the prefusion closed conformation is a potential target for inhibition, development of small-molecule leads has been stymied by difficulties in obtaining structural information. Here, we report crystal structures at 3.8-Å resolution of an HIV-1-Env trimer with BMS-378806 and a derivative BMS-626529 for which a prodrug version is currently in Phase III clinical trials. Both lead candidates recognized an induced binding pocket that was mostly excluded from solvent and comprised of Env elements from a conserved helix and the β20-21 hairpin. In both structures, the β20-21 region assumed a conformation distinct from prefusion-closed and CD4-bound states. Together with biophysical and antigenicity characterizations, the structures illuminate the allosteric and competitive mechanisms by which these small-molecule leads inhibit CD4-induced structural changes in Env.
Collapse
Affiliation(s)
- Marie Pancera
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Yen-Ting Lai
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Tatsiana Bylund
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Aliaksandr Druz
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Sijy O’Dell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Arne Schön
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - Robert T. Bailer
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Gwo-Yu Chuang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Hui Geng
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Mark K. Louder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Reda Rawi
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Djade I. Soumana
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Andrés Finzi
- Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Alon Herschhorn
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Navid Madani
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joseph Sodroski
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ernesto Freire
- Department of Biology, Johns Hopkins University, Baltimore, Maryland
| | - David R. Langley
- Computer Assisted Drug Design, Bristol-Myers Squibb, Research and Development, Wallingford, Connecticut
| | - John R. Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Adrian B. McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Peter D. Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
34
|
Purificação SI, Pires MJD, Rippel R, Santos AS, Marques MMB. One-Pot Synthesis of 1,2-Disubstituted 4-, 5-, 6-, and 7-Azaindoles from Amino-o-halopyridines via N-Arylation/Sonogashira/Cyclization Reaction. Org Lett 2017; 19:5118-5121. [DOI: 10.1021/acs.orglett.7b02403] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Sara I. Purificação
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Marina J. D. Pires
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Rafael Rippel
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - A. Sofia Santos
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - M. Manuel B. Marques
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| |
Collapse
|
35
|
Affiliation(s)
- Ming Yan
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Phil S. Baran
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
36
|
Fox RJ, Cohen B, La Cruz TE, Simpson JH, Freitag A, Saurer E, Tripp JC, Chen CK, Beutner GL, Rosso VW, Borgeson E, Glace AW, Eastgate MD, Schild RL, Sweeney JT, Conlon DA. Preparation of the HIV Attachment Inhibitor BMS-663068. Part 8. Installation of the Phosphonoxymethyl Prodrug Moiety. Org Process Res Dev 2017. [DOI: 10.1021/acs.oprd.7b00135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Richard J. Fox
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Benjamin Cohen
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Thomas E. La Cruz
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - James H. Simpson
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Adam Freitag
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Eric Saurer
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Jonathan C. Tripp
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Chien-Kuang Chen
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Gregory L. Beutner
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Victor W. Rosso
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Elizabeth Borgeson
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Andrew W. Glace
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Martin D. Eastgate
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Richard L. Schild
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Jason T. Sweeney
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - David A. Conlon
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| |
Collapse
|
37
|
Fox RJ, Tripp JC, Schultz MJ, Payack JF, Fanfair DD, Mudryk BM, Murugesan S, Chen CPH, La Cruz TE, Ivy SE, Broxer S, Cullen R, Erdemir D, Geng P, Xu Z, Fritz A, Doubleday WW, Conlon DA. Preparation of the HIV Attachment Inhibitor BMS-663068. Part 1. Evolution of Enabling Strategies. Org Process Res Dev 2017. [DOI: 10.1021/acs.oprd.7b00134] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Richard J. Fox
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Jonathan C. Tripp
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Mitchell J. Schultz
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Joseph F. Payack
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Dayne D. Fanfair
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Boguslaw M. Mudryk
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Saravanababu Murugesan
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Chung-Pin H. Chen
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Thomas E. La Cruz
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Sabrina E. Ivy
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Sévrine Broxer
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Ryan Cullen
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Deniz Erdemir
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Peng Geng
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Zhongmin Xu
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Alan Fritz
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - Wendel W. Doubleday
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| | - David A. Conlon
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey, 08903-0191, United States
| |
Collapse
|
38
|
La Cruz TE, Saurer EM, Engstrom J, Bultman MS, Forest R, Akpinar F, Ferreira G, Ho JW, Huang M, Soltani M, Murugesan S, Fanfair D, Ramirez A, Rosso VW, Erdemir D, Rosenbaum T, Haslam M, Grier S, Peddicord M, Pathirana C, Marshall J, Ding W, Huang Y, Ayers S, Braem A, Schild RL, Ivy SE, Payack J, McLeod DD, Nikitczuk W, Doubleday W, Shah S, Conlon DA. Preparation of the HIV Attachment Inhibitor BMS-663068. Part 9. Active Pharmaceutical Ingredient Process Development and Powder Properties. Org Process Res Dev 2017. [DOI: 10.1021/acs.oprd.7b00138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Thomas E. La Cruz
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Eric M. Saurer
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Joshua Engstrom
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Michael S. Bultman
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Robert Forest
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Fulya Akpinar
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Glenn Ferreira
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Jeanne W. Ho
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Masano Huang
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Michelle Soltani
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Saravanababu Murugesan
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Dayne Fanfair
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Antonio Ramirez
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Victor W. Rosso
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Deniz Erdemir
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Tamar Rosenbaum
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Michelle Haslam
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Stephen Grier
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Michael Peddicord
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Charles Pathirana
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Jonathan Marshall
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Wei Ding
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Yande Huang
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Sloan Ayers
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Alan Braem
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Richard L. Schild
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Sabrina E. Ivy
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Joseph Payack
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Douglas D. McLeod
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Whitney Nikitczuk
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Wendel Doubleday
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - Sapna Shah
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| | - David A. Conlon
- Chemical and Synthetic Development, ‡Drug Product Science and Technology, §API Operations, ∥Analytical and Bioanalytical
Operations, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903-0191, United States
| |
Collapse
|
39
|
Wang D, Wang Y, Zhao J, Shen M, Hu J, Liu Z, Li L, Xue F, Yu P. Strategic Approach to 8-Azacoumarins. Org Lett 2017; 19:984-987. [DOI: 10.1021/acs.orglett.6b03771] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Dong Wang
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Yuxi Wang
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Junjie Zhao
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Meng Shen
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Jianyong Hu
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhenlin Liu
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Linna Li
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Furen Xue
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Peng Yu
- China International
Science
and Technology Cooperation Base of Food Nutrition/Safety and Medicinal
Chemistry, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| |
Collapse
|
40
|
Pires MJD, Poeira DL, Purificação SI, Marques MMB. Synthesis of Substituted 4-, 5-, 6-, and 7-Azaindoles from Aminopyridines via a Cascade C–N Cross-Coupling/Heck Reaction. Org Lett 2016; 18:3250-3. [DOI: 10.1021/acs.orglett.6b01500] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Marina J. D. Pires
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Diogo L. Poeira
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - Sara I. Purificação
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| | - M. Manuel B. Marques
- LAQV@REQUIMTE, Departamento
de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Campus de Caparica, 2829-516 Caparica, Portugal
| |
Collapse
|
41
|
Hu HY, Yu XD, Wang F, Lin CR, Zeng JZ, Qiu YK, Fang MJ, Wu Z. Novel N-Substituted 2-(2-(Adamantan-1-yl)-1H-Indol-3-yl)-2-Oxoacetamide Derivatives: Synthesis and Biological Evaluation. Molecules 2016; 21:E530. [PMID: 27164070 PMCID: PMC6273615 DOI: 10.3390/molecules21050530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/03/2016] [Accepted: 04/16/2016] [Indexed: 11/16/2022] Open
Abstract
In this study, a series of novel N-substituted 2-(2-(adamantan-1-yl)-1H-indol-3-yl)-2-oxoacetamide derivatives were synthesized, and evaluated for their cytotoxicity in human cell lines including Hela (cervical cancer), MCF7 (breast cancer ) and HepG2 (liver cancer). Several compounds were found to have potent anti-proliferative activity against those human cancer cell lines and compound 5r showed the most potent biological activity against HepG2 cells with an IC50 value of 10.56 ± 1.14 μΜ. In addition, bioassays showed that compound 5r induced time-dependent and dose-dependent cleavage of poly ADP-ribose polymerase (PARP), and also induced a dose-dependent increase in caspase-3 and caspase-8 activity, but had little effect on caspase-9 protease activity in HepG2 cells. These results provide evidence that 5r-induced apoptosis in HepG2 cell is caspase-8-dependent.
Collapse
Affiliation(s)
- Hong-Yu Hu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Xu-Dong Yu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Fei Wang
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Chun-Rong Lin
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Jin-Zhang Zeng
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Ying-Kun Qiu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Mei-Juan Fang
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| | - Zhen Wu
- School of Pharmaceutical Sciences and the Key Laboratory for Chemical Biology of Fujian Province, Xiamen University, South Xiang-An Road, Xiamen 361102, China.
| |
Collapse
|
42
|
Pham NN, Dang TT, Ngo NT, Villinger A, Ehlers P, Langer P. Facile synthesis of 4- and 7-azaindoles from the corresponding imines by palladium-catalyzed cascade C-C and C-N coupling. Org Biomol Chem 2016; 13:6047-58. [PMID: 25947884 DOI: 10.1039/c5ob00720h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The cyclization of 2,3-dihalopyridines with readily available imines provides a convenient and regioselective approach to 4- and 7-azaindoles. The regioselectivity can be controlled by the choice of the halogen atoms at the pyridine ring (chlorine versus bromine).
Collapse
Affiliation(s)
- Ngo Nghia Pham
- Institut für Chemie, Universität Rostock, Albert-Einstein-Str. 3a, 18059 Rostock, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Ngo TN, Janert F, Ehlers P, Hoang DH, Dang TT, Villinger A, Lochbrunner S, Langer P. Pd(0)-catalyzed domino C-N coupling/hydroamination/C-H arylation reactions: efficient synthesis and photophysical properties of azaindolo[1,2-f]phenanthridines. Org Biomol Chem 2016; 14:1293-301. [PMID: 26647835 DOI: 10.1039/c5ob02274f] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
A series of new 7- and 4-azaindolo[1,2-f]phenanthridines were synthesized by a domino Pd(0)-catalyzed reaction, which involves three sequential steps: C-N coupling, hydroamination, and intramolecular C-H arylation. The products show promising fluorescence properties with high quantum yields (12-65%).
Collapse
Affiliation(s)
- Thang Ngoc Ngo
- Institut für Chemie, Universität Rostock, Albert-Einstein-Str. 3a, 18059 Rostock, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Yan J, He G, Yan F, Zhang J, Zhang G. The dicarbonylation of indoles via Friedel–Crafts reaction with dicarbonyl nitrile generated in situ and retro-cyanohydrination. RSC Adv 2016. [DOI: 10.1039/c6ra04016k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The reaction of indole and β-carbonyl nitrile to generate dicarbonyl indoles has been developed. This process involves α-oxonation of the β-carbonyl nitrile, Friedel–Crafts reaction with indoles and retro-cyanohydrination form dicarbonyl indoles.
Collapse
Affiliation(s)
- Jianwei Yan
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| | - Guangjie He
- Department of Forensic Medicine
- Xinxiang Medical University
- Xinxiang
- P. R. China
| | - Fulin Yan
- School of Pharmacy
- Xinxiang Medical University
- Xinxiang
- P. R. China
| | - Jixia Zhang
- School of Pharmacy
- Xinxiang Medical University
- Xinxiang
- P. R. China
| | - Guisheng Zhang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals
- Key Laboratory of Green Chemical Media and Reactions
- Ministry of Education
- School of Chemistry and Chemical Engineering
- Henan Normal University
| |
Collapse
|
45
|
Lu L, Yu F, Cai L, Debnath AK, Jiang S. Development of Small-molecule HIV Entry Inhibitors Specifically Targeting gp120 or gp41. Curr Top Med Chem 2016; 16:1074-90. [PMID: 26324044 PMCID: PMC4775441 DOI: 10.2174/1568026615666150901114527] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 05/17/2015] [Accepted: 05/27/2015] [Indexed: 12/31/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) envelope (Env) glycoprotein surface subunit gp120 and transmembrane subunit gp41 play important roles in HIV-1 entry, thus serving as key targets for the development of HIV-1 entry inhibitors. T20 peptide (enfuvirtide) is the first U.S. FDA-approved HIV entry inhibitor; however, its clinical application is limited by the lack of oral availability. Here, we have described the structure and function of the HIV-1 gp120 and gp41 subunits and reviewed advancements in the development of small-molecule HIV entry inhibitors specifically targeting these two Env glycoproteins. We then compared the advantages and disadvantages of different categories of HIV entry inhibitor candidates and further predicted the future trend of HIV entry inhibitor development.
Collapse
Affiliation(s)
| | | | | | | | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, Shanghai Medical College, Fudan University, 130 Dong An Road, Building #13, Shanghai 200032, China.
| |
Collapse
|
46
|
Synthesis and Anticancer Activity of 1-(1H-Indol-3-yl)-2-(4-diarylmethylpiperazine-1-yl)ethane-1,2-dione Derivatives. J CHEM-NY 2016. [DOI: 10.1155/2016/4617454] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Several new 1-(4-diarylmethylpiperazine-1-yl)-2-(1H-indol-3-yl)ethane-1,2-dione derivatives were synthesized by acylation of 1-diarylmethylpiperazine with 2-(1H-indol-3-yl)-2-oxoacetyl chloride. Their structures were confirmed by1H NMR, IR, mass spectra, and elemental analysis. These compounds were further evaluated for their anticancer activity, and most of them were found to have moderate-to-potent antiproliferative activities against Hela, A-549, and ECA-109 cancer cell linesin vitro.
Collapse
|
47
|
Inhibitors of HIV-1 attachment: The discovery and structure-activity relationships of tetrahydroisoquinolines as replacements for the piperazine benzamide in the 3-glyoxylyl 6-azaindole pharmacophore. Bioorg Med Chem Lett 2015; 26:160-7. [PMID: 26584882 DOI: 10.1016/j.bmcl.2015.11.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 11/22/2022]
Abstract
6,6-Fused ring systems including tetrahydroisoquinolines and tetrahydropyrido[3,4-d]pyrimidines have been explored as possible replacements for the piperazine benzamide portion of the HIV-1 attachment inhibitor BMS-663068. In initial studies, the tetrahydroisoquinoline compounds demonstrate sub-nanomolar activity in a HIV-1 pseudotype viral infection assay used as the initial screen for inhibitory activity. Analysis of SARs and approaches to optimization for an improved drug-like profile are examined herein.
Collapse
|
48
|
Discovery and optimization of novel small-molecule HIV-1 entry inhibitors using field-based virtual screening and bioisosteric replacement. Bioorg Med Chem Lett 2015; 24:5439-45. [PMID: 25454268 DOI: 10.1016/j.bmcl.2014.10.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/06/2014] [Accepted: 10/08/2014] [Indexed: 11/22/2022]
Abstract
With the emergence of drug-resistant strains and the cumulative toxicities associated with current therapies, demand remains for new inhibitors of HIV-1 replication. The inhibition of HIV-1 entry is an attractive, yet underexploited therapeutic approach with implications for salvage and preexposure prophylactic regimens, as well as topical microbicides. Using the combination of a field-derived bioactive conformation template to perform virtual screening and iterative bioisosteric replacements, coupled with in silico predictions of absorption, distribution, metabolism, and excretion, we have identified new leads for HIV-1 entry inhibitors.
Collapse
|
49
|
Patel RV, Park SW. Pyrroloaryls and pyrroloheteroaryls: Inhibitors of the HIV fusion/attachment, reverse transcriptase and integrase. Bioorg Med Chem 2015; 23:5247-63. [PMID: 26116177 DOI: 10.1016/j.bmc.2015.06.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Revised: 06/04/2015] [Accepted: 06/05/2015] [Indexed: 02/01/2023]
Abstract
Heterocyclic compounds execute a very important role in drug design and discovery. This article provides the basic milestones of the research for pyrroloaryl and pyrroloheteroaryl based components targeting HIV viral replication cycle. Anti-HIV activity is elaborated for several classes of pyrrolo-compounds as pyrrolopyridines, pyrrolopyrimidines, pyrrolopyridazines, pyrrolobenzodiazepinones, pyrrolobenzothiazepines, pyrrolobenzoxazepinones, pyrrolophenanthridines, pyrroloquinoxalines, pyrrolotriazines, pyrroloquinolines, pyrrolopyrazinones, pyrrolothiatriazines, arylthiopyrroles and pyrrolopyrazolones targeting two essential HIV enzymes, reverse transcriptase and integrase as well as attachment/fusion of HIV virons to the host CD-4 cell. Such attempts were resulted in a discovery of highly potent anti-HIV agents suitable for clinical trials, for example, BMS-378806, BMS-585248, BMS-626529, BMS-663068, BMS-488043 and BMS-663749, etc. as anti-HIV attachment agents, triciribine, QX432, BI-1 and BI-2 as HIV RT inhibitors which are in preclinical or clinical development. Mechanism of action of compounds presented in this article towards the suppression of HIV attachment/fusion as well as against the activities of HIV enzymes reverse transcriptase and integrase has been discussed. Relationships of new compounds' molecular framework and HIV viral target has been overviewed in order to facilitate further construction of promising anti-HIV agents in future drug discovery process.
Collapse
Affiliation(s)
- Rahul V Patel
- Laboratory of Growth Regulators, Centre of the Region Haná for Biotechnological and Agricultural Research, Institute of Experimental Botany ASCR & Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic.
| | - Se Won Park
- Organic Research Laboratory, Department of Bioresources and Food Science, College of Life and Environmental Sciences, Konkuk University, Seoul 143 701, South Korea
| |
Collapse
|
50
|
Abstract
In 1989, the reaction of vinyl magnesium halides with ortho-substituted nitroarenes leading to indoles was discovered. This reaction is now frequently reported as the "Bartoli reaction" or the "Bartoli indole synthesis" (BIS). It has rapidly become the shortest and most flexible route to 7-substituted indoles, because the classical indole syntheses generally fail in their preparation. The flexibility of the Bartoli reaction is great as it can be extended to heteroaromatic nitro derivatives and can be run on solid support. This review will focus on the use of the Bartoli indole synthesis as the key step in preparations of complex indoles, which appeared in the literature in the last few years.
Collapse
Affiliation(s)
- Giuseppe Bartoli
- Dipartimento Chimica Industriale 'Toso Montanari', Università di Bologna, Viale Risorgimento 4, I-40136 Bologna, Italy.
| | | | | |
Collapse
|