1
|
Zhan S, Zhang Y, Cao T, Yang R, Wang Q, Huang L, Cui R, Yu J, Meng H, Wang Y, Zhang S, Zheng M, Wu X. Discovery of Imidazo[1,2- a]pyrazine Derivatives as Potent ENPP1 Inhibitors. J Med Chem 2024; 67:18317-18333. [PMID: 39357030 DOI: 10.1021/acs.jmedchem.4c01634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
ENPP1 acts as a negative regulator of the cGAS-STING pathway through the hydrolysis of 2'3'-cGAMP. Inhibitors of ENPP1 are regarded as promising agents for stimulating the immune response in cancer immunotherapy. This study describes the identification and optimization of imidazo[1,2-a]pyrazine derivative 7 as a highly potent and selective ENPP1 inhibitor. Compound 7 demonstrated substantial inhibitory activity against ENPP1 with an IC50 value of 5.70 or 9.68 nM while showing weak inhibition against ENPP2 and ENPP3. Furthermore, compound 7 was shown to enhance the mRNA expression of cGAMP-induced STING pathway downstream target genes, such as IFNB1, CXCL10, and IL6. In vivo pharmacokinetic and antitumor studies showed promising results, with 7 not only exhibiting efficient pharmacokinetic properties but also enhancing the antitumor efficacy of the anti-PD-1 antibody. Treatment with 7 (80 mg/kg) combined with anti-PD-1 antibody achieved a tumor growth inhibition rate of 77.7% and improved survival in a murine model.
Collapse
Affiliation(s)
- Shiping Zhan
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Yingying Zhang
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tian Cao
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmacy, Guizhou Medical University, Guiyang 550014, China
| | - Ruirui Yang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qiang Wang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Lin Huang
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Rongrong Cui
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jie Yu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Haifang Meng
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
| | - Yitian Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Sulin Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Mingyue Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China
- The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaowei Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528400, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
2
|
Goswami A, Goyal S, Khurana P, Singh K, Deb B, Kulkarni A. Small molecule innate immune modulators in cancer therapy. Front Immunol 2024; 15:1395655. [PMID: 39318624 PMCID: PMC11419979 DOI: 10.3389/fimmu.2024.1395655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
Immunotherapy has proved to be a breakthrough in cancer treatment. So far, a bulk of the approved/late-stage cancer immunotherapy are antibody-based. Although these antibody-based drugs have demonstrated great promise, a majority of them are limited due to their access to extracellular targets, lack of oral bioavailability, tumor microenvironment penetration, induction of antibody dependent cytotoxicity etc. In recent times, there has been an increased research focus on the development of small molecule immunomodulators since they have the potential to overcome the aforementioned limitations posed by antibodies. Furthermore, while most biologics based therapeutics that are in clinical use are limited to modulating the adaptive immune system, very few clinically approved therapeutic modalities exist that modulate the innate immune system. The innate immune system, which is the body's first line of defense, has the ability to turn cold tumors hot and synergize strongly with existing adaptive immune modulators. In preclinical studies, small molecule innate immune modulators have demonstrated synergistic efficacy as combination modalities with current standard-of-care immune checkpoint antibodies. In this review, we highlight the recent advances made by small molecule innate immunomodulators in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Barnali Deb
- Aten Porus Lifesciences Pvt. Ltd., Bengaluru, India
| | - Aditya Kulkarni
- Aten Porus Lifesciences Pvt. Ltd., Bengaluru, India
- Avammune Therapeutics, Philadelphia, PA, United States
| |
Collapse
|
3
|
Sharafat RH, Saeed A. Ectonucleotidase inhibitors: targeting signaling pathways for therapeutic advancement-an in-depth review. Purinergic Signal 2024:10.1007/s11302-024-10031-0. [PMID: 38958821 DOI: 10.1007/s11302-024-10031-0] [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: 12/12/2023] [Accepted: 06/16/2024] [Indexed: 07/04/2024] Open
Abstract
Ectonucleotidase inhibitors are a family of pharmacological drugs that, by selectively targeting ectonucleotidases, are essential in altering purinergic signaling pathways. The hydrolysis of extracellular nucleotides and nucleosides is carried out by these enzymes, which include ectonucleoside triphosphate diphosphohydrolases (NTPDases) and ecto-5'-nucleotidase (CD73). Ectonucleotidase inhibitors can prevent the conversion of ATP and ADP into adenosine by blocking these enzymes and reduce extracellular adenosine. These molecules are essential for purinergic signaling, which is associated with a variability of physiological and pathological processes. By modifying extracellular nucleotide metabolism and improving purinergic signaling regulation, ectonucleotide pyrophosphatase/phosphodiesterase (ENPP) inhibitors have the potential to improve cancer treatment, inflammatory management, and immune response modulation. Purinergic signaling is affected by CD73 inhibitors because they prevent AMP from being converted to adenosine. These inhibitors are useful in cancer therapy and immunotherapy because they may improve chemotherapy effectiveness and alter immune responses. Purinergic signaling is controlled by NTPDase inhibitors, which specifically target enzymes involved in extracellular nucleotide breakdown. These inhibitors show promise in reducing immunological responses, thrombosis, and inflammation, perhaps assisting in the treatment of cardiovascular and autoimmune illnesses. Alkaline phosphatase (ALP) inhibitors alter the function of enzymes involved in dephosphorylation reactions, which has an impact on a variety of biological processes. By altering the body's phosphate levels, these inhibitors may be used to treat diseases including hyperphosphatemia and certain bone problems. This article provides a guide for researchers and clinicians looking to leverage the remedial capability of ectonucleotidase inhibitors in a variety of illness scenarios by illuminating their processes, advantages, and difficulties.
Collapse
Affiliation(s)
- R Huzaifa Sharafat
- Department of Chemistry, Quaid-I-Azam University, Islamabad, 45321, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-I-Azam University, Islamabad, 45321, Pakistan.
| |
Collapse
|
4
|
Tian X, Ai J, Tian X, Wei X. cGAS-STING pathway agonists are promising vaccine adjuvants. Med Res Rev 2024; 44:1768-1799. [PMID: 38323921 DOI: 10.1002/med.22016] [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: 10/17/2023] [Revised: 12/10/2023] [Accepted: 01/09/2024] [Indexed: 02/08/2024]
Abstract
Adjuvants are of critical value in vaccine development as they act on enhancing immunogenicity of antigen and inducing long-lasting immunity. However, there are only a few adjuvants that have been approved for clinical use, which highlights the need for exploring and developing new adjuvants to meet the growing demand for vaccination. Recently, emerging evidence demonstrates that the cGAS-STING pathway orchestrates innate and adaptive immunity by generating type I interferon responses. Many cGAS-STING pathway agonists have been developed and tested in preclinical research for the treatment of cancer or infectious diseases with promising results. As adjuvants, cGAS-STING agonists have demonstrated their potential to activate robust defense immunity in various diseases, including COVID-19 infection. This review summarized the current developments in the field of cGAS-STING agonists with a special focus on the latest applications of cGAS-STING agonists as adjuvants in vaccination. Potential challenges were also discussed in the hope of sparking future research interests to further the development of cGAS-STING as vaccine adjuvants.
Collapse
Affiliation(s)
- Xinyu Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Jiayuan Ai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Centre for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| |
Collapse
|
5
|
Bhakta A, Mukhtar S, Anwar S, Haider S, Alahmdi MI, Parveen H, Alsharif MA, Wani MY, Chakrabarty A, Hassan MI, Ahmed N. Design, synthesis, molecular docking and anti-proliferative activity of novel phenothiazine containing imidazo[1,2- a]pyridine derivatives against MARK4 protein. RSC Med Chem 2024; 15:1942-1958. [PMID: 38911173 PMCID: PMC11187548 DOI: 10.1039/d4md00059e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/10/2024] [Indexed: 06/25/2024] Open
Abstract
A series of novel phenothiazine-containing imidazo[1,2-a]pyridine derivatives were designed and synthesized under metal-free conditions in excellent yield. These derivatives were effectively transformed further into N-alkyl, sulfoxide, and sulfone derivatives. Derivatives were deployed against human microtubule affinity regulating kinase (MARK4), some molecules play crucial roles in cell-cycle progression such as G1/S transition and regulator of microtubule dynamics. Hence, molecules have shown excellent MARK4 inhibitory potential. Molecules with excellent IC50 values were selected for further studies such as ligand interactions using fluorescence quenching experiments for the binding constant. The highest binding constant was calculated as K = 0.79 × 105 and K = 0.1 × 107 for compounds 6a and 6h, respectively. Molecular docking, cell cytotoxicity, mitochondrial reactive oxygen species measurement and oxidative DNA damage were also studied to understand the mechanism of action of the molecules on cancer cells. It was found that the designed and synthesized compounds played anti-cancer roles by binding and inhibiting MARK4 protein.
Collapse
Affiliation(s)
- Avijit Bhakta
- Department of Chemistry, Indian Institute of Technology Roorkee Roorkee-247 667 U.K. India
| | - Sayeed Mukhtar
- Department of Chemistry, Faculty of Science, University of Tabuk Tabuk 71491 Saudi Arabia
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia New Delhi India
| | - Shaista Haider
- Department of Life Sciences, Shiv Nadar University Uttar Pradesh 201314 India
| | - Mohammed Issa Alahmdi
- Department of Chemistry, Faculty of Science, University of Tabuk Tabuk 71491 Saudi Arabia
| | - Humaira Parveen
- Department of Chemistry, Faculty of Science, University of Tabuk Tabuk 71491 Saudi Arabia
| | - Meshari A Alsharif
- Chemistry Department, Faculty of Applied Science, Umm Al-Qura University Makkah Saudi
| | - Mohmmad Younus Wani
- Department of Chemistry, College of Science, University of Jeddah 21589 Jeddah Saudi Arabia
| | | | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia New Delhi India
| | - Naseem Ahmed
- Department of Chemistry, Indian Institute of Technology Roorkee Roorkee-247 667 U.K. India
| |
Collapse
|
6
|
Tasleem M, Pelletier J, Sévigny J, Hussain Z, Khan A, Al-Harrasi A, El-Kott AF, Taslimi P, Negm S, Shafiq Z, Iqbal J. Synthesis, in vitro, and in silico studies of morpholine-based thiosemicarbazones as ectonucleotide pyrophosphatase/phosphodiesterase-1 and -3 inhibitors. Int J Biol Macromol 2024; 266:131068. [PMID: 38531526 DOI: 10.1016/j.ijbiomac.2024.131068] [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: 11/08/2023] [Revised: 03/06/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
An extensive range of new biologically active morpholine based thiosemicarbazones derivatives 3a-r were synthesized, characterized by spectral techniques and evaluated as inhibitors of ENPP isozymes. Most of the novel thiosemicarbazones exhibit potent inhibition towards NPP1 and NPP3 isozymes. Compound 3 h was potent inhibitor of NPP1 with IC50 value of 0.55 ± 0.02. However, the most powerful inhibitor of NPP3 was 3e with an IC50 value of 0.24 ± 0.02. Furthermore, Lineweaver-Burk plot for compound 3 h against NPP1 and for compound 3e against NPP3 was devised through enzymes kinetics studies. Molecular docking and in silico studies was also done for analysis of interaction pattern of all newly synthesized compounds. The results were further validated by molecular dynamic (MD) simulation where the stability of conformational transformation of the best protein-ligand complex (3e) were justified on the basis of RMSD and RMSF analysis.
Collapse
Affiliation(s)
- Mussarat Tasleem
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec-Université Laval, Québec G1V 4G2, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec-Université Laval, Québec G1V 4G2, Canada; Département de Microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec G1V 0A6, Canada
| | - Zahid Hussain
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
| | - Ajmal Khan
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Centre, University of Nizwa, P.O. Box 33, PC 616, Birkat Al Mauz, Nizwa, Sultanate of Oman.
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia; Department of Zoology, College of Science, Damanhour University, Egypt
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartin University, 74100 Bartin, Türkiye
| | - Sally Negm
- Department of Life Sciences, College of Science and Art Mahyel Aseer, King Khalid University, Abha 62529, Saudi Arabia
| | - Zahid Shafiq
- Institute of Chemical Sciences, Bahauddin Zakariya University, Multan 60800, Pakistan; Department of Pharmaceutical & Medicinal Chemistry, An der Immenburg 4, D-53121 Bonn, Germany.
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan.
| |
Collapse
|
7
|
Du B, Ru J, Zhan Z, Lin C, Liu Y, Mao W, Zhang J. Insight into small-molecule inhibitors targeting extracellular nucleotide pyrophosphatase/phosphodiesterase1 for potential multiple human diseases. Eur J Med Chem 2024; 268:116286. [PMID: 38432057 DOI: 10.1016/j.ejmech.2024.116286] [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: 12/31/2023] [Revised: 02/06/2024] [Accepted: 02/24/2024] [Indexed: 03/05/2024]
Abstract
Extracellular nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) has been identified as a type II transmembrane glycoprotein. It plays a crucial role in various biological processes, such as bone mineralization, cancer cell proliferation, and immune regulation. Consequently, ENPP1 has garnered attention as a promising target for pharmacological interventions. Despite its potential, the development of clinical-stage ENPP1 inhibitors for solid tumors, diabetes, and silent rickets remains limited. However, there are encouraging findings from preclinical trials involving small molecules exhibiting favorable therapeutic effects and safety profiles. This perspective aims to shed light on the structural properties, biological functions and the relationship between ENPP1 and diseases. Additionally, it focuses on the structure-activity relationship of ENPP1 inhibitors, with the intention of guiding the future development of new and effective ENPP1 inhibitors.
Collapse
Affiliation(s)
- Baochan Du
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinxiao Ru
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zixuan Zhan
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Congcong Lin
- Department of Medicinal Chemistry and Natural Medicine Chemistry, College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yang Liu
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, China
| | - Wuyu Mao
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
8
|
Sun Y, Chen M, Han Y, Li W, Ma X, Shi Z, Zhou Y, Xu L, Yu L, Wang Y, Yu J, Diao X, Meng L, Xu S. Discovery of Pyrido[2,3- d]pyrimidin-7-one Derivatives as Highly Potent and Efficacious Ectonucleotide Pyrophosphatase/Phosphodiesterase 1 (ENPP1) Inhibitors for Cancer Treatment. J Med Chem 2024; 67:3986-4006. [PMID: 38387074 DOI: 10.1021/acs.jmedchem.3c02288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) is an extracellular enzyme responsible for hydrolyzing cyclic guanosine monophosphate-adenosine monophosphate (cGAMP), the endogenous agonist for the stimulator of interferon genes (STING) pathway. Inhibition of ENPP1 can trigger STING and promote antitumor immunity, offering an attractive therapeutic target for cancer immunotherapy. Despite progress in the discovery of ENPP1 inhibitors, the diversity in chemical structures and the efficacy of the agents are far from desirable, emphasizing the demand for novel inhibitors. Herein, we describe the design, synthesis, and biological evaluation of a series of ENPP1 inhibitors based on the pyrido[2,3-d]pyrimidin-7-one scaffold. Optimization efforts led to compound 31 with significant potency in both ENPP1 inhibition and STING pathway stimulation in vitro. Notably, 31 demonstrated in vivo efficacy in a syngeneic 4T1 mouse triple negative breast cancer model. These findings provide a promising lead compound with a novel scaffold for further drug development in cancer immunotherapy.
Collapse
Affiliation(s)
- Yaoliang Sun
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Manman Chen
- Division of Antitumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuyan Han
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Weiqiang Li
- Center for Drug Metabolism and Pharmacokinetics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Ma
- Division of Antitumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zihan Shi
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Lan Xu
- Division of Antitumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yuxiang Wang
- Division of Antitumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jinghua Yu
- Center for Drug Metabolism and Pharmacokinetics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xingxing Diao
- Center for Drug Metabolism and Pharmacokinetics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linghua Meng
- Division of Antitumor Pharmacology, State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shilin Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Guan D, Fang L, Feng M, Guo S, Xie L, Chen C, Sun X, Wu Q, Yuan X, Xie Z, Zhou J, Zhang H. Ecto-nucleotide pyrophosphatase/phosphodiesterase 1 inhibitors: Research progress and prospects. Eur J Med Chem 2024; 267:116211. [PMID: 38359537 DOI: 10.1016/j.ejmech.2024.116211] [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: 11/13/2023] [Revised: 01/15/2024] [Accepted: 01/31/2024] [Indexed: 02/17/2024]
Abstract
The cancer immunotherapies involved in cGAS-STING pathway have been made great progress in recent years. STING agonists exhibit broad-spectrum anti-tumor effects with strong immune response. As a negative regulator of the cGAS-STING pathway, ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) can hydrolyze extracellular 2', 3'-cGAMP and reduce extracellular 2', 3'-cGAMP concentration. ENPP1 has been validated to play important roles in diabetes, cancers, and cardiovascular disease and now become a promising target for tumor immunotherapy. Several ENPP1 inhibitors under development have shown good anti-tumor effects alone or in combination with other agents in clinical and preclinical researches. In this review, the biological profiles of ENPP1 were described, and the structures and the structure-activity relationships (SAR) of the known ENPP1 inhibitors were summarized. This review also provided the prospects and challenges in the development of ENPP1 inhibitors.
Collapse
Affiliation(s)
- Dezhong Guan
- Department of Medicinal Chemistry, China Pharmaceutical University, TongjiaXiang 24, 210009, Nanjing, China
| | - Lincheng Fang
- Peking University Shenzhen Graduate School, Shenzhen, China
| | - Mingshun Feng
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shi Guo
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Lingfeng Xie
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Chao Chen
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Xue Sun
- Department of Medicinal Chemistry, China Pharmaceutical University, TongjiaXiang 24, 210009, Nanjing, China
| | - Qingyun Wu
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Xinrui Yuan
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China
| | - Zuoquan Xie
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.
| | - Jinpei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, TongjiaXiang 24, 210009, Nanjing, China.
| | - Huibin Zhang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, China.
| |
Collapse
|
10
|
Bashore FM, Katis VL, Du Y, Sikdar A, Wang D, Bradshaw WJ, Rygiel KA, Leisner TM, Chalk R, Mishra S, Williams CA, Gileadi O, Brennan PE, Wiley JC, Gockley J, Cary GA, Carter GW, Young JE, Pearce KH, Fu H, Axtman AD. Characterization of covalent inhibitors that disrupt the interaction between the tandem SH2 domains of SYK and FCER1G phospho-ITAM. PLoS One 2024; 19:e0293548. [PMID: 38359047 PMCID: PMC10868801 DOI: 10.1371/journal.pone.0293548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/15/2023] [Indexed: 02/17/2024] Open
Abstract
RNA sequencing and genetic data support spleen tyrosine kinase (SYK) and high affinity immunoglobulin epsilon receptor subunit gamma (FCER1G) as putative targets to be modulated for Alzheimer's disease (AD) therapy. FCER1G is a component of Fc receptor complexes that contain an immunoreceptor tyrosine-based activation motif (ITAM). SYK interacts with the Fc receptor by binding to doubly phosphorylated ITAM (p-ITAM) via its two tandem SH2 domains (SYK-tSH2). Interaction of the FCER1G p-ITAM with SYK-tSH2 enables SYK activation via phosphorylation. Since SYK activation is reported to exacerbate AD pathology, we hypothesized that disruption of this interaction would be beneficial for AD patients. Herein, we developed biochemical and biophysical assays to enable the discovery of small molecules that perturb the interaction between the FCER1G p-ITAM and SYK-tSH2. We identified two distinct chemotypes using a high-throughput screen (HTS) and orthogonally assessed their binding. Both chemotypes covalently modify SYK-tSH2 and inhibit its interaction with FCER1G p-ITAM, however, these compounds lack selectivity and this limits their utility as chemical tools.
Collapse
Affiliation(s)
- Frances M. Bashore
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Vittorio L. Katis
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Arunima Sikdar
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Dongxue Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - William J. Bradshaw
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Karolina A. Rygiel
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Tina M. Leisner
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Rod Chalk
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Swati Mishra
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - C. Andrew Williams
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Opher Gileadi
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | - Paul E. Brennan
- Nuffield Department of Medicine, Centre for Medicines Discovery, ARUK Oxford Drug Discovery Institute, University of Oxford, Headington, Oxford, United Kingdom
| | | | - Jake Gockley
- Sage Bionetworks, Seattle, WA, United States of America
| | - Gregory A. Cary
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States of America
| | - Gregory W. Carter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME, United States of America
| | - Jessica E. Young
- Department of Laboratory Medicine and Pathology, School of Medicine, University of Washington, Seattle, WA, United States of America
- Institute for Stem Cell and Regenerative Medicine, School of Medicine, University of Washington, Seattle, WA, United States of America
| | - Kenneth H. Pearce
- Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, United States of America
- Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, United States of America
| | | | - Alison D. Axtman
- Structural Genomics Consortium, Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
11
|
Rohilla A, Singh AK, Koleske B, Srikrishna G, Bishai WR. Structure-based virtual screening and in vitro validation of inhibitors of cyclic dinucleotide phosphodiesterases ENPP1 and CdnP. Microbiol Spectr 2024; 12:e0201223. [PMID: 38095464 PMCID: PMC10783014 DOI: 10.1128/spectrum.02012-23] [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: 05/12/2023] [Accepted: 11/21/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE In this paper, we describe novel inhibitors of cyclic dinucleotide phosphodiesterase enzymes from Mycobacterium tuberculosis (M.tb) (CdnP) and mammals (ENPP1). The phosphodiesterase enzymes hydrolyze cyclic dinucleotides, such as 2',3'-cyclic GMP-AMP and c-di-AMP, which are stimulator of interferon gene (STING) agonists. By blocking the hydrolysis of STING agonists, the cyclic GMP-AMP synthase (cGAS)-STING-IRF3 pathway is potentiated. There is strong evidence in tuberculosis and in cancer biology that potentiation of the cGAS-STING-IRF3 pathway leads to improved M.tb clearance and also improved antitumor responses in cancer. In addition to the identification of novel inhibitors and their biochemical characterization, we provide proof-of-concept evidence that our E-3 inhibitor potentiates the cGAS-STING-IRF3 pathway in both macrophage cell lines and also in primary human monocyte-derived macrophages.
Collapse
Affiliation(s)
- Akshay Rohilla
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alok Kumar Singh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benjamin Koleske
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Geetha Srikrishna
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William R. Bishai
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Center for Tuberculosis Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
12
|
Duangiad P, Nutho B, Chaijarasphong T, Morales NP, Pongtharangkul T, Hamachi I, Ojida A, Wongkongkatep J. Naturally occurring quercetin and myricetin as potent inhibitors for human ectonucleotide pyrophosphatase/phosphodiesterase 1. Sci Rep 2024; 14:125. [PMID: 38167594 PMCID: PMC10761680 DOI: 10.1038/s41598-023-50590-7] [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: 06/27/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024] Open
Abstract
Ecto-nucleotide pyrophosphatases/phosphodiesterases 1 (ENPP1) is a key enzyme in purinergic signaling pathways responsible for cell-to-cell communications and regulation of several fundamental pathophysiological processes. In this study, Kyoto Green, a rapid chemical sensor of pyrophosphate, was employed to screen for effective ENPP1 inhibitors among five representative flavonoids (quercetin, myricetin, morin, kaempferol, and quercetin-3-glucoside), five nucleosides (adenosine, guanosine, inosine, uridine, and cytidine), and five deoxynucleosides (2'- and 3'-deoxyadenosine, 2'-deoxyguanosine, 2'-deoxyinosine, and 2'-deoxyuridine). Conventional colorimetric, fluorescence, and bioluminescence assays revealed that ENPP1 was effectively inhibited by quercetin (Ki ~ 4 nM) and myricetin (Ki ~ 32 nM) when ATP was used as a substrate at pH 7.4. In silico analysis indicated that the presence of a chromone scaffold, particularly one containing a hydroxyl group at the 3' position on the B ring, may promote binding to the active site pocket of ENPP1 and enhance inhibition. This study demonstrated that the naturally derived quercetin and myricetin could effectively inhibit ENPP1 enzymatic activity and may offer health benefits in arthritis management.
Collapse
Affiliation(s)
- Peeradon Duangiad
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Bodee Nutho
- Department of Pharmacology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Thawatchai Chaijarasphong
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Noppawan Phumala Morales
- Department of Pharmacology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Thunyarat Pongtharangkul
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand
| | - Itaru Hamachi
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-Ku, Kyoto, 615-8510, Japan
| | - Akio Ojida
- Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Jirarut Wongkongkatep
- Department of Biotechnology, Faculty of Science, Mahidol University, Rama 6 Road, Bangkok, 10400, Thailand.
| |
Collapse
|
13
|
Cho Y, Kang M, Ji SH, Jeong HJ, Jung JE, Oh DH, Park S, Park YY, Choi J, Kim S, Kim NJ, Lee DH, Park CS, Han SJ, Lee S, Choi J. Discovery of Orally Bioavailable Phthalazinone Analogues as an ENPP1 Inhibitor for STING-Mediated Cancer Immunotherapy. J Med Chem 2023; 66:15141-15170. [PMID: 37963811 DOI: 10.1021/acs.jmedchem.3c01061] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
A lack of the T cell-inflamed tumor microenvironment limits the efficacy of immune checkpoint inhibitors (ICIs). Activation of stimulator of interferon genes (STING)-mediated innate immunity has emerged as a novel therapeutic approach in cancer therapy. 2',3'-Cyclic GMP-AMP (cGAMP) is a natural STING agonist; however, cGAMP is subjected to endogenous degradation by ecto-nucleotide pyrophosphatase phosphodiesterase 1 (ENPP1). To improve the ICI response rate, we developed 29f, a novel ENPP1 inhibitor with phthalazin-1(2H)-one as the core scaffold. 29f inhibited the cGAMP hydrolysis by ENPP1 in vitro (IC50 = 68 nM) and enhanced the STING-mediated type I interferon response in both immune and tumor cells. 29f demonstrated excellent metabolic stability and bioavailability (F = 65%). Orally administered 29f promoted tumor growth inhibition in a CT26 syngeneic model and increased the anti-PD-L1 response. Furthermore, 29f-induced immunological memory prevented the tumor relapse against tumor rechallenge, suggesting the promising therapeutic potential of 29f.
Collapse
Affiliation(s)
- Yeonguk Cho
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Miso Kang
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Su Hyun Ji
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Chemistry, Sogang University, Seoul 04107, Republic of Korea
| | - Hee Jin Jeong
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jae Eun Jung
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Do Hee Oh
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sunyoung Park
- Txinno Bioscience Inc., Yongin 16942, Republic of Korea
| | - Yong-Yea Park
- Txinno Bioscience Inc., Yongin 16942, Republic of Korea
| | - Junghwan Choi
- Txinno Bioscience Inc., Yongin 16942, Republic of Korea
| | - Sungjoon Kim
- Txinno Bioscience Inc., Yongin 16942, Republic of Korea
| | - Nam-Jung Kim
- Department of Fundamental Pharmaceutical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Duck-Hyung Lee
- Department of Chemistry, Sogang University, Seoul 04107, Republic of Korea
| | - Chan Sun Park
- Txinno Bioscience Inc., Yongin 16942, Republic of Korea
| | - Seo-Jung Han
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Sanghee Lee
- Brain Science Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department for HY-KIST Bio-convergence, Hanyang University, Seoul 04763, Republic of Korea
| | - Junwon Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
14
|
Castelli M, Yan P, Rodina A, Digwal CS, Panchal P, Chiosis G, Moroni E, Colombo G. How aberrant N-glycosylation can alter protein functionality and ligand binding: An atomistic view. Structure 2023; 31:987-1004.e8. [PMID: 37343552 PMCID: PMC10526633 DOI: 10.1016/j.str.2023.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/21/2023] [Accepted: 05/25/2023] [Indexed: 06/23/2023]
Abstract
Protein-assembly defects due to an enrichment of aberrant conformational protein variants are emerging as a new frontier in therapeutics design. Understanding the structural elements that rewire the conformational dynamics of proteins and pathologically perturb functionally oriented ensembles is important for inhibitor development. Chaperones are hub proteins for the assembly of multiprotein complexes and an enrichment of aberrant conformers can affect the cellular proteome, and in turn, phenotypes. Here, we integrate computational and experimental tools to investigte how N-glycosylation of specific residues in glucose-regulated protein 94 (GRP94) modulates internal dynamics and alters the conformational fitness of regions fundamental for the interaction with ATP and synthetic ligands and impacts substructures important for the recognition of interacting proteins. N-glycosylation plays an active role in modulating the energy landscape of GRP94, and we provide support for leveraging the knowledge on distinct glycosylation variants to design molecules targeting GRP94 disease-associated conformational states and assemblies.
Collapse
Affiliation(s)
- Matteo Castelli
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy
| | - Pengrong Yan
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chander S Digwal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Palak Panchal
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | | | - Giorgio Colombo
- Department of Chemistry, University of Pavia, via Taramelli 12, 27100 Pavia, Italy.
| |
Collapse
|
15
|
Bashore FM, Katis VL, Du Y, Sikdar A, Wang D, Bradshaw WJ, Rygiel KA, Leisner TM, Chalk R, Mishra S, Williams AC, Gileadi O, Brennan PE, Wiley JC, Gockley J, Cary GA, Carter GW, Young JE, Pearce KH, Fu H, Axtman AD. Characterization of covalent inhibitors that disrupt the interaction between the tandem SH2 domains of SYK and FCER1G phospho-ITAM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.28.551026. [PMID: 37547005 PMCID: PMC10402180 DOI: 10.1101/2023.07.28.551026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
RNA sequencing and genetic data support spleen tyrosine kinase (SYK) and high affinity immunoglobulin epsilon receptor subunit gamma (FCER1G) as putative targets to be modulated for Alzheimer's disease (AD) therapy. FCER1G is a component of Fc receptor complexes that contain an immunoreceptor tyrosine-based activation motif (ITAM). SYK interacts with the Fc receptor by binding to doubly phosphorylated ITAM (p-ITAM) via its two tandem SH2 domains (SYK-tSH2). Interaction of the FCER1G p-ITAM with SYK-tSH2 enables SYK activation via phosphorylation. Since SYK activation is reported to exacerbate AD pathology, we hypothesized that disruption of this interaction would be beneficial for AD patients. Herein, we developed biochemical and biophysical assays to enable the discovery of small molecules that perturb the interaction between the FCER1G p-ITAM and SYK-tSH2. We identified two distinct chemotypes using a high-throughput screen (HTS) and orthogonally assessed their binding. Both chemotypes covalently modify SYK-tSH2 and inhibit its interaction with FCER1G p-ITAM.
Collapse
Affiliation(s)
- Frances M Bashore
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, University of North Carolina, Chapel Hill, NC, USA
| | - Vittorio L Katis
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Yuhong Du
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Arunima Sikdar
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Dongxue Wang
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - William J Bradshaw
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Karolina A Rygiel
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | - Tina M Leisner
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Rod Chalk
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | | | | | - Opher Gileadi
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
- Current address: Structural Genomics Consortium, Department of Medicine, Karolinska Hospital and Karolinska Institute, 171 76 Stockholm, Sweden
| | - Paul E Brennan
- ARUK Oxford Drug Discovery Institute, Centre for Medicines Discovery, Nuffield Department of Medicine Research Building, Old Road Campus, University of Oxford, Headington, Oxford, OX3 7FZ, UK
| | | | | | | | | | | | - Kenneth H Pearce
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Center for Integrative Chemical Biology and Drug Discovery, University of North Carolina, Chapel Hill, NC, USA
| | - Haian Fu
- Department of Pharmacology and Chemical Biology, School of Medicine, Emory University, Atlanta, GA, USA; Emory Chemical Biology Discovery Center, School of Medicine, Emory University, Atlanta, GA, USA
| | - Alison D Axtman
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
16
|
3-arylthioimidazo[1,2-a]pyridine derivatives: A theoretical and experimental study of its photophysical properties. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2023.114675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/13/2023]
|
17
|
Choi J. Small molecule ectonucleotide pyrophosphatase/phosphodiesterase 1 inhibitors in cancer immunotherapy for harnessing innate immunity. B KOREAN CHEM SOC 2022. [DOI: 10.1002/bkcs.12646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Junwon Choi
- Department of Molecular Science and Technology Ajou University Suwon Gyeonggi Republic of Korea
| |
Collapse
|
18
|
Ullah S, Hamid K, Batool A, Pelletier J, Sévigny J, Khan AR, Langer P, Iqbal J. Synthesis of new sulphonate derivatives containing adamantane and 4-chlorophenyl moieties as nucleotide pyrophosphatase/phosphodiesterase-1 and -3 inhibitors. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.134494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
19
|
From Myricetin to the Discovery of Novel Natural Human ENPP1 Inhibitors: A Virtual Screening, Molecular Docking, Molecular Dynamics Simulation, and MM/GBSA Study. Molecules 2022; 27:molecules27196175. [PMID: 36234712 PMCID: PMC9573336 DOI: 10.3390/molecules27196175] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/10/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
It was recently revealed that naturally occurring myricetin can inhibit ectonucleotidase ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which, in turn, can treat ischemic cardiac injury. However, due to myricetin’s poor druggability, its further developments are relatively limited, which necessitates the discovery of novel ENPP1-inhibiting myricetin analogs as alternatives. In this study, the binding model of myricetin with ENPP1 was elucidated by molecular docking and molecular dynamics studies. Subsequently, virtual screening on the self-developed flavonoid natural product database (FNPD), led to the identification of two flavonoid glycosides (Cas No: 1397173-50-0 and 1169835-58-8), as potential ENPP1 inhibitors. Docking scores and MM/GBSA binding energies predicted that they might have higher inhibitory effects than myricetin. This study provides a strong foundation for the future development of ischemic cardiac injury drugs.
Collapse
|
20
|
Jung JE, Jang Y, Jeong HJ, Kim SJ, Park K, Oh DH, Yu A, Park CS, Han SJ. Discovery of 3,4-dihydropyrimido[4,5-d]pyrimidin-2(1H)-one and 3,4-dihydropyrido[2,3-d]pyrimidin-2(1H)-one derivatives as novel ENPP1 inhibitors. Bioorg Med Chem Lett 2022; 75:128947. [PMID: 35995398 DOI: 10.1016/j.bmcl.2022.128947] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 11/19/2022]
Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase-1 (ENPP1) negatively regulates the anti-cancer Stimulator of Interferon Genes (STING) pathway. We discovered that 3,4-dihydropyrimido[4,5-d]pyrimidin-2(1H)-one and 3,4-dihydropyrido[2,3-d]pyrimidin-2(1H)-one derivatives possessed inhibitory activities on ENPP1. A structure-activity relationship (SAR) study led to the identification of 46 and 23 as potent ENPP1 inhibitors. Also, compounds 46 and 23 possessed high microsomal stabilities in human, rat, and mouse liver microsome. Additionally, CYPs (1A2, 2C9, 2C19, 2D6, and 3A4) were not inhibited by 46 and 23. Molecular dynamics simulations provided an insight of binding modes between ENPP1 and compounds (46 and 23).
Collapse
Affiliation(s)
- Jae Eun Jung
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Yunseong Jang
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hee Jin Jeong
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sung Joon Kim
- Txinno Bioscience INC, 338 Gwanggyojungang-ro, Suji-gu, Yongin-si, Gyeonggi-do 16942, Republic of Korea
| | - Kichul Park
- OZIWORX. R&D Laboratory, 130-2, Donghwagongdan-ro, Wonju-si, Gangwon-do 26365, Republic of Korea
| | - Do Hee Oh
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Ahran Yu
- Txinno Bioscience INC, 338 Gwanggyojungang-ro, Suji-gu, Yongin-si, Gyeonggi-do 16942, Republic of Korea
| | - Chan Sun Park
- Txinno Bioscience INC, 338 Gwanggyojungang-ro, Suji-gu, Yongin-si, Gyeonggi-do 16942, Republic of Korea
| | - Seo-Jung Han
- Chemical & Biological Integrative Research Center, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
21
|
Arif M, Shabir G, Ejaz SA, Saeed A, Khan SU, Lecka J, Sévigny J, Iqbal J. Diacylhydrazine Derivatives of 2-(5-(Pyridin-3-yl)-2H-Tetrazol-2-yl)Acetohydrazide and 2-(5-(Pyridin-4-yl)-2H-Tetrazol-2-yl)Acetohydrazide as Potential Inhibitors of Nucleotide Pyrophosphatase. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2022. [DOI: 10.1134/s1068162022050053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Development of Novel Ecto-Nucleotide Pyrophosphatase/Phosphodiesterase 1 (ENPP1) Inhibitors for Tumor Immunotherapy. Int J Mol Sci 2022; 23:ijms23137104. [PMID: 35806118 PMCID: PMC9266353 DOI: 10.3390/ijms23137104] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/15/2022] Open
Abstract
The cyclic guanosine monophosphate–adenosine monophosphate synthase–stimulator of interferon genes–TANK-binding kinase 1–interferon regulating factor 3 (cGAS-STING-TBK1-IRF3) axis is now acknowledged as the major signaling pathway in innate immune responses. However, 2′,3′-cGAMP as a STING stimulator is easily recognized and degraded by ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which reduces the effect of tumor immunotherapy and promotes metastatic progression. In this investigation, the structure-based virtual screening strategy was adopted to discover eight candidate compounds containing zinc-binding quinazolin-4(3H)-one scaffold as ENPP1 inhibitors. Subsequently, these novel inhibitors targeting ENPP1 were synthesized and characterized by NMR and high-resolution mass spectra (HRMS). In bioassays, 7-fluoro-2-(((5-methoxy-1H-imidazo[4,5-b]pyridin-2-yl)thio)methyl)quina-zolin-4(3H)-one(compound 4e) showed excellent activity against the ENPP1 at the molecular and cellular levels, with IC50 values of 0.188 μM and 0.732 μM, respectively. Additionally, compound 4e had superior selectivity towards metastatic breast cancer cells (4T1) than towards normal cells (LO2 and 293T) in comparison with cisplatin, indicating that compound 4e can potentially be used in metastatic breast cancer therapy. On the other hand, compound 4e upgraded the expression levels of IFN-β in vivo by preventing the ENPP1 from hydrolyzing the cGAMP to stimulate a more potent innate immune response. Therefore, this compound might be applied to boost antitumor immunity for cancer immunotherapy. Overall, our work provides a strategy for the development of a promising drug candidate targeting ENPP1 for tumor immunotherapy.
Collapse
|
23
|
Bano S, Al-Rashida M, Alharthy RD, Khan IA, Iqbal J. Nucleotide pyrophosphatase/phosphodiesterases (NPPs) including NPP1 and NPP2/ ATX as important drug targets: A patent review (2015-2020). Expert Opin Ther Pat 2022; 32:743-751. [PMID: 35333684 DOI: 10.1080/13543776.2022.2058874] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Nucleoside triphosphate diphosphohydrolases (NTPDases), alkaline phosphatases (APs), and ecto-nucleotide pyrophosphatases/phosphodiesterases (NPPs) are nucleotidases found on the cell surface. It is a promising therapeutic target for a range of disorders, including fibrosis, tumour metastasis, pruritus, inflammation, multiple sclerosis, and autoimmune diseases. As a result, therapeutic intervention including selective inhibitors of NPPs is required. AREA COVERED Many chemical substances, including pyrazole, pyridine and bicyclic compounds have demonstrated promising inhibitory potential for ecto-nucleotide pyrophosphatase/phosphodiesterases. The chemistry and clinical applications of NPPs inhibitors patented between 2015 and 2020 are discussed in this review. EXPERT OPINION : In recent years, there has been a lot of effort put into finding effective and selective inhibitors of NPPs. Despite the fact that a variety of synthetic inhibitors have been created, only a few investigations on their in vivo activity have been published. In addition to IOA-289 which has passed Phase Ia clinical trials; potent ATX inhibitor compounds such as BLD-0409, IPF and BBT-877 have been placed in phase I clinical studies. Some of the most promising ATX inhibitors in recent years are closely related analogs of previously known inhibitors, such as PF-8380. Knowledge of the structure activity relationship of such promising inhibitors can potentially translate into the discovery of more potent and effective inhibitors of NPP with a variety of structural characteristics and favourable therapeutic activities.
Collapse
Affiliation(s)
- Sehrish Bano
- Center for Advanced Drug Research, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| | - Mariya Al-Rashida
- Department of Chemistry, Forman Christian College (A Chartered University), Ferozepur Road, Lahore 54600, Pakistan
| | - Rima D Alharthy
- Department of Chemistry, Science and Arts College, Rabigh Campus, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Imtiaz Ali Khan
- Department of Entomology, Agricultural University, Peshawar 25130, Khyber Pakhtunkhwa, Pakistan
| | - Jamshed Iqbal
- Center for Advanced Drug Research, COMSATS Institute of Information Technology, Abbottabad 22060, Pakistan
| |
Collapse
|
24
|
Zhou W, Tang J, Zhou X, Liu J. Tyrosinase inhibition by novel benzimidazole-thione Schiff base derivatives. LETT DRUG DES DISCOV 2022. [DOI: 10.2174/1570180819666220210100037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Tyrosinase is the enzyme responsible for the conversion of tyrosine to dopaquinone, which is related to melanoma, neurodegenerative disorders, freckles, pigmented acne and age spots. Controlling the tyrosinase activity could be an important way for treating overproduction of melanin.
Objective:
The development of safe and specific tyrosinase inhibitors could be used to treat hypermelanosis.
Methods:
5-nitro-1H-benzo[d]imidazole-2(3H)-thione was synthesized from 4-nitro-o-phenylenediamine and carbon disulfide. The nitro group of 5-nitro-1H- S-1 ESI-HRMS
benzo[d]imidazole-2(3H)-thione was reduced with iron powder. The 5-amino-1H-benzo[d]imidazole- 2(3H)-thione Schiff base derivatives were obtained by the reaction of 5-amino-1H-benzo[d]imidazole-2(3H)-thione with substituted benzaldehyde. The tyrosinase inhibitory activities were investigated. The studies of kinetic analysis, metal-chelating properties, docking and cytotoxicity were also performed.
Results:
All of the compounds showed strong tyrosinase inhibitory activities with 5-((4-nitrobenzylidene) amino)-1H-benzo [d]imidazole-2(3H)-thione (S-4) as the best tyrosinase inhibitor with an IC50 value of 4.8 ± 1.4 nM. Compound S-4 exhibited mixed type inhibition of mushroom tyrosinase, with Ki 15 nM and Kis 42 nM. Copper binding to S-4 was detected spectrophotometrically and 1-100 ìÌ S-4 displayed negligible cytotoxicity to murine B16 melanoma cells.
Conclusion:
Our results demonstrated that these benzimidazolethione Schiff base derivatives might be promising candidates as tyrosinase inhibitors.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Food and Chemical Engineering, Shaoyang University, Shao Shui Xi Road, Shaoyang 422100, China
| | - Junyuan Tang
- School of Pharmaceutical Sciences & State Key Laboratory of Respiratory Disease Guangzhou Medical University, Guangzhou,Guangdong 511436, China
| | - Xinchong Zhou
- Department of Food and Chemical Engineering, Shaoyang University, Shao Shui Xi Road, Shaoyang 422100, China
| | - Jinbing Liu
- Department of Food and Chemical Engineering, Shaoyang University, Shao Shui Xi Road, Shaoyang 422100, China
| |
Collapse
|
25
|
Zucolotto Cocca LH, Pelosi A, Sciuti LF, M. G. Abegão L, Kamada K, Piguel S, Renato Mendonça C, De Boni L. Two-photon brightness of highly fluorescent imidazopyridine derivatives: Two-photon and ultrafast transient absorption studies. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2021.118379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
26
|
Design, synthesis and biological evaluation studies of novel small molecule ENPP1 inhibitors for cancer immunotherapy. Bioorg Chem 2021; 119:105549. [PMID: 34929517 DOI: 10.1016/j.bioorg.2021.105549] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/25/2021] [Accepted: 12/05/2021] [Indexed: 01/07/2023]
Abstract
Ecto-nucleotide pyrophosphatase/phosphodiesterases 1 (ENPP1 or NPP1), is an attractive therapeutic target for various diseases, primarily cancer and mineralization disorders. The ecto-enzyme is located on the cell surface and has been implicated in the control of extracellular levels of nucleotide, nucleoside and (di) phosphate. Recently, it has emerged as a critical phosphodiesterase that hydrolyzes cyclic 2'3'- cGAMP, the endogenous ligand for STING (STimulator of INterferon Genes). STING plays an important role in innate immunity by activating type I interferon in response to cytosolic 2'3'-cGAMP. ENPP1 negatively regulates the STING pathway and hence its inhibition makes it an attractive therapeutic target for cancer immunotherapy. Herein, we describe the design, optimization and biological evaluation studies of a series of novel non-nucleotidic thioguanine based small molecule inhibitors of ENPP1. The lead compound 43 has shown good in vitro potency, stability in SGF/SIF/PBS, selectivity, ADME properties and pharmacokinetic profile and finally potent anti-tumor response in vivo. These compounds are a good starting point for the development of potentially effective cancer immunotherapy agents.
Collapse
|
27
|
Cao X, Cordova AF, Li L. Therapeutic Interventions Targeting Innate Immune Receptors: A Balancing Act. Chem Rev 2021; 122:3414-3458. [PMID: 34870969 DOI: 10.1021/acs.chemrev.1c00716] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The innate immune system is an organism's first line of defense against an onslaught of internal and external threats. The downstream adaptive immune system has been a popular target for therapeutic intervention, while there is a relative paucity of therapeutics targeting the innate immune system. However, the innate immune system plays a critical role in many human diseases, such as microbial infection, cancer, and autoimmunity, highlighting the need for ongoing therapeutic research. In this review, we discuss the major innate immune pathways and detail the molecular strategies underpinning successful therapeutics targeting each pathway as well as previous and ongoing efforts. We will also discuss any recent discoveries that could inform the development of novel therapeutic strategies. As our understanding of the innate immune system continues to develop, we envision that therapies harnessing the power of the innate immune system will become the mainstay of treatment for a wide variety of human diseases.
Collapse
|
28
|
Dorababu A. Pharmacological report of recently designed multifunctional coumarin and coumarin-heterocycle derivatives. Arch Pharm (Weinheim) 2021; 355:e2100345. [PMID: 34693550 DOI: 10.1002/ardp.202100345] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/04/2021] [Accepted: 10/06/2021] [Indexed: 12/23/2022]
Abstract
Coumarin is a naturally available molecule and has been identified as a potent pharmacophore due to its pharmacological activity. Because of this, coumarin has been exploited synthetically to prepare a wide range of derivatives. In fact, most coumarin derivatives have been found to be less toxic, which is the most essential property for a drug molecule. Such molecules are being prepared for therapeutic use as broad-spectrum pharmacological agents. Microbial diseases including viral diseases have become very common and are responsible for many deaths worldwide. In particular, microbial drug resistance is a problem that needs to be tackled in an effective manner. Also, for Alzheimer's disease, which affects most elderly persons, no efficient chemotherapy exists. In addition, although diabetes, a metabolic syndrome, can be treated with many drugs, there is no complete cure. Thus, more potent antidiabetic agents are required for the management of diabetes. Likewise, for the treatment of a wide range of ailments caused by microbes, genetic factors, or lifestyle-related factors, an efficient drug regimen is needed. In view of this, coumarin derivatives are designed and evaluated. Here, coumarin derivatives that have been reported recently are compiled, classified and evaluated critically. This study briefly takes the structure-activity relationship into consideration and suggests the next suitable step. With a focus on the most potent molecules, the pharmacological activity of the evaluated molecules is described.
Collapse
Affiliation(s)
- Atukuri Dorababu
- Department of Chemistry, SRMPP Government First Grade College, Huvinahadagali, Karnataka, India
| |
Collapse
|
29
|
Flemmich L, Moreno S, Micura R. Synthesis of O 6-alkylated preQ 1 derivatives. Beilstein J Org Chem 2021; 17:2295-2301. [PMID: 34621392 PMCID: PMC8450960 DOI: 10.3762/bjoc.17.147] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 08/27/2021] [Indexed: 12/23/2022] Open
Abstract
A naturally occurring riboswitch can utilize 7-aminomethyl-O 6-methyl-7-deazaguanine (m6preQ1) as cofactor for methyl group transfer resulting in cytosine methylation. This recently discovered riboswitch-ribozyme activity opens new avenues for the development of RNA labeling tools based on tailored O 6-alkylated preQ1 derivatives. Here, we report a robust synthesis for this class of pyrrolo[2,3-d]pyrimidines starting from readily accessible N 2-pivaloyl-protected 6-chloro-7-cyano-7-deazaguanine. Substitution of the 6-chloro atom with the alcoholate of interest proceeds straightforward. The transformation of the 7-cyano substituent into the required aminomethyl group turned out to be challenging and was solved by a hydration reaction sequence on a well-soluble dimethoxytritylated precursor via in situ oxime formation. The synthetic path now provides a solid foundation to access O 6-alkylated 7-aminomethyl-7-deazaguanines for the development of RNA labeling tools based on the preQ1 class-I riboswitch scaffold.
Collapse
Affiliation(s)
- Laurin Flemmich
- Institute of Organic Chemistry, Center for molecular Biosciences Innsbruck (CMBI), Innrain 80–82, 6020 Innsbruck, Austria
| | - Sarah Moreno
- Institute of Organic Chemistry, Center for molecular Biosciences Innsbruck (CMBI), Innrain 80–82, 6020 Innsbruck, Austria
| | - Ronald Micura
- Institute of Organic Chemistry, Center for molecular Biosciences Innsbruck (CMBI), Innrain 80–82, 6020 Innsbruck, Austria
| |
Collapse
|
30
|
Access to azolopyrimidine-6,7-diamines as a valuable “building-blocks” to develop new fused heteroaromatic systems. Tetrahedron 2021. [DOI: 10.1016/j.tet.2021.132172] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
31
|
Ullah S, El-Gamal MI, El-Gamal R, Pelletier J, Sévigny J, Shehata MK, Anbar HS, Iqbal J. Synthesis, biological evaluation, and docking studies of novel pyrrolo[2,3-b]pyridine derivatives as both ectonucleotide pyrophosphatase/phosphodiesterase inhibitors and antiproliferative agents. Eur J Med Chem 2021; 217:113339. [PMID: 33744686 DOI: 10.1016/j.ejmech.2021.113339] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 11/17/2022]
Abstract
Ecto-nucleotide pyrophosphatases/phosphodiesterases (NPPs) together with nucleoside triphosphate diphosphohydrolases (NTPDases) and alkaline phosphatases (APs) are nucleotidases located at the surface of the cells. NPP1 and NPP3 are important members of NPP family that are known as druggable targets for a number of disorders such as impaired calcification, type 2 diabetes, and cancer. Sulfonylurea derivatives have been reported as antidiabetic and anticancer agents, therefore, we synthesized and investigated series of sulfonylurea derivatives 1a-m possessing pyrrolo[2,3-b]pyridine core as inhibitors of NPP1 and NPP3 isozymes that are over-expressed in cancer and diabetes. The enzymatic evaluation highlighted compound 1a as selective NPP1 inhibitor, however, 1c was observed as the most potent inhibitor of NPP1 with an IC50 value of 0.80 ± 0.04 μM. Compound 1l was found to be the most potent and moderately selective inhibitor of NPP3 (IC50 = 0.55 ± 0.01 μM). Furthermore, in vitro cytotoxicity assays of compounds 1a-m against MCF-7 and HT-29 cancer cell lines exhibited compound 1c (IC50 = 4.70 ± 0.67 μM), and 1h (IC50 = 1.58 ± 0.20 μM) as the most cytotoxic compounds against MCF-7 and HT-29 cancer cell lines, respectively. Both of the investigated compounds showed high degree of selectivity towards cancer cells than normal cells (WI-38). Molecular docking studies of selective and potent enzyme inhibitors revealed promising mode of interactions with important binding sites residues of both isozymes i.e., Thr256, His380, Lys255, Asn277 residues of NPP1 and His329, Thr205, and Leu239 residues of NPP3. In addition, the most potent antiproliferative agent, compound 1h, doesn't produce hypoglycemia as a side effect when injected to mice. This is an additional merit of the promising compound 1h.
Collapse
Affiliation(s)
- Saif Ullah
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura, 35516, Egypt
| | - Randa El-Gamal
- Department of Medical Biochemistry, Faculty of Medicine, University of Mansoura, Mansoura, 35516, Egypt
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, G1V 4G2, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec - Université Laval, Québec, QC, G1V 4G2, Canada; Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Mahmoud K Shehata
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Hanan S Anbar
- Department of Clinical Pharmacy and Pharmacotherapeutics, Dubai Pharmacy College for Girls, Dubai, 19099, United Arab Emirates.
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan.
| |
Collapse
|
32
|
Kumar M, Lowery RG. Development of a High-Throughput Assay to Identify Inhibitors of ENPP1. SLAS DISCOVERY 2021; 26:740-746. [PMID: 33402044 DOI: 10.1177/2472555220982321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The innate immune response to cancer is initiated by cytosolic DNA, where it binds to cGAS and triggers type I interferon (IFN) expression via the STING receptor, leading to activation of tumor-specific T cells. Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1) has been identified as the primary enzyme responsible for degrading cGAMP, and therefore it is under intense investigation as a therapeutic target for cancer immunotherapy. ENPP1 hydrolyzes cGAMP to produce AMP and GMP, and hydrolyzes ATP and other nucleotides to monophosphates and pyrophosphate. We developed a robust, high-throughput screening (HTS)-compatible enzymatic assay method for ENPP1 using the Transcreener AMP2/GMP2 Assay, a competitive fluorescence polarization (FP) immunoassay that enables direct detection of AMP and GMP in a homogenous format. The monoclonal antibody used in the Transcreener AMP2/GMP2 Assay showed more than 104-fold selectivity for AMP and GMP versus cGAMP, and 3000-fold selectivity for AMP over ATP, indicating that the assay can be used for detection at initial velocity with either substrate. A working concentration of 100 pM ENPP1 was determined as optimal with a 60 min reaction period, enabling screening with very low quantities of enzyme. A Z' value of 0.72 was determined using ATP as substrate, indicating a high-quality assay. Consistent with previous studies, we found that ENPP1 preferred ATP as a substrate when compared with other nucleotides like GTP, ADP, and GDP. ENPP1 showed a 20-fold selectivity for 2'3'cGAMP compared with 2'3'c-diGMP and showed no activity with 3'3'c-diAMP. The Transcreener AMP2/GMP2 Assay should prove to be a valuable tool for the discovery of ENPP1 lead molecules.
Collapse
|
33
|
Kanwal A, Ullah S, Ahmad M, Pelletier J, Aslam S, Sultan S, Sévigny J, Iqbal M, Iqbal J. Synthesis and Nucleotide Pyrophosphatase/Phosphodiesterase Inhibition Studies of Carbohydrazides Based on Benzimidazole‐Benzothiazine Skeleton. ChemistrySelect 2020. [DOI: 10.1002/slct.202003479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Afshan Kanwal
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Saif Ullah
- Centre for Advanced Drug Research COMSATS University Islamabad, Abbottabad Campus Abbottabad 22060 Pakistan
| | - Matloob Ahmad
- Department of Chemistry Government College University Faisalabad 38000 Pakistan
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec- Université Laval Québec, QC G1 V 4G2 Canada
| | - Sana Aslam
- Department of Chemistry Government College Women University Faisalabad 38000 Pakistan
| | - Sadia Sultan
- Faculty of Pharmacy Universiti Teknologi MARA, Puncak Alam Campus 42300 Bandar Puncak Alam Selangor Darul Ehsan Malaysia
- Atta-ur-Rahman Institute for Natural Products Discovery (AuRIns) Universiti Teknologi MARA, Puncak Alam Campus 42300 Bandar Puncak Alam Selangor Darul Ehsan Malaysia
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec- Université Laval Québec, QC G1 V 4G2 Canada
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine Université Laval Québec, QC G1 V 0 A6 Canada
| | - Mazhar Iqbal
- Drug Discovery and Structural Biology Group Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering Faisalabad 38000 Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research COMSATS University Islamabad, Abbottabad Campus Abbottabad 22060 Pakistan
| |
Collapse
|
34
|
Carozza JA, Brown JA, Böhnert V, Fernandez D, AlSaif Y, Mardjuki RE, Smith M, Li L. Structure-Aided Development of Small-Molecule Inhibitors of ENPP1, the Extracellular Phosphodiesterase of the Immunotransmitter cGAMP. Cell Chem Biol 2020; 27:1347-1358.e5. [PMID: 32726585 PMCID: PMC7680421 DOI: 10.1016/j.chembiol.2020.07.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/28/2020] [Accepted: 07/08/2020] [Indexed: 11/23/2022]
Abstract
Cancer cells initiate an innate immune response by synthesizing and exporting the small-molecule immunotransmitter cGAMP, which activates the anti-cancer Stimulator of Interferon Genes (STING) pathway in the host. An extracellular enzyme, ectonucleotide pyrophosphatase phosphodiesterase 1 (ENPP1), hydrolyzes cGAMP and negatively regulates this anti-cancer immune response. Small-molecule ENPP1 inhibitors are much needed as tools to study the basic biology of extracellular cGAMP and as investigational cancer immunotherapy drugs. Here, we surveyed structure-activity relationships around a series of cell-impermeable and thus extracellular-targeting phosphonate inhibitors of ENPP1. In addition, we solved the crystal structure of an exemplary phosphonate inhibitor to elucidate the interactions that drive potency. This study yielded several best-in-class inhibitors with Ki < 2 nM and excellent physicochemical and pharmacokinetic properties. Finally, we demonstrate that an ENPP1 inhibitor delays tumor growth in a breast cancer mouse model. Together, we have developed ENPP1 inhibitors that are excellent tool compounds and potential therapeutics.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/chemical synthesis
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Cell Survival/drug effects
- Cells, Cultured
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Enzyme Inhibitors/chemical synthesis
- Enzyme Inhibitors/chemistry
- Enzyme Inhibitors/pharmacology
- Female
- Humans
- Mice
- Mice, Inbred C57BL
- Molecular Structure
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- Neurotransmitter Agents/chemistry
- Neurotransmitter Agents/isolation & purification
- Neurotransmitter Agents/metabolism
- Nucleotides, Cyclic/chemistry
- Nucleotides, Cyclic/isolation & purification
- Nucleotides, Cyclic/metabolism
- Phosphoric Diester Hydrolases/metabolism
- Pyrophosphatases/antagonists & inhibitors
- Pyrophosphatases/metabolism
- Small Molecule Libraries/chemical synthesis
- Small Molecule Libraries/chemistry
- Small Molecule Libraries/pharmacology
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Jacqueline A Carozza
- Department of Chemistry, Stanford University, Stanford, CA 93405, USA; Stanford ChEM-H, Stanford University, Stanford, CA 93405, USA
| | - Jenifer A Brown
- Stanford ChEM-H, Stanford University, Stanford, CA 93405, USA; Biophysics Program, Stanford University, Stanford, CA 93405, USA
| | - Volker Böhnert
- Stanford ChEM-H, Stanford University, Stanford, CA 93405, USA; Department of Biochemistry, Stanford University, Stanford, CA 93405, USA
| | - Daniel Fernandez
- Stanford ChEM-H Macromolecular Structure Knowledge Center, Stanford University, Stanford, CA 93405, USA
| | - Yasmeen AlSaif
- Stanford ChEM-H, Stanford University, Stanford, CA 93405, USA; Department of Biology, Stanford University, Stanford, CA 93405, USA
| | - Rachel E Mardjuki
- Department of Chemistry, Stanford University, Stanford, CA 93405, USA; Stanford ChEM-H, Stanford University, Stanford, CA 93405, USA
| | - Mark Smith
- Stanford ChEM-H Medicinal Chemistry Knowledge Center, Stanford, CA 93405, USA
| | - Lingyin Li
- Stanford ChEM-H, Stanford University, Stanford, CA 93405, USA; Department of Biochemistry, Stanford University, Stanford, CA 93405, USA.
| |
Collapse
|
35
|
Ahmad H, Ullah S, Rahman F, Saeed A, Pelletier J, Sévigny J, Hassan A, Iqbal J. Synthesis of biphenyl oxazole derivatives via Suzuki coupling and biological evaluations as nucleotide pyrophosphatase/phosphodiesterase-1 and -3 inhibitors. Eur J Med Chem 2020; 208:112759. [PMID: 32883636 DOI: 10.1016/j.ejmech.2020.112759] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/09/2020] [Accepted: 08/14/2020] [Indexed: 01/09/2023]
Abstract
Oxazole derivatives are important medicinal compounds which are inhibitors of various enzymes such as NPP1, NPP2, NPP3, tyrosine kinase, dipeptidyl-peptidase IV, cyclooxygenase-2, and protein tyrosine phosphatase. In this study, an extensive range of new biologically active biphenyl oxazole derivatives was synthesized in high to excellent yields (57-93%) through Suzuki-Miyaura cross-coupling of bromophenyloxazole with different boronic acids. The reaction was carried out in wet toluene under mild conditions. Overexpression of nucleotide pyrophosphatase/phosphodiesterase-1 (NPP1) and NPP3 has been associated with various health disorders including chondrocalcinosis, cancer, osteoarthritis, and type 2 diabetes. We evaluated the inhibitory potential and selectivity of the synthesized compounds (3a-3q) towards NPP1 and NPP3 at 100 μM concentrations. We found two compounds that were selective and potent inhibitors of these two enzymes on the artificial substrate thymidine 5'-monophosphate para-nitrophenyl ester: compound 3n inhibited NPP1 with an IC50 of 0.15 μM, and compound 3f inhibited NPP3 with an IC50 value of 0.17 μM. The compounds with promising inhibitory potential were docked inside the proteins of NPP1 and NPP3 isozymes to get insight into the plausible binding interactions with active site residues.
Collapse
Affiliation(s)
- Haseen Ahmad
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Saif Ullah
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Fouzia Rahman
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Julie Pelletier
- Centre de Recherche Du CHU de Québec - Université Laval, Québec, QC, G1V 4G2, Canada
| | - Jean Sévigny
- Centre de Recherche Du CHU de Québec - Université Laval, Québec, QC, G1V 4G2, Canada; Département de Microbiologie-infectiologie et D'immunologie, Faculté de Médecine, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Abbas Hassan
- Department of Chemistry, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan.
| |
Collapse
|
36
|
Synthesis, biological evaluation, and docking studies of new pyrazole-based thiourea and sulfonamide derivatives as inhibitors of nucleotide pyrophosphatase/phosphodiesterase. Bioorg Chem 2020; 99:103783. [DOI: 10.1016/j.bioorg.2020.103783] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 03/16/2020] [Accepted: 03/20/2020] [Indexed: 11/22/2022]
|
37
|
van der Zanden SY, Luimstra JJ, Neefjes J, Borst J, Ovaa H. Opportunities for Small Molecules in Cancer Immunotherapy. Trends Immunol 2020; 41:493-511. [PMID: 32381382 DOI: 10.1016/j.it.2020.04.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 01/08/2023]
Abstract
Cancer immunotherapy has proven remarkably successful through instigation of systemic antitumor T cell responses. Despite this achievement, further advancements are needed to expand the scope of susceptible cancer types and overcome variation in treatment outcomes between patients. Small-molecule drugs targeting defined pathways and/or cells capable of immune modulation are expected to substantially improve efficacy of cancer immunotherapy. Small-molecule drugs possess unique properties compatible with systemic administration and amenable to both extracellular and intracellular targets. These compounds can modify molecular pathways to overcome immune tolerance and suppression towards effective antitumor responses. Here, we provide an overview of how such effects might be achieved by combining immunotherapy with conventional and/or new small-molecule chemotherapeutics.
Collapse
Affiliation(s)
- Sabina Y van der Zanden
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Jolien J Luimstra
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| | - Jannie Borst
- Oncode Institute, Utrecht, The Netherlands; Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Huib Ovaa
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands; Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
38
|
Onyedibe KI, Wang M, Sintim HO. ENPP1, an Old Enzyme with New Functions, and Small Molecule Inhibitors-A STING in the Tale of ENPP1. Molecules 2019; 24:molecules24224192. [PMID: 31752288 PMCID: PMC6891441 DOI: 10.3390/molecules24224192] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/07/2019] [Accepted: 11/13/2019] [Indexed: 11/16/2022] Open
Abstract
Ectonucleotide pyrophosphatase/phosphodiesterase I (ENPP1) was identified several decades ago as a type II transmembrane glycoprotein with nucleotide pyrophosphatase and phosphodiesterase enzymatic activities, critical for purinergic signaling. Recently, ENPP1 has emerged as a critical phosphodiesterase that degrades the stimulator of interferon genes (STING) ligand, cyclic GMP-AMP (cGAMP). cGAMP or analogs thereof have emerged as potent immunostimulatory agents, which have potential applications in immunotherapy. This emerging role of ENPP1 has placed this "old" enzyme at the frontier of immunotherapy. This review highlights the roles played by ENPP1, the mechanism of cGAMP hydrolysis by ENPP1, and small molecule inhibitors of ENPP1 with potential applications in diverse disease states, including cancer.
Collapse
Affiliation(s)
- Kenneth I. Onyedibe
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; (K.I.O.); (M.W.)
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, IN 47907, USA
| | - Modi Wang
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; (K.I.O.); (M.W.)
| | - Herman O. Sintim
- Chemistry Department, Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA; (K.I.O.); (M.W.)
- Purdue Institute for Inflammation, Immunology, and Infectious Diseases, West Lafayette, IN 47907, USA
- Purdue University Center for Cancer Research, West Lafayette, IN 47907, USA
- Correspondence: ; Tel.: +1-(765)-496-6078; Fax: +1-(765)-494-0239
| |
Collapse
|
39
|
Structure-activity relationship study of NPP1 inhibitors based on uracil-N1-(methoxy)ethyl-β-phosphate scaffold. Eur J Med Chem 2019; 184:111754. [PMID: 31610377 DOI: 10.1016/j.ejmech.2019.111754] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 09/29/2019] [Accepted: 10/02/2019] [Indexed: 02/06/2023]
Abstract
Overexpression of ecto-nucleotide pyrophosphatase-1 (NPP1) is associated with diseases such as calcium pyrophosphate dihydrate deposition disease, calcific aortic valve disease, and type 2 diabetes. In this context, NPP1 inhibitors are potential drug candidates for the treatment of these diseases. The present study focuses on the analysis of the structure-activity relationship of NPP1 inhibitors based on acyclic uracil-nucleotides. For this purpose, we synthesized acyclic uridine-monophosphate analogs, 10-11, uridine-diphosphate analogs, 12-14, and uridine-Pα,α-dithio-triphosphate analogs, 15-17. We evaluated their inhibitory activity and selectivity towards NPP1, -3, NTPDase1, -2, -3, and -8, and P2Y2,4,6 receptors. Analogs 16 and 17 were the most selective and potent NPP1 inhibitors (Ki 0.94 and 0.73 μM, respectively) among the tested molecules. Analogs 10-17 had only minute effect on uracil-nucleotide responding P2Y2,4,6 receptors. Analog 17 (100 μM) displayed 96% inhibition of NPPase activity in osteoarthritic human chondrocytes. Analogs 14-17 displayed weak inhibitory effect on alkaline phosphatase activity at equimolar concentrations in human chondrocytes. All tested analogs showed no toxicity at human chondrocytes. We concluded that ribose-ring to chain transformation, as well as the type of the nucleobase, are parameters of minor significance to NPP1 inhibition, whereas the major parameter is Pα-dithio-substitution. In addition, the length of the phosphate chain also significantly affects inhibition. Overall, the experimental results were well reproduced by molecular docking. A correlation was observed between the activities of the compounds and the number of H-bonds and salt bridges formed between the inhibitors and NPP1 binding site residues. Uracil-N1-(methoxy)ethyl-β-Pα,α-dithio, Pβ,γ-methylene tri-phosphate, 17, was identified as the most potent, selective, and non-toxic NPP1 inhibitor among the tested analogs, and may be used as a lead structure for further drug development.
Collapse
|
40
|
Kawaguchi M, Han X, Hisada T, Nishikawa S, Kano K, Ieda N, Aoki J, Toyama T, Nakagawa H. Development of an ENPP1 Fluorescence Probe for Inhibitor Screening, Cellular Imaging, and Prognostic Assessment of Malignant Breast Cancer. J Med Chem 2019; 62:9254-9269. [DOI: 10.1021/acs.jmedchem.9b01213] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Mitsuyasu Kawaguchi
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Xiang Han
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Tomoka Hisada
- Graduate School of Medical Sciences, Department of Breast Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Sayaka Nishikawa
- Graduate School of Medical Sciences, Department of Breast Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Kuniyuki Kano
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Naoya Ieda
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Junken Aoki
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Tatsuya Toyama
- Graduate School of Medical Sciences, Department of Breast Surgery, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hidehiko Nakagawa
- Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| |
Collapse
|
41
|
Clausse V, Tao D, Debnath S, Fang Y, Tagad HD, Wang Y, Sun H, LeClair CA, Mazur SJ, Lane K, Shi ZD, Vasalatiy O, Eells R, Baker LK, Henderson MJ, Webb MR, Shen M, Hall MD, Appella E, Appella DH, Coussens NP. Physiologically relevant orthogonal assays for the discovery of small-molecule modulators of WIP1 phosphatase in high-throughput screens. J Biol Chem 2019; 294:17654-17668. [PMID: 31481464 PMCID: PMC6873202 DOI: 10.1074/jbc.ra119.010201] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/30/2019] [Indexed: 01/07/2023] Open
Abstract
WT P53-Induced Phosphatase 1 (WIP1) is a member of the magnesium-dependent serine/threonine protein phosphatase (PPM) family and is induced by P53 in response to DNA damage. In several human cancers, the WIP1 protein is overexpressed, which is generally associated with a worse prognosis. Although WIP1 is an attractive therapeutic target, no potent, selective, and bioactive small-molecule modulator with favorable pharmacokinetics has been reported. Phosphatase enzymes are among the most challenging targets for small molecules because of the difficulty of achieving both modulator selectivity and bioavailability. Another major obstacle has been the availability of robust and physiologically relevant phosphatase assays that are suitable for high-throughput screening. Here, we describe orthogonal biochemical WIP1 activity assays that utilize phosphopeptides from native WIP1 substrates. We optimized an MS assay to quantify the enzymatically dephosphorylated peptide reaction product in a 384-well format. Additionally, a red-shifted fluorescence assay was optimized in a 1,536-well format to enable real-time WIP1 activity measurements through the detection of the orthogonal reaction product, Pi. We validated these two optimized assays by quantitative high-throughput screening against the National Center for Advancing Translational Sciences (NCATS) Pharmaceutical Collection and used secondary assays to confirm and evaluate inhibitors identified in the primary screen. Five inhibitors were further tested with an orthogonal WIP1 activity assay and surface plasmon resonance binding studies. Our results validate the application of miniaturized physiologically relevant and orthogonal WIP1 activity assays to discover small-molecule modulators from high-throughput screens.
Collapse
Affiliation(s)
- Victor Clausse
- Synthetic Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Subrata Debnath
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhong Fang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Harichandra D Tagad
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Yuhong Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Hongmao Sun
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Christopher A LeClair
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Sharlyn J Mazur
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Kelly Lane
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Zhen-Dan Shi
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Olga Vasalatiy
- Imaging Probe Development Center, NHLBI, National Institutes of Health, Rockville, Maryland 20850
| | - Rebecca Eells
- Reaction Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355
| | - Lynn K Baker
- Reaction Biology Corporation, 1 Great Valley Parkway, Suite 2, Malvern, Pennsylvania 19355
| | - Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Martin R Webb
- Francis Crick Institute, 1 Midland Road, London NW1 AT, United Kingdom
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| | - Ettore Appella
- Laboratory of Cell Biology, Center for Cancer Research, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Daniel H Appella
- Synthetic Bioactive Molecules Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892
| | - Nathan P Coussens
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland 20850
| |
Collapse
|
42
|
Nassir M, Arad U, Lee SY, Journo S, Mirza S, Renn C, Zimmermann H, Pelletier J, Sévigny J, Müller CE, Fischer B. Identification of adenine-N9-(methoxy)ethyl-β-bisphosphonate as NPP1 inhibitor attenuates NPPase activity in human osteoarthritic chondrocytes. Purinergic Signal 2019; 15:247-263. [PMID: 31025169 DOI: 10.1007/s11302-019-09649-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/06/2019] [Indexed: 12/17/2022] Open
Abstract
Overproduction of extracellular diphosphate due to hydrolysis of ATP by NPP1 leads to pathological calcium diphosphate (pyrophosphate) dihydrate deposition (CPPD) in cartilage, resulting in a degenerative joint disease that today lacks a cure. Here, we targeted the identification of novel NPP1 inhibitors as potential therapeutic agents for CPPD deposition disease. Specifically, we synthesized novel analogs of AMP (NPP1 reaction product) and ADP (NPP1 inhibitor). These derivatives incorporate several chemical modifications of the natural nucleotides including (1) a methylene group replacing the Pα,β-bridging oxygen atom to provide metabolic resistance, (2) sulfonate group(s) replacing phosphonate(s) to improve binding to NPP1's catalytic zinc ions, (3) an acyclic nucleotide analog to allow flexible binding in the NPP1 catalytic site, and (4) a benzimidazole base replacing adenine. Among the investigated compounds, adenine-N9-(methoxy)ethyl-β-bisphosphonate, 10, was identified as an NPP1 inhibitor (Ki 16.3 μM vs. the artificial substrate p-nitrophenyl thymidine-5'-monophosphate (p-Nph-5'-TMP), and 9.60 μM vs. the natural substrate, ATP). Compound 10 was selective for NPP1 vs. human NPP3, human CD39, and tissue non-specific alkaline phosphatase (TNAP), but also inhibited human CD73 (Ki 12.6 μM). Thus, 10 is a dual NPP1/CD73 inhibitor, which could not only be of interest for treating CPPD deposition disease and calcific aortic valve disease but may also be considered for the immunotherapy of cancer. Compound 10 proved to be a promising inhibitor, which almost completely reduces NPPase activity in human osteoarthritic chondrocytes at a concentration of 100 μM.
Collapse
Affiliation(s)
- Molhm Nassir
- Department of Chemistry, Bar-Ilan University, 52900, Ramat-Gan, Israel
| | - Uri Arad
- Department of Rheumatology, Tel Aviv Medical Center and the Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Sang-Yong Lee
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Shani Journo
- Department of Rheumatology, Tel Aviv Medical Center and the Faculty of Medicine, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Salahuddin Mirza
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Christian Renn
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Herbert Zimmermann
- Institute of Cell Biology and Neuroscience, Goethe-University, 60438, Frankfurt am Main, Germany
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec - Université Laval, Québec City, QC, Canada.,Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, QC, Canada
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn, Bonn, Germany
| | - Bilha Fischer
- Department of Chemistry, Bar-Ilan University, 52900, Ramat-Gan, Israel.
| |
Collapse
|
43
|
Probing the high potency of pyrazolyl pyrimidinetriones and thioxopyrimidinediones as selective and efficient non-nucleotide inhibitors of recombinant human ectonucleotidases. Bioorg Chem 2019; 88:102893. [PMID: 30986550 DOI: 10.1016/j.bioorg.2019.03.067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 03/19/2019] [Accepted: 03/25/2019] [Indexed: 11/22/2022]
Abstract
With the aim to discover novel, efficient and selective inhibitors of human alkaline phosphatase and nucleotide pyrophosphatase enzymes, two new series of pyrazolyl pyrimidinetriones (PPTs) (6a-g) and thioxopyrimidinediones (PTPs) (6h-n) were synthesized in good chemical yields using Knoevenagel condensation reaction between pyrazole carbaldehydes (4a-g) and pharmacologically active N-alkylated pyrimidinetrione (5a) and thioxopyrimidinedione (5b). The inhibition potential of the synthesized hybrid compounds was evaluated against human alkaline phosphatase (h-TNAP and h-IAP) and ectonucleotidase (h-NPP1 and h-NPP3) enzymes. Most of the tested analogs were highly potent with a variable degree of inhibition depending on the functionalized hybrid structure. The detailed structure-activity relationship (SAR) of PPT and PTP derivatives suggested that the compound with unsubstituted phenyl ring from PPT series led to selective and potent inhibition (6a; IC50 = 0.33 ± 0.02 µM) of h-TNAP, whereas compound 6c selectively inhibited h-IAP isozyme with IC50 value of 0.86 ± 0.04 µM. Similarly, compounds 6b and 6h were identified as the lead scaffolds against h-NPP1 and h-NPP3, respectively. The probable binding modes for the most potent inhibitors were elucidated through molecular docking analysis. Structure-activity relationships, mechanism of action, cytotoxic effects and druglikeness properties are also discussed.
Collapse
|
44
|
Hwu JR, Huang WC, Lin SY, Tan KT, Hu YC, Shieh FK, Bachurin SO, Ustyugov A, Tsay SC. Chikungunya virus inhibition by synthetic coumarin-guanosine conjugates. Eur J Med Chem 2019; 166:136-143. [PMID: 30703657 DOI: 10.1016/j.ejmech.2019.01.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 10/18/2018] [Accepted: 01/15/2019] [Indexed: 01/09/2023]
Abstract
Since its discovery in Tanganyika, Africa in 1952, chikungunya virus (CHIKV) outbreaks have occurred in Africa, Asia, Europe, and America. Till now chikungunya fever has spread in nearly 40 countries. Because of lack of effective vaccines and antiviral drugs to intervene this disease, 21 new conjugated compounds were designed and synthesized by coupling of 6,8-dithioguanosine at its C-6 position with 3-(chloromethyl)coumarins bearing an F, Cl, Br, Me, or -OMe substituent through the -SCH2- joint. Meanwhile, an organic "dummy" ligand (e.g., methyl, benzyl, and naphthylmethyl) or a coumarinyl moiety was attached at the C-8 position. By high through-put screening, three of these new conjugates were found to inhibit CHIKV in Vero cells with significant potency (EC50 = 9.9-13.9 μM) and showed low toxicity (CC50 = 96.5-212 μM). The selectivity index values were 9.37-21.7. Their structure-activity relationship was deduced, which indicates that the coumarin moiety is essential and the presence of a -OMe group enhances the antiviral activity.
Collapse
Affiliation(s)
- Jih Ru Hwu
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300, Taiwan; Department of Chemistry, National Central University, Jhongli City, Taoyuan, 320, Taiwan.
| | - Wen-Chieh Huang
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Shu-Yu Lin
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Kui-Thong Tan
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Yu-Chen Hu
- Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300, Taiwan; Department of Chemical Engineering, National Tsing Hua University, Hsinchu, 300, Taiwan
| | - Fa-Kuen Shieh
- Department of Chemistry, National Central University, Jhongli City, Taoyuan, 320, Taiwan
| | - Sergey O Bachurin
- The Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Alexey Ustyugov
- The Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, 142432, Russia
| | - Shwu-Chen Tsay
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300, Taiwan; Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, 300, Taiwan; Department of Chemistry, National Central University, Jhongli City, Taoyuan, 320, Taiwan.
| |
Collapse
|
45
|
Design, synthesis, biological evaluation and cellular imaging of imidazo[4,5-b]pyridine derivatives as potent and selective TAM inhibitors. Bioorg Med Chem 2018; 26:5510-5530. [PMID: 30309671 DOI: 10.1016/j.bmc.2018.09.031] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 09/17/2018] [Accepted: 09/23/2018] [Indexed: 12/14/2022]
Abstract
The TAM kinase family arises as a new effective and attractive therapeutic target for cancer therapy, autoimmune and viral diseases. A series of 2,6-disubstituted imidazo[4,5-b]pyridines were designed, synthesized and identified as highly potent TAM inhibitors. Despite remarkable structural similarities within the TAM family, compounds 28 and 25 demonstrated high activity and selectivity in vitro against AXL and MER, with IC50 value of 0.77 nM and 9 nM respectively and a 120- to 900-fold selectivity. We also observed an unexpected nuclear localization for compound 10Bb, thanks to nanoSIMS technology, which could be correlated to the absence of cytotoxicity on three different cancer cell lines being sensitive to TAM inhibition.
Collapse
|
46
|
Deazapurine Analogues Bearing a 1 H-Pyrazolo[3,4- b]pyridin-3(2 H)-one Core: Synthesis and Biological Activity. European J Org Chem 2018. [DOI: 10.1002/ejoc.201800163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
47
|
Jiang H, Dong X, Jin X, Zhu D, Yin R, Yu R, Wan S, Zhang L, Jiang T. Efficient Synthesis of Five Types of Heterocyclic Compounds via Intramolecular Elimination Using Ultrasound-Static Heating Technique. Chem Asian J 2018; 13:2009-2013. [PMID: 29858560 DOI: 10.1002/asia.201800682] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/29/2018] [Indexed: 11/10/2022]
Abstract
An experimental technique, ultrasound-static heating, has been developed for the efficient synthesis of heterocyclic compounds. The technique involves ultrasonic irradiation and static heating processes. First, the ultrasonic irradiation process is performed to form an intermediate of the heterocyclic compound under mild conditions and the subsequent static heating process (heating the intermediate under solvent-free conditions without stirring) produces the target heterocyclic compounds via intramolecular elimination.
Collapse
Affiliation(s)
- Hongfei Jiang
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
| | - Xueyang Dong
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
| | - Xin Jin
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
| | - Danyang Zhu
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
| | - Ruijuan Yin
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Rilei Yu
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| | - Shengbiao Wan
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
| | - Lijuan Zhang
- Institute of Cerebrovascular Diseases, Affiliated Hospital of Qingdao University Medical College, Qingdao, 266500, P. R. China
| | - Tao Jiang
- Key Laboratory of Marine Drugs, Chinese, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, P. R. China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266237, P. R. China
| |
Collapse
|
48
|
Lee SY, Müller CE. Nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1) and its inhibitors. MEDCHEMCOMM 2017; 8:823-840. [PMID: 30108800 PMCID: PMC6072468 DOI: 10.1039/c7md00015d] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 02/08/2017] [Indexed: 01/22/2023]
Abstract
Ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1, EC 3.1.4.1) is a metalloenzyme that belongs to the NPP family, which comprises seven subtypes (NPP1-7). NPP1 hydrolyzes a wide range of phosphodiester bonds, e.g. in nucleoside triphosphates, (cyclic) dinucleotides, and nucleotide sugars yielding nucleoside 5'-monophosphates as products. Its main substrate is ATP which is cleaved to AMP and diphosphate. The enzyme is involved in various biological processes including bone mineralization, soft-tissue calcification, insulin receptor signalling, cancer cell proliferation and immune modulation. Therefore, NPP1 inhibitors have potential as novel drugs, e.g. for (immuno)oncology. In the last two decades several inhibitors of NPP1 derived from nucleotide- or non-nucleotide scaffolds have been developed. The most potent and selective NPP1-inhibitory substrate analog is adenosine 5'-α,β-methylene-γ-thiotriphosphate (Ki = 20 nM vs. p-Nph-5'-TMP, human membrane-bound NPP1). Non-nucleotide-derived NPP1 inhibitors comprise polysulfonates, polysaccharides, polyoxometalates and small heterocyclic compounds. The polyoxometalate [TiW11CoO40]8- (PSB-POM141) is the most potent and selective NPP1 inhibitor described to date (Ki = 1.46 nM vs. ATP, human soluble NPP1); it displays an allosteric mechanism of inhibition and represents a useful pharmacological tool for evaluating the potential of NPP1 as a novel drug target.
Collapse
Affiliation(s)
- Sang-Yong Lee
- PharmaCenter Bonn , Pharmaceutical Institute , Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany . ; ; Tel: +49 228 73 2480
| | - Christa E Müller
- PharmaCenter Bonn , Pharmaceutical Institute , Pharmaceutical Chemistry I , University of Bonn , An der Immenburg 4 , D-53121 Bonn , Germany . ; ; Tel: +49 228 73 2480
| |
Collapse
|
49
|
Lee SY, Sarkar S, Bhattarai S, Namasivayam V, De Jonghe S, Stephan H, Herdewijn P, El-Tayeb A, Müller CE. Substrate-Dependence of Competitive Nucleotide Pyrophosphatase/Phosphodiesterase1 (NPP1) Inhibitors. Front Pharmacol 2017; 8:54. [PMID: 28261095 PMCID: PMC5309242 DOI: 10.3389/fphar.2017.00054] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/25/2017] [Indexed: 01/10/2023] Open
Abstract
Nucleotide pyrophosphatase/phosphodiesterase type 1 (NPP1) is a membrane glycoprotein involved in the hydrolysis of extracellular nucleotides. Its major substrate is ATP which is converted to AMP and diphosphate. NPP1 was proposed as a new therapeutic target in brain cancer and immuno-oncology. Several NPP1 inhibitors have been reported to date, most of which were evaluated vs. the artificial substrate p-nitrophenyl 5′-thymidine monophosphate (p-Nph-5′-TMP). Recently, we observed large discrepancies in inhibitory potencies for a class of competitive NPP1 inhibitors when tested vs. the artificial substrate p-Nph-5′-TMP as compared to the natural substrate ATP. Therefore, the goal of the present study was to investigate whether inhibitors of human NPP1 generally display substrate-dependent inhibitory potency. Systematic evaluation of nucleotidic as well as non-nucleotidic NPP1 inhibitors revealed significant differences in determined Ki values for competitive, but not for non- and un-competitive inhibitors when tested vs. the frequently used artificial substrate p-Nph-5′-TMP as compared to ATP. Allosteric modulation of NPP1 by p-Nph-5′-TMP may explain these discrepancies. Results obtained using the AMP derivative p-nitrophenyl 5′-adenosine monophosphate (p-Nph-5′-AMP) as an alternative artificial substrate correlated much better with those employing the natural substrate ATP.
Collapse
Affiliation(s)
- Sang-Yong Lee
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn Bonn, Germany
| | - Soumya Sarkar
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn Bonn, Germany
| | - Sanjay Bhattarai
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn Bonn, Germany
| | - Vigneshwaran Namasivayam
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn Bonn, Germany
| | - Steven De Jonghe
- Laboratory of Medicinal Chemistry, KU Leuven, Rega Institute for Medical Research Leuven, Belgium
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Dresden, Germany
| | - Piet Herdewijn
- Laboratory of Medicinal Chemistry, KU Leuven, Rega Institute for Medical Research Leuven, Belgium
| | - Ali El-Tayeb
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Chemistry I, University of Bonn Bonn, Germany
| |
Collapse
|
50
|
Barbeau X, Mathieu P, Paquin JF, Lagüe P. Characterization of the structure, dynamics and allosteric pathways of human NPP1 in its free form and substrate-bound complex from molecular modeling. MOLECULAR BIOSYSTEMS 2017; 13:1058-1069. [DOI: 10.1039/c7mb00095b] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Here we report 3D structure modeling and extensive molecular dynamics simulations of NPP1 complemented with a dynamical network analysis.
Collapse
Affiliation(s)
- Xavier Barbeau
- Department of Chemistry
- Faculty of Science and Engineering
- Université Laval
- Québec (Québec)
- Canada
| | | | - Jean-François Paquin
- Department of Chemistry
- Faculty of Science and Engineering
- Université Laval
- Québec (Québec)
- Canada
| | - Patrick Lagüe
- PROTEO
- The Quebec Network for Research on Protein Function
- Engineering
- and Applications
- Canada
| |
Collapse
|