1
|
de Keijzer MJ, de Klerk DJ, de Haan LR, van Kooten RT, Franchi LP, Dias LM, Kleijn TG, van Doorn DJ, Heger M. Inhibition of the HIF-1 Survival Pathway as a Strategy to Augment Photodynamic Therapy Efficacy. Methods Mol Biol 2022; 2451:285-403. [PMID: 35505024 DOI: 10.1007/978-1-0716-2099-1_19] [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] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) is a non-to-minimally invasive treatment modality that utilizes photoactivatable drugs called photosensitizers to disrupt tumors with locally photoproduced reactive oxygen species (ROS). Photosensitizer activation by light results in hyperoxidative stress and subsequent tumor cell death, vascular shutdown and hypoxia, and an antitumor immune response. However, sublethally afflicted tumor cells initiate several survival mechanisms that account for decreased PDT efficacy. The hypoxia inducible factor 1 (HIF-1) pathway is one of the most effective cell survival pathways that contributes to cell recovery from PDT-induced damage. Several hundred target genes of the HIF-1 heterodimeric complex collectively mediate processes that are involved in tumor cell survival directly and indirectly (e.g., vascularization, glucose metabolism, proliferation, and metastasis). The broad spectrum of biological ramifications culminating from the activation of HIF-1 target genes reflects the importance of HIF-1 in the context of therapeutic recalcitrance. This chapter elaborates on the involvement of HIF-1 in cancer biology, the hypoxic response mechanisms, and the role of HIF-1 in PDT. An overview of inhibitors that either directly or indirectly impede HIF-1-mediated survival signaling is provided. The inhibitors may be used as pharmacological adjuvants in combination with PDT to augment therapeutic efficacy.
Collapse
Affiliation(s)
- Mark J de Keijzer
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Daniel J de Klerk
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Lianne R de Haan
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Robert T van Kooten
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Leonardo P Franchi
- Departamento de Bioquímica e Biologia Molecular, Instituto de Ciências Biológicas (ICB) 2, Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
- Faculty of Philosophy, Sciences, and Letters of Ribeirão Preto, epartment of Chemistry, Center of Nanotechnology and Tissue Engineering-Photobiology and Photomedicine Research Group,University of São Paulo, São Paulo, Brazil
| | - Lionel M Dias
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Tony G Kleijn
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands
| | - Diederick J van Doorn
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Michal Heger
- Jiaxing Key Laboratory for Photonanomedicine and Experimental Therapeutics, Department of Pharmaceutics, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, People's Republic of China.
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.
- Laboratory of Experimental Oncology, Department of Pathology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Gutiérrez JR, Salgadoa ARM, Arias MDÁ, Vergara HSJ, Rada WR, Gómez CMM. Epigenetic Modulators as Treatment Alternative to Diverse Types of Cancer. Curr Med Chem 2021; 29:1503-1542. [PMID: 34963430 DOI: 10.2174/0929867329666211228111036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 01/10/2023]
Abstract
DNA is packaged in rolls in an octamer of histones forming a complex of DNA and proteins called chromatin. Chromatin as a structural matrix of a chromosome and its modifications are nowadays considered relevant aspects for regulating gene expression, which has become of high interest in understanding genetic mechanisms regulating various diseases, including cancer. In various types of cancer, the main modifications are found to be DNA methylation in the CpG dinucleotide as a silencing mechanism in transcription, post-translational histone modifications such as acetylation, methylation and others that affect the chromatin structure, the ATP-dependent chromatin remodeling and miRNA-mediated gene silencing. In this review we analyze the main alterations in gene expression, the epigenetic modification patterns that cancer cells present, as well as the main modulators and inhibitors of each epigenetic mechanism and the molecular evolution of the most representative inhibitors, which have opened a promising future in the study of HAT, HDAC, non-glycoside DNMT inhibitors and domain inhibitors.
Collapse
Affiliation(s)
- Jorseth Rodelo Gutiérrez
- Organic and Biomedical Chemistry Research Group, Faculty of Basic Sciences, Universidad del Atlántico, Barranquilla, Colombia
| | - Arturo René Mendoza Salgadoa
- Organic and Biomedical Chemistry Research Group, Faculty of Basic Sciences, Universidad del Atlántico, Barranquilla, Colombia
| | - Marcio De Ávila Arias
- Department of Medicine, Biotechnology Research Group, Health Sciences Division, Universidad del Norte, Barranquilla, Colombia
| | - Homero San- Juan- Vergara
- Department of Medicine, Biotechnology Research Group, Health Sciences Division, Universidad del Norte, Barranquilla, Colombia
| | - Wendy Rosales Rada
- Advanced Biomedicine Research Group. Faculty of Exact and Natural Sciences, Universidad Libre Seccional, Barranquilla, Colombia
- Advanced Biomedicine Research Group. Faculty of Exact and Natural Sciences, Universidad Libre Seccional, Barranquilla, Colombia
| | - Carlos Mario Meléndez Gómez
- Organic and Biomedical Chemistry Research Group, Faculty of Basic Sciences, Universidad del Atlántico, Barranquilla, Colombia
| |
Collapse
|
3
|
Kaluz S, Zhang Q, Kuranaga Y, Yang H, Osuka S, Bhattacharya D, Devi NS, Mun J, Wang W, Zhang R, Goodman MM, Grossniklaus HE, Van Meir EG. Targeting HIF-activated collagen prolyl 4-hydroxylase expression disrupts collagen deposition and blocks primary and metastatic uveal melanoma growth. Oncogene 2021; 40:5182-5191. [PMID: 34218269 PMCID: PMC8887959 DOI: 10.1038/s41388-021-01919-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 02/06/2023]
Abstract
Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, and patients that develop metastases (~50%) survive <1 year, highlighting the urgent need for new therapies. TCGA has recently revealed that a hypoxia gene signature is associated with poor UM patient prognosis. Here we show that expression of hypoxia-regulated collagen prolyl-4-hydroxylase genes P4HA1 and P4HA2 is significantly upregulated in UM patients with metastatic disease and correlates with poor prognosis, suggesting these enzymes might be key tumor drivers. We targeted hypoxia-induced expression of P4HA1/2 in UM with KCN1, a hypoxia inducible factor-1 (HIF-1) pathway inhibitor and found potent inhibition of primary and metastatic disease and extension of animal survival, without overt side effects. At the molecular level, KCN1 antagonized hypoxia-induced expression of P4HA1 and P4HA2, which regulate collagen maturation and deposition in the extracellular matrix. The treatment decreased prolyl hydroxylation, induced proteolytic cleavage and rendered a disordered structure to collagen VI, the main collagen produced by UM, and reduced UM cell invasion. Together, these data demonstrate that extracellular collagen matrix formation can be targeted in UM by inhibiting hypoxia-induced P4HA1 and P4HA2 expression, warranting further development of this strategy in patients with uveal melanoma.
Collapse
Affiliation(s)
- Stefan Kaluz
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Qing Zhang
- Department of Ophthalmology, Emory University, Atlanta, GA, USA
- Department of Ophthalmology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuki Kuranaga
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hua Yang
- Department of Ophthalmology, Emory University, Atlanta, GA, USA
| | - Satoru Osuka
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Narra S Devi
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
| | - Jiyoung Mun
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Wei Wang
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
- Drug Discovery Institute, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Ruiwen Zhang
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, USA
- Drug Discovery Institute, College of Pharmacy, University of Houston, Houston, TX, USA
| | - Mark M Goodman
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Hans E Grossniklaus
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
- Department of Ophthalmology, Emory University, Atlanta, GA, USA.
- Department of Pathology, Emory University, Atlanta, GA, USA.
| | - Erwin G Van Meir
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, USA.
- Department of Neurosurgery, University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
4
|
Rehman Z, Jabeen I, Fahim A, Bhatti A, John P. Molecular docking and pharmacophore models to probe binding hypothesis of inhibitors of hypoxia inducible factor-1. J Biomol Struct Dyn 2021; 40:7714-7725. [PMID: 33896358 DOI: 10.1080/07391102.2021.1914167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Hypoxia inducible factor-1 is a heterodimeric transcription factor that regulates cellular responses to hypoxia and is involved in tumor progression and resistance to chemotherapy. Dimerization between HIF-1α and β subunits has been recognized crucial for DNA binding and transcriptional activity of HIF-1. Therefore, inhibitors of α and β dimerization subunits of HIF-1 may potentially evade HIF-1-mediated chemotherapy resistance. In the current study, ligand-based pharmacophore model was developed to determine 3 D binding features of HIF-1 inhibitors. The selected pharmacophore model comprises of one hydrogen bond donor, one hydrogen bond acceptor and one hydrophobic feature. The selected model was used for virtual screening of publically available data base by ChemBridge Corporation. Overall, six potential hits against HIF-1α and β dimerization have been identified. These include, Hit 1 (4-(4-chlorophenyl)-2,6-dimethyl-3,5-pyridinedicarboxylic acid), 3 (2-[2-(2-hydroxybenzoyl)carbonohydrazonoyl]benzoic acid) and 5 (3-(4-methoxyphenyl)-2,4-quinolinedicarboxylic acid) nicotonic acid derivatives, Hit 2 (3-[(1-adamantylamino)sulfonyl]benzoic acid), 4 (5-{[(2-fluorophenyl)amino]sulfonyl}-2-methylbenzoic acid), and 6 (4-({[2-(trifluoromethyl)phenyl]sulfonyl}amino)benzoic acid) sulfonamide derivatives. Additionally, adamantyl moiety of compound 2 shows interactions with the experimentally known hydrophobic amino acid residues (V336, C334, E245) of HIF-1α and β dimerization site. The identified hits showed lower to higher µM biological activity (IC50) values and thus, after further structure optimization may serve as potential inhibitor of HIF-1 dimerization in cancer chemotherapy.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Zaira Rehman
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Ishrat Jabeen
- Research Centre for Modeling and Simulation, National University of Sciences and Technology, Islamabad, Pakistan
| | - Ammad Fahim
- Department of Multidisciplinary Sciences, National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | - Attya Bhatti
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Peter John
- Atta ur Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| |
Collapse
|
5
|
Design and Synthesis of Sulfonamide‐Attached 2‐(Isoxazol‐3‐yl)‐1
H
‐imidazoles as Anticancer Agents. ChemistrySelect 2020. [DOI: 10.1002/slct.202001449] [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]
|
6
|
Zhao C, Rakesh KP, Ravidar L, Fang WY, Qin HL. Pharmaceutical and medicinal significance of sulfur (S VI)-Containing motifs for drug discovery: A critical review. Eur J Med Chem 2019; 162:679-734. [PMID: 30496988 PMCID: PMC7111228 DOI: 10.1016/j.ejmech.2018.11.017] [Citation(s) in RCA: 304] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/17/2018] [Accepted: 11/07/2018] [Indexed: 01/04/2023]
Abstract
Sulfur (SVI) based moieties, especially, the sulfonyl or sulfonamide based analogues have showed a variety of pharmacological properties, and its derivatives propose a high degree of structural diversity that has established useful for the finding of new therapeutic agents. The developments of new less toxic, low cost and highly active sulfonamides containing analogues are hot research topics in medicinal chemistry. Currently, more than 150 FDA approved Sulfur (SVI)-based drugs are available in the market, and they are widely used to treat various types of diseases with therapeutic power. This comprehensive review highlights the recent developments of sulfonyl or sulfonamides based compounds in huge range of therapeutic applications such as antimicrobial, anti-inflammatory, antiviral, anticonvulsant, antitubercular, antidiabetic, antileishmanial, carbonic anhydrase, antimalarial, anticancer and other medicinal agents. We believe that, this review article is useful to inspire new ideas for structural design and developments of less toxic and powerful Sulfur (SVI) based drugs against the numerous death-causing diseases.
Collapse
Affiliation(s)
- Chuang Zhao
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR, China
| | - K P Rakesh
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR, China.
| | - L Ravidar
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR, China
| | - Wan-Yin Fang
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR, China
| | - Hua-Li Qin
- Department of Pharmaceutical Engineering, School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, 205 Luoshi Road, Wuhan, 430070, PR, China.
| |
Collapse
|
7
|
Dong L, You S, Zhang Q, Osuka S, Devi NS, Kaluz S, Ferguson JH, Yang H, Chen G, Wang B, Grossniklaus HE, Van Meir EG. Arylsulfonamide 64B Inhibits Hypoxia/HIF-Induced Expression of c-Met and CXCR4 and Reduces Primary Tumor Growth and Metastasis of Uveal Melanoma. Clin Cancer Res 2018; 25:2206-2218. [PMID: 30563937 DOI: 10.1158/1078-0432.ccr-18-1368] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 10/02/2018] [Accepted: 12/14/2018] [Indexed: 01/02/2023]
Abstract
PURPOSE Uveal melanoma (UM) is the most prevalent and lethal intraocular malignancy in adults. Here, we examined the importance of hypoxia in UM growth and tested the antitumor effects of arylsulfonamide 64B, an inhibitor of the hypoxia-induced factor (HIF) pathway in animal models of UM and investigated the related mechanisms. EXPERIMENTAL DESIGN UM cells were implanted in the uvea of mice eyes and mice systemically treated with 64B. Drug effect on primary eye tumor growth, circulating tumor cells, metastasis formation in liver, and survival were examined. 64B effects on UM cell growth, invasion and hypoxia-induced expression of C-X-C chemokine receptor type 4 (CXCR4) and mesenchymal-epithelial transition factor (c-Met) were measured. Luciferase reporter assays, chromatin immunoprecipitation, co-immunoprecipitation, and cellular thermal shift assays were used to determine how 64B interferes with the HIF transcriptional complex. RESULTS Systemic administration of 64B had potent antitumor effects against UM in several orthotopic mouse models, suppressing UM growth in the eye (∼70% reduction) and spontaneous liver metastasis (∼50% reduction), and extending mice survival (P < 0.001) while being well tolerated. 64B inhibited hypoxia-induced expression of CXCR4 and c-Met, 2 key drivers of tumor invasion and metastasis. 64B disrupted the HIF-1 complex by interfering with HIF-1α binding to p300/CBP co-factors, thus reducing p300 recruitment to the MET and CXCR4 gene promoters. 64B could thermostabilize p300, supporting direct 64B binding to p300. CONCLUSIONS Our preclinical efficacy studies support the further optimization of the 64B chemical scaffold toward a clinical candidate for the treatment of UM.
Collapse
Affiliation(s)
- Lei Dong
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, Georgia
| | - Shuo You
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, Georgia
| | - Qing Zhang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia
| | - Satoru Osuka
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, Georgia
| | - Narra S Devi
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, Georgia
| | - Stefan Kaluz
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | | | - Hua Yang
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drugs Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, PR China
| | - Binghe Wang
- Winship Cancer Institute, Emory University, Atlanta, Georgia.,Department of Chemistry, Georgia State University, Atlanta, Georgia
| | - Hans E Grossniklaus
- Department of Ophthalmology, School of Medicine, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Erwin G Van Meir
- Department of Neurosurgery, School of Medicine, Emory University, Atlanta, Georgia. .,Winship Cancer Institute, Emory University, Atlanta, Georgia.,Department of Hematology and Medical Oncology, School of Medicine, Emory University, Atlanta, Georgia
| |
Collapse
|
8
|
Sabt A, Abdelhafez OM, El-Haggar RS, Madkour HMF, Eldehna WM, El-Khrisy EEDAM, Abdel-Rahman MA, Rashed LA. Novel coumarin-6-sulfonamides as apoptotic anti-proliferative agents: synthesis, in vitro biological evaluation, and QSAR studies. J Enzyme Inhib Med Chem 2018; 33:1095-1107. [PMID: 29944015 PMCID: PMC6022226 DOI: 10.1080/14756366.2018.1477137] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 11/07/2022] Open
Abstract
Herein, we report the synthesis of different novel sets of coumarin-6-sulfonamide derivatives bearing different functionalities (4a, b, 8a-d, 11a-d, 13a, b, and 15a-c), and in vitro evaluation of their growth inhibitory activity towards the proliferation of three cancer cell lines; HepG2 (hepatocellular carcinoma), MCF-7 (breast cancer), and Caco-2 (colon cancer). HepG2 cells were the most sensitive cells to the influence of the target coumarins. Compounds 13a and 15a emerged as the most active members against HepG2 cells (IC50 = 3.48 ± 0.28 and 5.03 ± 0.39 µM, respectively). Compounds 13a and 15a were able to induce apoptosis in HepG2 cells, as assured by the upregulation of the Bax and downregulation of the Bcl-2, besides boosting caspase-3 levels. Besides, compound 13a induced a significant increase in the percentage of cells at Pre-G1 by 6.4-folds, with concurrent significant arrest in the G2-M phase by 5.4-folds compared to control. Also, 13a displayed significant increase in the percentage of annexin V-FITC positive apoptotic cells from 1.75-13.76%. Moreover, QSAR models were established to explore the structural requirements controlling the anti-proliferative activities.
Collapse
Affiliation(s)
- Ahmed Sabt
- Chemistry of Natural Compounds Department, National Research Centre, Dokki, Egypt
| | - Omaima M. Abdelhafez
- Chemistry of Natural Compounds Department, National Research Centre, Dokki, Egypt
| | - Radwan S. El-Haggar
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | | | - Wagdy M. Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, Egypt
| | | | - Mohamed A. Abdel-Rahman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, Egypt
| | - Laila. A. Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
9
|
Abstract
Hypoxia-inducible factors (HIFs), a family of transcription factors activated by hypoxia, consist of three α-subunits (HIF1α, HIF2α and HIF3α) and one β-subunit (HIF1β), which serves as a heterodimerization partner of the HIFα subunits. HIFα subunits are stabilized from constitutive degradation by hypoxia largely through lowering the activity of the oxygen-dependent prolyl hydroxylases that hydroxylate HIFα, leading to their proteolysis. HIF1α and HIF2α are expressed in different tissues and regulate target genes involved in angiogenesis, cell proliferation and inflammation, and their expression is associated with different disease states. HIFs have been widely studied because of their involvement in cancer, and HIF2α-specific inhibitors are being investigated in clinical trials for the treatment of kidney cancer. Although cancer has been the major focus of research on HIF, evidence has emerged that this pathway has a major role in the control of metabolism and influences metabolic diseases such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease. Notably increased HIF1α and HIF2α signalling in adipose tissue and small intestine, respectively, promotes metabolic diseases in diet-induced disease models. Inhibition of HIF1α and HIF2α decreases the adverse diet-induced metabolic phenotypes, suggesting that they could be drug targets for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.
| |
Collapse
|
10
|
Panda P, Nayak S, Bhakta S, Mohapatra S, Murthy TR. Design and synthesis of (Z/E)-2-phenyl/H-3-styryl-2H-chromene derivatives as antimicrotubule agents. J CHEM SCI 2018. [DOI: 10.1007/s12039-018-1520-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
11
|
Bhattarai D, Xu X, Lee K. Hypoxia-inducible factor-1 (HIF-1) inhibitors from the last decade (2007 to 2016): A "structure-activity relationship" perspective. Med Res Rev 2017; 38:1404-1442. [PMID: 29278273 DOI: 10.1002/med.21477] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/20/2017] [Accepted: 11/27/2017] [Indexed: 12/19/2022]
Abstract
Tumor hypoxia is a common feature in most solid tumors and is associated with overexpression of the hypoxia response pathway. Overexpression of the hypoxia-inducible factor (HIF-1) protein leads to angiogenesis, metastasis, apoptosis resistance, and many other pro-tumorigenic responses in cancer development. HIF-1 is a promising target in cancer drug development to increase the patient's response to chemotherapy and radiotherapy as well as the survival rate of cancer patients. Since up to 1% of genes are hypoxia-sensitive, a target-specific HIF-1 inhibitor may be a better clinical candidate in cancer drug discovery. Though no HIF-1 inhibitor is clinically available to date, a lot of effort has been applied during the last decade in search of potent HIF-1 inhibitors. In this review, we will summarize the structure-activity relationship of ten different chemotypes reported to be HIF-1 inhibitors in the last decade (2007-2016), their mechanisms of action for HIF-1 inhibition, progress in the way of target-specific inhibitors, and problems associated with current inhibitors. It is anticipated that the results of these research on the medicinal chemistry of HIF-1 inhibitors will provide decent information in the design and development of future inhibitors.
Collapse
Affiliation(s)
- Deepak Bhattarai
- College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| | - Xuezhen Xu
- College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| |
Collapse
|
12
|
Ferguson JH, De Los Santos Z, Devi SN, Kaluz S, Van Meir EG, Zingales SK, Wang B. Design and synthesis of benzopyran-based inhibitors of the hypoxia-inducible factor-1 pathway with improved water solubility. J Enzyme Inhib Med Chem 2017; 32:992-1001. [PMID: 28766956 PMCID: PMC6009948 DOI: 10.1080/14756366.2017.1347784] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 06/23/2017] [Accepted: 06/24/2017] [Indexed: 12/31/2022] Open
Abstract
While progress has been made in treating cancer, cytotoxic chemotherapeutic agents are still the most widely used drugs and are associated with severe side-effects. Drugs that target unique molecular signalling pathways are needed for treating cancer with low or no intrinsic toxicity to normal cells. Our goal is to target hypoxic tumours and specifically the hypoxia inducible factor (HIF) pathway for the development of new cancer therapies. To this end, we have previously developed benzopyran-based HIF-1 inhibitors such as arylsulfonamide KCN1. However, KCN1 and its earlier analogs have poor water solubility, which hamper their applications. Herein, we describe a series of KCN1 analogs that incorporate a morpholine moiety at various positions. We found that replacing the benzopyran group of KCN1 with a phenyl group with a morpholinomethyl moiety at the para positions had minimal effect on potency and improved the water solubility of two new compounds by more than 10-fold compared to KCN1, the lead compound.
Collapse
Affiliation(s)
| | | | - Saroja N. Devi
- Departments of Neurosurgery and Hematology and Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Stefan Kaluz
- Departments of Neurosurgery and Hematology and Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Erwin G. Van Meir
- Departments of Neurosurgery and Hematology and Medical Oncology, School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sarah K. Zingales
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
- Department of Chemistry and Physics, Armstrong State University, Savannah, GA, USA
| | - Binghe Wang
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
| |
Collapse
|
13
|
Dai X, Kaluz S, Jiang Y, Shi L, Mckinley D, Wang Y, Wang B, Van Meir EG, Tan C. A novel small-molecule arylsulfonamide causes energetic stress and suppresses breast and lung tumor growth and metastasis. Oncotarget 2017; 8:99245-99260. [PMID: 29245898 PMCID: PMC5725089 DOI: 10.18632/oncotarget.22104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/10/2017] [Indexed: 12/19/2022] Open
Abstract
Neoplastic cells display reprogrammed metabolism due to the heightened energetic demands and the need for biomass synthesis of a growing tumor. Targeting metabolic vulnerabilities is thus an important goal for cancer therapy. Here, we describe a novel small-molecule arylsulfonamide (N-cyclobutyl-N-((2,2-dimethyl-2H-pyrano[3,2-b]pyridin-6-yl)methyl)-3,4-dimethoxybenzenesulfonamide) that exerts potent cytotoxicity and energetic stress on tumor cells while largely sparing non-cancerous human cells. In tumor cells, it stimulates glycolysis and accelerates glucose consumption. Consequently, intracellular ATP levels plummet, triggering activation of AMP-activated protein kinase (AMPK), and diminishing the mammalian target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor 1 (HIF-1) signaling. In orthotopic triple-negative breast cancer and subcutaneous lung cancer mouse models, this arylsulfonamide robustly suppresses primary tumor growth, inhibits the formation of distant metastases to the lung, and extends mouse survival while being very well tolerated. These therapeutic effects are further potentiated by co-administration of 2-deoxy-D-glucose (2-DG), a glucose analog and glycolysis inhibitor. Collectively, our findings provide preclinical proof of concept for the further development of this arylsulfonamide in combination with 2-DG towards cancer treatment.
Collapse
Affiliation(s)
- Xin Dai
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS, USA
| | - Stefan Kaluz
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Ying Jiang
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA, USA
| | - Lei Shi
- Department of Biology, Georgia State University, Atlanta, GA, USA
| | - DeAngelo Mckinley
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA, USA
| | - Yingzhe Wang
- Department of Pharmaceutical Sciences, Mercer University, Atlanta, GA, USA
| | - Binghe Wang
- Department of Chemistry, Georgia State University, Atlanta, GA, USA
| | - Erwin G Van Meir
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.,Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Chalet Tan
- Department of Pharmaceutics and Drug Delivery, University of Mississippi, Oxford, MS, USA
| |
Collapse
|
14
|
Perán M, López-Ruiz E, García MÁ, Nadaraia-Hoke S, Brandt R, Marchal JA, Kenyon J. A formulation of pancreatic pro-enzymes provides potent anti-tumour efficacy: a pilot study focused on pancreatic and ovarian cancer. Sci Rep 2017; 7:13998. [PMID: 29070896 PMCID: PMC5656641 DOI: 10.1038/s41598-017-14571-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 10/12/2017] [Indexed: 12/13/2022] Open
Abstract
Proteolytic enzymes have shown efficacy in cancer therapy. We present a combination of the two pro-enzymes Trypsinogen and Chymotrypsinogen A with potent in vitro and in vivo anti-tumour efficacy. A synergetic anti-tumour effect for Trypsinogen and Chymotrypsinogen A was determined at a ratio 1:6 (named PRP) using 24 human cancer cell lines. The antiangiogenic effect of PRP was analysed by matrigel-based tube formation and by fibrous capsule formation assays. Furthermore, cell invasion and wound healing assays together with qRT-PCR determination of epithelial-to-mesenchymal transition (EMT) markers were performed on human cancer cells treated with PRP. Additionally, in vivo pharmacokinetic studies were implemented and the PRP's anti-tumour efficacy was explored against orthotopic pancreatic and ovarian cancer tumours. PRP formulation was proven to inhibit in vitro angiogenesis, tumour growth, cancer cell migration and invasiveness; and to be an effective and well tolerated in vivo anti-tumour treatment. Finally, the clinical efficacy of a suppository formulation containing both pancreatic pro-enzymes in the context of a UK Pharmaceuticals Special Scheme was evaluated in advanced cancer patients. Consequently, PRP could have relevant oncological clinical applications for the treatment of advanced or metastatic pancreatic adenocarcinoma and advanced epithelial ovarian cancer.
Collapse
Affiliation(s)
- Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén, Spain.
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain.
| | - Elena López-Ruiz
- Department of Health Sciences, University of Jaén, Jaén, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - María Ángel García
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Department of Oncology, University Hospital Virgen de las Nieves, Granada, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain
| | | | - Ralf Brandt
- vivoPharm LLC, 1214 Research Boulevard 17036, Hummelstown PA, United States
| | - Juan A Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada-University of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Julian Kenyon
- The Dove Clinic for Integrated Medicine, Twyford, SO21 1RG, UK.
| |
Collapse
|
15
|
Liu H, Qu M, Xu L, Han X, Wang C, Shu X, Yao J, Liu K, Peng J, Li Y, Ma X. Design and synthesis of sulfonamide-substituted diphenylpyrimidines (SFA-DPPYs) as potent Bruton's tyrosine kinase (BTK) inhibitors with improved activity toward B-cell lymphoblastic leukemia. Eur J Med Chem 2017; 135:60-69. [PMID: 28432946 DOI: 10.1016/j.ejmech.2017.04.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/31/2022]
Abstract
A new series of diphenylpyrimidine derivatives (SFA-DPPYs) were synthesized by introducing a functional sulfonamide into the C-2 aniline moiety of pyrimidine template, and then were biologically evaluated as potent Bruton's tyrosine kinase (BTK) inhibitors. Among these molecules, inhibitors 10c, 10i, 10j and 10k displayed high potency against the BTK enzyme, with IC50 values of 1.18 nM, 0.92 nM, 0.42 nM and 1.05 nM, respectively. In particular, compound 10c could remarkably inhibit the proliferation of the B lymphoma cell lines at concentrations of 6.49 μM (Ramos cells) and 13.2 μM (Raji cells), and was stronger than the novel agent spebrutinib. In addition, the inhibitory potency toward the normal PBMC cells showed that inhibitor 10c possesses low cell cytotoxicity. All these explorations indicated that molecule 10c could serve as a valuable inhibitor for B-cell lymphoblastic leukemia treatment.
Collapse
Affiliation(s)
- He Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Menghua Qu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Changyuan Wang
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Xiaohong Shu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Jihong Yao
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China
| | - Yanxia Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, PR China
| | - Xiaodong Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, PR China.
| |
Collapse
|
16
|
Synthesis of new dihydropyrrol-2-one derivatives bearing sulfonamide groups and studies their antibacterial activity. MONATSHEFTE FUR CHEMIE 2016. [DOI: 10.1007/s00706-016-1847-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
17
|
Subhani S, Vavilala DT, Mukherji M. HIF inhibitors for ischemic retinopathies and cancers: options beyond anti-VEGF therapies. Angiogenesis 2016; 19:257-73. [DOI: 10.1007/s10456-016-9510-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 04/16/2016] [Indexed: 12/15/2022]
|
18
|
Ban HS, Uto Y, Won M, Nakamura H. Hypoxia-inducible factor (HIF) inhibitors: a patent survey (2011-2015). Expert Opin Ther Pat 2016; 26:309-22. [DOI: 10.1517/13543776.2016.1146252] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
19
|
Konda S, Raparthi S, Bhaskar K, Munaganti RK, Guguloth V, Nagarapu L, Akkewar DM. Synthesis and antimicrobial activity of novel benzoxazine sulfonamide derivatives. Bioorg Med Chem Lett 2015; 25:1643-6. [PMID: 25754493 DOI: 10.1016/j.bmcl.2015.01.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 12/02/2014] [Accepted: 01/13/2015] [Indexed: 11/25/2022]
Abstract
A new series of benzoxazine-6-sulfonamide derivatives were synthesized in excellent yields and the resulting compounds were evaluated for their antimicrobial activities. All the synthesized compounds were assessed for their antibacterial and antifungal activities. Among them 1a, 1b, 1c, 1e, 1h, 2c, 2d, 2e, 2g, 2h, 2i, 2j, 2k and 2l showed low inhibitory concentration (MIC of 31.25 and 62.5 μg/mL) against Gram-positive bacteria, Gram-negative bacteria and fungi, which are comparable to the inhibitory effect of standard drugs.
Collapse
Affiliation(s)
- Saidulu Konda
- Organic Chemistry Division-II (CPC), CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, A.P., India
| | - Srujana Raparthi
- Organic Chemistry Division-II (CPC), CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, A.P., India
| | - K Bhaskar
- Organic Chemistry Division-II (CPC), CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, A.P., India
| | - Rajesh Kumar Munaganti
- Organic Chemistry Division-II (CPC), CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, A.P., India
| | - Vijayacharan Guguloth
- Organic Chemistry Division-II (CPC), CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, A.P., India
| | - Lingaiah Nagarapu
- Organic Chemistry Division-II (CPC), CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, A.P., India.
| | - Dattatray M Akkewar
- Organic Chemistry Division-II (CPC), CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, A.P., India.
| |
Collapse
|
20
|
Nag S, Qin JJ, Voruganti S, Wang MH, Sharma H, Patil S, Buolamwini JK, Wang W, Zhang R. Development and validation of a rapid HPLC method for quantitation of SP-141, a novel pyrido[b]indole anticancer agent, and an initial pharmacokinetic study in mice. Biomed Chromatogr 2014; 29:654-63. [PMID: 25294254 DOI: 10.1002/bmc.3327] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 05/31/2014] [Accepted: 08/15/2014] [Indexed: 11/07/2022]
Abstract
There is an increasing interest in targeting the MDM2 oncogene for cancer therapy. SP-141, a novel designed small molecule MDM2 inhibitor, exerts excellent in vitro and in vivo anticancer activity. To facilitate the preclinical development of this candidate anticancer agent, we have developed an HPLC method for the quantitative analysis of SP-141. The method was validated to be precise, accurate, and specific, with a linear range of 16.2-32,400 ng/mL in plasma, 16.2-6480 ng/mL in homogenates of brain, heart, liver, kidneys, lungs, muscle and tumor, and 32.4-6480 ng/mL in spleen homogenates. The lower limit of quantification was 16.2 ng/mL in plasma and all the tissue homogenates, except for spleen homogenates, where it was 32.4 ng/mL. The intra- and inter-assay precisions (coefficient of variation) were between 0.86 and 13.39%, and accuracies (relative errors) ranged from -8.50 to 13.92%. The relative recoveries were 85.6-113.38%. SP-141 was stable in mouse plasma, modestly plasma bound and metabolized by S9 microsomal enzymes. We performed an initial pharmacokinetic study in tumor-bearing nude mice, demonstrating that SP-141 has a short half-life in plasma and wide tissue distribution. In summary, this HPLC method can be used in future preclinical and clinical investigations of SP-141.
Collapse
Affiliation(s)
- Subhasree Nag
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, 79106, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Wang W, Qin JJ, Voruganti S, Wang MH, Sharma H, Patil S, Zhou J, Wang H, Mukhopadhyay D, Buolamwini JK, Zhang R. Identification of a new class of MDM2 inhibitor that inhibits growth of orthotopic pancreatic tumors in mice. Gastroenterology 2014; 147:893-902.e2. [PMID: 25016295 PMCID: PMC4170027 DOI: 10.1053/j.gastro.2014.07.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Revised: 06/02/2014] [Accepted: 07/07/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND & AIMS The oncogene MDM2, which encodes an E3 ubiquitin ligase, is overexpressed in pancreatic cancers and is therefore a therapeutic target. Current inhibitors of MDM2 target the interaction between MDM2 and P53; these would have no effect on cancer cells that do not express full-length P53, including many pancreatic cancer cells. We searched for a compound that specifically inhibits MDM2 itself. METHODS We performed a virtual screen and structure-based design to identify specific inhibitors of MDM2. We tested the activities of compounds identified on viability, proliferation, and protein levels of HPAC, Panc-1, AsPC-1, and Mia-Paca-2 pancreatic cancer cell lines. We tested whether intraperitoneal injections of one of the compounds identified affected growth of xenograft tumors from Panc-1 cells, or orthotopic tumors from Panc-1 and AsPC-1 cells (injected into pancreata), in nude mice. RESULTS We identified a compound, called SP141, which bound directly to MDM2, promoting its auto-ubiquitination and degradation by the proteasome. The compound reduced levels of MDM2 in pancreatic cancer cell lines, as well as their proliferation, with 50% inhibitory concentrations <0.5 μM (0.38-0.50 μM). Increasing concentrations of SP141 induced increasing levels of apoptosis and G2-M-phase arrest of pancreatic cancer cell lines, whether or not they expressed functional P53. Injection of nude mice with SP141 (40 mg/kg/d) inhibited growth of xenograft tumors (by 75% compared with control mice), and led to regression of orthotopic tumors. CONCLUSIONS In a screen for specific inhibitors of MDM2, we identified a compound called SP141 that reduces levels of MDM2 in pancreatic cancer cell lines, as well as their proliferation and ability to form tumors in nude mice. SP141 is a new class of MDM2 inhibitor that promotes MDM2 auto-ubiquitination and degradation. It might be further developed as a therapeutic agent for pancreatic cancer.
Collapse
Affiliation(s)
- Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Jiang-Jiang Qin
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Sukesh Voruganti
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Ming-Hai Wang
- Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas; Department of Biomedical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Horrick Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Shivaputra Patil
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, Cancer Center, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hui Wang
- Key Laboratory of Food Safety Research Center, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - John K Buolamwini
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee.
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas.
| |
Collapse
|
22
|
Barcelos RC, Pelizzaro-Rocha KJ, Pastre JC, Dias MP, Ferreira-Halder CV, Pilli RA. A new goniothalamin N-acylated aza-derivative strongly downregulates mediators of signaling transduction associated with pancreatic cancer aggressiveness. Eur J Med Chem 2014; 87:745-58. [PMID: 25305718 DOI: 10.1016/j.ejmech.2014.09.085] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/05/2014] [Accepted: 09/08/2014] [Indexed: 12/20/2022]
Abstract
In this study, a novel concise series of molecules based on the structure of goniothalamin (1) was synthesized and evaluated against a highly metastatic human pancreatic cancer cell line (Panc-1). Among them, derivative 8 displayed a low IC50 value (2.7 μM) and its concentration for decreasing colony formation was 20-fold lower than goniothalamin (1). Both compounds reduced the levels of the receptor tyrosine kinase (AXL) and cyclin D1 which are known to be overexpressed in pancreatic cancer cells. Importantly, despite the fact that goniothalamin (1) and derivative 8 caused pancreatic cancer cell cycle arrest and cell death, only derivative 8 was able to downregulate pro-survival and proliferation pathways mediated by mitogen activated protein kinase ERK1/2. Another interesting finding was that Panc-1 cells treated with derivative 8 displayed a strong decrease in the transcription factor (c-Myc), hypoxia-inducible factor-1α (HIF-1α) and vascular endothelial growth factor (VEGF) protein levels. Notably, the molecular effects caused by derivative 8 might not be related to ROS generation, since no significant production of ROS was observed in low concentrations of this compound (from 1.5 up to 3 μM). Therefore, the downregulation of important mediators of pancreatic cancer aggressiveness by derivative 8 reveals its great potential for the development of new chemotherapeutic agents for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Rosimeire Coura Barcelos
- Department of Organic Chemistry, Chemistry Institute, University of Campinas, CP 6154, 13083-970, Campinas, SP, Brazil
| | | | - Julio Cezar Pastre
- Department of Organic Chemistry, Chemistry Institute, University of Campinas, CP 6154, 13083-970, Campinas, SP, Brazil
| | - Marina Pereira Dias
- Department of Biochemistry, Biology Institute, University of Campinas, 13083-862, Campinas, São Paulo, Brazil
| | | | - Ronaldo Aloise Pilli
- Department of Organic Chemistry, Chemistry Institute, University of Campinas, CP 6154, 13083-970, Campinas, SP, Brazil.
| |
Collapse
|
23
|
Luke JJ, Triozzi PL, McKenna KC, Van Meir EG, Gershenwald JE, Bastian BC, Gutkind JS, Bowcock AM, Streicher HZ, Patel PM, Sato T, Sossman JA, Sznol M, Welch J, Thurin M, Selig S, Flaherty KT, Carvajal RD. Biology of advanced uveal melanoma and next steps for clinical therapeutics. Pigment Cell Melanoma Res 2014; 28:135-47. [PMID: 25113308 DOI: 10.1111/pcmr.12304] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/05/2014] [Indexed: 01/03/2023]
Abstract
Uveal melanoma is the most common intraocular malignancy although it is a rare subset of all melanomas. Uveal melanoma has distinct biology relative to cutaneous melanoma, with widely divergent patient outcomes. Patients diagnosed with a primary uveal melanoma can be stratified for risk of metastasis by cytogenetics or gene expression profiling, with approximately half of patients developing metastatic disease, predominately hepatic in location, over a 15-yr period. Historically, no systemic therapy has been associated with a clear clinical benefit for patients with advanced disease, and median survival remains poor. Here, as a joint effort between the Melanoma Research Foundation's ocular melanoma initiative, CURE OM and the National Cancer Institute, the current understanding of the molecular and immunobiology of uveal melanoma is reviewed, and on-going laboratory research into the disease is highlighted. Finally, recent investigations relevant to clinical management via targeted and immunotherapies are reviewed, and next steps in the development of clinical therapeutics are discussed.
Collapse
Affiliation(s)
- Jason J Luke
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Hypoxia is a significant feature of solid tumor cancers. Hypoxia leads to a more malignant phenotype that is resistant to chemotherapy and radiation, is more invasive and has greater metastatic potential. Hypoxia activates the hypoxia inducible factor (HIF) pathway, which mediates the biological effects of hypoxia in tissues. The HIF complex acts as a transcription factor for many genes that increase tumor survival and proliferation. To date, many HIF pathway inhibitors indirectly affect HIF but there have been no clinically approved direct HIF inhibitors. This can be attributed to the complexity of the HIF pathway, as well as to the challenges of inhibiting protein-protein interactions.
Collapse
|
25
|
Yin S, Kaluz S, Devi NS, Jabbar AA, de Noronha RG, Mun J, Zhang Z, Boreddy PR, Wang W, Wang Z, Abbruscato T, Chen Z, Olson JJ, Zhang R, Goodman MM, Nicolaou KC, Van Meir EG. Arylsulfonamide KCN1 inhibits in vivo glioma growth and interferes with HIF signaling by disrupting HIF-1α interaction with cofactors p300/CBP. Clin Cancer Res 2012; 18:6623-33. [PMID: 22923450 DOI: 10.1158/1078-0432.ccr-12-0861] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
PURPOSE The hypoxia-inducible factor-1 (HIF-1) plays a critical role in tumor adaptation to hypoxia, and its elevated expression correlates with poor prognosis and treatment failure in patients with cancer. In this study, we determined whether 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide, KCN1, the lead inhibitor in a novel class of arylsulfonamide inhibitors of the HIF-1 pathway, had antitumorigenic properties in vivo and further defined its mechanism of action. EXPERIMENTAL DESIGN We studied the inhibitory effect of systemic KCN1 delivery on the growth of human brain tumors in mice. To define mechanisms of KCN1 anti-HIF activities, we examined its influence on the assembly of a functional HIF-1α/HIF-1β/p300 transcription complex. RESULTS KCN1 specifically inhibited HIF reporter gene activity in several glioma cell lines at the nanomolar level. KCN1 also downregulated transcription of endogenous HIF-1 target genes, such as VEGF, Glut-1, and carbonic anhydrase 9, in a hypoxia-responsive element (HRE)-dependent manner. KCN1 potently inhibited the growth of subcutaneous malignant glioma tumor xenografts with minimal adverse effects on the host. It also induced a temporary survival benefit in an intracranial model of glioma but had no effect in a model of melanoma metastasis to the brain. Mechanistically, KCN1 did not downregulate the levels of HIF-1α or other components of the HIF transcriptional complex; rather, it antagonized hypoxia-inducible transcription by disrupting the interaction of HIF-1α with transcriptional coactivators p300/CBP. CONCLUSIONS Our results suggest that the new HIF pathway inhibitor KCN1 has antitumor activity in mouse models, supporting its further translation for the treatment of human tumors displaying hypoxia or HIF overexpression.
Collapse
Affiliation(s)
- Shaoman Yin
- Departments of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shi Q, Yin S, Kaluz S, Ni N, Devi NS, Mun J, Wang D, Damera K, Chen W, Burroughs S, Mooring SR, Goodman MM, Van Meir EG, Wang B, Snyder JP. Binding Model for the Interaction of Anticancer Arylsulfonamides with the p300 Transcription Cofactor. ACS Med Chem Lett 2012; 3:620-5. [PMID: 24936238 DOI: 10.1021/ml300042k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Accepted: 06/21/2012] [Indexed: 02/02/2023] Open
Abstract
Hypoxia inducible factors (HIFs) are transcription factors that activate expression of multiple gene products and promote tumor adaptation to a hypoxic environment. To become transcriptionally active, HIFs associate with cofactors p300 or CBP. Previously, we found that arylsulfonamides can antagonize HIF transcription in a bioassay, block the p300/HIF-1α interaction, and exert potent anticancer activity in several animal models. In the present work, KCN1-bead affinity pull down, (14)C-labeled KCN1 binding, and KCN1-surface plasmon resonance measurements provide initial support for a mechanism in which KCN1 can bind to the CH1 domain of p300 and likely prevent the p300/HIF-1α assembly. Using a previously reported NMR structure of the p300/HIF-1α complex, we have identified potential binding sites in the p300-CH1 domain. A two-site binding model coupled with IC50 values has allowed establishment of a modest ROC-based enrichment and creation of a guide for future analogue synthesis.
Collapse
Affiliation(s)
- Qi Shi
- Department
of Chemistry, Emory University, Atlanta,
Georgia 30322, United States
| | - Shaoman Yin
- Laboratory of Molecular Neuro-Oncology,
Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Stefan Kaluz
- Laboratory of Molecular Neuro-Oncology,
Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
| | - Nanting Ni
- Department of Chemistry and Center
for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30302-4098, United States
| | - Narra Sarojini Devi
- Laboratory of Molecular Neuro-Oncology,
Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Jiyoung Mun
- Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30322, United States
| | - Danzhu Wang
- Department of Chemistry and Center
for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30302-4098, United States
| | - Krishna Damera
- Department of Chemistry and Center
for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30302-4098, United States
| | - Weixuan Chen
- Department of Chemistry and Center
for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30302-4098, United States
| | - Sarah Burroughs
- Department of Chemistry and Center
for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30302-4098, United States
| | - Suazette Reid Mooring
- Department of Chemistry and Center
for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30302-4098, United States
| | - Mark M. Goodman
- Department of Hematology and
Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
- Radiology and Imaging Sciences, Emory University, Atlanta, Georgia 30322, United States
| | - Erwin G. Van Meir
- Laboratory of Molecular Neuro-Oncology,
Department of Neurosurgery, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Department of Hematology and
Medical Oncology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, United States
| | - Binghe Wang
- Department of Chemistry and Center
for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia 30302-4098, United States
| | - James P. Snyder
- Department
of Chemistry, Emory University, Atlanta,
Georgia 30322, United States
- Emory Institute for Drug Discovery, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
27
|
Mun J, Jabbar AA, Devi NS, Yin S, Wang Y, Tan C, Culver D, Snyder JP, Van Meir EG, Goodman MM. Design and in vitro activities of N-alkyl-N-[(8-R-2,2-dimethyl-2H-chromen-6-yl)methyl]heteroarylsulfonamides, novel, small-molecule hypoxia inducible factor-1 pathway inhibitors and anticancer agents. J Med Chem 2012; 55:6738-50. [PMID: 22746274 PMCID: PMC3756490 DOI: 10.1021/jm300752n] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The hypoxia inducible factor (HIF) pathway is an attractive target for cancer, as it controls tumor adaptation to growth under hypoxia and mediates chemotherapy and radiation resistance. We previously discovered 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide as a novel, small-molecule HIF-1 pathway inhibitor in a high-throughput cell-based assay, but its in vivo delivery is hampered by poor aqueous solubility (0.009 μM in water; log P(7.4) = 3.7). Here we describe the synthesis of 12 N-alkyl-N-[(8-R-2,2-dimethyl-2H-chromen-6-yl)methyl]heteroarylsulfonamides, which were designed to possess optimal lipophilicities and aqueous solubilities by in silico calculations. Experimental log P(7.4) values of 8 of the 12 new analogs ranged from 1.2-3.1. Aqueous solubilities of three analogs were measured, among which the most soluble N-[(8-methoxy-2,2-dimethyl-2H-chromen-6-yl)methyl]-N-(propan-2-yl)pyridine-2-sulfonamide had an aqueous solubility of 80 μM, e.g., a solubility improvement of ∼9000-fold. The pharmacological optimization had limited impact on drug efficacy as the compounds retained IC(50) values at or below 5 μM in our HIF-dependent reporter assay.
Collapse
Affiliation(s)
- Jiyoung Mun
- Department of Radiology and imaging sciences, Emory University CSI, Wesley Woods Health Center, 1841 Clifton Road, NE, Atlanta, GA 30329
| | - Adnan Abdul Jabbar
- Hematology and Medical Oncology, Emory University School of Medicine, Emory University, Atlanta, GA 30322
| | | | - Shaoman Yin
- Department of Neurosurgery, Emory University, Atlanta, GA 30322
| | - Yingzhe Wang
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Atlanta, GA 30341
| | - Chalet Tan
- Department of Pharmaceutical Sciences, Mercer University College of Pharmacy, Atlanta, GA 30341
| | - Deborah Culver
- Emory Institute for Drug Discovery, Emory University, Atlanta, Georgia 30322
| | - James P. Snyder
- Emory Institute for Drug Discovery, Emory University, Atlanta, Georgia 30322
- Department of Chemistry, Emory University, Atlanta, Georgia, 30322
| | - Erwin G. Van Meir
- Department of Neurosurgery, Emory University, Atlanta, GA 30322
- Hematology and Medical Oncology, Emory University School of Medicine, Emory University, Atlanta, GA 30322
- Winship Cancer Institute, Emory University, Atlanta, GA 30322
| | - Mark M. Goodman
- Department of Radiology and imaging sciences, Emory University CSI, Wesley Woods Health Center, 1841 Clifton Road, NE, Atlanta, GA 30329
- Winship Cancer Institute, Emory University, Atlanta, GA 30322
| |
Collapse
|