1
|
Goher SS, Abdrabo WS, Veerakanellore GB, Elgendy B. 2,5-Diketopiperazines (DKPs): Promising Scaffolds for Anticancer Agents. Curr Pharm Des 2024; 30:597-623. [PMID: 38343054 DOI: 10.2174/0113816128291798240201112916] [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: 11/07/2023] [Accepted: 01/22/2024] [Indexed: 05/25/2024]
Abstract
2,5-Diketopiperazine (2,5-DKP) derivatives represent a family of secondary metabolites widely produced by bacteria, fungi, plants, animals, and marine organisms. Many natural products with DKP scaffolds exhibited various pharmacological activities such as antiviral, antifungal, antibacterial, and antitumor. 2,5-DKPs are recognized as privileged structures in medicinal chemistry, and compounds that incorporate the 2,5-DKP scaffold have been extensively investigated for their anticancer properties. This review is a thorough update on the anti-cancer activity of natural and synthesized 2,5-DKPs from 1997 to 2022. We have explored various aspects of 2,5-DKPs modifications and summarized their structure-activity relationships (SARs) to gain insight into their anticancer activities. We have also highlighted the novel approaches to enhance the specificity and pharmacokinetics of 2,5-DKP-based anticancer agents.
Collapse
Affiliation(s)
- Shaimaa S Goher
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
- Nanotechnology Research Centre (NTRC), The British University in Egypt (BUE), Suez Desert Road, El Sherouk City, Cairo 1183, Egypt
| | - Wessam S Abdrabo
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
| | - Giri Babu Veerakanellore
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences and Pharmacy, St. Louis, Missouri 63110, United States
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| | - Bahaa Elgendy
- Chemistry Department, Faculty of Science, Benha University, Benha 13518, Egypt
- Center for Clinical Pharmacology, Washington University School of Medicine and University of Health Sciences and Pharmacy, St. Louis, Missouri 63110, United States
- Department of Anesthesiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri 63110, United States
| |
Collapse
|
2
|
Gomari MM, Abkhiz S, Pour TG, Lotfi E, Rostami N, Monfared FN, Ghobari B, Mosavi M, Alipour B, Dokholyan NV. Peptidomimetics in cancer targeting. Mol Med 2022; 28:146. [PMID: 36476230 PMCID: PMC9730693 DOI: 10.1186/s10020-022-00577-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/16/2022] [Indexed: 12/12/2022] Open
Abstract
The low efficiency of treatment strategies is one of the main obstacles to developing cancer inhibitors. Up to now, various classes of therapeutics have been developed to inhibit cancer progression. Peptides due to their small size and easy production compared to proteins are highly regarded in designing cancer vaccines and oncogenic pathway inhibitors. Although peptides seem to be a suitable therapeutic option, their short lifespan, instability, and low binding affinity for their target have not been widely applicable against malignant tumors. Given the peptides' disadvantages, a new class of agents called peptidomimetic has been introduced. With advances in physical chemistry and biochemistry, as well as increased knowledge about biomolecule structures, it is now possible to chemically modify peptides to develop efficient peptidomimetics. In recent years, numerous studies have been performed to the evaluation of the effectiveness of peptidomimetics in inhibiting metastasis, angiogenesis, and cancerous cell growth. Here, we offer a comprehensive review of designed peptidomimetics to diagnose and treat cancer.
Collapse
Affiliation(s)
- Mohammad Mahmoudi Gomari
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shadi Abkhiz
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Taha Ghantab Pour
- grid.411746.10000 0004 4911 7066Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ehsan Lotfi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Neda Rostami
- grid.411425.70000 0004 0417 7516Department of Chemical Engineering, Faculty of Engineering, Arak University, Arak, Iran
| | - Fatemeh Nafe Monfared
- grid.411705.60000 0001 0166 0922Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Ghobari
- grid.412831.d0000 0001 1172 3536Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mona Mosavi
- grid.411746.10000 0004 4911 7066Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behruz Alipour
- grid.411705.60000 0001 0166 0922Medical Biotechnology Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikolay V. Dokholyan
- grid.240473.60000 0004 0543 9901Department of Pharmacology, Penn State College of Medicine, Hershey, PA USA ,grid.240473.60000 0004 0543 9901Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA USA
| |
Collapse
|
3
|
Targeting RGD-binding integrins as an integrative therapy for diabetic retinopathy and neovascular age-related macular degeneration. Prog Retin Eye Res 2021; 85:100966. [PMID: 33775825 DOI: 10.1016/j.preteyeres.2021.100966] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/15/2021] [Accepted: 03/19/2021] [Indexed: 12/14/2022]
Abstract
Integrins are a class of transmembrane receptors that are involved in a wide range of biological functions. Dysregulation of integrins has been implicated in many pathological processes and consequently, they are attractive therapeutic targets. In the ophthalmology arena, there is extensive evidence suggesting that integrins play an important role in diabetic retinopathy (DR), age-related macular degeneration (AMD), glaucoma, dry eye disease and retinal vein occlusion. For example, there is extensive evidence that arginyl-glycyl-aspartic acid (Arg-Gly-Asp; RGD)-binding integrins are involved in key disease hallmarks of DR and neovascular AMD (nvAMD), specifically inflammation, vascular leakage, angiogenesis and fibrosis. Based on such evidence, drugs that engage integrin-linked pathways have received attention for their potential to block all these vision-threatening pathways. This review focuses on the pathophysiological role that RGD-binding integrins can have in complex multifactorial retinal disorders like DR, diabetic macular edema (DME) and nvAMD, which are leading causes of blindness in developed countries. Special emphasis will be given on how RGD-binding integrins can modulate the intricate molecular pathways and regulate the underlying pathological mechanisms. For instance, the interplay between integrins and key molecular players such as growth factors, cytokines and enzymes will be summarized. In addition, recent clinical advances linked to targeting RGD-binding integrins in the context of DME and nvAMD will be discussed alongside future potential for limiting progression of these diseases.
Collapse
|
4
|
Panzeri S, Arosio D, Gazzola S, Belvisi L, Civera M, Potenza D, Vasile F, Kemker I, Ertl T, Sewald N, Reiser O, Piarulli U. Cyclic RGD and isoDGR Integrin Ligands Containing cis-2-amino-1-cyclopentanecarboxylic ( cis-β-ACPC) Scaffolds. Molecules 2020; 25:molecules25245966. [PMID: 33339382 PMCID: PMC7766232 DOI: 10.3390/molecules25245966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Integrin ligands containing the tripeptide sequences Arg-Gly-Asp (RGD) and iso-Asp-Gly- Arg (isoDGR) were actively investigated as inhibitors of tumor angiogenesis and directing unit in tumor-targeting drug conjugates. Reported herein is the synthesis, of two RGD and one isoDGR cyclic peptidomimetics containing (1S,2R) and (1R,2S) cis-2-amino-1-cyclopentanecarboxylic acid (cis-β-ACPC), using a mixed solid phase/solution phase synthetic protocol. The three ligands were examined in vitro in competitive binding assays to the purified αvβ3 and α5β1 receptors using biotinylated vitronectin (αvβ3) and fibronectin (α5β1) as natural displaced ligands. The IC50 values of the ligands ranged from nanomolar (the two RGD ligands) to micromolar (the isoDGR ligand) with a pronounced selectivity for αvβ3 over α5β1. In vitro cell adhesion assays were also performed using the human skin melanoma cell line WM115 (rich in integrin αvβ3). The two RGD ligands showed IC50 values in the same micromolar range as the reference compound (cyclo[RGDfV]), while for the isoDGR derivative an IC50 value could not be measured for the cell adhesion assay. A conformational analysis of the free RGD and isoDGR ligands by NMR (VT-NMR and NOESY experiments) and computational studies (MC/EM and MD), followed by docking simulations performed in the αVβ3 integrin active site, provided a rationale for the behavior of these ligands toward the receptor.
Collapse
Affiliation(s)
- Silvia Panzeri
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell’Insubria, Via Valleggio 11, 22100 Como, Italy; (S.P.); (S.G.)
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany; (T.E.); (O.R.)
| | - Daniela Arosio
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Scienze e Tecnologie Chimiche (SCITEC), Giulio Natta, Via C. Golgi 19, 20133 Milan, Italy;
| | - Silvia Gazzola
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell’Insubria, Via Valleggio 11, 22100 Como, Italy; (S.P.); (S.G.)
| | - Laura Belvisi
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Monica Civera
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Donatella Potenza
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Francesca Vasile
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Isabell Kemker
- Department of Chemistry, Organic and Bioorganic Chemistry, University of Bielefeld, Universitätsstraße 25, 33615 Bielefeld, Germany; (I.K.); (N.S.)
| | - Thomas Ertl
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany; (T.E.); (O.R.)
| | - Norbert Sewald
- Department of Chemistry, Organic and Bioorganic Chemistry, University of Bielefeld, Universitätsstraße 25, 33615 Bielefeld, Germany; (I.K.); (N.S.)
| | - Oliver Reiser
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany; (T.E.); (O.R.)
| | - Umberto Piarulli
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell’Insubria, Via Valleggio 11, 22100 Como, Italy; (S.P.); (S.G.)
- Correspondence:
| |
Collapse
|
5
|
Zanella S, Bocchinfuso G, De Zotti M, Arosio D, Marino F, Raniolo S, Pignataro L, Sacco G, Palleschi A, Siano AS, Piarulli U, Belvisi L, Formaggio F, Gennari C, Stella L. Rational Design of Antiangiogenic Helical Oligopeptides Targeting the Vascular Endothelial Growth Factor Receptors. Front Chem 2019; 7:170. [PMID: 30984741 PMCID: PMC6449863 DOI: 10.3389/fchem.2019.00170] [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: 01/11/2019] [Accepted: 03/05/2019] [Indexed: 01/25/2023] Open
Abstract
Tumor angiogenesis, essential for cancer development, is regulated mainly by vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs), which are overexpressed in cancer cells. Therefore, the VEGF/VEGFR interaction represents a promising pharmaceutical target to fight cancer progression. The VEGF surface interacting with VEGFRs comprises a short α-helix. In this work, helical oligopeptides mimicking the VEGF-C helix were rationally designed based on structural analyses and computational studies. The helical conformation was stabilized by optimizing intramolecular interactions and by introducing helix-inducing Cα,α-disubstituted amino acids. The conformational features of the synthetic peptides were characterized by circular dichroism and nuclear magnetic resonance, and their receptor binding properties and antiangiogenic activity were determined. The best hits exhibited antiangiogenic activity in vitro at nanomolar concentrations and were resistant to proteolytic degradation.
Collapse
Affiliation(s)
- Simone Zanella
- Department of Chemistry, University of Milan, Milan, Italy
| | - Gianfranco Bocchinfuso
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Marta De Zotti
- Padova Unit, Department of Chemistry, Institute of Biomolecular Chemistry, CNR, University of Padova, Padova, Italy
| | - Daniela Arosio
- National Research Council, Institute of Molecular Science and Technologies, Milan, Italy
| | - Franca Marino
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Stefano Raniolo
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Luca Pignataro
- Department of Chemistry, University of Milan, Milan, Italy
| | - Giovanni Sacco
- Department of Chemistry, University of Milan, Milan, Italy
| | - Antonio Palleschi
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Alvaro S Siano
- Departamento de Química Organica, Facultad de Bioquímica y Ciencias Biologicas, Universidad Nacional del Litoral, Santa Fe, Argentina
| | - Umberto Piarulli
- Center for Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Laura Belvisi
- Department of Chemistry, University of Milan, Milan, Italy.,National Research Council, Institute of Molecular Science and Technologies, Milan, Italy
| | - Fernando Formaggio
- Padova Unit, Department of Chemistry, Institute of Biomolecular Chemistry, CNR, University of Padova, Padova, Italy
| | - Cesare Gennari
- Department of Chemistry, University of Milan, Milan, Italy.,National Research Council, Institute of Molecular Science and Technologies, Milan, Italy
| | - Lorenzo Stella
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
6
|
Hamdan F, Bigdeli Z, Asghari SM, Sadremomtaz A, Balalaie S. Synthesis of Modified RGD-Based Peptides and Their in vitro Activity. ChemMedChem 2019; 14:282-288. [PMID: 30506622 DOI: 10.1002/cmdc.201800704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Indexed: 11/07/2022]
Abstract
Arg-Gly-Asp (RGD) peptides represent the most outstanding recognition motif involved in cell adhesion that binds to the αv β3 integrin, which has been targeted for cancer therapy. Various RGD-containing peptides and peptidomimetics have been designed and synthesized to selectively inhibit this integrin. In this study, the synthesis of RGD-based peptides through the incorporation of the short bioactive peptide Phe-Ala-Lys-Leu-Phe (FAKLF) at the C and N termini of RGD has been achieved by using a solid-phase peptide synthesis approach. The peptides were purified by means of preparative RP-HPLC and their structures were confirmed through HRMS (ESI). The MTT assay revealed that the RGD and FAKLF peptides inhibited the proliferation of human umbilical vein endothelial cells (HUVECs) in a dose-dependent manner, with IC50 values of 3000 and 500 ng mL-1 , respectively. Interestingly, a drastic improvement was observed in the antiproliferative activity of the combined structures of the FAKLFRGD and RGDFAKLF peptides, leading to IC50 values of 200 and 136.7 ng mL-1 , respectively. Meanwhile, based on apoptosis results, the potential of peptides to induce apoptosis, in accordance with their antiproliferative activity, indicated that the RGD and FAKLF peptides, and the peptides synthesized based on their combinations induced cell apoptosis in a dose-dependent manner followed by inhibition of the proliferation of endothelial cells. Moreover, the incorporation of d-leucine increased the induction of apoptosis by these peptides.
Collapse
Affiliation(s)
- Fatima Hamdan
- Peptide Chemistry Research Center, K.N. Toosi University of Technology, P.O. Box 15875-4416, Tehran, Iran
| | - Zahra Bigdeli
- Peptide Chemistry Research Center, K.N. Toosi University of Technology, P.O. Box 15875-4416, Tehran, Iran
| | - S Mohsen Asghari
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Afsaneh Sadremomtaz
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Center, K.N. Toosi University of Technology, P.O. Box 15875-4416, Tehran, Iran.,Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
7
|
Investigating the Interaction of Cyclic RGD Peptidomimetics with α Vβ₆ Integrin by Biochemical and Molecular Docking Studies. Cancers (Basel) 2017; 9:cancers9100128. [PMID: 28934103 PMCID: PMC5664067 DOI: 10.3390/cancers9100128] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 01/15/2023] Open
Abstract
The interaction of a small library of cyclic RGD (Arg-Gly-Asp) peptidomimetics with αVβ6 integrin has been investigated by means of competitive solid phase binding assays to the isolated receptor and docking calculations in the crystal structure of the αVβ6 binding site. To this aim, a rigid receptor-flexible ligand docking protocol has been set up and then applied to predict the binding mode of the cyclic RGD peptidomimetics to αVβ6 integrin. Although the RGD interaction with αVβ6 recapitulates the RGD binding mode observed in αVβ3, differences between the integrin binding pockets can strongly affect the ligand binding ability. In general, the peptidomimetics exhibited IC50 values for integrin αVβ6 (i.e., the concentration of compound required for 50% inhibition of biotinylated fibronectin binding to isolated αVβ6 integrin) in the nanomolar range (77–345 nM), about 10–100 times higher than those for the related αVβ3 receptor, with a single notable ligand displaying a low nanomolar IC50 value (2.3 nM). Insights from the properties of the binding pocket combined with the analysis of the docking poses provided a rationale for ligand recognition and selectivity.
Collapse
|
8
|
Liu HT, Xing AY, Chen X, Ma RR, Wang YW, Shi DB, Zhang H, Li P, Chen HF, Li YH, Gao P. MicroRNA-27b, microRNA-101 and microRNA-128 inhibit angiogenesis by down-regulating vascular endothelial growth factor C expression in gastric cancers. Oncotarget 2016; 6:37458-70. [PMID: 26460960 PMCID: PMC4741941 DOI: 10.18632/oncotarget.6059] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 09/23/2015] [Indexed: 01/30/2023] Open
Abstract
Vascular Endothelial Growth Factor C (VEGF-C) has critical roles in angiogenesis in human cancers; however, the underlying mechanisms regulating VEGF-C expression remain largely unknown. In the present study, VEGF-C protein expression and the density of blood vessels or lymphatic vessels were determined by immunohistochemistry in 103 cases of gastric cancer tissues. Suppression of VEGF-C by miR-27b, miR-101 and miR-128 was investigated by luciferase assays, Western blot and ELISA. The miRNAs expression levels were detected in human gastric cancers by real-time quantitative PCR. Cell proliferation, migration and invasion assays were performed to assess the effect of miRNAs on gastric cancer cells and human umbilical vascular endothelial cells (HUVECs). Our data showed that high VEGF-C expression was significantly associated with increased tumor size, advanced TNM classification and clinical stage, higher microvessel density (MVD) and lymphatic density (LVD), as well as poor survival in patients with gastric cancer. Furthermore, VEGF-C was found to be a direct target gene of miR-27b, miR-101, and miR-128. The expression levels of the three miRNAs were inversely correlated with MVD. Overexpression of miR-27b, miR-101, or miR-128 suppressed migration, proliferation activity, and tube formation in HUVECs by repressing VEGF-C secretion in gastric cancer cells. We conclude that miR-27b, miR-101 and miR-128 inhibit angiogenesis by down-regulating VEGF-C expression in gastric cancers.
Collapse
Affiliation(s)
- Hai-Ting Liu
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ai-Yan Xing
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Xu Chen
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ran-Ran Ma
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ya-Wen Wang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Duan-Bo Shi
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Hui Zhang
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Peng Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Hong-Fang Chen
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China.,Department of Pathology, Qingzhou Center Hospital, Weifang, P.R. China
| | - Yu-Hong Li
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China.,Department of Pathology, Liaocheng Peoples Hospital, Liaocheng, P.R. China
| | - Peng Gao
- Department of Pathology, Qilu Hospital, Shandong University, Jinan, P.R. China
| |
Collapse
|
9
|
Cui YF, Liu AH, An DZ, Sun RB, Shi Y, Shi YX, Shi M, Zhang Q, Wang LL, Feng Q, Pan GL, Wang Q. Claudin-4 is required for vasculogenic mimicry formation in human breast cancer cells. Oncotarget 2016; 6:11087-97. [PMID: 25871476 PMCID: PMC4484441 DOI: 10.18632/oncotarget.3571] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/22/2015] [Indexed: 11/29/2022] Open
Abstract
Vasculogenic mimicry (VM) refers to the unique capability of aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks. Claudins are aberrantly expressed in aggressive breast cancer. However, the relationship between claudins and VM formation is not clear. We examined VM in two human breast cancer cell lines with different aggressive capabilities (MDA-MB-231 and MCF-7 cells) and one human umbilical vein endothelial cell line (HUVEC). Both HUVEC and MDA-MB-231 cells formed vascular channels in Matrigel cultures, while MCF-7 cells did not. Western blot analysis revealed a possible correlation between claudin-4 and -6 expression in breast cancer cell lines and tumor aggressiveness, with protein levels correlating with the ability to form vascular channels. Treatment of MDA-MB-231 and HUVEC cells with claudin-4 monoclonal antibodies completely inhibited the ability of cells to form vascular channels. Moreover, knockdown of claudin-4 by short hairpin RNA completely inhibited tubule formation in MDA-MB-231 cells. Overexpression of claudin-4 in MCF-7 cells induced formation of vascular channels. Immunocytochemistry revealed that membranous claudin-4 protein was significantly associated with vascular channel formation. Collectively, these results indicate that claudin-4 may play a critical role in VM in human breast cancer cells, opening new opportunities to improve aggressive breast cancer therapy.
Collapse
Affiliation(s)
- Yong-Feng Cui
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - An-Heng Liu
- Cardiovascular Medicine, Affiliated Hospital of Academy of Military Medical Sciences, Beijing, China
| | - Dai-Zhi An
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Ru-Bao Sun
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Yun Shi
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Yun-Xiang Shi
- Department of Physiology, BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Miao Shi
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Qiang Zhang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Li-Li Wang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| | - Qiong Feng
- Department of Physiology, BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Gui-Lan Pan
- Department of Physiology, BaoTou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Qiang Wang
- Center of Hygiene Assessment and Research, Institute of Disease Control and Prevention, Academy of Military Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Zanella S, Mingozzi M, Dal Corso A, Fanelli R, Arosio D, Cosentino M, Schembri L, Marino F, De Zotti M, Formaggio F, Pignataro L, Belvisi L, Piarulli U, Gennari C. Synthesis, Characterization, and Biological Evaluation of a Dual-Action Ligand Targeting αvβ3 Integrin and VEGF Receptors. ChemistryOpen 2015; 4:633-41. [PMID: 26491644 PMCID: PMC4608532 DOI: 10.1002/open.201500062] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Indexed: 12/17/2022] Open
Abstract
A dual-action ligand targeting both integrin αVβ3 and vascular endothelial growth factor receptors (VEGFRs), was synthesized via conjugation of a cyclic peptidomimetic αVβ3 Arg-Gly-Asp (RGD) ligand with a decapentapeptide. The latter was obtained from a known VEGFR antagonist by acetylation at the Lys13 side chain. Functionalization of the precursor ligands was carried out in solution and in the solid phase, affording two fragments: an alkyne VEGFR ligand and the azide integrin αVβ3 ligand, which were conjugated by click chemistry. Circular dichroism studies confirmed that both the RGD and VEGFR ligand portions of the dual-action compound substantially adopt the biologically active conformation. In vitro binding assays on isolated integrin αVβ3 and VEGFR-1 showed that the dual-action conjugate retains a good level of affinity for both its target receptors, although with one order of magnitude (10/20 times) decrease in potency. The dual-action ligand strongly inhibited the VEGF-induced morphogenesis in Human Umbilical Vein Endothelial Cells (HUVECs). Remarkably, its efficiency in preventing the formation of new blood vessels was similar to that of the original individual ligands, despite the worse affinity towards integrin αVβ3 and VEGFR-1.
Collapse
Affiliation(s)
- Simone Zanella
- Dipartimento di Chimica, Università degli Studi di MilanoVia C. Golgi 19, 20133, Milan, Italy
| | - Michele Mingozzi
- Dipartimento di Chimica, Università degli Studi di MilanoVia C. Golgi 19, 20133, Milan, Italy
| | - Alberto Dal Corso
- Dipartimento di Chimica, Università degli Studi di MilanoVia C. Golgi 19, 20133, Milan, Italy
| | - Roberto Fanelli
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell'InsubriaVia Valleggio 11, 22100, Como, Italy
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari (ISTM), National Research Council (CNR)Via C. Golgi 19, 20133, Milan, Italy
| | - Marco Cosentino
- Center for Research in Medical Pharmacology, Università degli Studi dell'InsubriaVia Ottorino Rossi 9, 21100, Varese, Italy
| | - Laura Schembri
- Center for Research in Medical Pharmacology, Università degli Studi dell'InsubriaVia Ottorino Rossi 9, 21100, Varese, Italy
| | - Franca Marino
- Center for Research in Medical Pharmacology, Università degli Studi dell'InsubriaVia Ottorino Rossi 9, 21100, Varese, Italy
| | - Marta De Zotti
- Istituto di Chimica Biomolecolare, CNR, Unità di Padova, Dipartimento di Chimica, Università degli Studi di PadovaVia Marzolo 1, 35131, Padova, Italy
| | - Fernando Formaggio
- Istituto di Chimica Biomolecolare, CNR, Unità di Padova, Dipartimento di Chimica, Università degli Studi di PadovaVia Marzolo 1, 35131, Padova, Italy
| | - Luca Pignataro
- Dipartimento di Chimica, Università degli Studi di MilanoVia C. Golgi 19, 20133, Milan, Italy
| | - Laura Belvisi
- Dipartimento di Chimica, Università degli Studi di MilanoVia C. Golgi 19, 20133, Milan, Italy
| | - Umberto Piarulli
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell'InsubriaVia Valleggio 11, 22100, Como, Italy
| | - Cesare Gennari
- Dipartimento di Chimica, Università degli Studi di MilanoVia C. Golgi 19, 20133, Milan, Italy
| |
Collapse
|
11
|
Panzeri S, Zanella S, Arosio D, Vahdati L, Dal Corso A, Pignataro L, Paolillo M, Schinelli S, Belvisi L, Gennari C, Piarulli U. Cyclic isoDGR and RGD peptidomimetics containing bifunctional diketopiperazine scaffolds are integrin antagonists. Chemistry 2015; 21:6265-71. [PMID: 25761230 DOI: 10.1002/chem.201406567] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Indexed: 01/18/2023]
Abstract
The cyclo[DKP-isoDGR] peptidomimetics 2-5, containing bifunctional diketopiperazine (DKP) scaffolds that differ in the configuration of the two DKP stereocenters and in the substitution at the DKP nitrogen atoms, were prepared and examined in vitro in competitive binding assays with purified αv β3 and αv β5 integrin receptors. IC50 values ranged from low nanomolar (ligand 3) to submicromolar with αv β3 integrin. The biological activities of ligands cyclo[DKP3-RGD] 1 and cyclo[DKP3-isoDGR] 3, bearing the same bifunctional DKP scaffold and showing similar αV β3 integrin binding values, were compared in terms of their cellular effects in human U373 glioblastoma cells. Compounds 1 and 3 displayed overlapping inhibitory effects on the FAK/Akt integrin activated transduction pathway and on integrin-mediated cell infiltration processes, and qualify therefore, despite the different RGD and isoDGR sequences, as integrin antagonists. Both compounds induced apoptosis in glioma cells after 72 hour treatment.
Collapse
Affiliation(s)
- Silvia Panzeri
- Università degli Studi dell'Insubria, Dipartimento di Scienza e Alta Tecnologia, Via Valleggio 11, 22100 Como (Italy)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|