1
|
Anand U, Bandyopadhyay A, Jha NK, Pérez de la Lastra JM, Dey A. Translational aspect in peptide drug discovery and development: An emerging therapeutic candidate. Biofactors 2022; 49:251-269. [PMID: 36326181 DOI: 10.1002/biof.1913] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 08/11/2022] [Indexed: 11/06/2022]
Abstract
In the last two decades, protein-protein interactions (PPIs) have been used as the main target for drug development. However, with larger or superficial binding sites, it has been extremely difficult to disrupt PPIs with small molecules. On the other hand, intracellular PPIs cannot be targeted by antibodies that cannot penetrate the cell membrane. Peptides that have a combination of conformational rigidity and flexibility can be used to target difficult binding interfaces with appropriate binding affinity and specificity. Since the introduction of insulin nearly a century ago, more than 80 peptide drugs have been approved to treat a variety of diseases. These include deadly diseases such as cancer and human immunodeficiency virus infection. It is also useful against diabetes, chronic pain, and osteoporosis. Today, more research is being done on these drugs as lessons learned from earlier approaches, which are still valid today, complement newer approaches such as peptide display libraries. At the same time, integrated genomics and peptide display libraries are new strategies that open new avenues for peptide drug discovery. The purpose of this review is to examine the problems in elucidating the peptide-protein recognition mechanism. This is important to develop peptide-based interventions that interfere with endogenous protein interactions. New approaches are being developed to improve the binding affinity and specificity of existing approaches and to develop peptide agents as potentially useful drugs. We also highlight the key challenges that must be overcome in peptide drug development to realize their potential and provide an overview of recent trends in peptide drug development. In addition, we take an in-depth look at early efforts in human hormone discovery, smart medicinal chemistry and design, natural peptide drugs, and breakthrough advances in molecular biology and peptide chemistry.
Collapse
Affiliation(s)
- Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, Punjab, India
- Department of Biotechnology, School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - José M Pérez de la Lastra
- Biotechnology of Macromolecules Research Group, Instituto de Productos Naturales y Agrobiología, IPNA-CSIC, Tenerife, Spain
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
2
|
Agamennone M, Fantacuzzi M, Vivenzio G, Scala MC, Campiglia P, Superti F, Sala M. Antiviral Peptides as Anti-Influenza Agents. Int J Mol Sci 2022; 23:11433. [PMID: 36232735 PMCID: PMC9569631 DOI: 10.3390/ijms231911433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Influenza viruses represent a leading cause of high morbidity and mortality worldwide. Approaches for fighting flu are seasonal vaccines and some antiviral drugs. The development of the seasonal flu vaccine requires a great deal of effort, as careful studies are needed to select the strains to be included in each year's vaccine. Antiviral drugs available against Influenza virus infections have certain limitations due to the increased resistance rate and negative side effects. The highly mutative nature of these viruses leads to the emergence of new antigenic variants, against which the urgent development of new approaches for antiviral therapy is needed. Among these approaches, one of the emerging new fields of "peptide-based therapies" against Influenza viruses is being explored and looks promising. This review describes the recent findings on the antiviral activity, mechanism of action and therapeutic capability of antiviral peptides that bind HA, NA, PB1, and M2 as a means of countering Influenza virus infection.
Collapse
Affiliation(s)
- Mariangela Agamennone
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Marialuigia Fantacuzzi
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| | - Giovanni Vivenzio
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Maria Carmina Scala
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Fabiana Superti
- National Centre for Innovative Technologies in Public Health, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marina Sala
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| |
Collapse
|
3
|
Vasco AV, Ceballos LG, Wessjohann LA, Rivera DG. Multicomponent Functionalization of the Octreotide Peptide Macrocyclic Scaffold. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Aldrin V. Vasco
- Leibniz Institute of Plant Biochemistry: Leibniz-Institut fur Pflanzenbiochemie Bioorganic Chemistry GERMANY
| | | | - Ludger A. Wessjohann
- Leibniz Institute of Plant Biochemistry: Leibniz-Institut fur Pflanzenbiochemie Bioorganic Chemistry GERMANY
| | - Daniel García Rivera
- Universidad de la Habana Laboratory of Synthetic and Biomolecular Chemistry Zapata y G 10400 La Habana CUBA
| |
Collapse
|
4
|
Hydrophilic Interaction Liquid Chromatography Coupled with Fluorescence Detection (HILIC-FL) for the Quantitation of Octreotide in Injection Forms. ANALYTICA 2021. [DOI: 10.3390/analytica2040012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Octreotide is a synthetic cyclic octapeptide analogue of somatostatin-14. It is mainly administered for the treatment of acromegaly, severe diarrhea, and neuroendocrine neoplasias. In this work, a hydrophilic interaction liquid chromatography (HILIC) method with fluorescence (FL) detection was developed and validated for the quantitation of octreotide in solutions for injection. Chromatographic separation was performed on an XBridge®-HILIC analytical column under isocratic elution with a short chromatographic run time of less than 10 min. The mobile phase consisted of ammonium bicarbonate 8.6 mM (pH 8.1)/acetonitrile 35/65 (v/v). The high sensitivity and selectivity of the fluorescence detection, with the excitation wavelength (λexcitation) set at 280 nm and the emission wavelength set at (λemission) 330 nm, enabled a simple sample preparation procedure that included only dilution steps. The calibration curve showed good linearity with a correlation coefficient greater than 0.998. The method was successfully applied to the analysis of commercially available octreotide injection forms.
Collapse
|
5
|
Abstract
Since the introduction of insulin almost a century ago, more than 80 peptide drugs have reached the market for a wide range of diseases, including diabetes, cancer, osteoporosis, multiple sclerosis, HIV infection and chronic pain. In this Perspective, we summarize key trends in peptide drug discovery and development, covering the early efforts focused on human hormones, elegant medicinal chemistry and rational design strategies, peptide drugs derived from nature, and major breakthroughs in molecular biology and peptide chemistry that continue to advance the field. We emphasize lessons from earlier approaches that are still relevant today as well as emerging strategies such as integrated venomics and peptide-display libraries that create new avenues for peptide drug discovery. We also discuss the pharmaceutical landscape in which peptide drugs could be particularly valuable and analyse the challenges that need to be addressed for them to reach their full potential.
Collapse
|
6
|
Paramonov VM, Gerstenberg M, Sahlgren C, Lindén M, Rivero-Müller A. In vitro Targetability Validation of Peptide-Functionalized Mesoporous Silica Nanoparticles in the Presence of Serum Proteins. Front Chem 2020; 8:603616. [PMID: 33282845 PMCID: PMC7691633 DOI: 10.3389/fchem.2020.603616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 10/12/2020] [Indexed: 11/13/2022] Open
Abstract
Demonstration of receptor-mediated targeting of nanoparticles to specific organs and/or cell types is an integral aim in many bionanomedicine development projects. However, engagement of targeted receptors with ligands on nanocarriers, which is the cornerstone of the active targeting concept, is challenging to study under biologically relevant conditions and thus often stays overlooked. In this work, we utilize an in-house established bioassay for in vitro targetability validation of mesoporous silica nanoparticles (MSNs), functionalized with high-affinity peptide ligands to somatostatin receptors via protective group chemistry, ensuring the correct orientation of the peptide's pharmacophore. We demonstrate that targeted nanoparticles, but not scrambled peptide-decorated counterparts, specifically engage the targeted receptors in living cells in culture media containing serum protein. The importance of being able to exclude false positives originating from the premature detachment of targeting peptides from the MSNs is highlighted.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland
| | | | - Cecilia Sahlgren
- Turku Bioscience, University of Turku and Åbo Akademi University, Turku, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Turku, Finland.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Mika Lindén
- Department of Inorganic Chemistry II, Ulm University, Ulm, Germany
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Turku, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
7
|
Avdeev DV, Sidorova MV, Ovchinnikov MV, Moiseeva NI, Osipov VN, Balaev AN, Khachatryan DS. Synthesis and Antitumor Activity of Conjugates Based on the Phe-D-Trp-Lys-Thr Peptide Fragment of Somatostatin. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2019. [DOI: 10.1134/s1068162019040034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Solomonov AV, Marfin YS, Rumyantsev EV, Ragozin E, Zahavi TS, Gellerman G, Tesler AB, Muench F, Kumagai A, Miyawaki A. Self-assembled micellar clusters based on Triton-X-family surfactants for enhanced solubilization, encapsulation, proteins permeability control, and anticancer drug delivery. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 99:794-804. [DOI: 10.1016/j.msec.2019.02.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Revised: 02/03/2019] [Accepted: 02/04/2019] [Indexed: 12/11/2022]
|
9
|
Rovira A, Gandioso A, Goñalons M, Galindo A, Massaguer A, Bosch M, Marchán V. Solid-Phase Approaches for Labeling Targeting Peptides with Far-Red Emitting Coumarin Fluorophores. J Org Chem 2019; 84:1808-1817. [PMID: 30628454 DOI: 10.1021/acs.joc.8b02624] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Fluorophores based on organic molecules hold great potential for ligand-targeted imaging applications, particularly those operating in the optical window in biological tissues. In this work, we have developed three straightforward solid-phase approaches based on amide-bond formation or a Cu(I)-catalyzed azide-alkyne click (CuAAC) reaction for labeling an octreotide peptide with far-red emitting coumarin-based COUPY dyes. First, the conjugatable versions of COUPY fluorophores incorporating the required functional groups (e.g., carboxylic acid, azide, or alkyne) were synthesized and characterized. All of them were found fully compatible with Fmoc/ tBu solid-phase peptide synthesis, which allowed for the labeling of octreotide either through amide-bond formation or by CuAAC reaction. A near quantitative conversion was obtained after only 1 h of reaction at RT when using CuSO4 and sodium ascorbate independently of the click chemistry approach used (azido-COUPY/alkynyl-peptide resin or alkynyl-COUPY/azido-peptide resin). COUPY-octreotide conjugates were found stable in cell culture medium as well as noncytotoxic in HeLa cells, and their spectroscopic and photophysical properties were found similar to those of their parent coumarin dyes. Finally, the potential bioimaging applications of COUPY-octreotide conjugates were demonstrated by confocal microscopy through the visualization of living HeLa cells overexpressing the somatostatin subtype-2 receptor.
Collapse
Affiliation(s)
- Anna Rovira
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB , Universitat de Barcelona , Martí i Franquès 1-11 , E-08028 Barcelona , Spain
| | - Albert Gandioso
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB , Universitat de Barcelona , Martí i Franquès 1-11 , E-08028 Barcelona , Spain
| | - Marina Goñalons
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB , Universitat de Barcelona , Martí i Franquès 1-11 , E-08028 Barcelona , Spain
| | - Alex Galindo
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB , Universitat de Barcelona , Martí i Franquès 1-11 , E-08028 Barcelona , Spain
| | - Anna Massaguer
- Departament de Biologia , Universitat de Girona , E-17071 Girona , Spain
| | - Manel Bosch
- Unitat de Microscòpia Òptica Avançada, Centres Científics i Tecnològics , Universitat de Barcelona , E-08028 Barcelona , Spain
| | - Vicente Marchán
- Departament de Química Inorgànica i Orgànica, Secció de Química Orgànica, IBUB , Universitat de Barcelona , Martí i Franquès 1-11 , E-08028 Barcelona , Spain
| |
Collapse
|
10
|
Paramonov VM, Desai D, Kettiger H, Mamaeva V, Rosenholm JM, Sahlgren C, Rivero-Müller A. Targeting Somatostatin Receptors By Functionalized Mesoporous Silica Nanoparticles - Are We Striking Home? Nanotheranostics 2018; 2:320-346. [PMID: 30148051 PMCID: PMC6107779 DOI: 10.7150/ntno.23826] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 06/25/2018] [Indexed: 12/02/2022] Open
Abstract
The concept of delivering nanoformulations to desired tissues by means of targeting membrane receptors of high local abundance by ligands anchored to the nanocarrier has gained a lot of attention over the last decade. Currently, there is no unanimous opinion on whether surface functionalization of nanocarriers by targeting ligands translates into any real benefit in terms of pharmacokinetics or treatment outcomes. Having examined the published nanocarriers designed to engage with somatostatin receptors, we realized that in the majority of cases targetability claims were not supported by solid evidence of targeting ligand-targeted receptor coupling, which is the very crux of a targetability concept. Here, we present an approach to characterize targetability of mesoporous silica-based nanocarriers functionalized with ligands of somatostatin receptors. The targetability proof in our case comes from a functional assay based on a genetically-encoded cAMP probe, which allows for real-time capture of receptor activation in living cells, triggered by targeting ligands on nanoparticles. We elaborate on the development and validation of the assay, highlighting the power of proper functional tests in the characterization pipeline of targeted nanoformulations.
Collapse
Affiliation(s)
- Valeriy M Paramonov
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Diti Desai
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Helene Kettiger
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Veronika Mamaeva
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Finland
| | - Cecilia Sahlgren
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Adolfo Rivero-Müller
- Institute of Biomedicine, Research Centre for Integrative Physiology and Pharmacology, University of Turku, Finland.,Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Finland.,Faculty of Science and Engineering, Cell Biology, Åbo Akademi University, Finland.,Department of Biochemistry and Molecular Biology, Medical University of Lublin, Poland
| |
Collapse
|
11
|
Gandioso A, Massaguer A, Villegas N, Salvans C, Sánchez D, Brun-Heath I, Marchán V, Orozco M, Terrazas M. Efficient siRNA-peptide conjugation for specific targeted delivery into tumor cells. Chem Commun (Camb) 2018; 53:2870-2873. [PMID: 28218319 DOI: 10.1039/c6cc10287e] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Despite the broad applicability of the Huisgen cycloaddition reaction, the click functionalization of RNAs with peptides still remains a challenge. Here we describe a straightforward method for the click functionalization of siRNAs with peptides of different sizes and complexities. Among them, a promising peptide carrier for the selective siRNA delivery into HER2+ breast cancer cell lines has been reported.
Collapse
Affiliation(s)
- Albert Gandioso
- Department of Inorganic and Organic Chemistry, Section of Organic Chemistry, IBUB, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain
| | - Anna Massaguer
- Department of Biology, University of Girona, Campus Montilivi, 17071 Girona, Spain
| | - Núria Villegas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Joint IRB-BSC Program in Computational Biology, Baldiri Reixac 10-12, 08028 Barcelona, Spain. and The Join IRB-BSC Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Spain
| | - Cándida Salvans
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Joint IRB-BSC Program in Computational Biology, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| | - Dani Sánchez
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Joint IRB-BSC Program in Computational Biology, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| | - Isabelle Brun-Heath
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Joint IRB-BSC Program in Computational Biology, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| | - Vicente Marchán
- Department of Inorganic and Organic Chemistry, Section of Organic Chemistry, IBUB, University of Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain
| | - Modesto Orozco
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Joint IRB-BSC Program in Computational Biology, Baldiri Reixac 10-12, 08028 Barcelona, Spain. and The Join IRB-BSC Program in Computational Biology, Institute for Research in Biomedicine (IRB Barcelona), Spain and Department of Biochemistry and Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Montserrat Terrazas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Joint IRB-BSC Program in Computational Biology, Baldiri Reixac 10-12, 08028 Barcelona, Spain.
| |
Collapse
|
12
|
Novohradsky V, Zamora A, Gandioso A, Brabec V, Ruiz J, Marchán V. Somatostatin receptor-targeted organometallic iridium(iii) complexes as novel theranostic agents. Chem Commun (Camb) 2017; 53:5523-5526. [PMID: 28466888 DOI: 10.1039/c7cc01946g] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
A novel somatostatin receptor-targeted anticancer agent based on the conjugation of a highly cytotoxic and luminescent cyclometalated iridium(iii) complex to tumor-targeting vectors based on octreotide peptide has been described, and its potential for targeted theranostic applications has been demonstrated.
Collapse
Affiliation(s)
- Vojtech Novohradsky
- Institute of Biophysics, Academy of Sciences of the Czech Republic, v.v.i. Kralovopolska 135, 612 65 Brno, Czech Republic.
| | | | | | | | | | | |
Collapse
|
13
|
Pekošak A, Rotstein BH, Collier TL, Windhorst AD, Vasdev N, Poot AJ. Stereoselective11C Labeling of a “Native” Tetrapeptide by Using Asymmetric Phase-Transfer Catalyzed Alkylation Reactions. European J Org Chem 2017. [DOI: 10.1002/ejoc.201601641] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Aleksandra Pekošak
- Department of Radiology and Nuclear Medicine; VU University Medical Center; De Boelelaan 1085 c 1081HV Amsterdam Netherland
- Division of Nuclear Medicine and Molecular Imaging Institution; Massachusetts General Hospital; 02114 Boston MA USA
- Department of Radiology; Harvard Medical School; 02115 Boston MA USA
| | - Benjamin H. Rotstein
- Division of Nuclear Medicine and Molecular Imaging Institution; Massachusetts General Hospital; 02114 Boston MA USA
- Department of Radiology; Harvard Medical School; 02115 Boston MA USA
| | - Thomas L. Collier
- Division of Nuclear Medicine and Molecular Imaging Institution; Massachusetts General Hospital; 02114 Boston MA USA
- Department of Radiology; Harvard Medical School; 02115 Boston MA USA
- Advion Inc.; 14850 Ithaca NY USA
| | - Albert D. Windhorst
- Department of Radiology and Nuclear Medicine; VU University Medical Center; De Boelelaan 1085 c 1081HV Amsterdam Netherland
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging Institution; Massachusetts General Hospital; 02114 Boston MA USA
- Department of Radiology; Harvard Medical School; 02115 Boston MA USA
| | - Alex J. Poot
- Department of Radiology and Nuclear Medicine; VU University Medical Center; De Boelelaan 1085 c 1081HV Amsterdam Netherland
| |
Collapse
|
14
|
Bandholtz S, Erdmann S, von Hacht JL, Exner S, Krause G, Kleinau G, Grötzinger C. Urolinin: The First Linear Peptidic Urotensin-II Receptor Agonist. J Med Chem 2016; 59:10100-10112. [PMID: 27791374 DOI: 10.1021/acs.jmedchem.6b00164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
This study investigated the role of individual U-II amino acid positions and side chain characteristics important for U-IIR activation. A complete permutation library of 209 U-II variants was studied in an activity screen that contained single substitution variants of each position with one of the other 19 proteinogenic amino acids. Receptor activation was measured using a cell-based high-throughput fluorescence calcium mobilization assay. We generated the first complete U-II substitution map for U-II receptor activation, resulting in a detailed view into the structural features required for receptor activation, accompanied by complementary information from receptor modeling and ligand docking studies. On the basis of the systematic SAR study of U-II, we created 33 further short and linear U-II variants from eight to three amino acids in length, including d- and other non-natural amino acids. We identified the first high-potency linear U-II analogues. Urolinin, a linear U-II agonist (nWWK-Tyr(3-NO2)-Abu), shows low nanomolar potency as well as improved metabolic stability.
Collapse
Affiliation(s)
- Sebastian Bandholtz
- Campus Virchow-Klinikum, Department of Hepatology and Gastroenterology and Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin , Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Sarah Erdmann
- Campus Virchow-Klinikum, Department of Hepatology and Gastroenterology and Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin , Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Jan Lennart von Hacht
- Campus Virchow-Klinikum, Department of Hepatology and Gastroenterology and Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin , Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Samantha Exner
- Campus Virchow-Klinikum, Department of Hepatology and Gastroenterology and Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin , Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Gerd Krause
- Leibniz-Institut für Molekulare Pharmakologie , 13125 Berlin, Germany
| | - Gunnar Kleinau
- Institute of Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin , D-13353 Berlin, Germany
| | - Carsten Grötzinger
- Campus Virchow-Klinikum, Department of Hepatology and Gastroenterology and Molecular Cancer Research Center (MKFZ), Charité-Universitätsmedizin Berlin , Augustenburger Platz 1, D-13353 Berlin, Germany
| |
Collapse
|
15
|
Van der Poorten O, Knuhtsen A, Sejer Pedersen D, Ballet S, Tourwé D. Side Chain Cyclized Aromatic Amino Acids: Great Tools as Local Constraints in Peptide and Peptidomimetic Design. J Med Chem 2016; 59:10865-10890. [PMID: 27690430 DOI: 10.1021/acs.jmedchem.6b01029] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Constraining the conformation of flexible peptides is a proven strategy to increase potency, selectivity, and metabolic stability. The focus has mostly been on constraining the backbone dihedral angles; however, the correct orientation of the amino acid side chains (χ-space) that constitute the peptide pharmacophore is equally important. Control of χ-space utilizes conformationally constrained amino acids that favor, disfavor, or exclude the gauche (-), the gauche (+), or the trans conformation. In this review we focus on cyclic aromatic amino acids in which the side chain is connected to the peptide backbone to provide control of χ1- and χ2-space. The manifold applications for cyclized analogues of the aromatic amino acids Phe, Tyr, Trp, and His within peptide medicinal chemistry are showcased herein with examples of enzyme inhibitors and ligands for G protein-coupled receptors.
Collapse
Affiliation(s)
- Olivier Van der Poorten
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-Engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| | - Astrid Knuhtsen
- Department of Drug Design and Pharmacology, University of Copenhagen , Jagtvej 162, 2100 Copenhagen, Denmark
| | - Daniel Sejer Pedersen
- Department of Drug Design and Pharmacology, University of Copenhagen , Jagtvej 162, 2100 Copenhagen, Denmark
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-Engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| | - Dirk Tourwé
- Research Group of Organic Chemistry, Departments of Chemistry and Bio-Engineering Sciences, Vrije Universiteit Brussel , Pleinlaan 2, 1050 Brussels, Belgium
| |
Collapse
|
16
|
Fridén-Saxin M, Seifert T, Malo M, da Silva Andersson K, Pemberton N, Dyrager C, Friberg A, Dahlén K, Wallén EA, Grøtli M, Luthman K. Chroman-4-one and chromone based somatostatin β-turn mimetics. Eur J Med Chem 2016; 114:59-64. [DOI: 10.1016/j.ejmech.2016.02.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 11/28/2022]
|
17
|
Sabatino G, Guryanov I, Rombecchi A, Zanon J, Ricci A, Cabri W, Papini AM, Rovero P. Production of peptides as generic drugs: a patent landscape of octreotide. Expert Opin Ther Pat 2016; 26:485-95. [DOI: 10.1517/13543776.2016.1158810] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
18
|
Lesma G, Cecchi R, Cagnotto A, Gobbi M, Meneghetti F, Musolino M, Sacchetti A, Silvani A. Tetrahydro-β-carboline-based spirocyclic lactam as type II' β-turn: application to the synthesis and biological evaluation of somatostatine mimetics. J Org Chem 2013; 78:2600-10. [PMID: 23409740 DOI: 10.1021/jo302737j] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The synthesis of novel spirocyclic lactams, embodying D-tryptophan (Trp) amino acid as the central core and acting as peptidomimetics, is presented. It relies on the strategic combination of Seebach's self-reproduction of chirality chemistry and Pictet-Spengler condensation as key steps. Investigation of the conformational behavior by molecular modeling, X-ray crystallography, and NMR and IR spectroscopies suggests very stable and highly predictable type II' β-turn conformations for all compounds. Relying on this feature, we also pursued their application to two potential mimetics of the hormone somatostatin, a pharmaceutically relevant natural peptide, which contains a Trp-based type II' β-turn pharmacophore.
Collapse
Affiliation(s)
- Giordano Lesma
- Dipartimento di Chimica, Università degli Studi di Milano, via C. Golgi 19, 20133 Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Banerjee S, Wiggins WJ, Geoghegan JL, Anthony CT, Woltering EA, Masterson DS. Novel synthesis of various orthogonally protected Cα-methyllysine analogues and biological evaluation of a Vapreotide analogue containing (S)-α-methyllysine. Org Biomol Chem 2013; 11:6307-19. [DOI: 10.1039/c3ob41282b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
20
|
Barragán F, Carrion-Salip D, Gómez-Pinto I, González-Cantó A, Sadler PJ, de Llorens R, Moreno V, González C, Massaguer A, Marchán V. Somatostatin subtype-2 receptor-targeted metal-based anticancer complexes. Bioconjug Chem 2012; 23:1838-55. [PMID: 22871231 DOI: 10.1021/bc300173h] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Conjugates of a dicarba analogue of octreotide, a potent somatostatin agonist whose receptors are overexpressed on tumor cells, with [PtCl(2)(dap)] (dap = 1-(carboxylic acid)-1,2-diaminoethane) (3), [(η(6)-bip)Os(4-CO(2)-pico)Cl] (bip = biphenyl, pico = picolinate) (4), [(η(6)-p-cym)RuCl(dap)](+) (p-cym = p-cymene) (5), and [(η(6)-p-cym)RuCl(imidazole-CO(2)H)(PPh(3))](+) (6), were synthesized by using a solid-phase approach. Conjugates 3-5 readily underwent hydrolysis and DNA binding, whereas conjugate 6 was inert to ligand substitution. NMR spectroscopy and molecular dynamics calculations showed that conjugate formation does not perturb the overall peptide structure. Only 6 exhibited antiproliferative activity in human tumor cells (IC(50) = 63 ± 2 μM in MCF-7 cells and IC(50) = 26 ± 3 μM in DU-145 cells) with active participation of somatostatin receptors in cellular uptake. Similar cytotoxic activity was found in a normal cell line (IC(50) = 45 ± 2.6 μM in CHO cells), which can be attributed to a similar level of expression of somatostatin subtype-2 receptor. These studies provide new insights into the effect of receptor-binding peptide conjugation on the activity of metal-based anticancer drugs, and demonstrate the potential of such hybrid compounds to target tumor cells specifically.
Collapse
Affiliation(s)
- Flavia Barragán
- Departament de Química Orgànica and IBUB, Universitat de Barcelona, Martí i Franquès 1-11, E-08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Chen S, Morales-Sanfrutos J, Angelini A, Cutting B, Heinis C. Structurally diverse cyclisation linkers impose different backbone conformations in bicyclic peptides. Chembiochem 2012; 13:1032-8. [PMID: 22492661 DOI: 10.1002/cbic.201200049] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Indexed: 11/07/2022]
Abstract
Combinatorial libraries of structurally diverse peptide macrocycles offer a rich source for the development of high-affinity ligands to targets of interest. In this work we have developed linkers for the generation of genetically encoded bicyclic peptides and tested whether the peptides cyclised by them have significant variations in their backbone conformations. Two new cyclisation reagents, each containing three thiol-reactive groups, efficiently and selectively cyclised linear peptides containing three cysteine moieties. When the mesitylene linker of the bicyclic peptide PK15, a potent inhibitor of plasma kallikrein (K(i)=2 nM), was replaced by the new linkers, its inhibitory activity dropped by a factor of more than 1000, suggesting that the linkers impose different conformations on the peptide. Indeed, structural analysis by solution-state NMR revealed different NOE constraints in the three bicyclic peptides, indicating that these relatively small linkers at the centres of bicyclic peptide structures significantly influence the conformations of the peptides. These results demonstrate the prominent structural role of linkers in peptide macrocycles and suggest that application of different cyclisation linkers in a combinatorial fashion could be an attractive means to generate topologically diverse macrocycle libraries.
Collapse
Affiliation(s)
- Shiyu Chen
- Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
22
|
Erchegyi J, Cescato R, Waser B, Rivier JE, Reubi JC. N-imidazolebenzyl-histidine substitution in somatostatin and in its octapeptide analogue modulates receptor selectivity and function. J Med Chem 2011; 54:5981-7. [PMID: 21806016 DOI: 10.1021/jm200307v] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Despite 3 decades of focused chemical, biological, structural, and clinical developments, unusual properties of somatostatin (SRIF, 1) analogues are still being uncovered. Here we report the unexpected functional properties of 1 and the octapeptide cyclo(3-14)H-Cys-Phe-Phe-Trp(8)-Lys-Thr-Phe-Cys-OH (somatostatin numbering; OLT-8, 9) substituted by imBzl-l- or -d-His at position 8. These analogues were tested for their binding affinity to the five human somatostatin receptors (sst(1-5)), as well as for their functional properties (or functionalities) in an sst(3) internalization assay and in an sst(3) luciferase reporter gene assay. While substitution of Trp(8) in somatostatin by imBzl-l- or -d-His(8) results in sst(3) selectivity, substitution of Trp(8) in the octapeptide 9 by imBzl-l- or -d-His(8) results in loss of binding affinity for sst(1,2,4,5) and a radical functional switch from agonist to antagonist.
Collapse
Affiliation(s)
- Judit Erchegyi
- The Clayton Foundation Laboratories for Peptide Biology, The Salk Institute, 10010 N. Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
23
|
Barragán F, López-Senín P, Salassa L, Betanzos-Lara S, Habtemariam A, Moreno V, Sadler PJ, Marchán V. Photocontrolled DNA binding of a receptor-targeted organometallic ruthenium(II) complex. J Am Chem Soc 2011; 133:14098-108. [PMID: 21797210 DOI: 10.1021/ja205235m] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A photoactivated ruthenium(II) arene complex has been conjugated to two receptor-binding peptides, a dicarba analogue of octreotide and the Arg-Gly-Asp (RGD) tripeptide. These peptides can act as "tumor-targeting devices" since their receptors are overexpressed on the membranes of tumor cells. Both ruthenium-peptide conjugates are stable in aqueous solution in the dark, but upon irradiation with visible light, the pyridyl-derivatized peptides were selectively photodissociated from the ruthenium complex, as inferred by UV-vis and NMR spectroscopy. Importantly, the reactive aqua species generated from the conjugates, [(η(6)-p-cym)Ru(bpm)(H(2)O)](2+), reacted with the model DNA nucleobase 9-ethylguanine as well as with guanines of two DNA sequences, (5')dCATGGCT and (5')dAGCCATG. Interestingly, when irradiation was performed in the presence of the oligonucleotides, a new ruthenium adduct involving both guanines was formed as a consequence of the photodriven loss of p-cymene from the two monofunctional adducts. The release of the arene ligand and the formation of a ruthenated product with a multidentate binding mode might have important implications for the biological activity of such photoactivated ruthenium(II) arene complexes. Finally, photoreactions with the peptide-oligonucleotide hybrid, Phac-His-Gly-Met-linker-p(5')dCATGGCT, also led to arene release and to guanine adducts, including a GG chelate. The lack of interaction with the peptide fragment confirms the preference of such organometallic ruthenium(II) complexes for guanine over other potential biological ligands, such as histidine or methionine amino acids.
Collapse
Affiliation(s)
- Flavia Barragán
- Departament de Química Orgànica and IBUB, Universitat de Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Robertson CR, Flynn SP, White HS, Bulaj G. Anticonvulsant neuropeptides as drug leads for neurological diseases. Nat Prod Rep 2011; 28:741-62. [PMID: 21340067 DOI: 10.1039/c0np00048e] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Anticonvulsant neuropeptides are best known for their ability to suppress seizures and modulate pain pathways. Galanin, neuropeptide Y, somatostatin, neurotensin, dynorphin, among others, have been validated as potential first-in-class anti-epileptic or/and analgesic compounds in animal models of epilepsy and pain, but their therapeutic potential extends to other neurological indications, including neurodegenerative and psychatric disorders. Disease-modifying properties of neuropeptides make them even more attractive templates for developing new-generation neurotherapeutics. Arguably, efforts to transform this class of neuropeptides into drugs have been limited compared to those for other bioactive peptides. Key challenges in developing neuropeptide-based anticonvulsants are: to engineer optimal receptor-subtype selectivity, to improve metabolic stability and to enhance their bioavailability, including penetration across the blood–brain barrier (BBB). Here, we summarize advances toward developing systemically active and CNS-penetrant neuropeptide analogs. Two main objectives of this review are: (1) to provide an overview of structural and pharmacological properties for selected anticonvulsant neuropeptides and their analogs and (2) to encourage broader efforts to convert these endogenous natural products into drug leads for pain, epilepsy and other neurological diseases.
Collapse
Affiliation(s)
- Charles R Robertson
- College of Pharmacy, Department of Medicinal Chemistry, 421 Wakara Way, STE. 360 Salt Lake City, UT 84108, USA
| | | | | | | |
Collapse
|
25
|
Fridkin G, Maina T, Nock BA, Blat D, Lev-Goldman V, Scolnik Y, Kapitkovski A, Vachutinsky Y, Shechter Y, Levy Y. Intramolecular azo-bridge as a cystine disulfide bond surrogate: Somatostatin-14 and brain natriuretic peptide (BNP) analogs. Bioorg Med Chem 2011; 19:798-806. [DOI: 10.1016/j.bmc.2010.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Revised: 11/26/2010] [Accepted: 12/04/2010] [Indexed: 02/06/2023]
|
26
|
Barragán F, Moreno V, Marchán V. Solid-phase synthesis and DNA binding studies of dichloroplatinum(ii) conjugates of dicarba analogues of octreotide as new anticancer drugs. Chem Commun (Camb) 2009:4705-7. [PMID: 19641816 DOI: 10.1039/b909698a] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The first dichloroplatinum(ii) conjugates of dicarba analogues of octreotide, which is expected to act as a 'tumour-targeting device', have been efficiently synthesized following a stepwise solid-phase approach; these compounds emulate the mechanism of cisplatin since they form a 1,2-intrastrand cross-link with two consecutive guanines of an oligonucleotide.
Collapse
Affiliation(s)
- Flavia Barragán
- Departament de Química Orgànica-IBUB, Universitat de Barcelona, Barcelona, E-08028, Spain
| | | | | |
Collapse
|
27
|
Erchegyi J, Cescato R, Grace CRR, Waser B, Piccand V, Hoyer D, Riek R, Rivier JE, Reubi JC. Novel, potent, and radio-iodinatable somatostatin receptor 1 (sst1) selective analogues. J Med Chem 2009; 52:2733-46. [PMID: 19351180 PMCID: PMC2779710 DOI: 10.1021/jm801314f] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The proposed sst(1) pharmacophore (J. Med. Chem. 2005, 48, 523-533) derived from the NMR structures of a family of mono- and dicyclic undecamers was used to design octa-, hepta-, and hexamers with high affinity and selectivity for the somatostatin sst(1) receptor. These compounds were tested for their in vitro binding properties to all five somatostatin (SRIF) receptors using receptor autoradiography; those with high SRIF receptor subtype 1 (sst(1)) affinity and selectivity were shown to be agonists when tested functionally in a luciferase reporter gene assay. Des-AA(1,4-6,10,12,13)-[DTyr(2),DAgl(NMe,2naphthoyl)(8),IAmp(9)]-SRIF-Thr-NH(2) (25) was radio-iodinated ((125)I-25) and specifically labeled sst(1)-expressing cells and tissues. 3D NMR structures were calculated for des-AA(1,4-6,10,12,13)-[DPhe(2),DTrp(8),IAmp(9)]-SRIF-Thr-NH(2) (16), des-AA(1,2,4-6,10,12,13)-[DAgl(NMe,2naphthoyl)(8),IAmp(9)]-SRIF-Thr-NH(2) (23), and des-AA(1,2,4-6,10,12,13)-[DAgl(NMe,2naphthoyl)(8),IAmp(9),Tyr(11)]-SRIF-NH(2) (27) in DMSO. Though the analogues have the sst(1) pharmacophore residues at the previously determined distances from each other, the positioning of the aromatic residues in 16, 23, and 27 is different from that described earlier, suggesting an induced fit mechanism for sst(1) binding of these novel, less constrained sst(1)-selective family members.
Collapse
Affiliation(s)
- Judit Erchegyi
- The Clayton Foundation Laboratories for Peptide Biology, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Somatostatin as an Anti-Inflammatory Neuropeptide. ACTA ACUST UNITED AC 2009. [DOI: 10.1016/s1567-7443(08)10406-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
29
|
Feytens D, De Vlaeminck M, Cescato R, Tourwé D, Reubi JC. Highly Potent 4-Amino-indolo[2,3-c]azepin-3-one-Containing Somatostatin Mimetics with a Range of sst Receptor Selectivities. J Med Chem 2008; 52:95-104. [DOI: 10.1021/jm801205x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Debby Feytens
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Magali De Vlaeminck
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Renzo Cescato
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Dirk Tourwé
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| | - Jean Claude Reubi
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050 Brussels, Belgium, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland
| |
Collapse
|
30
|
Moore SB, Grant M, Rew Y, Bosa E, Fabbri M, Kumar U, Goodman M. Synthesis and biologic activity of conformationally constrained analogs of L-363,301. ACTA ACUST UNITED AC 2008; 66:404-22. [PMID: 16316457 DOI: 10.1111/j.1399-3011.2005.00309.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report the synthesis, biological activity and conformational analysis of analogs of the cyclic hexapeptide L-363,301, c[Pro6-Phe7-D-Trp8-Lys9-Thr10-Phe11] (numbering as in the native hormone somatostatin-14). The d-Trp in position 8 was replaced with (2R,3S)- and (2R,3R)-beta-MeTrp respectively, with an added methyl group in the beta position of Trp. The objective of our study was to determine the potency and selectivity generated by the added constraint in the beta position of the d-Trp upon binding to human somatostatin receptors hsst1-5. We synthesized the building blocks enantioselectively and incorporated them into the peptides by SPPS. Competition binding assays revealed that both compounds 2 and 3 were selective for hsst2 over hsst5. The (2R,3S) analog 2 was approximately 30 times more potent at hsst2 than the (2R,3R) analog 3. Interestingly, the (2R,3R) compound showed no binding affinity at hsst5.
Collapse
Affiliation(s)
- S B Moore
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093-0343, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Seebach D, Dubost E, Mathad R, Jaun B, Limbach M, Löweneck M, Flögel O, Gardiner J, Capone S, Beck A, Widmer H, Langenegger D, Monna D, Hoyer D. New Open-Chain and Cyclic Tetrapeptides, Consisting ofα-,β2-, andβ3-Amino-Acid Residues, as Somatostatin Mimics - A Survey. Helv Chim Acta 2008. [DOI: 10.1002/hlca.200890190] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
32
|
Noy K, Kalisman N, Keasar C. Prediction of structural stability of short beta-hairpin peptides by molecular dynamics and knowledge-based potentials. BMC STRUCTURAL BIOLOGY 2008; 8:27. [PMID: 18510728 PMCID: PMC2427033 DOI: 10.1186/1472-6807-8-27] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2007] [Accepted: 05/29/2008] [Indexed: 11/10/2022]
Abstract
BACKGROUND The structural stability of peptides in solution strongly affects their binding affinities and specificities. Thus, in peptide biotechnology, an increase in the structural stability is often desirable. The present work combines two orthogonal computational techniques, Molecular Dynamics and a knowledge-based potential, for the prediction of structural stability of short peptides (< 20 residues) in solution. RESULTS We tested the new approach on four families of short beta-hairpin peptides: TrpZip, MBH, bhpW and EPO, whose structural stabilities have been experimentally measured in previous studies. For all four families, both computational techniques show considerable correlation (r > 0.65) with the experimentally measured stabilities. The consensus of the two techniques shows higher correlation (r > 0.82). CONCLUSION Our results suggest a prediction scheme that can be used to estimate the relative structural stability within a peptide family. We discuss the applicability of this predictive approach for in-silico screening of combinatorial peptide libraries.
Collapse
Affiliation(s)
- Karin Noy
- Department of Life Sciences, Ben-Gurion University, Beer-Sheva, Israel.
| | | | | |
Collapse
|
33
|
Erchegyi J, Grace CRR, Samant M, Cescato R, Piccand V, Riek R, Reubi JC, Rivier JE. Ring size of somatostatin analogues (ODT-8) modulates receptor selectivity and binding affinity. J Med Chem 2008; 51:2668-75. [PMID: 18410084 PMCID: PMC2782568 DOI: 10.1021/jm701444y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The synthesis, biological testing, and NMR studies of several analogues of H-c[Cys (3)-Phe (6)-Phe (7)-DTrp (8)-Lys (9)-Thr (10)-Phe (11)-Cys (14)]-OH (ODT-8, a pan-somatostatin analogue, 1) have been performed to assess the effect of changing the stereochemistry and the number of atoms in the disulfide bridge on binding affinity. Cysteine at positions 3 and/or 14 (somatostatin numbering) were/was substituted with d-cysteine, norcysteine, D-norcysteine, homocysteine, and/or D-homocysteine. The 3D structure analysis of selected partially selective, bioactive analogues (3, 18, 19, and 21) was carried out in dimethylsulfoxide. Interestingly and not unexpectedly, the 3D structures of these analogues comprised the pharmacophore for which the analogues had the highest binding affinities (i.e., sst 4 in all cases).
Collapse
Affiliation(s)
- Judit Erchegyi
- The Clayton Foundation Laboratories for Peptide Biology and Structural Biology Laboratory, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Sako Y, Goto Y, Murakami H, Suga H. Ribosomal synthesis of peptidase-resistant peptides closed by a nonreducible inter-side-chain bond. ACS Chem Biol 2008; 3:241-9. [PMID: 18338852 DOI: 10.1021/cb800010p] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Here we report a new enabling technology for the synthesis of peptidase-resistant cyclic peptides by means of genetic code reprogramming involving the flexizyme (a tRNA acylation ribozyme) and PURE (a reconstituted cell-free translation) systems. In this work, we have developed a new nonproteinogenic amino acid bearing a chloroacetyl group in the side chain, which forms a physiologically stable thioether bond by intramolecular reaction with the sulfhydryl group of a Cys residue in the peptide chain upon translation. Significantly, this chemistry takes place spontaneously in situ of the translation solution, giving the corresponding cyclic peptides independent of ring sizes. We have used this method to convert human urotensin II, known as a potent vasoconstrictor, to its analogue containing a thioether bond, showing that this new analogue retains biological activity. Moreover, this peptide exhibits remarkable resistance against peptidases under reducing conditions. Thus, this technology offers a new means to accelerate the discovery of therapeutic peptidic drugs.
Collapse
Affiliation(s)
- Yusuke Sako
- Research Center for Advanced
Science and Technology, The University of Tokyo, 4-6-1, Komaba, Meguro,
Tokyo, 153-8904, Japan
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, 113-8656, Japan
| | - Yuki Goto
- Research Center for Advanced
Science and Technology, The University of Tokyo, 4-6-1, Komaba, Meguro,
Tokyo, 153-8904, Japan
- Department of Advanced Interdisciplinary Studies, Graduate School of Engineering, The University of Tokyo, 4-6-1, Komaba, Meguro, Tokyo, 153-8904, Japan
| | - Hiroshi Murakami
- Research Center for Advanced
Science and Technology, The University of Tokyo, 4-6-1, Komaba, Meguro,
Tokyo, 153-8904, Japan
| | - Hiroaki Suga
- Research Center for Advanced
Science and Technology, The University of Tokyo, 4-6-1, Komaba, Meguro,
Tokyo, 153-8904, Japan
- Department of Chemistry and Biotechnology, Graduate School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo, Tokyo, 113-8656, Japan
- Department of Advanced Interdisciplinary Studies, Graduate School of Engineering, The University of Tokyo, 4-6-1, Komaba, Meguro, Tokyo, 153-8904, Japan
| |
Collapse
|
35
|
Valéry C, Pouget E, Pandit A, Verbavatz JM, Bordes L, Boisdé I, Cherif-Cheikh R, Artzner F, Paternostre M. Molecular origin of the self-assembly of lanreotide into nanotubes: a mutational approach. Biophys J 2008; 94:1782-95. [PMID: 17993497 PMCID: PMC2242760 DOI: 10.1529/biophysj.107.108175] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2007] [Accepted: 10/01/2007] [Indexed: 11/18/2022] Open
Abstract
Lanreotide, a synthetic, therapeutic octapeptide analog of somatostatin, self-assembles in water into perfectly hollow and monodisperse (24-nm wide) nanotubes. Lanreotide is a cyclic octapeptide that contains three aromatic residues. The molecular packing of the peptide in the walls of a nanotube has recently been characterized, indicating four hierarchical levels of organization. This is a fascinating example of spontaneous self-organization, very similar to the formation of the gas vesicle walls of Halobacterium halobium. However, this unique peptide self-assembly raises important questions about its molecular origin. We adopted a directed mutation approach to determine the molecular parameters driving the formation of such a remarkable peptide architecture. We have modified the conformation by opening the cycle and by changing the conformation of a Lys residue, and we have also mutated the aromatic side chains of the peptide. We show that three parameters are essential for the formation of lanreotide nanotubes: i), the specificity of two of the three aromatic side chains, ii), the spatial arrangement of the hydrophilic and hydrophobic residues, and iii), the aromatic side chain in the beta-turn of the molecule. When these molecular characteristics are modified, either the peptides lose their self-assembling capability or they form less-ordered architectures, such as amyloid fibers and curved lamellae. Thus we have determined key elements of the molecular origins of lanreotide nanotube formation.
Collapse
Affiliation(s)
- Céline Valéry
- Ipsen Pharma, 08980 Sant Feliu de Llobregat, Barcelona, Spain
| | - Emilie Pouget
- Unité mixte de Recherche du Centre National pour la Recherche Scientifique 6626, Université Rennes 1, F-35042 Rennes, France
| | - Anjali Pandit
- Institut de Bio Technologies de Saclay, Commisariat à l’Energie Atomique et Centre National pour la Recherche Scientifique, F-91191 Gif-sur-Yvette, France
| | - Jean-Marc Verbavatz
- Institut de Bio Technologies de Saclay, Commisariat à l’Energie Atomique et Centre National pour la Recherche Scientifique, F-91191 Gif-sur-Yvette, France
| | - Luc Bordes
- Institut de Bio Technologies de Saclay, Commisariat à l’Energie Atomique et Centre National pour la Recherche Scientifique, F-91191 Gif-sur-Yvette, France
| | - Isabelle Boisdé
- Institut de Bio Technologies de Saclay, Commisariat à l’Energie Atomique et Centre National pour la Recherche Scientifique, F-91191 Gif-sur-Yvette, France
| | | | - Franck Artzner
- Unité mixte de Recherche du Centre National pour la Recherche Scientifique 6626, Université Rennes 1, F-35042 Rennes, France
| | - Maité Paternostre
- Institut de Bio Technologies de Saclay, Commisariat à l’Energie Atomique et Centre National pour la Recherche Scientifique, F-91191 Gif-sur-Yvette, France
| |
Collapse
|
36
|
|
37
|
Jaggi M, Prasad S, Singh AT, Praveen R, Dutt S, Mathur A, Sharma R, Gupta N, Ahuja R, Mukherjee R, Burman AC. Anticancer activity of a peptide combination in gastrointestinal cancers targeting multiple neuropeptide receptors. Invest New Drugs 2008; 26:489-504. [PMID: 18217205 DOI: 10.1007/s10637-008-9117-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2007] [Accepted: 01/10/2008] [Indexed: 01/31/2023]
Abstract
A novel peptide combination consisting of four synthetic neuropeptide analogs of Vasoactive Intestinal Peptide (VIP), Bombesin, Substance P and Somatostatin has been found to have potent anticancer activity in vitro and in vivo. The receptors of these four neuropeptides are known to be over expressed in various cancers. We have found the presence of native neuropeptides in the culture supernatant of the primary tumor cells of human colon adenocarcinomas. It was further demonstrated by receptor-ligand assays that not only do these tumor cells synthesize and secrete four peptide hormones but also possess specific high affinity receptors on their surface. Screening a large panel of analogs to the four peptide hormones on tumor cell proliferation led to the identification of four cytotoxic analogs, the combination of which was code-named DRF7295. The design and synthesis of the peptide analogs have been described in this paper. In vitro anticancer activity of DRF7295 was studied in a large panel of human tumor cells. Gastrointestinal tumor cells of the colon, pancreas and duodenum were found to be most sensitive to DRF7295 with moderate activity seen in glioblastoma, prostate, leukemia and those of oral cancer cells. Efficacy studies in xenograft models of colon and duodenum resulted in T/C% of less than 40%, which is indicative of strong tumor regressing potential of DRF7295 in gastrointestinal cancers. Acute and long-term toxicity studies as well as safety pharmacology studies conducted indicate the safety of the drug upon systemic administration with no significant adverse pharmacological effects.
Collapse
Affiliation(s)
- Manu Jaggi
- Dabur Research Foundation, 22 Site IV, Sahibabad, Ghaziabad, 201010, India.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
D’Addona D, Carotenuto A, Novellino E, Piccand V, Reubi JC, Di Cianni A, Gori F, Papini AM, Ginanneschi M. Novel sst5-Selective Somatostatin Dicarba-Analogues: Synthesis and Conformation−Affinity Relationships. J Med Chem 2008; 51:512-20. [DOI: 10.1021/jm070886i] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Debora D’Addona
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Alfonso Carotenuto
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Ettore Novellino
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Véronique Piccand
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Jean Claude Reubi
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Alessandra Di Cianni
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Francesca Gori
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Anna Maria Papini
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| | - Mauro Ginanneschi
- Laboratory of Peptides and Proteins, Chemistry and Biology, Department of Organic Chemistry, University of Firenze, Via Lastruccia 13 I-50019, Sesto Fiorentino, Italy, Department of Pharmaceutical Chemistry and Toxicology, University of Napoli, Via Domenico Montesano 49, Italy, Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Murtenstrasse 31 CH-3010 Berne, Switzerland, and Laboratory of Peptides and Proteins, Chemistry and Biology, Department of
| |
Collapse
|
39
|
Green BR, Catlin P, Zhang MM, Fiedler B, Bayudan W, Morrison A, Norton RS, Smith BJ, Yoshikami D, Olivera BM, Bulaj G. Conotoxins containing nonnatural backbone spacers: cladistic-based design, chemical synthesis, and improved analgesic activity. ACTA ACUST UNITED AC 2007; 14:399-407. [PMID: 17462575 DOI: 10.1016/j.chembiol.2007.02.009] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2006] [Revised: 02/09/2007] [Accepted: 02/20/2007] [Indexed: 11/23/2022]
Abstract
Disulfide-rich neurotoxins from venomous animals continue to provide compounds with therapeutic potential. Minimizing neurotoxins often results in removal of disulfide bridges or critical amino acids. To address this drug-design challenge, we explored the concept of disulfide-rich scaffolds consisting of isostere polymers and peptidic pharmacophores. Flexible spacers, such as amino-3-oxapentanoic or 6-aminohexanoic acids, were used to replace conformationally constrained parts of a three-disulfide-bridged conotoxin, SIIIA. The peptide-polymer hybrids, polytides, were designed based on cladistic identification of nonconserved loci in related peptides. After oxidative folding, the polytides appeared to be better inhibitors of sodium currents in dorsal root ganglia and sciatic nerves in mice. Moreover, the polytides appeared to be significantly more potent and longer-lasting analgesics in the inflammatory pain model in mice, when compared to SIIIA. The resulting polytides provide a promising strategy for transforming disulfide-rich peptides into therapeutics.
Collapse
Affiliation(s)
- Brad R Green
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
De León-Rodríguez LM, Kovacs Z. The Synthesis and Chelation Chemistry of DOTA−Peptide Conjugates. Bioconjug Chem 2007; 19:391-402. [DOI: 10.1021/bc700328s] [Citation(s) in RCA: 200] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Luis M. De León-Rodríguez
- University of Texas Southwestern Medical Center, Advanced Imaging Research Center, 2201 Inwood Road NE 4.2, Dallas, Texas 75390-8568
| | - Zoltan Kovacs
- University of Texas Southwestern Medical Center, Advanced Imaging Research Center, 2201 Inwood Road NE 4.2, Dallas, Texas 75390-8568
| |
Collapse
|
41
|
Prasad S, Mathur A, Jaggi M, Mukherjee R. Delivering multiple anticancer peptides as a single prodrug using lysyl-lysine as a facile linker. J Pept Sci 2007; 13:458-67. [PMID: 17559067 DOI: 10.1002/psc.867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
A large 40-residue precursor peptide (propeptide 5) was synthesized by linking together four designed anticancer peptide analogs to the neuropeptides: vasoactive intestinal peptide, somatostatin, bombesin and substance P, using enzyme cleavable lysyl-lysine linkers. On incubation with the enzyme trypsin, propeptide 5 was cleaved in a sequence-specific manner at the lysyl-lysine residues in the linker to release the individual peptide fragments which were identified by LC-MS. Another precursor peptide (propeptide 5a), consisting of two of the peptide analogs linked through lysyl-lysine linker, was also preferentially cleaved at the Lys-Lys site on incubation with the enzyme trypsin. Propeptide 5 showed potent anticancer activity, both in vitro and in vivo, which was greater than that of the individual component peptides. The enhanced activity suggests that the propeptide is possibly cleaved in the biological system at the lysyl-lysine site to yield the individual peptide analogs, which together show a synergistic effect. On the basis of these experimental findings, it can be concluded that pairs of basic amino acids such as Lys-Lys can be used as facile linkers for delivering multiple biologically active peptides.
Collapse
|
42
|
Nikiforovich GV, Marshall GR, Achilefu S. Molecular modeling suggests conformational scaffolds specifically targeting five subtypes of somatostatin receptors. Chem Biol Drug Des 2007; 69:163-9. [PMID: 17441902 DOI: 10.1111/j.1747-0285.2007.00493.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Several analogs of somatostatin with conformational constraints in their peptide backbones have been modeled to determine energetically feasible conformations. Comparison of low-energy backbone structures of these peptides suggested unique conformations of the central Phe/Ala(i)-D-Trp(i+1)-Lys(i+2)-Thr(i+3) fragment characteristic for specific interactions of somatostatin with each of the five distinct subtypes of somatostatin receptors (SSTRs). The conformations obtained were in good agreement with experimental data obtained earlier by NMR measurements and/or X-ray crystallography. The results help rationalize experimental observations on the specificity of binding of various somatostatin analogs with different subtypes of the SSTRs. They also serve as templates for the design of conformationally constrained non-peptide scaffolds that effectively and selectively interact with different subtypes of SSTRs. Such scaffolds can be convenient carriers of radiolabels and near-infrared labels in specific agents for imaging tumors expressing different SSTR subtypes.
Collapse
Affiliation(s)
- Gregory V Nikiforovich
- Department of Biochemistry and Molecular Biology, Washington University Medical School, St. Louis, MO 63110, USA.
| | | | | |
Collapse
|
43
|
Feytens D, Cescato R, Reubi JC, Tourwé D. New sst4/5-selective somatostatin peptidomimetics based on a constrained tryptophan scaffold. J Med Chem 2007; 50:3397-401. [PMID: 17559206 DOI: 10.1021/jm070246f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The synthesis and biological evaluation of four peptidomimetic analogs of somatostatin based on a constrained Trp residue, 3-amino-indolo[2,3-c]azepin-2-one (Aia), are reported. It is shown that dipeptidomimetics with a D-Aia-Lys sequence, functionalized with N- and C-terminal aromatic substituents, display a good selectivity for both sst4 and sst5. This study allowed us to identify a new highly potent sst5 agonist with good selectivity over the other receptors, except versus sst4.
Collapse
Affiliation(s)
- Debby Feytens
- Department of Organic Chemistry, Vrije Universiteit Brussel, Pleinlaan 2, B-1050, Brussels, Belgium
| | | | | | | |
Collapse
|
44
|
Pintér E, Helyes Z, Szolcsányi J. Inhibitory effect of somatostatin on inflammation and nociception. Pharmacol Ther 2006; 112:440-56. [PMID: 16764934 DOI: 10.1016/j.pharmthera.2006.04.010] [Citation(s) in RCA: 181] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2006] [Accepted: 04/27/2006] [Indexed: 10/24/2022]
Abstract
The present review focuses on promising new opportunities for anti-inflammatory and analgesic therapy. The theoretical background is an original observation based on our own experimental results. These data demonstrate that somatostatin is released from capsaicin-sensitive, peptidergic sensory nerve endings in response to noxious heat and chemical stimuli such as vanilloids, protons or lipoxygenase products. It reaches distant parts of the body via the circulation and exerts systemic anti-inflammatory and analgesic effects. Somatostatin binds to G-protein-coupled membrane receptors (sst(1)-sst(5)) and diminishes neurogenic inflammation by prejunctional action on sensory-efferent nerve terminals, as well as by postjunctional mechanisms on target cells. It decreases the release of pro-inflammatory neuropeptides from sensory nerve endings and also acts on receptors of vascular endothelial, inflammatory and immune cells. Analgesic effect is mediated by an inhibitory action on peripheral terminals of nociceptive neurons, since circulating somatostatin cannot exert central action. Somatostatin itself is not suitable for drug development because of its broad spectrum and short elimination half life, stable, receptor-selective agonists have been synthesized and investigated. The present overview is aimed at summarizing the physiological importance of somatostatin and sst receptors, pharmacological significance of synthetic agonists and their potential in the development of novel anti-inflammatory and analgesic drugs. These compounds might provide novel perspectives in the pharmacotherapy of acute and chronic painful inflammatory diseases, as well as neuropathic conditions.
Collapse
Affiliation(s)
- Erika Pintér
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Pécs, H-7624 Pécs, Szigeti u. 12, Hungary.
| | | | | |
Collapse
|
45
|
Gairí M, Saiz P, Madurga S, Roig X, Erchegyi J, Koerber SC, Reubi JC, Rivier JE, Giralt E. Conformational analysis of a potent SSTR3-selective somatostatin analogue by NMR in water solution. J Pept Sci 2006; 12:82-91. [PMID: 16365912 DOI: 10.1002/psc.743] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The three-dimensional structure of a potent SSTR3-selective analogue of somatostatin, cyclo(3-14)H-Cys(3)-Phe(6)-Tyr(7)-D-Agl(8)(N(beta) Me, 2-naphthoyl)-Lys(9)-Thr(10)-Phe(11)-Cys(14)-OH (des-AA(1, 2, 4, 5, 12, 13)[Tyr(7), D-Agl(8)(N(beta) Me, 2-naphthoyl)]-SRIF) (peptide 1) has been determined by (1)H NMR in water and molecular dynamics (MD) simulations. The peptide exists in two conformational isomers differing mainly by the cis/trans isomerization of the side chain in residue 8. The structure of 1 is compared with the consensus structural motifs of other somatostatin analogues that bind predominantly to SSTR1, SSTR2/SSTR5 and SSTR4 receptors, and to the 3D structure of a non-selective SRIF analogue, cyclo(3-14)H-Cys(3)-Phe(6)-Tyr(7)-D-2Nal(8)-Lys(9)-Thr(10)-Phe(11)-Cys(14)-OH (des-AA(1, 2, 4, 5, 12, 13)[Tyr(7), D-2Nal(8)]-SRIF) (peptide 2). The structural determinant factors that could explain selectivity of peptide 1 for SSTR3 receptors are discussed.
Collapse
Affiliation(s)
- Margarida Gairí
- NMR Facility, Serveis Cientificotècnics, University of Barcelona, Barcelona Science Park, Josep Samitier 1-5, 08028 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Prasad S, Mathur A, Sharma R, Gupta N, Ahuja R, Jaggi M, Singh AT, Mukherjee R. Octapeptide Analogs of Somatostatin Containing α,α-Dialkylated Amino Acids with Potent Anticancer Activity. Int J Pept Res Ther 2006. [DOI: 10.1007/s10989-005-9005-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
47
|
Contour-Galcéra MO, Sidhu A, Plas P, Roubert P. 3-Thio-1,2,4-triazoles, novel somatostatin sst2/sst5 agonists. Bioorg Med Chem Lett 2005; 15:3555-9. [PMID: 15982879 DOI: 10.1016/j.bmcl.2005.05.061] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2005] [Revised: 05/12/2005] [Accepted: 05/17/2005] [Indexed: 11/16/2022]
Abstract
Novel 3-thio-1,2,4-triazoles have been obtained via a solution-phase parallel synthesis strategy, affording potent non-peptidic human somatostatin receptor subtypes 2 and 5 agonists.
Collapse
|
48
|
Balraju V, Reddy DS, Periasamy M, Iqbal J. Synthesis of Conformationally Constrained Cyclic Peptides Using an Intramolecular Sonogashira Coupling. J Org Chem 2005; 70:9626-8. [PMID: 16268647 DOI: 10.1021/jo051412z] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
[Reaction: see text]. Small peptides having a 3-bromobenzyl group at the C-termini and n-alkynoyl group at the N-termini undergo a smooth copper-free intramolecular Sonogashira coupling reaction to afford the corresponding cyclic peptides in moderate yields. Scope and limitations of this macrocyclization is demonstrated with di-, tri-, and tetrapeptides.
Collapse
Affiliation(s)
- V Balraju
- Discovery Research, Dr. Reddy's Laboratories Ltd., Bollaram Road, Miyapur, Hyderabad, 500 049, A.P., India
| | | | | | | |
Collapse
|
49
|
Zapf CW, Del Valle JR, Goodman M. Utilizing the intramolecular Fukuyama–Mitsunobu reaction for a flexible synthesis of novel heterocyclic scaffolds for peptidomimetic drug design. Bioorg Med Chem Lett 2005; 15:4033-6. [PMID: 16002286 DOI: 10.1016/j.bmcl.2005.06.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Revised: 06/06/2005] [Accepted: 06/06/2005] [Indexed: 11/28/2022]
Abstract
We report the synthesis of the novel scaffolds pyrazino[1,2-b]isoquinoline and pyrrolo[1,2-a]pyrazine displaying the somatostatin pharmacophores. Both classes of compounds contain a pyrazine heterocycle, which can be prepared in a straightforward manner utilizing an intramolecular Fukuyama-Mitsunobu reaction. As both the families derive from amino acids, they can be accessed in high optical purity.
Collapse
Affiliation(s)
- Christoph W Zapf
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093-0343, USA.
| | | | | |
Collapse
|
50
|
Meis S, Sosulina L, Schulz S, Höllt V, Pape HC. Mechanisms of somatostatin-evoked responses in neurons of the rat lateral amygdala. Eur J Neurosci 2005; 21:755-62. [PMID: 15733093 DOI: 10.1111/j.1460-9568.2005.03922.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The effects of somatostatin in the rat lateral amygdala (LA) in vitro were investigated through whole cell recording techniques. Somatostatin induced an inwardly rectifying K+ current in approximately 98% of LA projection neurons. Half-maximal effects were obtained by 189 nM somatostatin. The effects of somatostatin were insensitive to tetrodotoxin, reduced by Ba2+, occluded or abolished by the presence of nonhydrolysable GTP or GDP analogues, respectively, and blocked or mimicked by a somatostatin receptor type 2 antagonist (BIM-23627) or somatostatin receptor type 2 agonist (L-779,976), respectively, while somatostatin receptor type 1, 3 and 4 agonists were ineffective (L-797,591, L-796,778, L-803,087). Responses to somatostatin were associated with membrane hyperpolarization and decrease in input resistance, resulting in a dampening of cell excitability. It is suggested that these cellular mechanisms contribute to the role of somatostatin in decreasing anxiety behaviour as well as to anticonvulsant and antiepileptogenic actions of somatostatin or somatostatin agonists in the amygdala.
Collapse
Affiliation(s)
- Susanne Meis
- Institut für Physiologie, Medizinische Fakultät, Otto-von-Guericke-Universität, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | | | | | | | | |
Collapse
|