1
|
Ludwig BS, Krautkremer N, Tomassi S, Di Maro S, Di Leva FS, Benge A, Nieberler M, Kessler H, Marinelli L, Kossatz S, Reuning U. Switching Roles─Exploring Concentration-Dependent Agonistic versus Antagonistic Behavior of Integrin Ligands. J Med Chem 2025; 68:4334-4351. [PMID: 39908297 DOI: 10.1021/acs.jmedchem.4c02111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/07/2025]
Abstract
Identification of integrins as cancer targets has stimulated the development of specific inhibitory ligands. However, following cilengitide's unexpected clinical failure by promoting angiogenesis at low concentrations, pure ligand antagonism was soon scrutinized. We evaluated αvβ3, αvβ6, or α5β1 ligands for concentration-dependent functional switches in respective integrin subtype-overexpressing cancer cells. Cilengitide (L2) or L1 provoked minor transient changes in (p)-FAK and (p)-p44/42(erk-1/2) predominantly at low concentrations and antagonized cell migration at high concentrations, while agonistically accelerating it at low concentrations. L5 (α5β1) showed bell-shaped FAK activation at both concentrations, blocking cell migration at high concentrations only in α5β1+ OV-MZ-6 cells, not acting agonistically. L3 (αvβ6) did not alter signaling upon long exposure but transiently and early activated FAK in αvβ6+ HN cells at both concentrations, with neither antagonistic nor agonistic consequences on cell motility. These data underscore the need for in-depth evaluation of ligand actions to ensure their most promising medical use.
Collapse
Affiliation(s)
- Beatrice Stefanie Ludwig
- Department of Nuclear Medicine, School of Medicine & Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine & Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Nils Krautkremer
- Department of Oral and Maxillofacial Surgery, School of Medicine & Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Stefano Tomassi
- UNINA - Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Salvatore Di Maro
- SUN - Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, Università degli Studi della Campania "Luigi Vanvitelli", Viale Abramo Lincoln, 5, Caserta 81100, Italy
| | - Francesco Saverio Di Leva
- UNINA - Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Anke Benge
- Department of Obstetrics & Gynecology, School of Medicine & Health, Clinical Research Unit, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Markus Nieberler
- Department of Oral and Maxillofacial Surgery, School of Medicine & Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Horst Kessler
- Department of Chemistry, School of Natural Sciences and Bavarian NMR Center (BNMRZ), Institute for Advanced Study, Technical University Munich, Lichtenbergstrasse 2a, Garching 85748, Germany
| | - Luciana Marinelli
- UNINA - Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano 49, Naples 80131, Italy
| | - Susanne Kossatz
- Department of Nuclear Medicine, School of Medicine & Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine & Health, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
- Department of Chemistry, School of Natural Sciences, Technical University Munich, Ismaninger Strasse 22, Munich 81675, Germany
| | - Ute Reuning
- Department of Obstetrics & Gynecology, School of Medicine & Health, Clinical Research Unit, Klinikum rechts der Isar, TUM University Hospital, Technical University of Munich, Ismaninger Strasse 22, Munich 81675, Germany
| |
Collapse
|
2
|
Paulus J, Nachtigall B, Meyer P, Sewald N. RGD Peptidomimetic MMAE-Conjugate Addressing Integrin αVβ3-Expressing Cells with High Targeting Index. Chemistry 2023; 29:e202203476. [PMID: 36454662 DOI: 10.1002/chem.202203476] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/08/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/03/2022]
Abstract
Small molecule-drug conjugates (SMDCs) mimicking the RGD sequence (-Arg-Gly-Asp-) with a non-peptide moiety require a pharmacophore-independent attachment site. A library of 36 sulfonamide-modified RGD mimetics with nM to pM affinity for integrin αV β3 was synthesized and analysed via DAD mapping. The best structure of the conjugable RGD mimetic was used and a linker was attached to an aromatic ring by Negishi cross-coupling. The product retained high affinity and selectivity for integrin αV β3 . The conjugable RGD mimetic was then attached to an enzymatically cleavable GKGEVA linker equipped with a self-immolative PABC and the antimitotic drug monomethyl auristatin E (MMAE). The resulting SMDC preferred binding to integrin αV β3 over α5 β1 in a ratio of 1 : 4519 (ELISA) and showed selectivity for αV β3 -positive WM115 cells over αV β3 -negative M21-L cells in the in vitro cell adhesion assay as well as in cell viability assays with a targeting index of 134 (M21-L/WM115).
Collapse
Affiliation(s)
- Jannik Paulus
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Beate Nachtigall
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Peter Meyer
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Universitätsstraße 25, 33615, Bielefeld, Germany
| |
Collapse
|
3
|
Li J, Peng L, Chen Q, Ye Z, Zhao T, Hou S, Gu J, Hang Q. Integrin β1 in Pancreatic Cancer: Expressions, Functions, and Clinical Implications. Cancers (Basel) 2022; 14:cancers14143377. [PMID: 35884437 PMCID: PMC9318555 DOI: 10.3390/cancers14143377] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/02/2022] [Revised: 07/02/2022] [Accepted: 07/07/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pancreatic cancer (PC) is a highly aggressive malignant tumor with an extremely poor prognosis. Early diagnosis and treatment are key to improving the survival rate of PC patients. Emerging studies show that integrins might contribute to the pathogenesis of PC. This review presents the various signaling pathways that are mediated by integrins in PC and emphasizes the multiple functions of integrin β1 in malignant behaviors of PC. It also discusses the clinical significance of integrin β1 as well as integrin β1-based therapy in PC patients. Abstract Pancreatic cancer (PC) is characterized by rapid progression and a high mortality rate. The current treatment is still based on surgical treatment, supplemented by radiotherapy and chemotherapy, and new methods of combining immune and molecular biological treatments are being explored. Despite this, the survival rate of PC patients is still very disappointing. Therefore, clarifying the molecular mechanism of PC pathogenesis and developing precisely targeted drugs are key to improving PC prognosis. As the most common β subunit of the integrin family, integrin β1 has been proved to be closely related to the vascular invasion, distant metastasis, and survival of PC patients, and treatment targeting integrin β1 in PC has gained initial success in animal models. In this review, we summarize the various signaling pathways by which integrins are involved in PC, focusing on the roles of integrin β1 in the malignant behaviors of PC. Additionally, recent studies regarding the feasibility of integrin β1 as a diagnostic and prognostic biomarker in PC are also discussed. Finally, we present the progress of several integrin β1-based clinical trials to highlight the potential of integrin β1 as a target for personalized therapy in PC.
Collapse
Affiliation(s)
- Jiajia Li
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou 225009, China; (J.L.); (S.H.)
| | - Liyao Peng
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210000, China;
| | - Qun Chen
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China;
| | - Ziping Ye
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China;
| | - Tiantian Zhao
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou 225001, China;
| | - Sicong Hou
- Department of Gastroenterology, The Affiliated Hospital of Yangzhou University, Yangzhou 225009, China; (J.L.); (S.H.)
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou 225001, China;
| | - Jianguo Gu
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 81-8558, Japan
- Correspondence: (J.G.); (Q.H.); Tel.: +86-13-8145-8885 (Q.H.)
| | - Qinglei Hang
- Division of Regulatory Glycobiology, Institute of Molecular Biomembrane and Glycobiology, Tohoku Medical and Pharmaceutical University, Sendai 81-8558, Japan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence: (J.G.); (Q.H.); Tel.: +86-13-8145-8885 (Q.H.)
| |
Collapse
|
4
|
Collyer SE, Stack GD, Walsh JJ. Selective delivery of clinically approved tubulin binding agents through covalent conjugation to an active targeting moiety. Curr Med Chem 2022; 29:5179-5211. [DOI: 10.2174/0929867329666220401105929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/25/2021] [Revised: 01/19/2022] [Accepted: 01/24/2022] [Indexed: 11/22/2022]
Abstract
Abstract:
The efficacy and tolerability of tubulin binding agents are hampered by their low specificity for cancer cells, like most clinically used anticancer agents. To improve specificity, tubulin binding agents have been covalently conjugated to agents which target cancer cells to give actively targeted drug conjugates. These conjugates are designed to increase uptake of the drug by cancer cells, while having limited uptake by normal cells thereby improving efficacy and tolerability.
Approaches used include attachment to small molecules, polysaccharides, peptides, proteins and antibodies that exploit the overexpression of receptors for these substances. Antibody targeted strategies have been the most successful to date with six such examples having gained clinical approval. Many other conjugate types, especially those targeting the folate receptor, have shown promising efficacy and toxicity profiles in pre-clinical models and in early-stage clinical studies. Presented herein is a discussion of the success or otherwise of the recent strategies used to form these actively targeted conjugates.
Collapse
Affiliation(s)
- Samuel E. Collyer
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| | - Gary D. Stack
- Department of Nursing and Healthcare, Technological University of the Shannon: Midlands Midwest, Athlone, Ireland
| | - John J. Walsh
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
5
|
Baiula M, Cirillo M, Martelli G, Giraldi V, Gasparini E, Anelli AC, Spampinato SM, Giacomini D. Selective Integrin Ligands Promote Cell Internalization of the Antineoplastic Agent Fluorouracil. ACS Pharmacol Transl Sci 2021; 4:1528-1542. [PMID: 34661072 PMCID: PMC8506610 DOI: 10.1021/acsptsci.1c00094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/29/2021] [Indexed: 02/08/2023]
Abstract
Drug conjugates consisting of an antineoplastic drug and a targeting receptor ligand could be effective to overcome the heavy side effects of unselective anticancer agents. To address this need, we report here the results of a project aimed to study agonist and antagonist integrin ligands as targeting head of molecular cargoes for the selective delivery of 5-fluorouracil (5-FU) to cancer or noncancer cells. Initially, two fluorescent β-lactam-based integrin ligands were synthesized and tested for an effective and selective internalization mediated by α4β1 or α5β1 integrins in Jurkat and K562 cells, respectively. No cellular uptake was observed for both fluorescent compounds in HEK293 noncancerous control cells. Afterward, three conjugates composed of the β-lactam-based integrin ligand, suitable linkers, and 5-FU were realized. The best compound E, acting as α5β1 integrin agonist, is able to selectively deliver 5-FU into tumor cells, successfully leading to cancer cell death.
Collapse
Affiliation(s)
- Monica Baiula
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Martina Cirillo
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giulia Martelli
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | | | - Elisa Gasparini
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | | | - Santi Mario Spampinato
- Department
of Pharmacy and Biotechnology, University
of Bologna, Via Irnerio, 48, 40126, Bologna, Italy
| | - Daria Giacomini
- Department
of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| |
Collapse
|
6
|
Abstract
Integrin molecules are transmembrane αβ heterodimers involved in cell adhesion, trafficking, and signaling. Upon activation, integrins undergo dynamic conformational changes that regulate their affinity to ligands. The physiological functions and activation mechanisms of integrins have been heavily discussed in previous studies and reviews, but the fluorescence imaging techniques -which are powerful tools for biological studies- have not. Here we review the fluorescence labeling methods, imaging techniques, as well as Förster resonance energy transfer assays used to study integrin expression, localization, activation, and functions.
Collapse
Affiliation(s)
- Chen Cai
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| | - Hao Sun
- Department of Medicine, University of California, San Diego, La Jolla, 92093, USA
| | - Liang Hu
- Cardiovascular Institute of Zhengzhou University, Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450051, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, 06030, USA
| |
Collapse
|
7
|
Baiula M, Caligiana A, Bedini A, Zhao J, Santino F, Cirillo M, Gentilucci L, Giacomini D, Spampinato S. Leukocyte Integrin Antagonists as a Novel Option to Treat Dry Age-Related Macular Degeneration. Front Pharmacol 2021; 11:617836. [PMID: 33584300 PMCID: PMC7878375 DOI: 10.3389/fphar.2020.617836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/15/2020] [Accepted: 12/30/2020] [Indexed: 11/28/2022] Open
Abstract
Age-related macular degeneration (AMD) is a complex multifactorial degenerative disease that leads to irreversible blindness. AMD affects the macula, the central part of the retina responsible for sharp central vision. Retinal pigment epithelium (RPE) is the main cellular type affected in dry AMD. RPE cells form a monolayer between the choroid and the neuroretina and are in close functional relationship with photoreceptors; moreover, RPE cells are part of the blood retina barrier that is disrupted in ocular diseases such as AMD. During ocular inflammation lymphocytes and macrophages are recruited, contact RPE and produce pro-inflammatory cytokines, which play an important role in AMD pathogenesis. The interaction between RPE and immune cells is mediated by leukocyte integrins, heterodimeric transmembrane receptors, and adhesion molecules, including VCAM-1 and ICAM-1. Within this frame, this study aimed to characterize RPE-leukocytes interaction and to investigate any potentially beneficial effects induced by integrin antagonists (DS-70, MN27 and SR714), developed in previous studies. ARPE-19 cells were co-cultured for different incubation times with Jurkat cells and apoptosis and necrosis levels were analyzed by flow cytometry. Moreover, we measured the mRNA levels of the pro-inflammatory cytokine IL-1β and the expression of adhesion molecules VCAM-1 and ICAM-1. We found that RPE-lymphocyte interaction increased apoptosis and necrosis levels in RPE cells and the expression of IL-1β. This interaction was mediated by the binding of α4β1 and αLβ2 integrins to VCAM-1 and ICAM-1, respectively. The blockade of RPE-lymphocyte interaction with blocking antibodies highlighted the pivotal role played by integrins. Therefore, α4β1 and αLβ2 integrin antagonists were employed to disrupt RPE-lymphocyte crosstalk. Small molecule integrin antagonists proved to be effective in reducing RPE cell death and expression of IL-1β, demonstrating that integrin antagonists could protect RPE cells from detrimental effects induced by the interaction with immune cells recruited to the retina. Overall, the leukocyte integrin antagonists employed in the present study may represent a novel opportunity to develop new drugs to fight dry AMD.
Collapse
Affiliation(s)
- Monica Baiula
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Alberto Caligiana
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Andrea Bedini
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Junwei Zhao
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Federica Santino
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Martina Cirillo
- Laboratory of Design and Synthesis of Biologically Active Compounds, Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Daria Giacomini
- Laboratory of Design and Synthesis of Biologically Active Compounds, Department of Chemistry "G. Ciamician", University of Bologna, Bologna, Italy
| | - Santi Spampinato
- Laboratory of Cellular and Molecular Pharmacology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Specilization School of Hospital Pharmacy, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
8
|
Cirillo M, Giacomini D. Molecular Delivery of Cytotoxic Agents via Integrin Activation. Cancers (Basel) 2021; 13:299. [PMID: 33467465 PMCID: PMC7830197 DOI: 10.3390/cancers13020299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/23/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022] Open
Abstract
Integrins are cell adhesion receptors overexpressed in tumor cells. A direct inhibition of integrins was investigated, but the best inhibitors performed poorly in clinical trials. A gained attention towards these receptors arouse because they could be target for a selective transport of cytotoxic agents. Several active-targeting systems have been developed to use integrins as a selective cell entrance for some antitumor agents. The aim of this review paper is to report on the most recent results on covalent conjugates between integrin ligands and antitumor drugs. Cytotoxic drugs thus conjugated through specific linker to integrin ligands, mainly RGD peptides, demonstrated that the covalent conjugates were more selective against tumor cells and hopefully with fewer side effects than the free drugs.
Collapse
Affiliation(s)
| | - Daria Giacomini
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum University of Bologna, Via Selmi 2, 40126 Bologna, Italy;
| |
Collapse
|
9
|
Understanding the role of integrins in breast cancer invasion, metastasis, angiogenesis, and drug resistance. Oncogene 2021; 40:1043-1063. [PMID: 33420366 DOI: 10.1038/s41388-020-01588-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/30/2020] [Revised: 11/11/2020] [Accepted: 11/26/2020] [Indexed: 12/13/2022]
Abstract
Integrins are cell adhesion receptors, which are typically transmembrane glycoproteins that connect to the extracellular matrix (ECM). The function of integrins regulated by biochemical events within the cells. Understanding the mechanisms of cell growth by integrins is important in elucidating their effects on tumor progression. One of the major events in integrin signaling is integrin binding to extracellular ligands. Another event is distant signaling that gathers chemical signals from outside of the cell and transmit the signals upon cell adhesion to the inside of the cell. In normal breast tissue, integrins function as checkpoints to monitor effects on cell proliferation, while in cancer tissue these functions altered. The combination of tumor microenvironment and its associated components determines the cell fate. Hypoxia can increase the expression of several integrins. The exosomal integrins promote the growth of metastatic cells. Expression of certain integrins is associated with increased metastasis and decreased prognosis in cancers. In addition, integrin-binding proteins promote invasion and metastasis in breast cancer. Targeting specific integrins and integrin-binding proteins may provide new therapeutic approaches for breast cancer therapies. This review will examine the current knowledge of integrins' role in breast cancer.
Collapse
|
10
|
Baiula M. An in Vitro Assay to Study the Role of Opioids in Modulating Immune Cell Adhesion. Methods Mol Biol 2021; 2201:163-169. [PMID: 32975797 DOI: 10.1007/978-1-0716-0884-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 03/25/2023]
Abstract
Opioids play a pivotal role in pain transmission but are also able to modulate immune cell functions. In the last decades a connection between opioids and integrins-adhesion molecules involved, among many other processes, in leukocyte recruitment at inflamed site-has been established. To study immune cell integrin-mediated adhesion, cell adhesion assay is a simple, reproducible, and valuable tool capable of unraveling concentration-dependent effects of a test candidate on integrin-mediated cell adhesion.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy.
| |
Collapse
|
11
|
Sani S, Messe M, Fuchs Q, Pierrevelcin M, Laquerriere P, Entz-Werle N, Reita D, Etienne-Selloum N, Bruban V, Choulier L, Martin S, Dontenwill M. Biological Relevance of RGD-Integrin Subtype-Specific Ligands in Cancer. Chembiochem 2020; 22:1151-1160. [PMID: 33140906 DOI: 10.1002/cbic.202000626] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/02/2020] [Revised: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Integrins are heterodimeric transmembrane proteins able to connect cells with the micro-environment. They represent a family of receptors involved in almost all the hallmarks of cancer. Integrins recognizing the Arg-Gly-Asp (RGD) peptide in their natural extracellular matrix ligands have been particularly investigated as tumoral therapeutic targets. In the last 30 years, intense research has been dedicated to designing specific RGD-like ligands able to discriminate selectively the different RGD-recognizing integrins. Chemists' efforts have led to the proposition of modified peptide or peptidomimetic libraries to be used for tumor targeting and/or tumor imaging. Here we review, from the biological point of view, the rationale underlying the need to clearly delineate each RGD-integrin subtype by selective tools. We describe the complex roles of RGD-integrins (mainly the most studied αvβ3 and α5β1 integrins) in tumors, the steps towards selective ligands and the current usefulness of such ligands. Although the impact of integrins in cancer is well acknowledged, the biological characteristics of each integrin subtype in a specific tumor are far from being completely resolved. Selective ligands might help us to reconsider integrins as therapeutic targets in specific clinical settings.
Collapse
Affiliation(s)
- Saidu Sani
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Cancer and Diabetic Research Group, Department of Biochemistry and Molecular Biology, Faculty of Science, Federal University Ndufu-Alike Ikwo, P.M.B, 1010, Abakaliki, Ebonyi State, Nigeria
| | - Mélissa Messe
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Institut Pluridisciplinaire Hubert Curien (IPHC), UMR CNRS 7178, Université de Strasbourg, 67000, Strasbourg, France
| | - Quentin Fuchs
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Marina Pierrevelcin
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Patrice Laquerriere
- Institut Pluridisciplinaire Hubert Curien (IPHC), UMR CNRS 7178, Université de Strasbourg, 67000, Strasbourg, France
| | - Natacha Entz-Werle
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Pediatric Onco-Hematology Department, Pediatrics, University Hospital of Strasbourg, 1 avenue Molière, 67098, Strasbourg, France
| | - Damien Reita
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Department of Oncobiology, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, France
| | - Nelly Etienne-Selloum
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Institut du Cancer Strasbourg Europe (ICANS), Service de Pharmacie, 17 rue Albert Calmette, 67200 Strasbourg, France
| | - Véronique Bruban
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Laurence Choulier
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Sophie Martin
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Monique Dontenwill
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| |
Collapse
|
12
|
Marusak C, Thakur V, Li Y, Freitas JT, Zmina PM, Thakur VS, Chang M, Gao M, Tan J, Xiao M, Lu Y, Mills GB, Flaherty K, Frederick DT, Miao B, Sullivan RJ, Moll T, Boland GM, Herlyn M, Zhang G, Bedogni B. Targeting Extracellular Matrix Remodeling Restores BRAF Inhibitor Sensitivity in BRAFi-resistant Melanoma. Clin Cancer Res 2020; 26:6039-6050. [PMID: 32820016 PMCID: PMC7669662 DOI: 10.1158/1078-0432.ccr-19-2773] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/21/2019] [Revised: 07/07/2020] [Accepted: 08/17/2020] [Indexed: 12/16/2022]
Abstract
PURPOSE The extracellular matrix (ECM) is an intriguing, yet understudied component of therapy resistance. Here, we investigated the role of ECM remodeling by the collagenase, MT1-MMP, in conferring resistance of v-Raf murine sarcoma viral oncogene homolog B1 (BRAF)-mutant melanoma to BRAF inhibitor (BRAFi) therapy. EXPERIMENTAL DESIGN Publicly available RNA-sequencing data and reverse phase protein array were used to determine the relevance of MT1-MMP upregulation in BRAFi-resistant melanoma in patients, patient-derived xenografts, and cell line-derived tumors. Short hairpin RNA (shRNA)-mediated knockdown of MT1-MMP, inhibition via the selective MT1-MMP/MMP2 inhibitor, ND322, or overexpression of MT1-MMP was used to assess the role of MT1-MMP in mediating resistance to BRAFi. RESULTS MT1-MMP was consistently upregulated in posttreatment tumor samples derived from patients upon disease progression and in melanoma xenografts and cell lines that acquired resistance to BRAFi. shRNA- or ND322-mediated inhibition of MT1-MMP synergized with BRAFi leading to resensitization of resistant cells and tumors to BRAFi. The resistant phenotype depends on the ability of cells to cleave the ECM. Resistant cells seeded in MT1-MMP uncleavable matrixes were resensitized to BRAFi similarly to MT1-MMP inhibition. This is due to the inability of cells to activate integrinβ1 (ITGB1)/FAK signaling, as restoration of ITGB1 activity is sufficient to maintain resistance to BRAFi in the context of MT1-MMP inhibition. Finally, the increase in MT1-MMP in BRAFi-resistant cells is TGFβ dependent, as inhibition of TGFβ receptors I/II dampens MT1-MMP overexpression and restores sensitivity to BRAF inhibition. CONCLUSIONS BRAF inhibition results in a selective pressure toward higher expression of MT1-MMP. MT1-MMP is pivotal to an ECM-based signaling pathway that confers resistance to BRAFi therapy.
Collapse
Affiliation(s)
- Charles Marusak
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Varsha Thakur
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Yuan Li
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Juliano T Freitas
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Patrick M Zmina
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida
| | - Vijay S Thakur
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
| | - Ming Gao
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana
| | - Jiufeng Tan
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Min Xiao
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Yiling Lu
- Department of Genomic Medicine, Division of Cancer Medicine, MD Anderson Cancer Center, Houston, Texas
| | - Gordon B Mills
- The Knight Cancer Institute, Oregon Health Sciences University, Portland, Oregon
| | - Keith Flaherty
- Department of Medicine, Harvard Medical School, Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | | | - Benchun Miao
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Ryan J Sullivan
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Tabea Moll
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Gao Zhang
- Department of Neurosurgery, Duke University, Durham, North Carolina
| | - Barbara Bedogni
- Department of Dermatology, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
13
|
Altei WF, Pachane BC, dos Santos PK, Ribeiro LNM, Sung BH, Weaver AM, Selistre-de-Araújo HS. Inhibition of αvβ3 integrin impairs adhesion and uptake of tumor-derived small extracellular vesicles. Cell Commun Signal 2020; 18:158. [PMID: 32988382 PMCID: PMC7520983 DOI: 10.1186/s12964-020-00630-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/06/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) are lipid-bound particles that are naturally released from cells and mediate cell-cell communication. Integrin adhesion receptors are enriched in small EVs (SEVs) and SEV-carried integrins have been shown to promote cancer cell migration and to mediate organ-specific metastasis; however, how integrins mediate these effects is not entirely clear and could represent a combination of EV binding to extracellular matrix and cells. METHODS To probe integrin role in EVs binding and uptake, we employed a disintegrin inhibitor (DisBa-01) of integrin binding with specificity for αvβ3 integrin. EVs were purified from MDA-MB-231 cells conditioned media by serial centrifugation method. Isolated EVs were characterized by different techniques and further employed in adhesion, uptake and co-culture experiments. RESULTS We find that SEVs secreted from MDA-MB-231 breast cancer cells carry αvβ3 integrin and bind directly to fibronectin-coated plates, which is inhibited by DisBa-01. SEV coating on tissue culture plates also induces adhesion of MDA-MB-231 cells, which is inhibited by DisBa-01 treatment. Analysis of EV uptake and interchange between cells reveals that the amount of CD63-positive EVs delivered from malignant MDA-MB-231 breast cells to non-malignant MCF10A breast epithelial cells is reduced by DisBa-01 treatment. Inhibition of αvβ3 integrin decreases CD63 expression in cancer cells suggesting an effect on SEV content. CONCLUSION In summary, our findings demonstrate for the first time a key role of αvβ3 integrin in cell-cell communication through SEVs. Video Abstract.
Collapse
Affiliation(s)
- Wanessa F. Altei
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Bianca C. Pachane
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Patty K. dos Santos
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| | - Lígia N. M. Ribeiro
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas-UNICAMP, Campinas, São Paulo Brazil
| | - Bong Hwan Sung
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
| | - Alissa M. Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, USA
| | - Heloisa S. Selistre-de-Araújo
- Biochemistry and Molecular Biology Laboratory, Department of Physiological Sciences, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|
14
|
Baiula M, Greco R, Ferrazzano L, Caligiana A, Hoxha K, Bandini D, Longobardi P, Spampinato S, Tolomelli A. Integrin-mediated adhesive properties of neutrophils are reduced by hyperbaric oxygen therapy in patients with chronic non-healing wound. PLoS One 2020; 15:e0237746. [PMID: 32810144 PMCID: PMC7433869 DOI: 10.1371/journal.pone.0237746] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2020] [Accepted: 07/31/2020] [Indexed: 12/30/2022] Open
Abstract
In recent years, several studies suggested that the ability of hyperbaric oxygen therapy (HBOT) to promote healing in patients with diabetic ulcers and chronic wounds is due to the reduction of inflammatory cytokines and to a significant decrease in neutrophils recruitment to the damaged area. α4 and β2 integrins are receptors mediating the neutrophil adhesion to the endothelium and the comprehension of the effects of hyperbaric oxygenation on their expression and functions in neutrophils could be of great importance for the design of novel therapeutic protocols focused on anti-inflammatory agents. In this study, the α4 and β2 integrins' expression and functions have been evaluated in human primary neutrophils obtained from patients with chronic non-healing wounds and undergoing a prolonged HBOT (150 kPa per 90 minutes). The effect of a peptidomimetic α4β1 integrin antagonist has been also analyzed under these conditions. A statistically significant decrease (68%) in β2 integrin expression on neutrophils was observed during the treatment with HBO and maintained one month after the last treatment, while α4 integrin levels remained unchanged. However, cell adhesion function of both neutrophilic integrins α4β1 and β2 was significantly reduced 70 and 67%, respectively), but α4β1 integrin was still sensitive to antagonist inhibition in the presence of fibronectin, suggesting that a combined therapy between HBOT and integrin antagonists could have greater antinflammatory efficacy.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Roberto Greco
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum -University of Bologna, Bologna, Italy
| | - Lucia Ferrazzano
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum -University of Bologna, Bologna, Italy
| | - Alberto Caligiana
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | | | | | | | - Santi Spampinato
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Alessandra Tolomelli
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum -University of Bologna, Bologna, Italy
| |
Collapse
|
15
|
Martelli G, Bloise N, Merlettini A, Bruni G, Visai L, Focarete ML, Giacomini D. Combining Biologically Active β-Lactams Integrin Agonists with Poly(l-lactic acid) Nanofibers: Enhancement of Human Mesenchymal Stem Cell Adhesion. Biomacromolecules 2020; 21:1157-1170. [PMID: 32011862 PMCID: PMC7997109 DOI: 10.1021/acs.biomac.9b01550] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
Abstract
![]()
Regulating
stem cell adhesion and growth onto functionalized biomaterial
scaffolds is an important issue in the field of tissue engineering
and regenerative medicine. In this study, new electrospun scaffolds
of poly(l-lactic acid) (PLLA), as bioresorbable polymer,
and β-lactam compounds agonists of selected integrins, as functional
components with cell adhesive properties, are designed. The new β-lactam-PLLA
scaffolds contribute significantly in guiding protein translation
involved in human bone marrow mesenchymal stem cells (hBM-MSC) adhesion
and integrin gene expression. Scanning electron microscopy, confocal
laser scanning microscopy, and Western Blot analyses reveal that GM18-PLLA
shows the best results, promoting cell adhesion by significantly driving
changes in focal adhesion proteins distribution (β1 integrin and vinculin) and activation (pFAK), with a notable increase
of GM18-targets subunits integrin gene expression, α4 and β1. These novel functionalized submicrometric
fibrous scaffolds demonstrate, for the first time, the powerful combination
of selective β-lactams agonists of integrins with biomimetic
scaffolds, suggesting a designed rule that could be suitably applied
to tissue repair and regeneration.
Collapse
Affiliation(s)
- Giulia Martelli
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Nora Bloise
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Via S. Boezio 28, 27100 Pavia, Italy
| | - Andrea Merlettini
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Giovanna Bruni
- Department of Chemistry, Section of Physical Chemistry, University of Pavia, Viale Taramelli 16, 27100 Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine (DMM), Biochemistry Unit, Center for Health Technologies (CHT), UdR INSTM University of Pavia, Viale Taramelli 3/B, 27100 Pavia, Italy.,Department of Occupational Medicine, Toxicology and Environmental Risks, Istituti Clinici Scientifici Maugeri S.p.A, IRCCS, Via S. Boezio 28, 27100 Pavia, Italy
| | - Maria Letizia Focarete
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Daria Giacomini
- Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| |
Collapse
|
16
|
Zheng Y, Leftheris K. Insights into Protein–Ligand Interactions in Integrin Complexes: Advances in Structure Determinations. J Med Chem 2020; 63:5675-5696. [DOI: 10.1021/acs.jmedchem.9b01869] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yajun Zheng
- Pliant Therapeutics, South San Francisco, California 94080, United States
| | - Katerina Leftheris
- Pliant Therapeutics, South San Francisco, California 94080, United States
| |
Collapse
|
17
|
López Rivas P, Müller C, Breunig C, Hechler T, Pahl A, Arosio D, Belvisi L, Pignataro L, Dal Corso A, Gennari C. β-Glucuronidase triggers extracellular MMAE release from an integrin-targeted conjugate. Org Biomol Chem 2020; 17:4705-4710. [PMID: 31020985 DOI: 10.1039/c9ob00617f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
A non-internalizing αvβ3 integrin ligand was conjugated to the anticancer drug MMAE through a β-glucuronidase-responsive linker. In the presence of β-glucuronidase, only the conjugate bearing a PEG4 spacer inhibited the proliferation of integrin-expressing cancer cells at low nanomolar concentrations, indicating important structural requirements for the efficacy of these therapeutics.
Collapse
Affiliation(s)
- Paula López Rivas
- Università degli Studi di Milano, Dipartimento di Chimica, Via C. Golgi, 19 I-20133, Milan, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Martelli G, Baiula M, Caligiana A, Galletti P, Gentilucci L, Artali R, Spampinato S, Giacomini D. Could Dissecting the Molecular Framework of β-Lactam Integrin Ligands Enhance Selectivity? J Med Chem 2019; 62:10156-10166. [DOI: 10.1021/acs.jmedchem.9b01000] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022]
Affiliation(s)
- Giulia Martelli
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Alberto Caligiana
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Paola Galletti
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| | | | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Daria Giacomini
- Department of Chemistry “G. Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy
| |
Collapse
|
19
|
Sun Y, Hu C, Yang Y, Dong B, Deng Y. Fibroin/peptide co-functionalized calcium titanate nanorods improve osteoinductivity of titanium via mimicking osteogenic niche. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 103:109836. [DOI: 10.1016/j.msec.2019.109836] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 01/23/2019] [Revised: 03/25/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
|
20
|
Are Integrins Still Practicable Targets for Anti-Cancer Therapy? Cancers (Basel) 2019; 11:cancers11070978. [PMID: 31336983 PMCID: PMC6678560 DOI: 10.3390/cancers11070978] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/01/2019] [Revised: 06/27/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023] Open
Abstract
Correlative clinical evidence and experimental observations indicate that integrin adhesion receptors, in particular those of the αV family, are relevant to cancer cell features, including proliferation, survival, migration, invasion, and metastasis. In addition, integrins promote events in the tumor microenvironment that are critical for tumor progression and metastasis, including tumor angiogenesis, matrix remodeling, and the recruitment of immune and inflammatory cells. In spite of compelling preclinical results demonstrating that the inhibition of integrin αVβ3/αVβ5 and α5β1 has therapeutic potential, clinical trials with integrin inhibitors targeting those integrins have repeatedly failed to demonstrate therapeutic benefits in cancer patients. Here, we review emerging integrin functions and their proposed contribution to tumor progression, discuss preclinical evidence of therapeutic significance, revisit clinical trial results, and consider alternative approaches for their therapeutic targeting in oncology, including targeting integrins in the other cells of the tumor microenvironment, e.g., cancer-associated fibroblasts and immune/inflammatory cells. We conclude that integrins remain a valid target for cancer therapy; however, agents with better pharmacological properties, alternative models for their preclinical evaluation, and innovative combination strategies for clinical testing (e.g., together with immuno-oncology agents) are needed.
Collapse
|
21
|
Baiula M, Spampinato S, Gentilucci L, Tolomelli A. Novel Ligands Targeting α 4β 1 Integrin: Therapeutic Applications and Perspectives. Front Chem 2019; 7:489. [PMID: 31338363 PMCID: PMC6629825 DOI: 10.3389/fchem.2019.00489] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/14/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Among the other members of the adhesion molecules' family, α4β1 integrin, a heterodimeric receptor, plays a crucial role in inflammatory diseases, cancer development, metastasis and stem cell mobilization or retention. In many cases, its function in pathogenesis is not yet completely understood and investigations on ligand binding and related stabilization of active/inactive conformations still represent an important goal. For this reason, starting from the highlight of α4β1 functions in human pathologies, we report an overview of synthetic α4β1 integrin ligands under development as potential therapeutic agents. The small molecule library that we have selected represents a collection of lead compounds. These molecules are the object of future refinement in academic and industrial research, in order to achieve a fine tuning of α4β1 integrin regulation for the development of novel agents against pathologies still eluding an effective solution.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician,” University of Bologna, Bologna, Italy
| | | |
Collapse
|
22
|
Martelli G, Galletti P, Baiula M, Calcinari L, Boschi G, Giacomini D. Chiral β-lactam-based integrin ligands through Lipase-catalysed kinetic resolution and their enantioselective receptor response. Bioorg Chem 2019; 88:102975. [DOI: 10.1016/j.bioorg.2019.102975] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/22/2019] [Revised: 05/03/2019] [Accepted: 05/04/2019] [Indexed: 12/22/2022]
|
23
|
Raposo Moreira Dias A, Pina A, Dean A, Lerchen H, Caruso M, Gasparri F, Fraietta I, Troiani S, Arosio D, Belvisi L, Pignataro L, Dal Corso A, Gennari C. Neutrophil Elastase Promotes Linker Cleavage and Paclitaxel Release from an Integrin-Targeted Conjugate. Chemistry 2019; 25:1696-1700. [PMID: 30452790 PMCID: PMC6471013 DOI: 10.1002/chem.201805447] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/31/2018] [Revised: 11/16/2018] [Indexed: 12/16/2022]
Abstract
This work takes advantage of one of the hallmarks of cancer, that is, the presence of tumor infiltrating cells of the immune system and leukocyte-secreted enzymes, to promote the activation of an anticancer drug at the tumor site. The peptidomimetic integrin ligand cyclo(DKP-RGD) was found to accumulate on the surface of αv β3 integrin-expressing human renal cell carcinoma 786-O cells. The ligand was conjugated to the anticancer drug paclitaxel through a Asn-Pro-Val (NPV) tripeptide linker, which is a substrate of neutrophil-secreted elastase. In vitro linker cleavage assays and cell antiproliferative experiments demonstrate the efficacy of this tumor-targeting conjugate, opening the way to potential therapeutic applications.
Collapse
Affiliation(s)
| | - Arianna Pina
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | - Amelia Dean
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | | | - Michele Caruso
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Fabio Gasparri
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Ivan Fraietta
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Sonia Troiani
- Nerviano Medical SciencesViale Pasteur, 10I-20014NervianoItaly
| | - Daniela Arosio
- CNR, Istituto di Scienze e Tecnologie Molecolari (ISTM)Via C. Golgi, 19I-20133MilanItaly
| | - Laura Belvisi
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
- CNR, Istituto di Scienze e Tecnologie Molecolari (ISTM)Via C. Golgi, 19I-20133MilanItaly
| | - Luca Pignataro
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | - Alberto Dal Corso
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
| | - Cesare Gennari
- Università degli Studi di MilanoDipartimento di ChimicaVia C. Golgi, 19I-20133MilanItaly
- CNR, Istituto di Scienze e Tecnologie Molecolari (ISTM)Via C. Golgi, 19I-20133MilanItaly
| |
Collapse
|
24
|
Cesar PHS, Braga MA, Trento MVC, Menaldo DL, Marcussi S. Snake Venom Disintegrins: An Overview of their Interaction with Integrins. Curr Drug Targets 2019; 20:465-477. [DOI: 10.2174/1389450119666181022154737] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/21/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Disintegrins are non-enzymatic proteins that interfere on cell–cell interactions and signal transduction, contributing to the toxicity of snake venoms and play an essential role in envenomations. Most of their pharmacological and toxic effects are the result of the interaction of these molecules with cell surface ligands, which has been widely described and studied. These proteins may act on platelets, leading to hemorrhage, and may also induce apoptosis and cytotoxicity, which highlights a high pharmacological potential for the development of thrombolytic and antitumor agents. Additionally, these molecules interfere with the functions of integrins by altering various cellular processes such as migration, adhesion and proliferation. This review gathers information on functional characteristics of disintegrins isolated from snake venoms, emphasizing a comprehensive view of the possibility of direct use of these molecules in the development of new drugs, or even indirectly as structural models.
Collapse
Affiliation(s)
- Pedro Henrique Souza Cesar
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| | - Mariana Aparecida Braga
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| | - Marcus Vinicius Cardoso Trento
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| | - Danilo Luccas Menaldo
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of Sao Paulo (FCFRP-USP), Ribeirão Preto-SP, Brazil
| | - Silvana Marcussi
- Department of Chemistry, Biochemistry Laboratory, Federal University of Lavras (UFLA), Lavras, Minas Gerais, 37200-000, Brazil
| |
Collapse
|
25
|
Kuroda H, Tachikawa M, Yagi Y, Umetsu M, Nurdin A, Miyauchi E, Watanabe M, Uchida Y, Terasaki T. Cluster of Differentiation 46 Is the Major Receptor in Human Blood-Brain Barrier Endothelial Cells for Uptake of Exosomes Derived from Brain-Metastatic Melanoma Cells (SK-Mel-28). Mol Pharm 2018; 16:292-304. [PMID: 30452273 DOI: 10.1021/acs.molpharmaceut.8b00985] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/28/2022]
Abstract
Brain metastasis is a frequent complication of cancer and may be mediated, at least in part, by the internalization of cancer-cell-derived exosomes into brain capillary endothelial cells. Clarifying the mechanism(s) of this internalization is of interest because it could help us to develop ways to block brain metastasis, as well as affording a potential new route for drug delivery into the brain. Therefore, the purpose of the present study was to address this issue by identifying the receptors involved in the internalization of exosomes derived from a brain-metastatic cancer cell line (SK-Mel-28) into human blood-brain barrier endothelial cells (hCMEC/D3 cells). The combination of sulfo-SBED-based cross-linking and comprehensive proteomics yielded 20 proteins as exosome receptor candidates in hCMEC/D3 cells. The uptake of PKH67-labeled exosomes by hCMEC/D3 cells measured at 37 °C was significantly reduced by 95.6% at 4 °C and by 15.3% in the presence of 1 mM RGD peptide, an integrin ligand. Therefore, we focused on the identified RGD receptors, integrin α5 and integrin αV, and CD46, which is reported to act as an adenovirus receptor, together with integrin αV. A mixture of neutralizing antibodies against integrin α5 and integrin αV significantly decreased the exosome uptake by 11.8%, while application of CD46 siRNA reduced it by 39.0%. Immunohistochemical analysis confirmed the presence of CD46 in human brain capillary endothelial cells. These results suggest that CD46 is a major receptor for the uptake of SK-Mel-28-derived exosomes by human blood-brain barrier endothelial cells (hCMEC/D3 cells).
Collapse
Affiliation(s)
- Hiroki Kuroda
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Yuta Yagi
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Mina Umetsu
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Armania Nurdin
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Eisuke Miyauchi
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Michitoshi Watanabe
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Yasuo Uchida
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , Sendai 980-8578 , Japan
| |
Collapse
|
26
|
Alimbetov D, Askarova S, Umbayev B, Davis T, Kipling D. Pharmacological Targeting of Cell Cycle, Apoptotic and Cell Adhesion Signaling Pathways Implicated in Chemoresistance of Cancer Cells. Int J Mol Sci 2018; 19:ijms19061690. [PMID: 29882812 PMCID: PMC6032165 DOI: 10.3390/ijms19061690] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/02/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/11/2022] Open
Abstract
Chemotherapeutic drugs target a physiological differentiating feature of cancer cells as they tend to actively proliferate more than normal cells. They have well-known side-effects resulting from the death of highly proliferative normal cells in the gut and immune system. Cancer treatment has changed dramatically over the years owing to rapid advances in oncology research. Developments in cancer therapies, namely surgery, radiotherapy, cytotoxic chemotherapy and selective treatment methods due to better understanding of tumor characteristics, have significantly increased cancer survival. However, many chemotherapeutic regimes still fail, with 90% of the drug failures in metastatic cancer treatment due to chemoresistance, as cancer cells eventually develop resistance to chemotherapeutic drugs. Chemoresistance is caused through genetic mutations in various proteins involved in cellular mechanisms such as cell cycle, apoptosis and cell adhesion, and targeting those mechanisms could improve outcomes of cancer therapy. Recent developments in cancer treatment are focused on combination therapy, whereby cells are sensitized to chemotherapeutic agents using inhibitors of target pathways inducing chemoresistance thus, hopefully, overcoming the problems of drug resistance. In this review, we discuss the role of cell cycle, apoptosis and cell adhesion in cancer chemoresistance mechanisms, possible drugs to target these pathways and, thus, novel therapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Dauren Alimbetov
- Laboratory of bioengineering and regenerative medicine, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave, Z05H0P9 Astana, Kazakhstan.
| | - Sholpan Askarova
- Laboratory of bioengineering and regenerative medicine, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave, Z05H0P9 Astana, Kazakhstan.
| | - Bauyrzhan Umbayev
- Laboratory of bioengineering and regenerative medicine, Center for Life Sciences, National Laboratory Astana, Nazarbayev University, 53 Kabanbay Batyr Ave, Z05H0P9 Astana, Kazakhstan.
| | - Terence Davis
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| | - David Kipling
- Division of Cancer and Genetics, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
27
|
Nair RV, Farrukh A, del Campo A. A Photoactivatable α5
β1
-Specific Integrin Ligand. Chembiochem 2018; 19:1280-1287. [DOI: 10.1002/cbic.201800180] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/06/2018] [Indexed: 01/16/2023]
Affiliation(s)
- Roshna V. Nair
- INM-Leibniz Institute for New Materials; Campus D2 2 66123 Saarbrücken Germany
| | - Aleeza Farrukh
- INM-Leibniz Institute for New Materials; Campus D2 2 66123 Saarbrücken Germany
| | - Aránzazu del Campo
- INM-Leibniz Institute for New Materials; Campus D2 2 66123 Saarbrücken Germany
- Chemistry Department; Saarland University; Campus C4 2 66123 Saarbrücken Germany
| |
Collapse
|
28
|
Abstract
While integrins were originally discovered as cell adhesion receptors, recent studies have reinforced the concept that integrins have central roles in cancer that extend far beyond controlling cell adhesion and migration. Indeed, as transmembrane cell surface receptors that occupy a critical position at the interface of cellular and extracellular interactions and are capable of both "inside-out" and "outside-in" signaling, integrins are uniquely poised to regulate the cell's ability to promote, sense, and react to changes in the tumor microenvironment. Moreover, integrins are present on all cell types in the tumor microenvironment, and they have important roles in regulating intercellular communication. Decades of promising pre-clinical studies have implicated certain integrins as attractive therapeutic targets in the cancer clinic. Nevertheless, results of the few clinical trials that target integrins in cancer have thus far been disappointing. Importantly, these clinical failures likely reflect the emerging complexity of individual and combinatorial integrin function within both tumor cells and other cell types of the tumor microenvironment, together with a need to explore integrin-targeting agents not just as monotherapies but also as adjuvants to more conventional radiotherapies or chemotherapies. In this review, we will examine recent advances toward understanding how integrins regulate cancer progression, including their roles in intercellular communication and modulation of the tumor microenvironment. Additionally, we will discuss factors that underlie the limited efficacy of current efforts to target integrins in the cancer clinic as well as potential strategies to overcome these challenges.
Collapse
Affiliation(s)
- Whitney Longmate
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA
| | - C Michael DiPersio
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, New York, USA.,Department of Surgery, Albany Medical College, Albany , New York, USA
| |
Collapse
|