1
|
Le Roy MM, Métivier C, Rbah-Vidal L, Saëc PL, Bouhsine H, Chérel M, Faivre-Chauvet A, Troadec T, Tripier R. Grafting a chromophore on AMD070 analogues for CXCR4 bioimaging: Chemical synthesis and in vitro assessment of the inhibition properties of the CXCR4 receptor. Bioorg Med Chem Lett 2025; 115:130027. [PMID: 39551220 DOI: 10.1016/j.bmcl.2024.130027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/19/2024]
Abstract
Thank to their particular pharmacokinetics, the use of small organic molecules can be a very promising alternative to macromolecular targeting biomolecules (i.e. antibodies, peptides…) for specific imaging of tumours. Herein, the potential of two AMD070-like inhibitors as CXCR4-targeting units for specific imaging of cancer cells, and the influence of chromophore-grafting on their recognition properties was investigated.
Collapse
Affiliation(s)
- Marie M Le Roy
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29200 Brest, France
| | - Cassandra Métivier
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI(2)NA, 8 quai Moncousu, 44007 Nantes Cedex, France
| | - Latifa Rbah-Vidal
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI(2)NA, 8 quai Moncousu, 44007 Nantes Cedex, France
| | - Patricia Le Saëc
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI(2)NA, 8 quai Moncousu, 44007 Nantes Cedex, France
| | - Hela Bouhsine
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI(2)NA, 8 quai Moncousu, 44007 Nantes Cedex, France
| | - Michel Chérel
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI(2)NA, 8 quai Moncousu, 44007 Nantes Cedex, France
| | - Alain Faivre-Chauvet
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI(2)NA, 8 quai Moncousu, 44007 Nantes Cedex, France.
| | - Thibault Troadec
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29200 Brest, France. https://twitter.com/@TTroadecChem
| | - Raphaël Tripier
- Univ. Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29200 Brest, France
| |
Collapse
|
2
|
Korbecki J, Bosiacki M, Kupnicka P, Barczak K, Chlubek D, Baranowska-Bosiacka I. CXCR4 as a therapeutic target in acute myeloid leukemia. Leukemia 2024; 38:2303-2317. [PMID: 39261603 DOI: 10.1038/s41375-024-02326-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 09/13/2024]
Abstract
Extensive research on the CXCL12-CXCR4 axis in acute myeloid leukemia (AML) has resulted in the incorporation of novel anti-leukemia drugs targeting this axis into therapeutic strategies. However, despite this progress, a comprehensive and up-to-date review addressing the role of the CXCL12-CXCR4 axis in AML's oncogenic processes is lacking. In this review, we examine its molecular aspects influencing cancer progression, such as its impact on autonomous proliferation, apoptotic regulation, chemoresistance mechanisms, and interactions with non-leukemic cells such as MSCs and Treg cells. Additionally, we explore clinical implications, including prognosis, correlation with WBC count, blast count in the bone marrow and peripheral blood, as well as its association with FLT3-ITD, NPM1 mutations, and FAB classification. Finally, this paper extensively discusses drugs that specifically target the CXCL12-CXCR4 axis, including plerixafor/AMD3100, ulocuplumab, peptide E5, and motixafortide, shedding light on their potential therapeutic value in the treatment of AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Receptors, CXCR4/metabolism
- Receptors, CXCR4/antagonists & inhibitors
- Receptors, CXCR4/genetics
- Nucleophosmin
- Molecular Targeted Therapy
- Chemokine CXCL12/metabolism
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
- Drug Resistance, Neoplasm
- Mutation
- Animals
Collapse
Affiliation(s)
- Jan Korbecki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, Zyty 28, 65-046, Zielona Góra, Poland
| | - Mateusz Bosiacki
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Katarzyna Barczak
- Department of Conservative Dentistry and Endodontics, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Dariusz Chlubek
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland
| | - Irena Baranowska-Bosiacka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111, Szczecin, Poland.
| |
Collapse
|
3
|
Alexandru I, Davidescu L, Motofelea AC, Ciocarlie T, Motofelea N, Costachescu D, Marc MS, Suppini N, Șovrea AS, Coșeriu RL, Bondor DA, Bobeică LG, Crintea A. Emerging Nanomedicine Approaches in Targeted Lung Cancer Treatment. Int J Mol Sci 2024; 25:11235. [PMID: 39457017 PMCID: PMC11508987 DOI: 10.3390/ijms252011235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Lung cancer, the leading cause of cancer-related deaths worldwide, is characterized by its aggressive nature and poor prognosis. As traditional chemotherapy has the disadvantage of non-specificity, nanomedicine offers innovative approaches for targeted therapy, particularly through the development of nanoparticles that can deliver therapeutic agents directly to cancer cells, minimizing systemic toxicity and enhancing treatment efficacy. VEGF and VEGFR are shown to be responsible for activating different signaling cascades, which will ultimately enhance tumor development, angiogenesis, and metastasis. By inhibiting VEGF and VEGFR signaling pathways, these nanotherapeutics can effectively disrupt tumor angiogenesis and proliferation. This review highlights recent advancements in nanoparticle design, including lipid-based, polymeric, and inorganic nanoparticles, and their clinical implications in improving lung cancer outcomes, exploring the role of nanomedicine in lung cancer diagnoses and treatment.
Collapse
Affiliation(s)
- Isaic Alexandru
- Department X of General Surgery, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Lavinia Davidescu
- Department of Medical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania
| | - Alexandru Cătălin Motofelea
- Department of Internal Medicine, Faculty of Medicine, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Tudor Ciocarlie
- Department VII Internal Medicine II, Discipline of Cardiology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Nadica Motofelea
- Department of Obstetrics and Gynecology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania;
| | - Dan Costachescu
- Radiology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Monica Steluta Marc
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Noemi Suppini
- Discipline of Pulmonology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania; (M.S.M.); (N.S.)
| | - Alina Simona Șovrea
- Department of Morphological Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania;
| | - Răzvan-Lucian Coșeriu
- Department of Microbiology, University of Medicine, Pharmacy, Science and Technology “George Emil Palade”, 540142 Târgu-Mures, Romania;
| | - Daniela-Andreea Bondor
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Laura-Gabriela Bobeică
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| | - Andreea Crintea
- Department of Medical Biochemistry, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (D.-A.B.); (L.-G.B.); (A.C.)
| |
Collapse
|
4
|
Renard I, D’huys T, Burke BP, Ajoleza T, Cain AN, Funwie NL, Khan A, Maples DL, Maples RD, Matz DL, McRobbie G, Ullom R, Prior TJ, Linder DP, Van Loy T, Hubin TJ, Schols D, Archibald SJ. Rigid Macrocycle Metal Complexes as CXCR4 Chemokine Receptor Antagonists: Influence of Ring Size. Pharmaceutics 2024; 16:1000. [PMID: 39204345 PMCID: PMC11360128 DOI: 10.3390/pharmaceutics16081000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/13/2024] [Accepted: 07/19/2024] [Indexed: 09/04/2024] Open
Abstract
Understanding the role of chemokine receptors in health and disease has been of increasing interest in recent years. Chemokine receptor CXCR4 has been extensively studied because of its defined role in immune cell trafficking, HIV infection, inflammatory diseases, and cancer progression. We have developed high affinity rigidified CXCR4 antagonists that incorporate metal ions to optimize the binding interactions with the aspartate side chains at the extracellular surface of the CXCR4 chemokine receptor and increase the residence time. Cross- and side-bridged tetraazamacrocylic complexes offer significant advantages over the non-bridged molecular structures in terms of receptor affinity, potential for radiolabelling, and use in therapeutic applications. Our investigation has been extended to the influence of the ring size on bridged tetraazamacrocyclic compounds with the addition of two novel chelators (bis-cross-bridged homocyclen and bis-cross-bridged cyclen) to compare to the bis-bridged cyclam, along with novel metal complexes formed with copper(II) or zinc(II). The in vitro biological assays showed that all of the zinc(II) complexes are high affinity antagonists with a marked increase in CXCR4 selectivity for the bis-cross-bridged cyclen complex, whereas the properties of the copper(II) complexes are highly dependent on metal ion geometry. X-ray crystal structural data and DFT computational studies allow for the rationalisation of the relative affinities and the aspartate residue interactions on the protein surface. Changing the ring size from 14-membered can increase the selectivity for the CXCR4 receptor whilst retaining potent inhibitory activity, improving the key pharmacological characteristics.
Collapse
Affiliation(s)
- Isaline Renard
- Centre for Biomedicine and Positron Emission Tomography Research Centre, The University of Hull, Cottingham Road, Hull HU6 7RX, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| | - Thomas D’huys
- Division of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium
| | - Benjamin P. Burke
- Centre for Biomedicine and Positron Emission Tomography Research Centre, The University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Trisha Ajoleza
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Amy N. Cain
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Neil L. Funwie
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Abid Khan
- Centre for Biomedicine and Positron Emission Tomography Research Centre, The University of Hull, Cottingham Road, Hull HU6 7RX, UK
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Danny L. Maples
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Randall D. Maples
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Dallas L. Matz
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Graeme McRobbie
- Centre for Biomedicine and Positron Emission Tomography Research Centre, The University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Robert Ullom
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Timothy J. Prior
- Chemistry, Faculty of Science and Engineering, The University of Hull, Cottingham Road, Hull HU6 7RX, UK
| | - Douglas P. Linder
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Tom Van Loy
- Division of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium
| | - Timothy J. Hubin
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Dominique Schols
- Division of Virology and Chemotherapy, Rega Institute for Medical Research, KU Leuven, B-3000 Leuven, Belgium
| | - Stephen J. Archibald
- Centre for Biomedicine and Positron Emission Tomography Research Centre, The University of Hull, Cottingham Road, Hull HU6 7RX, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor Lambeth Wing, St Thomas’ Hospital, London SE1 7EH, UK
| |
Collapse
|
5
|
Alcantara KP, Malabanan JWT, Vajragupta O, Rojsitthisak P, Rojsitthisak P. A promising strategy of surface-modified nanoparticles targeting CXCR4 for precision cancer therapy. J Drug Target 2024; 32:587-605. [PMID: 38634290 DOI: 10.1080/1061186x.2024.2345235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/11/2024] [Indexed: 04/19/2024]
Abstract
Nanoparticle (NP) functionalization with specific ligands enhances targeted cancer therapy and imaging by promoting receptor recognition and improving cellular uptake. This review focuses on recent research exploring the interaction between cancer cell-expressed chemokine receptor 4 (CXCR4) and ligand-conjugated NPs, utilising small molecules, peptides, and antibodies. Active NP targeting has shown improved tumour targeting and reduced toxicity, enabling precision therapy and diagnosis. However, challenges persist in the clinical translation of targeted NPs due to issues with biological response, tumour accumulation, and maintaining NP quality at an industrial scale. Biological and intratumoral barriers further hinder efficient NP accumulation in tumours, hampering translatability. To address these challenges, the academic community is refocusing efforts on understanding NP biological fate and establishing robust preclinical models. Future studies should investigate NP-body interactions, develop computational models, and identify optimal preclinical models. Establishing central NP research databases and fostering collaboration across disciplines is crucial to expediting clinical translation. Overcoming these hurdles will unlock the transformative potential of CXCR4-ligand-NP conjugates in revolutionising cancer treatment.
Collapse
Affiliation(s)
- Khent Primo Alcantara
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - John Wilfred T Malabanan
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Opa Vajragupta
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok, Thailand
- Molecular Probes for Imaging Research Network, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Pornchai Rojsitthisak
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Pranee Rojsitthisak
- Center of Excellence in Natural Products for Ageing and Chronic Diseases, Chulalongkorn University, Bangkok, Thailand
- Metallurgy and Materials Science Research Institute, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
6
|
Le Roy MM, Claes S, Saffon-Merceron N, Schols D, Troadec T, Tripier R. Selective synthesis of an elusive C-functional bis-cyclam and study of its inhibition of the CXCR4 chemokine receptor. Org Biomol Chem 2024; 22:3059-3067. [PMID: 38545887 DOI: 10.1039/d3ob02050a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2024]
Abstract
This article presents the controlled synthesis of a rare example of C,C'-linked bis-cyclam architecture in mild conditions through the "bis-aminal" route previously used for the advantageous synthesis of cyclam, N- and C-functional cyclams and N,N'-bis-cyclams. Two synthetic pathways were explored with the smart design of α,β-unsaturated ketones or alkyl halides bis-cyclizing agents. The first led to the isolation of a key intermediate for the future design of N-functionalized bis-cyclams, whereas the second allowed the preparation of the targeted C,C'-xylylene-bis-cyclam under mild conditions with decent yield. This compound was then studied as a CXCR4 receptor inhibitor, one of the main applications known for bis-macrocyclic compounds, in particular in the context of HIV (human immunodeficiency virus) infection. Although results demonstrated that its potency is lower (i.e. 137-fold higher IC50) than the gold standard AMD3100 against HIV infection, clear evidence of CXCR4 inhibition is presented, confirming the potential of this novel architecture and related compounds in this research field.
Collapse
Affiliation(s)
- Marie M Le Roy
- Univ Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France.
| | - Sandra Claes
- Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | | | - Dominique Schols
- Rega Institute for Medical Research, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Thibault Troadec
- Univ Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France.
| | - Raphaël Tripier
- Univ Brest, UMR CNRS 6521 CEMCA, 6 Avenue Victor le Gorgeu, 29200 Brest, France.
| |
Collapse
|
7
|
Alzahrani SO, McRobbie G, Khan A, D'huys T, Van Loy T, Walker AN, Renard I, Hubin TJ, Schols D, Burke BP, Archibald SJ. trans-IV restriction: a new configuration for metal bis-cyclam complexes as potent CXCR4 inhibitors. Dalton Trans 2024; 53:5616-5623. [PMID: 38439632 PMCID: PMC10949960 DOI: 10.1039/d3dt01729j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
The chemokine receptor CXCR4 is implicated in multiple diseases including inflammatory disorders, cancer growth and metastasis, and HIV/AIDS. CXCR4 targeting has been evaluated in treating cancer metastasis and therapy resistance. Cyclam derivatives, most notably AMD3100 (Plerixafor™), are a common motif in small molecule CXCR4 antagonists. However, AMD3100 has not been shown to be effective in cancer treatment as an individual agent. Configurational restriction and transition metal complex formation increases receptor binding affinity and residence time. In the present study, we have synthesized novel trans-IV locked cyclam-based CXCR4 inhibitors, a previously unexploited configuration, and demonstrated their higher affinity for CXCR4 binding and CXCL12-mediated signaling inhibition compared to AMD3100. These results pave the way for even more potent CXCR4 inhibitors that may provide significant efficacy in cancer therapy.
Collapse
Affiliation(s)
- Seraj O Alzahrani
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Graeme McRobbie
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Abid Khan
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
- The University of Manchester, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester, UK
| | - Thomas D'huys
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Tom Van Loy
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Ashlie N Walker
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Isaline Renard
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK
| | - Timothy J Hubin
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK 73096, USA
| | - Dominique Schols
- KU Leuven, Department of Microbiology, Immunology and Transplantation, Rega Institute, Laboratory of Virology and Chemotherapy, Leuven, Belgium
| | - Benjamin P Burke
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
| | - Stephen J Archibald
- Centre for Biomedicine and Positron Emission Tomography Research Centre, Hull York Medical School and University of Hull, Cottingham Road, Hull, HU6 7RX, UK.
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK
| |
Collapse
|
8
|
Nadai M, Doria F, Frasson I, Perrone R, Pirota V, Bergamaschi G, Freccero M, Richter SN. Naphthalene Diimide-Tetraazacycloalkane Conjugates Are G-Quadruplex-Based HIV-1 Inhibitors with a Dual Mode of Action. ACS Infect Dis 2024; 10:489-499. [PMID: 38175706 PMCID: PMC10862543 DOI: 10.1021/acsinfecdis.3c00453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 01/05/2024]
Abstract
Human immunodeficiency virus 1 (HIV-1) therapeutic regimens consist of three or more drugs targeting different steps of the viral life cycle to limit the emergence of viral resistance. In line with the multitargeting strategy, here we conjugated a naphthalene diimide (NDI) moiety with a tetraazacycloalkane to obtain novel naphthalene diimide (NDI)-tetraazacycloalkane conjugates. The NDI inhibits the HIV-1 promoter activity by binding to LTR G-quadruplexes, and the tetraazacycloalkane mimics AMD3100, which blocks HIV entry into cells by interfering with the CXCR4 coreceptor. We synthesized, purified, and tested the metal-free NDI-tetraazacycloalkane conjugate and the two derived metal-organic complexes (MOCs) that incorporate Cu2+ and Zn2+. The NDI-MOCs showed enhanced binding to LTR G4s as assessed by FRET and CD assays in vitro. They also showed enhanced activity in cells where they dose-dependently reduced LTR promoter activity and inhibited viral entry only of the HIV-1 strain that exploited the CXCR4 coreceptor. The time of addition assay confirmed the dual targeting at the different HIV-1 steps. Our results indicate that the NDI-MOC conjugates can simultaneously inhibit viral entry, by targeting the CXCR4 coreceptor, and LTR promoter activity, by stabilizing the LTR G-quadruplexes. The approach of combining multiple targets in a single compound may streamline treatment regimens and improve the overall patient outcomes.
Collapse
Affiliation(s)
- Matteo Nadai
- Department
of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Filippo Doria
- Department
of Chemistry, University of Pavia, V.le Taramelli 10, 27100 Pavia, Italy
| | - Ilaria Frasson
- Department
of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
| | - Rosalba Perrone
- Buck
Institute for Research on Aging, Novato, California 94945, United States
| | - Valentina Pirota
- Department
of Chemistry, University of Pavia, V.le Taramelli 10, 27100 Pavia, Italy
| | - Greta Bergamaschi
- National
Research Council of Italy, Istituto di Scienze e Tecnologie Chimiche
“Giulio Natta” (SCITEC–CNR), Via Mario Bianco 9, 20131 Milano, Italy
| | - Mauro Freccero
- Department
of Chemistry, University of Pavia, V.le Taramelli 10, 27100 Pavia, Italy
| | - Sara N. Richter
- Department
of Molecular Medicine, University of Padua, Via Gabelli 63, 35121 Padua, Italy
- Microbiology
and Virology Unit, Padua University Hospital, 35121 Padua, Italy
| |
Collapse
|
9
|
Meng Q, Zhu R, Mao Y, Zhu S, Wu Y, Huang L, Ciechanover A, An J, Xu Y, Huang Z. Biological and mutational analyses of CXCR4-antagonist interactions and design of new antagonistic analogs. Biosci Rep 2023; 43:BSR20230981. [PMID: 38131305 PMCID: PMC10987480 DOI: 10.1042/bsr20230981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/05/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
The chemokine receptor CXCR4 has become an attractive therapeutic target for HIV-1 infection, hematopoietic stem cell mobilization, and cancer metastasis. A wide variety of synthetic antagonists of CXCR4 have been developed and studied for a growing list of clinical applications. To compare the biological effects of different antagonists on CXCR4 functions and their common and/or distinctive molecular interactions with the receptor, we conducted head-to-head comparative cell-based biological and mutational analyses of the interactions with CXCR4 of eleven reported antagonists, including HC4319, DV3, DV1, DV1 dimer, V1, vMIP-II, CVX15, LY2510924, IT1t, AMD3100, and AMD11070 that were representative of different structural classes of D-peptides, L-peptide, natural chemokine, cyclic peptides, and small molecules. The results were rationalized by molecular modeling of CXCR4-antagonist interactions from which the common as well as different receptor binding sites of these antagonists were derived, revealing a number of important residues such as W94, D97, H113, D171, D262, and E288, mostly of negative charge. To further examine this finding, we designed and synthesized new antagonistic analogs by adding positively charged residues Arg to a D-peptide template to enhance the postulated charge-charge interactions. The newly designed analogs displayed significantly increased binding to CXCR4, which supports the notion that negatively charged residues of CXCR4 can engage in interactions with moieties of positive charge of the antagonistic ligands. The results from these mutational, modeling and new analog design studies shed new insight into the molecular mechanisms of different types of antagonists in recognizing CXCR4 and guide the development of new therapeutic agents.
Collapse
Affiliation(s)
- Qian Meng
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ruohan Zhu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yujia Mao
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Siyu Zhu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yi Wu
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Lina S.M. Huang
- Division of Infectious Diseases and Global Public Heath, Department of Medicine, School of Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093, U.S.A
| | - Aaron Ciechanover
- The Rapport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Jing An
- Division of Infectious Diseases and Global Public Heath, Department of Medicine, School of Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093, U.S.A
| | - Yan Xu
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, Chinese University of Hong Kong, Shenzhen 518172, China
| | - Ziwei Huang
- School of Life Sciences, Tsinghua University, Beijing 100084, China
- Division of Infectious Diseases and Global Public Heath, Department of Medicine, School of Medicine, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093, U.S.A
- Ciechanover Institute of Precision and Regenerative Medicine, School of Medicine, Chinese University of Hong Kong, Shenzhen 518172, China
| |
Collapse
|
10
|
Jouve M, Carpentier R, Kraiem S, Legrand N, Sobolewski C. MiRNAs in Alcohol-Related Liver Diseases and Hepatocellular Carcinoma: A Step toward New Therapeutic Approaches? Cancers (Basel) 2023; 15:5557. [PMID: 38067261 PMCID: PMC10705678 DOI: 10.3390/cancers15235557] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/15/2023] [Accepted: 11/17/2023] [Indexed: 06/29/2024] Open
Abstract
Alcohol-related Liver Disease (ALD) is the primary cause of chronic liver disorders and hepatocellular carcinoma (HCC) development in developed countries and thus represents a major public health concern. Unfortunately, few therapeutic options are available for ALD and HCC, except liver transplantation or tumor resection for HCC. Deciphering the molecular mechanisms underlying the development of these diseases is therefore of major importance to identify early biomarkers and to design efficient therapeutic options. Increasing evidence indicate that epigenetic alterations play a central role in the development of ALD and HCC. Among them, microRNA importantly contribute to the development of this disease by controlling the expression of several genes involved in hepatic metabolism, inflammation, fibrosis, and carcinogenesis at the post-transcriptional level. In this review, we discuss the current knowledge about miRNAs' functions in the different stages of ALD and their role in the progression toward carcinogenesis. We highlight that each stage of ALD is associated with deregulated miRNAs involved in hepatic carcinogenesis, and thus represent HCC-priming miRNAs. By using in silico approaches, we have uncovered new miRNAs potentially involved in HCC. Finally, we discuss the therapeutic potential of targeting miRNAs for the treatment of these diseases.
Collapse
Affiliation(s)
- Mickaël Jouve
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Rodolphe Carpentier
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Sarra Kraiem
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Noémie Legrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| | - Cyril Sobolewski
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France
| |
Collapse
|
11
|
Crees ZD, Rettig MP, Bashey A, Devine SM, Jaglowski S, Wan F, Zhou A, Harding M, Vainstein-Haras A, Sorani E, Gliko-Kabir I, Grossman BJ, Westervelt P, DiPersio JF, Uy GL. Hematopoietic stem cell mobilization for allogeneic stem cell transplantation by motixafortide, a novel CXCR4 inhibitor. Blood Adv 2023; 7:5210-5214. [PMID: 37327120 PMCID: PMC10500469 DOI: 10.1182/bloodadvances.2023010407] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/18/2023] Open
Affiliation(s)
- Zachary D. Crees
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Michael P. Rettig
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Asad Bashey
- Blood and Marrow Transplant Program, Northside Hospital, Atlanta, GA
| | - Steven M. Devine
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program, Minneapolis, MN
| | - Samantha Jaglowski
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Fei Wan
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Amy Zhou
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Melinda Harding
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | | | | | | | - Brenda J. Grossman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Peter Westervelt
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - John F. DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| | - Geoffrey L. Uy
- Division of Oncology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
12
|
Yu J, Zhou X, Shen L. CXCR4-Targeted Radiopharmaceuticals for the Imaging and Therapy of Malignant Tumors. Molecules 2023; 28:4707. [PMID: 37375261 DOI: 10.3390/molecules28124707] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/31/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
C-X-C chemokine receptor type 4 (CXCR4), also known as fusin or CD184, is a 7-transmembrane helix G-protein-coupled receptor that is encoded by the CXCR4 gene. Involved in various physiological processes, CXCR4 could form an interaction with its endogenous partner, chemokine ligand 12 (CXCL12), which is also named SDF-1. In the past several decades, the CXCR4/CXCL12 couple has attracted a large amount of research interest due to its critical functions in the occurrence and development of refractory diseases, such as HIV infection, inflammatory diseases, and metastatic cancer, including breast cancer, gastric cancer, and non-small cell lung cancer. Furthermore, overexpression of CXCR4 in tumor tissues was shown to have a high correlation with tumor aggressiveness and elevated risks of metastasis and recurrence. The pivotal roles of CXCR4 have encouraged an effort around the world to investigate CXCR4-targeted imaging and therapeutics. In this review, we would like to summarize the implementation of CXCR4-targeted radiopharmaceuticals in the field of various kinds of carcinomas. The nomenclature, structure, properties, and functions of chemokines and chemokine receptors are briefly introduced. Radiopharmaceuticals that could target CXCR4 will be described in detail according to their structure, such as pentapeptide-based structures, heptapeptide-based structures, nonapeptide-based structures, etc. To make this review a comprehensive and informative article, we would also like to provide the predictive prospects for the CXCR4-targeted species in future clinical development.
Collapse
Affiliation(s)
- Jingjing Yu
- HTA Co., Ltd., Beijing 102413, China
- Department of Nuclear Technology Application, China Institute of Atomic Energy, Beijing 102413, China
| | - Xu Zhou
- HTA Co., Ltd., Beijing 102413, China
| | - Langtao Shen
- HTA Co., Ltd., Beijing 102413, China
- National Isotope Center of Engineering and Technology, China Institute of Atomic Energy, Beijing 102413, China
| |
Collapse
|
13
|
Leusmann S, Ménová P, Shanin E, Titz A, Rademacher C. Glycomimetics for the inhibition and modulation of lectins. Chem Soc Rev 2023; 52:3663-3740. [PMID: 37232696 PMCID: PMC10243309 DOI: 10.1039/d2cs00954d] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Indexed: 05/27/2023]
Abstract
Carbohydrates are essential mediators of many processes in health and disease. They regulate self-/non-self- discrimination, are key elements of cellular communication, cancer, infection and inflammation, and determine protein folding, function and life-times. Moreover, they are integral to the cellular envelope for microorganisms and participate in biofilm formation. These diverse functions of carbohydrates are mediated by carbohydrate-binding proteins, lectins, and the more the knowledge about the biology of these proteins is advancing, the more interfering with carbohydrate recognition becomes a viable option for the development of novel therapeutics. In this respect, small molecules mimicking this recognition process become more and more available either as tools for fostering our basic understanding of glycobiology or as therapeutics. In this review, we outline the general design principles of glycomimetic inhibitors (Section 2). This section is then followed by highlighting three approaches to interfere with lectin function, i.e. with carbohydrate-derived glycomimetics (Section 3.1), novel glycomimetic scaffolds (Section 3.2) and allosteric modulators (Section 3.3). We summarize recent advances in design and application of glycomimetics for various classes of lectins of mammalian, viral and bacterial origin. Besides highlighting design principles in general, we showcase defined cases in which glycomimetics have been advanced to clinical trials or marketed. Additionally, emerging applications of glycomimetics for targeted protein degradation and targeted delivery purposes are reviewed in Section 4.
Collapse
Affiliation(s)
- Steffen Leusmann
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Petra Ménová
- University of Chemistry and Technology, Prague, Technická 5, 16628 Prague 6, Czech Republic
| | - Elena Shanin
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany.
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), Standort Hannover-Braunschweig, Germany
| | - Christoph Rademacher
- Department of Pharmaceutical Sciences, University of Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria.
- Department of Microbiology, Immunobiology and Genetics, Max F. Perutz Laboratories, University of Vienna, Biocenter 5, 1030 Vienna, Austria
| |
Collapse
|
14
|
Crees ZD, Rettig MP, Jayasinghe RG, Stockerl-Goldstein K, Larson SM, Arpad I, Milone GA, Martino M, Stiff P, Sborov D, Pereira D, Micallef I, Moreno-Jiménez G, Mikala G, Coronel MLP, Holtick U, Hiemenz J, Qazilbash MH, Hardy N, Latif T, García-Cadenas I, Vainstein-Haras A, Sorani E, Gliko-Kabir I, Goldstein I, Ickowicz D, Shemesh-Darvish L, Kadosh S, Gao F, Schroeder MA, Vij R, DiPersio JF. Motixafortide and G-CSF to mobilize hematopoietic stem cells for autologous transplantation in multiple myeloma: a randomized phase 3 trial. Nat Med 2023; 29:869-879. [PMID: 37069359 PMCID: PMC10115633 DOI: 10.1038/s41591-023-02273-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 02/22/2023] [Indexed: 04/19/2023]
Abstract
Autologous hematopoietic stem cell transplantation (ASCT) improves survival in multiple myeloma (MM). However, many individuals are unable to collect optimal CD34+ hematopoietic stem and progenitor cell (HSPC) numbers with granulocyte colony-stimulating factor (G-CSF) mobilization. Motixafortide is a novel cyclic-peptide CXCR4 inhibitor with extended in vivo activity. The GENESIS trial was a prospective, phase 3, double-blind, placebo-controlled, multicenter study with the objective of assessing the superiority of motixafortide + G-CSF over placebo + G-CSF to mobilize HSPCs for ASCT in MM. The primary endpoint was the proportion of patients collecting ≥6 × 106 CD34+ cells kg-1 within two apheresis procedures; the secondary endpoint was to achieve this goal in one apheresis. A total of 122 adult patients with MM undergoing ASCT were enrolled at 18 sites across five countries and randomized (2:1) to motixafortide + G-CSF or placebo + G-CSF for HSPC mobilization. Motixafortide + G-CSF enabled 92.5% to successfully meet the primary endpoint versus 26.2% with placebo + G-CSF (odds ratio (OR) 53.3, 95% confidence interval (CI) 14.12-201.33, P < 0.0001). Motixafortide + G-CSF also enabled 88.8% to meet the secondary endpoint versus 9.5% with placebo + G-CSF (OR 118.0, 95% CI 25.36-549.35, P < 0.0001). Motixafortide + G-CSF was safe and well tolerated, with the most common treatment-emergent adverse events observed being transient, grade 1/2 injection site reactions (pain, 50%; erythema, 27.5%; pruritis, 21.3%). In conclusion, motixafortide + G-CSF mobilized significantly greater CD34+ HSPC numbers within two apheresis procedures versus placebo + G-CSF while preferentially mobilizing increased numbers of immunophenotypically and transcriptionally primitive HSPCs. Trial Registration: ClinicalTrials.gov , NCT03246529.
Collapse
Affiliation(s)
- Zachary D Crees
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| | - Michael P Rettig
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Reyka G Jayasinghe
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | | | - Sarah M Larson
- Division of Hematology-Oncology, UCLA School of Medicine, Los Angeles, CA, USA
| | - Illes Arpad
- Division of Hematology, Department of Internal Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Giulio A Milone
- Unità di Trapianto Emopoietico, Azienda Ospedaliero Universitaria 'Policlinico-San Marco', Catania, Italy
| | - Massimo Martino
- Unit of Stem Cell Transplantation and Cellular Therapies, Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy
| | | | - Douglas Sborov
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Denise Pereira
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, USA
| | | | | | - Gabor Mikala
- Center Hospital of Southern Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | | | - Udo Holtick
- Department I of Internal Medicine, Medical Faculty and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - John Hiemenz
- Division of Hematology-Oncology, University of Florida, Gainesville, FL, USA
| | - Muzaffar H Qazilbash
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nancy Hardy
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tahir Latif
- Division of Hematology-Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Irene García-Cadenas
- Department of Hematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | | | | | | | | | | | | | - Feng Gao
- Division of Public Health Sciences, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Mark A Schroeder
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Ravi Vij
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - John F DiPersio
- Division of Oncology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| |
Collapse
|
15
|
Chang MR, Rusanov DA, Arakelyan J, Alshehri M, Asaturova AV, Kireeva GS, Babak MV, Ang WH. Targeting emerging cancer hallmarks by transition metal complexes: Cancer stem cells and tumor microbiome. Part I. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2022.214923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
16
|
Discovery of Bis-Imidazoline Derivatives as New CXCR4 Ligands. Molecules 2023; 28:molecules28031156. [PMID: 36770826 PMCID: PMC9920567 DOI: 10.3390/molecules28031156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
The chemokine receptor CXCR4 and its ligand CXCL12 regulate leukocyte trafficking, homeostasis and functions and are potential therapeutic targets in many diseases such as HIV-1 infection and cancers. Here, we identified new CXCR4 ligands in the CERMN chemical library using a FRET-based high-throughput screening assay. These are bis-imidazoline compounds comprising two imidazole rings linked by an alkyl chain. The molecules displace CXCL12 binding with submicromolar potencies, similarly to AMD3100, the only marketed CXCR4 ligand. They also inhibit anti-CXCR4 mAb 12G5 binding, CXCL12-mediated chemotaxis and HIV-1 infection. Further studies with newly synthesized derivatives pointed out to a role of alkyl chain length on the bis-imidazoline properties, with molecules with an even number of carbons equal to 8, 10 or 12 being the most potent. Interestingly, these differ in the functions of CXCR4 that they influence. Site-directed mutagenesis and molecular docking predict that the alkyl chain folds in such a way that the two imidazole groups become lodged in the transmembrane binding cavity of CXCR4. Results also suggest that the alkyl chain length influences how the imidazole rings positions in the cavity. These results may provide a basis for the design of new CXCR4 antagonists targeting specific functions of the receptor.
Collapse
|
17
|
Steiner N, Göbel G, Mauser L, Mühlnikel L, Fischinger M, Künz T, Willenbacher W, Hetzenauer G, Rudzki J, Nussbaumer W, Mayer W, Gunsilius E, Kircher B, Wolf D, Nachbaur D. Poor Mobilizers in Lymphoma but Not Myeloma Patients Had Significantly Poorer Progression-Free Survival after Autologous Stem Cell Transplantation: Results of a Large Retrospective, Single-Center Observational Study. Cancers (Basel) 2023; 15:cancers15030608. [PMID: 36765566 PMCID: PMC9913576 DOI: 10.3390/cancers15030608] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/11/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
In our single-center study, 357 myeloma and lymphoma patients between 2009 and 2019 were mobilized with granulocyte colony-stimulating factor (G-CSF 7.5 µg/kg bid for four days) plus a fixed dose of 24 mg Plerixafor when indicated (Plerixafor Group, n = 187) or G-CSF alone (G-CSF Group, n = 170). The target CD34 cell yields were ≥2.0 × 106 CD34+ cells/kg in lymphoma and ≥4.0 × 106 CD34+ cells/kg in myeloma patients to enable putative second transplants in the latter. There were no significant differences in engraftment kinetics or transfusion requirements between the Plerixafor Group and the control group in the myeloma cohort, with lymphoma patients not requiring Plerixafor showing significantly faster neutrophil recovery, a trend to faster platelet recovery, and a significantly lower need for platelet transfusions, probably due to the significantly lower number of CD34-positive cells re-transfused. While in myeloma patients the outcome (overall survival, progression-free survival) following autologous stem cell transplantation (ASCT) was similar between the Plerixafor Group and the control group, hard to mobilize lymphoma patients had significantly poorer progression-free survival (47% vs. 74% at 36 months after ASCT, p = 0.003) with a trend also to poorer overall survival (71% vs. 84%). In conclusion, while there seem to be no differences in stemness capacity and long-term engraftment efficiency between the Plerixafor and the G-CSF Group in lymphoma as well as myeloma patients, poor mobilizing lymphoma patients per se constitute a high-risk population with a poorer outcome after ASCT. Whether disease characteristics and/or a more intense or stem cell-toxic pre-mobilization chemo-/radiotherapy burden in this cohort are responsible for this observation remains to be shown in future studies.
Collapse
Affiliation(s)
- Normann Steiner
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
- Correspondence: ; Tel.: +43-(0)-512504-24003; Fax: +43-(0)-512504-25615
| | - Georg Göbel
- Department of Medical Statistics, Informatics and Health Economics, Medical University of Innsbruck, Schöpfstrasse 41/1, A-6020 Innsbruck, Austria
| | - Leonie Mauser
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Lena Mühlnikel
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Marie Fischinger
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Tina Künz
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Wolfgang Willenbacher
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Gabriele Hetzenauer
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Jakob Rudzki
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Walter Nussbaumer
- Central Institute for Blood Transfusion and Department of Immunology, University Hospital of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Wolfgang Mayer
- Central Institute for Blood Transfusion and Department of Immunology, University Hospital of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Eberhard Gunsilius
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Brigitte Kircher
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - Dominik Wolf
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| | - David Nachbaur
- Department of Internal Medicine V (Hematology and Medical Oncology), Medical University of Innsbruck, Anichstrasse 35, A-6020 Innsbruck, Austria
| |
Collapse
|
18
|
Holmberg LA, Linenberger M, Connelly-Smith L. Successful Mobilization of Autologous Hematopoietic Peripheral Blood Stem Cells after Salvage Chemotherapy in Patients with Low CD34 Blood Cell Counts. Transplant Cell Ther 2022; 28:754-759. [DOI: 10.1016/j.jtct.2022.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 08/11/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
|
19
|
Ng C, Lee KL, Muthiah MD, Wu KX, Chioh FWJ, Tan K, Soon GST, Shabbir A, Loo WM, Low ZS, Chen Q, Tan NS, Ng HH, Dan YY, Cheung C. Endothelial‐immune crosstalk contributes to vasculopathy in nonalcoholic fatty liver disease. EMBO Rep 2022; 23:e54271. [PMID: 35403791 PMCID: PMC9171677 DOI: 10.15252/embr.202154271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 11/17/2022] Open
Abstract
The top cause of mortality in patients with nonalcoholic fatty liver disease (NAFLD) is cardiovascular complications. However, mechanisms of NAFLD‐associated vasculopathy remain understudied. Here, we show that blood outgrowth endothelial cells (BOECs) from NAFLD subjects exhibit global transcriptional upregulation of chemokines and human leukocyte antigens. In mouse models of diet‐induced NAFLD, we confirm heightened endothelial expressions of CXCL12 in the aortas and the liver vasculatures, and increased retention of infiltrated leukocytes within the vessel walls. To elucidate endothelial‐immune crosstalk, we performed immunoprofiling by single‐cell analysis, uncovering T cell intensification in NAFLD patients. Functionally, treatment with a CXCL12‐neutralizing antibody is effective at moderating the enhanced chemotactic effect of NAFLD BOECs in recruiting CD8+ T lymphocytes. Interference with the CXCL12‐CXCR4 axis using a CXCR4 antagonist also averts the impact of immune cell transendothelial migration and restores endothelial barrier integrity. Clinically, we detect threefold more circulating damaged endothelial cells in NAFLD patients than in healthy controls. Our work provides insight into the modulation of interactions with effector immune cells to mitigate endothelial injury in NAFLD.
Collapse
Affiliation(s)
- Chun‐Yi Ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Khang Leng Lee
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Mark Dhinesh Muthiah
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Medicine National University Health System Singapore Singapore
| | - Kan Xing Wu
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | | | - Konstanze Tan
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | | | - Asim Shabbir
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Surgery University Surgical Cluster National University Health System Singapore Singapore
| | - Wai Mun Loo
- Department of Medicine National University Health System Singapore Singapore
| | - Zun Siong Low
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Nguan Soon Tan
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- School of Biological Sciences Nanyang Technological University Singapore Singapore
| | - Huck Hui Ng
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
- School of Biological Sciences Nanyang Technological University Singapore Singapore
- Genome Institute of Singapore Agency for Science Technology and Research (A*STAR) Singapore Singapore
| | - Yock Young Dan
- Yong Loo Lin School of Medicine National University of Singapore Singapore Singapore
- Department of Medicine National University Health System Singapore Singapore
| | - Christine Cheung
- Lee Kong Chian School of Medicine Nanyang Technological University Singapore Singapore
- Institute of Molecular and Cell Biology Agency for Science Technology and Research (A*STAR) Singapore Singapore
| |
Collapse
|
20
|
Zhang C, Hang Y, Tang W, Sil D, Jensen-Smith HC, Bennett RG, McVicker BL, Oupický D. Dually Active Polycation/miRNA Nanoparticles for the Treatment of Fibrosis in Alcohol-Associated Liver Disease. Pharmaceutics 2022; 14:pharmaceutics14030669. [PMID: 35336043 PMCID: PMC8949580 DOI: 10.3390/pharmaceutics14030669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
Alcohol-associated liver disease (AALD) is a major cause of liver disorders worldwide. Current treatment options are limited, especially for AALD-associated fibrosis. Promising approaches include RNA interference for miR-155 overexpression in Kupffer cells (KCs), as well as the use of CXCR4 antagonists that inhibit the activation of hepatic stellate cells (HSCs) through the CXCL12/CXCR4 axis. The development of dual-functioning nanoparticles for the effective delivery of antifibrotic RNA together with a CXCR4 inhibitor thus promises to improve the treatment of AALD fibrosis. In this study, cholesterol-modified polymeric CXCR4 inhibitor (Chol-PCX) was synthesized and used to encapsulate anti-miR-155 or non-coding (NC) miRNA in the form of Chol-PCX/miRNA nanoparticles. The results indicate that the nanoparticles induce a significant miR-155 silencing effect both in vitro and in vivo. Treatment with the Chol-PCX/anti-miR-155 particles in a model of moderate alcohol consumption with secondary liver insult resulted in a significant reduction in aminotransferase enzymes as well as collagen content in the liver parenchyma. Overall, our data support the use of Chol-PCX as a carrier for anti-miR-155 for the combined therapeutic inhibition of CXCR4 and miR-155 expression as a way to improve fibrotic damage in the liver.
Collapse
Affiliation(s)
- Chuhan Zhang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.Z.); (Y.H.); (W.T.); (D.S.)
| | - Yu Hang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.Z.); (Y.H.); (W.T.); (D.S.)
| | - Weimin Tang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.Z.); (Y.H.); (W.T.); (D.S.)
| | - Diptesh Sil
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.Z.); (Y.H.); (W.T.); (D.S.)
| | - Heather C. Jensen-Smith
- Eppley Institute for Cancer Research & Fred and Pamela Buffer Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA;
| | - Robert G. Bennett
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.G.B.); (B.L.M.)
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - Benita L. McVicker
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.G.B.); (B.L.M.)
- VA Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; (C.Z.); (Y.H.); (W.T.); (D.S.)
- Correspondence:
| |
Collapse
|
21
|
Barreca M, Spanò V, Raimondi MV, Bivacqua R, Giuffrida S, Montalbano A, Cavalli A, Bertoni F, Barraja P. GPCR Inhibition in Treating Lymphoma. ACS Med Chem Lett 2022; 13:358-364. [PMID: 38239337 PMCID: PMC10796172 DOI: 10.1021/acsmedchemlett.1c00600] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are important classes of cell surface receptors involved in multiple physiological functions. Aberrant expression, upregulation, and mutation of GPCR signaling pathways are frequent in many types of cancers, promoting hyperproliferation, angiogenesis, and metastasis. Recent studies showed that alterations of GPCRs are involved in different lymphoma types. Herein, we review the synthetic strategies to obtain GPCR inhibitors, focusing on CXCR4 inhibitors which represent most of the GPCR inhibitors available in the market or under preclinical investigations for these diseases.
Collapse
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Virginia Spanò
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Maria V Raimondi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Roberta Bivacqua
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Stefano Giuffrida
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Andrea Cavalli
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, USI, Via Francesco Chiesa 5, 6500 Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Quartier Sorge - Batiment Amphipole, 1015 Lausanne, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Via Francesco Chiesa 5, 6500 Bellinzona, Switzerland
- Oncology Institute of Southern Switzerland, Via Vincenzo Vela 6, 6500 Bellinzona, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| |
Collapse
|
22
|
Sabet A, Mader N, Bittenbring JT, Khreish F, Grünwald F, Biersack HJ, Ezziddin S. Prophylactic Peripheral Blood Stem Cell Collection in Patients with Extensive Bone-Marrow Infiltration of Neuroendocrine Tumours Prior to Peptide Receptor Radionuclide Therapy with 177Lu-DOTATATE. Pharmaceuticals (Basel) 2021; 14:ph14101022. [PMID: 34681247 PMCID: PMC8539404 DOI: 10.3390/ph14101022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 09/20/2021] [Accepted: 09/27/2021] [Indexed: 11/16/2022] Open
Abstract
Peptide receptor radionuclide therapy (PRRT) of metastatic neuroendocrine tumors (NET) can be successfully repeated but may eventually be dose-limited. Since 177Lu-DOTATATE dose limitation may come from hematological rather than renal function, hematological peripheral blood stem cell backup might be desirable. Here, we report our initial experience of peripheral blood stem-cell collection (PBSC) in patients with treatment-related cytopenia and therefore high risk of bone-marrow failure. Five patients with diffuse bone-marrow infiltration of NET and relevant myelosuppression (≥grade 2) received PBSC before one PRRT cycle with 177Lu-DOTATATE (7.6 ± 0.8 GBq/cycle). Standard stem-cell mobilization with Granulocyte-colony stimulating factor (G-CSF) was applied, and successful PBSC was defined as a collection of >2 × 106/kg CD34+ cells. In case of initial failure, Plerixafor was administered in addition to G-CSF prior to apheresis. PBSC was successfully performed in all patients with no adverse events. Median cumulative activity was 44.8 GBq (range, 21.3-62.4). Three patients had been previously treated with PRRT, two of which needed the addition of Plerixafor for stem-cell mobilization. Only one of five patients required autologous peripheral blood stem-cell transplantation during the median follow up time of 28 months. PBSC collection seems to be feasible in NET with bone-marrow involvement and might be worth considering as a backup strategy prior to PRRT, in order to overcome dose-limiting bone-marrow toxicity.
Collapse
Affiliation(s)
- Amir Sabet
- Department of Nuclear Medicine, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany; (N.M.); (F.G.)
- Correspondence:
| | - Nicolai Mader
- Department of Nuclear Medicine, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany; (N.M.); (F.G.)
| | - Jörg Thomas Bittenbring
- Department of Haematology and Oncology, Caritas Hospital Saarbrücken, 66123 Saarbrücken, Germany;
| | - Fadi Khreish
- Department of Nuclear Medicine, Saarland University Medical Center, 66421 Homburg, Germany; (F.K.); (S.E.)
| | - Frank Grünwald
- Department of Nuclear Medicine, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany; (N.M.); (F.G.)
| | | | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University Medical Center, 66421 Homburg, Germany; (F.K.); (S.E.)
| |
Collapse
|
23
|
Bhunia SS, Saxena AK. Efficiency of Homology Modeling Assisted Molecular Docking in G-protein Coupled Receptors. Curr Top Med Chem 2021; 21:269-294. [PMID: 32901584 DOI: 10.2174/1568026620666200908165250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Molecular docking is in regular practice to assess ligand affinity on a target protein crystal structure. In the absence of protein crystal structure, the homology modeling or comparative modeling is the best alternative to elucidate the relationship details between a ligand and protein at the molecular level. The development of accurate homology modeling (HM) and its integration with molecular docking (MD) is essential for successful, rational drug discovery. OBJECTIVE The G-protein coupled receptors (GPCRs) are attractive therapeutic targets due to their immense role in human pharmacology. The GPCRs are membrane-bound proteins with the complex constitution, and the understanding of their activation and inactivation mechanisms is quite challenging. Over the past decade, there has been a rapid expansion in the number of solved G-protein-coupled receptor (GPCR) crystal structures; however, the majority of the GPCR structures remain unsolved. In this context, HM guided MD has been widely used for structure-based drug design (SBDD) of GPCRs. METHODS The focus of this review is on the recent (i) developments on HM supported GPCR drug discovery in the absence of GPCR crystal structures and (ii) application of HM in understanding the ligand interactions at the binding site, virtual screening, determining receptor subtype selectivity and receptor behaviour in comparison with GPCR crystal structures. RESULTS The HM in GPCRs has been extremely challenging due to the scarcity in template structures. In such a scenario, it is difficult to get accurate HM that can facilitate understanding of the ligand-receptor interactions. This problem has been alleviated to some extent by developing refined HM based on incorporating active /inactive ligand information and inducing protein flexibility. In some cases, HM proteins were found to outscore crystal structures. CONCLUSION The developments in HM have been highly operative to gain insights about the ligand interaction at the binding site and receptor functioning at the molecular level. Thus, HM guided molecular docking may be useful for rational drug discovery for the GPCRs mediated diseases.
Collapse
Affiliation(s)
- Shome S Bhunia
- Global Institute of Pharmaceutical Education and Research, Kashipur, Uttarakhand, India
| | - Anil K Saxena
- Division of Medicinal and Process Chemistry, CSIR-CDRI, Lucknow 226031, India
| |
Collapse
|
24
|
Boulad F, Zhang J, Yazdanbakhsh K, Sadelain M, Shi PA. Evidence for continued dose escalation of plerixafor for hematopoietic progenitor cell collections in sickle cell disease. Blood Cells Mol Dis 2021; 90:102588. [PMID: 34166998 DOI: 10.1016/j.bcmd.2021.102588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 11/19/2022]
Abstract
We present data from our study of plerixafor mobilization (NCT02193191) relevant to the question of whether further dose escalation of plerixafor can address inconsistent adequacy of CD34+ mobilization for gene therapy of sickle cell disease (SCD). We found that, in the same patient, higher plerixafor dosing was associated with higher fold increases in PB CD34+ count, but not necessarily higher absolute CD34+ counts. Variation in pre-apheresis absolute CD34+ counts was related to intra-individual variation in baseline PB CD34+ counts and inter-individual variation in responsiveness to plerixafor. Overall, our results support further studies of continued dose escalation of plerixafor for autologous HPC collection in SCD.
Collapse
Affiliation(s)
- Farid Boulad
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Jiahao Zhang
- New York Blood Center, New York, NY, United States of America
| | | | - Michel Sadelain
- Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Patricia A Shi
- New York Blood Center, New York, NY, United States of America; Sickle Cell Program, Division of Hematology, Albert Einstein College of Medicine, Bronx, NY, United States of America.
| |
Collapse
|
25
|
Jørgensen AS, Daugvilaite V, De Filippo K, Berg C, Mavri M, Benned-Jensen T, Juzenaite G, Hjortø G, Rankin S, Våbenø J, Rosenkilde MM. Biased action of the CXCR4-targeting drug plerixafor is essential for its superior hematopoietic stem cell mobilization. Commun Biol 2021; 4:569. [PMID: 33980979 PMCID: PMC8115334 DOI: 10.1038/s42003-021-02070-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 03/31/2021] [Indexed: 01/14/2023] Open
Abstract
Following the FDA-approval of the hematopoietic stem cell (HSC) mobilizer plerixafor, orally available and potent CXCR4 antagonists were pursued. One such proposition was AMD11070, which was orally active and had superior antagonism in vitro; however, it did not appear as effective for HSC mobilization in vivo. Here we show that while AMD11070 acts as a full antagonist, plerixafor acts biased by stimulating β-arrestin recruitment while fully antagonizing G protein. Consequently, while AMD11070 prevents the constitutive receptor internalization, plerixafor allows it and thereby decreases receptor expression. These findings are confirmed by the successful transfer of both ligands' binding sites and action to the related CXCR3 receptor. In vivo, plerixafor exhibits superior HSC mobilization associated with a dramatic reversal of the CXCL12 gradient across the bone marrow endothelium, which is not seen for AMD11070. We propose that the biased action of plerixafor is central for its superior therapeutic effect in HSC mobilization.
Collapse
Affiliation(s)
- Astrid S Jørgensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Viktorija Daugvilaite
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Katia De Filippo
- Department of Medicine, National Heart and Lung Institute (NHLI), Imperial College, London, United Kingdom
| | - Christian Berg
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
- Unit for Infectious Diseases, Department of Medicine, Herlev-Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Masa Mavri
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Tau Benned-Jensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
- Lundbeck A/S, Copenhagen, Denmark
| | - Goda Juzenaite
- Department of Medicine, National Heart and Lung Institute (NHLI), Imperial College, London, United Kingdom
| | - Gertrud Hjortø
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Sara Rankin
- Department of Medicine, National Heart and Lung Institute (NHLI), Imperial College, London, United Kingdom
| | - Jon Våbenø
- Helgeland Hospital Trust, Sandnessjøen, Norway.
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
26
|
Mai CL, Tan Z, Xu YN, Zhang JJ, Huang ZH, Wang D, Zhang H, Gui WS, Zhang J, Lin ZJ, Meng YT, Wei X, Jie YT, Grace PM, Wu LJ, Zhou LJ, Liu XG. CXCL12-mediated monocyte transmigration into brain perivascular space leads to neuroinflammation and memory deficit in neuropathic pain. Theranostics 2021; 11:1059-1078. [PMID: 33391521 PMCID: PMC7738876 DOI: 10.7150/thno.44364] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Emerging clinical and experimental evidence demonstrates that neuroinflammation plays an important role in cognitive impairment associated with neuropathic pain. However, how peripheral nerve challenge induces remote inflammation in the brain remains largely unknown. Methods: The circulating leukocytes and plasma C-X-C motif chemokine 12 (CXCL12) and brain perivascular macrophages (PVMs) were analyzed by flow cytometry, Western blotting, ELISA, and immunostaining in spared nerve injury (SNI) mice. The memory function was evaluated with a novel object recognition test (NORT) in mice and with Montreal Cognitive Assessment (MoCA) in chronic pain patients. Results: The classical monocytes and CXCL12 in the blood, PVMs in the perivascular space, and gliosis in the brain, particularly in the hippocampus, were persistently increased following SNI in mice. Using the transgenic CCR2RFP/+ and CX3CR1GFP/+ mice, we discovered that at least some of the PVMs were recruited from circulating monocytes. The SNI-induced increase in hippocampal PVMs, gliosis, and memory decline were substantially prevented by either depleting circulating monocytes via intravenous injection of clodronate liposomes or blockade of CXCL12-CXCR4 signaling. On the contrary, intravenous injection of CXCL12 at a pathological concentration in naïve mice mimicked SNI effects. Significantly, we found that circulating monocytes and plasma CXCL12 were elevated in chronic pain patients, and both of them were closely correlated with memory decline. Conclusion: CXCL12-mediated monocyte recruitment into the perivascular space is critical for neuroinflammation and the resultant cognitive impairment in neuropathic pain.
Collapse
Affiliation(s)
- Chun-Lin Mai
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhi Tan
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ya-Nan Xu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing-Jun Zhang
- Department of Anesthesiology and Pain Clinic, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zhen-Hua Huang
- Division of Emergency Medicine, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dong Wang
- Department of Clinical Laboratory, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hui Zhang
- Department of Anesthesiology, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China
| | - Wen-Shan Gui
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jun Zhang
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhen-Jia Lin
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Tong Meng
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao Wei
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Tao Jie
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Peter M. Grace
- Department of Critical Care & Respiratory Care Research (PMG), University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN 55905, USA
| | - Li-Jun Zhou
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou 510080, China
| | - Xian-Guo Liu
- Department of Physiology and Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Guangzhou 510080, China
| |
Collapse
|
27
|
Ullah A, Chen G, Hussain A, Khan H, Abbas A, Zhou Z, Shafiq M, Ahmad S, Ali U, Usman M, Raza F, Ahmed A, Qiu Z, Zheng M, Liu D. Cyclam-Modified Polyethyleneimine for Simultaneous TGFβ siRNA Delivery and CXCR4 Inhibition for the Treatment of CCl 4-Induced Liver Fibrosis. Int J Nanomedicine 2021; 16:4451-4470. [PMID: 34234436 PMCID: PMC8257077 DOI: 10.2147/ijn.s314367] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Liver fibrosis is a chronic liver disease with excessive production of extracellular matrix proteins, leading to cirrhosis, hepatocellular carcinoma, and death. PURPOSE This study aimed at the development of a novel derivative of polyethyleneimine (PEI) that can effectively deliver transforming growth factor β (TGFβ) siRNA and inhibit chemokine receptor 4 (CXCR4) for TGFβ silencing and CXCR4 Inhibition, respectively, to treat CCl4-induced liver fibrosis in a mouse model. METHODS Cyclam-modified PEI (PEI-Cyclam) was synthesized by incorporating cyclam moiety into PEI by nucleophilic substitution reaction. Gel electrophoresis confirmed the PEI-Cyclam polyplex formation and stability against RNAase and serum degradation. Transmission electron microscopy and zeta sizer were employed for the morphology, particle size, and zeta potential, respectively. The gene silencing and CXCR4 targeting abilities of PEI-Cyclam polyplex were evaluated by luciferase and CXCR4 redistribution assays, respectively. The histological and immunohistochemical staining determined the anti-fibrotic activity of PEI-Cyclam polyplex. The TGFβ silencing of PEI-Cyclam polyplex was authenticated by Western blotting. RESULTS The 1H NMR of PEI-Cyclam exhibited successful incorporation of cyclam content onto PEI. The PEI-Cyclam polyplex displayed spherical morphology, positive surface charge, and stability against RNAse and serum degradation. Cyclam modification decreased the cytotoxicity and demonstrated CXCR4 antagonistic and luciferase gene silencing efficiency. PEI-Cyclam/siTGFβ polyplexes decreased inflammation, collagen deposition, apoptosis, and cell proliferation, thus ameliorating liver fibrosis. Also, PEI-Cyclam/siTGFβ polyplex significantly downregulated α-smooth muscle actin, TGFβ, and collagen type III. CONCLUSION Our findings validate the feasibility of using PEI-Cyclam as a siRNA delivery vector for simultaneous TGFβ siRNA delivery and CXCR4 inhibition for the combined anti-fibrotic effects in a setting of CCl4-induced liver fibrosis.
Collapse
Affiliation(s)
- Aftab Ullah
- Department of Pharmacy, Shantou University Medical College, Shantou, 515041, Guangdong, People’s Republic of China
- Correspondence: Aftab Ullah; Daojun Liu Email ;
| | - Gang Chen
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, People’s Republic of China
| | - Abid Hussain
- School of Life Science, Key Laboratory of Molecular Medicine and Biotherapy, Beijing Institute of Technology, Beijing, 100081, People’s Republic of China
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China
| | - Hanif Khan
- Department of Pharmacy, Shantou University Medical College, Shantou, 515041, Guangdong, People’s Republic of China
| | - Azar Abbas
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210028, Jiangsu, People’s Republic of China
| | - Zhanwei Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210028, Jiangsu, People’s Republic of China
| | - Muhammad Shafiq
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, 515041, people's Republic of China
| | - Saleem Ahmad
- Department of Medicine, Shantou University Medical College Cancer Hospital, Shantou, People’s Republic of China
| | - Usman Ali
- School of Pharmacy, Shanghai Jiaotong University, Shanghai, 200240, Shanghai, People’s Republic of China
| | - Muhammad Usman
- Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong, 515041, people's Republic of China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiaotong University, Shanghai, 200240, Shanghai, People’s Republic of China
| | - Abrar Ahmed
- School of Pharmacy, Shanghai Jiaotong University, Shanghai, 200240, Shanghai, People’s Republic of China
| | - Zijie Qiu
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210028, Jiangsu, People’s Republic of China
| | - Maochao Zheng
- Department of Pharmacy, Shantou University Medical College, Shantou, 515041, Guangdong, People’s Republic of China
| | - Daojun Liu
- Department of Pharmacy, Shantou University Medical College, Shantou, 515041, Guangdong, People’s Republic of China
| |
Collapse
|
28
|
Göçer M, Kurtoğlu E. Is absence of CD56 a predictive factor for peripheral blood stem cell mobilization failure in patients with multiple myeloma? J Clin Apher 2020; 36:332-339. [PMID: 33333593 DOI: 10.1002/jca.21864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/22/2020] [Accepted: 12/06/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND CD56 is believed to play a major role in MM pathogenesis with a 70% to 80% expression rate in malignant plasma cells at the time of diagnosis. Our objective in this study was to investigate the relationship between the characteristics of CD56 expression in bone marrow aspiration material at the time of diagnosis and the success of stem cell mobilization in patients diagnosed with MM. METHODS This monocenter study included 94 patients who were diagnosed with MM and had a stem cell mobilization procedure for autologous hematopoietic stem cell transplantation. The primary endpoint of the study was to compare the mobilization success between the groups with and without CD56 expression. The secondary endpoint was to identify other factors affecting mobilization failure outside CD56. RESULTS At the time of diagnosis, 49 (52.1%) patients had CD56 expression and 45 (47.9%) did not. Mobilization failed in 11 (11.7%) patients. Age, gender, ISS stage and the number of premobilization treatment regimens were not found predictive of mobilization failure. CD56 negativity was 42.2% in the group that had mobilization success and 90.9% in the group that had mobilization failure (P = .001). CONCLUSIONS The fact that CD56 residing on the membrane enables interaction between bone marrow cells and ECM and functions as a signal molecule increases sensitivity to the chemotherapy and G-CSF that are used for mobilization. We found that absence of CD56 can be used as a predictive factor for mobilization failure at the time of diagnosis.
Collapse
Affiliation(s)
- Mesut Göçer
- Department of Internal Medicine, Division of Hematology, Antalya Training and Research Hospital, Antalya, Turkey
| | - Erdal Kurtoğlu
- Department of Internal Medicine, Division of Hematology, Antalya Training and Research Hospital, Antalya, Turkey
| |
Collapse
|
29
|
Chang CC, Liou JW, Dass KTP, Li YT, Jiang SJ, Pan SF, Yeh YC, Hsu HJ. Internal water channel formation in CXCR4 is crucial for G i-protein coupling upon activation by CXCL12. Commun Chem 2020; 3:133. [PMID: 36703316 PMCID: PMC9814148 DOI: 10.1038/s42004-020-00383-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 09/15/2020] [Indexed: 01/29/2023] Open
Abstract
Chemokine receptor CXCR4 is a major drug target for numerous diseases because of its involvement in the regulation of cell migration and the developmental process. In this study, atomic-level molecular dynamics simulations were used to determine the activation mechanism and internal water formation of CXCR4 in complex with chemokine CXCL12 and Gi-protein. The results indicated that CXCL12-bound CXCR4 underwent transmembrane 6 (TM6) outward movement and a decrease in tyrosine toggle switch by eliciting the breakage of hydrophobic layer to form a continuous internal water channel. In the GDP-bound Gαi-protein state, the rotation and translation of the α5-helix of Gαi-protein toward the cytoplasmic pocket of CXCR4 induced an increase in interdomain distance for GDP leaving. Finally, an internal water channel formation model was proposed based on our simulations for CXCL12-bound CXCR4 in complex with Gαi-protein upon activation for downstream signaling. This model could be useful in anticancer drug development.
Collapse
Affiliation(s)
- Chun-Chun Chang
- Department of Laboratory Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 97004, Taiwan
- Department of Laboratory Medicine and Biotechnology, College of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Je-Wen Liou
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | | | - Ya-Tzu Li
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Sheng-Feng Pan
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Yu-Chen Yeh
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
- Department of Life Sciences, Tzu Chi University, Hualien, 97004, Taiwan
| | - Hao-Jen Hsu
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan.
- Department of Life Sciences, Tzu Chi University, Hualien, 97004, Taiwan.
| |
Collapse
|
30
|
Stephens BS, Ngo T, Kufareva I, Handel TM. Functional anatomy of the full-length CXCR4-CXCL12 complex systematically dissected by quantitative model-guided mutagenesis. Sci Signal 2020; 13:eaay5024. [PMID: 32665413 PMCID: PMC7437921 DOI: 10.1126/scisignal.aay5024] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Because of their prominent roles in development, cancer, and HIV, the chemokine receptor CXCR4 and its ligand CXCL12 have been the subject of numerous structural and functional studies, but the determinants of ligand binding, selectivity, and signaling are still poorly understood. Here, building on our latest structural model, we used a systematic mutagenesis strategy to dissect the functional anatomy of the CXCR4-CXCL12 complex. Key charge swap mutagenesis experiments provided evidence for pairwise interactions between oppositely charged residues in the receptor and chemokine, confirming the accuracy of the predicted orientation of the chemokine relative to the receptor and providing insight into ligand selectivity. Progressive deletion of N-terminal residues revealed an unexpected contribution of the receptor N terminus to chemokine signaling. This finding challenges a longstanding "two-site" hypothesis about the essential features of the receptor-chemokine interaction in which the N terminus contributes only to binding affinity. Our results suggest that although the interaction of the chemokine N terminus with the receptor-binding pocket is the key driver of signaling, the signaling amplitude depends on the extent to which the receptor N terminus binds the chemokine. Together with systematic characterization of other epitopes, these data enable us to propose an experimentally consistent structural model for how CXCL12 binds CXCR4 and initiates signal transmission through the receptor transmembrane domain.
Collapse
Affiliation(s)
- Bryan S Stephens
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Tony Ngo
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
31
|
Pathway and mechanism of drug binding to chemokine receptors revealed by accelerated molecular simulations. Future Med Chem 2020; 12:1213-1225. [PMID: 32515227 DOI: 10.4155/fmc-2020-0044] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Background: Chemokine GPCRs play key roles in biology and medicine. Particularly, CXCR4 promotes cancer metastasis and facilitate HIV entry into host cells. Plerixafor (PLX) is a CXCR4 drug, but the pathway and binding site of PLX in CXCR4 remain unknown. Results & methodology: We have performed molecular docking and all-atom simulations using Gaussian accelerated molecular dynamics (GaMD), which are consistent with previous mutation experiments, suggesting that PLX binds to the orthosteric site of CXCR4 as an antagonist. The GaMD simulations further revealed an intermediate allosteric binding site at the extracellular mouth of CXCR4. Conclusion: The newly identified allosteric site can be targeted for novel drug design targeting CXCR4 and other chemokine receptors.
Collapse
|
32
|
Discovery of novel aminopiperidinyl amide CXCR4 modulators through virtual screening and rational drug design. Eur J Med Chem 2020; 201:112479. [PMID: 32534343 DOI: 10.1016/j.ejmech.2020.112479] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/08/2020] [Accepted: 05/16/2020] [Indexed: 12/29/2022]
Abstract
The C-X-C chemokine receptor type 4 (CXCR4) is a potential therapeutic target for HIV infection, metastatic cancer, and inflammatory autoimmune diseases. In this study, we screened the ZINC chemical database for novel CXCR4 modulators through a series of in silico guided processes. After evaluating the screened compounds for their binding affinities to CXCR4 and inhibitory activities against the chemoattractant CXCL12, we identified a hit compound (ZINC 72372983) showing 100 nM affinity and 69% chemotaxis inhibition at the same concentration (100 nM). To increase the potency of our hit compound, we explored the protein-ligand interactions at an atomic level using molecular dynamics simulation which enabled us to design and synthesize a novel compound (Z7R) with nanomolar affinity (IC50 = 1.25 nM) and improved chemotaxis inhibition (78.5%). Z7R displays promising anti-inflammatory activity (50%) in a mouse edema model by blocking CXCR4-expressed leukocytes, being supported by our immunohistochemistry study.
Collapse
|
33
|
Eckardt V, Miller MC, Blanchet X, Duan R, Leberzammer J, Duchene J, Soehnlein O, Megens RT, Ludwig AK, Dregni A, Faussner A, Wichapong K, Ippel H, Dijkgraaf I, Kaltner H, Döring Y, Bidzhekov K, Hackeng TM, Weber C, Gabius HJ, von Hundelshausen P, Mayo KH. Chemokines and galectins form heterodimers to modulate inflammation. EMBO Rep 2020; 21:e47852. [PMID: 32080959 PMCID: PMC7132340 DOI: 10.15252/embr.201947852] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 01/14/2023] Open
Abstract
Chemokines and galectins are simultaneously upregulated and mediate leukocyte recruitment during inflammation. Until now, these effector molecules have been considered to function independently. Here, we tested the hypothesis that they form molecular hybrids. By systematically screening chemokines for their ability to bind galectin‐1 and galectin‐3, we identified several interacting pairs, such as CXCL12 and galectin‐3. Based on NMR and MD studies of the CXCL12/galectin‐3 heterodimer, we identified contact sites between CXCL12 β‐strand 1 and Gal‐3 F‐face residues. Mutagenesis of galectin‐3 residues involved in heterodimer formation resulted in reduced binding to CXCL12, enabling testing of functional activity comparatively. Galectin‐3, but not its mutants, inhibited CXCL12‐induced chemotaxis of leukocytes and their recruitment into the mouse peritoneum. Moreover, galectin‐3 attenuated CXCL12‐stimulated signaling via its receptor CXCR4 in a ternary complex with the chemokine and receptor, consistent with our structural model. This first report of heterodimerization between chemokines and galectins reveals a new type of interaction between inflammatory mediators that can underlie a novel immunoregulatory mechanism in inflammation. Thus, further exploration of the chemokine/galectin interactome is warranted.
Collapse
Affiliation(s)
- Veit Eckardt
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Michelle C Miller
- Department of Biochemistry, Molecular Biology & Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, USA
| | - Xavier Blanchet
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Rundan Duan
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Julian Leberzammer
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Johan Duchene
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Oliver Soehnlein
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Remco Ta Megens
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Anna-Kristin Ludwig
- Faculty of Veterinary Medicine, Institute of Physiological Chemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Aurelio Dregni
- Department of Biochemistry, Molecular Biology & Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Faussner
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Kanin Wichapong
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Hans Ippel
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Ingrid Dijkgraaf
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Herbert Kaltner
- Faculty of Veterinary Medicine, Institute of Physiological Chemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Yvonne Döring
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Kiril Bidzhekov
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - Tilman M Hackeng
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Christian Weber
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany.,Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.,German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Hans-Joachim Gabius
- Faculty of Veterinary Medicine, Institute of Physiological Chemistry, Ludwig-Maximilians-University, Munich, Germany
| | - Philipp von Hundelshausen
- Faculty of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany.,German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Munich, Germany
| | - Kevin H Mayo
- Department of Biochemistry, Molecular Biology & Biophysics, Health Sciences Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
34
|
Bobkov V, Arimont M, Zarca A, De Groof TWM, van der Woning B, de Haard H, Smit MJ. Antibodies Targeting Chemokine Receptors CXCR4 and ACKR3. Mol Pharmacol 2019; 96:753-764. [PMID: 31481460 DOI: 10.1124/mol.119.116954] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/03/2019] [Indexed: 12/19/2022] Open
Abstract
Dysregulation of the chemokine system is implicated in a number of autoimmune and inflammatory diseases, as well as cancer. Modulation of chemokine receptor function is a very promising approach for therapeutic intervention. Despite interest from academic groups and pharmaceutical companies, there are currently few approved medicines targeting chemokine receptors. Monoclonal antibodies (mAbs) and antibody-based molecules have been successfully applied in the clinical therapy of cancer and represent a potential new class of therapeutics targeting chemokine receptors belonging to the class of G protein-coupled receptors (GPCRs). Besides conventional mAbs, single-domain antibodies and antibody scaffolds are also gaining attention as promising therapeutics. In this review, we provide an extensive overview of mAbs, single-domain antibodies, and other antibody fragments targeting CXCR4 and ACKR3, formerly referred to as CXCR7. We discuss their unique properties and advantages over small-molecule compounds, and also refer to the molecules in preclinical and clinical development. We focus on single-domain antibodies and scaffolds and their utilization in GPCR research. Additionally, structural analysis of antibody binding to CXCR4 is discussed. SIGNIFICANCE STATEMENT: Modulating the function of GPCRs, and particularly chemokine receptors, draws high interest. A comprehensive review is provided for monoclonal antibodies, antibody fragments, and variants directed at CXCR4 and ACKR3. Their advantageous functional properties, versatile applications as research tools, and use in the clinic are discussed.
Collapse
Affiliation(s)
- Vladimir Bobkov
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Marta Arimont
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Aurélien Zarca
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Timo W M De Groof
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Bas van der Woning
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Hans de Haard
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| | - Martine J Smit
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules Medicines and Systems, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (V.B., M.A., A.Z., T.W.M.D.G., M.J.S.); and argenx BVBA, Zwijnaarde, Belgium (V.B., B.W., H.H.)
| |
Collapse
|
35
|
Alves LG, Portel JF, Sousa SA, Ferreira O, Almada S, Silva ER, Martins AM, Leitão JH. Investigations into the Structure/Antibacterial Activity Relationships of Cyclam and Cyclen Derivatives. Antibiotics (Basel) 2019; 8:antibiotics8040224. [PMID: 31739454 PMCID: PMC6963676 DOI: 10.3390/antibiotics8040224] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 11/16/2022] Open
Abstract
A series of cyclam- and cyclen-derived salts are described in the present work; they were designed specifically to gain insights into their structure and antibacterial activity towards Staphylococcus aureus and Escherichia coli, used respectively, as Gram-positive and Gram-negative model organisms. The newly synthesized compounds are monosubstituted and trans-disubstituted tetraazamacrocycles that display benzyl, methylbenzyl, trifluoromethylbenzyl, or trifluoroethylbenzyl substituents appended on the nitrogen atoms of the macrocyclic ring. The results obtained show that the chemical nature, polarity, and substitution patterns of the benzyl groups, as well as the number of pendant arms, are critical parameters for the antibacterial activity of the cyclam-based salts. The most active compounds against both bacterial strains were the trans-disubstituted cyclam salts displaying CF3 groups in the para-position of the aromatic rings of the macrocyclic pendant arms. The analogous cyclen species presents a lower activity, revealing that the size of the macrocyclic backbone is an important requirement for the antibacterial activity of the tetraazamacrocycles. The nature of the anionic counterparts present on the salts was found to play a minor role in the antibacterial activity.
Collapse
Affiliation(s)
- Luis G. Alves
- Centro de Química Estrutural, Associação do Instituto Superior Técnico para a Investigação e Desenvolvimento, 1049-003 Lisboa, Portugal
- Correspondence: (L.G.A.); (J.H.L.)
| | - João F. Portel
- IBB-Instituto de Bioengenharia e Biociências, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal (S.A.S.)
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal (A.M.M.)
| | - Sílvia A. Sousa
- IBB-Instituto de Bioengenharia e Biociências, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal (S.A.S.)
| | - Olga Ferreira
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (O.F.)
- CERENA—Centro de Recursos Naturais e Ambiente, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Stephanie Almada
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal (A.M.M.)
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (O.F.)
| | - Elisabete R. Silva
- BioISI—Biosystems & Integrative Sciences Institute, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal; (O.F.)
- CERENA—Centro de Recursos Naturais e Ambiente, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal
| | - Ana M. Martins
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal (A.M.M.)
| | - Jorge H. Leitão
- IBB-Instituto de Bioengenharia e Biociências, Instituto Superior Técnico, Universidade de Lisboa, 1049-001 Lisboa, Portugal (S.A.S.)
- Correspondence: (L.G.A.); (J.H.L.)
| |
Collapse
|
36
|
C-terminal-modified LY2510924: a versatile scaffold for targeting C-X-C chemokine receptor type 4. Sci Rep 2019; 9:15284. [PMID: 31653903 PMCID: PMC6814797 DOI: 10.1038/s41598-019-51754-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 10/08/2019] [Indexed: 01/28/2023] Open
Abstract
C-X-C chemokine receptor type 4 (CXCR4) constitutes a promising target for tumor diagnosis and therapy. Herein, we evaluate a new 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA)-conjugated CXCR4 antagonist derived from LY2510924, FRM001, and its metal complexes as CXCR4-targeting probes. FRM001 was synthesized by modifying the C-terminus of LY2510924 with maleimido-mono-amide-DOTA via a cysteine linker. FRM001 exhibited CXCR4-specific binding with an affinity similar to that of the parental LY2510924. The binding affinity of FRM001 remained unchanged after complexation with Ga, Lu, and Y. The internalization of 67Ga-FRM001 into the cells was hardly observed. In mice biodistribution studies, 67Ga-FRM001 exhibited high accumulation in the tumor and the liver with rapid elimination rates from the blood. The hepatic accumulation of 67Ga-FRM001 was preferentially and significantly reduced by co-injecting a CXCR4 antagonist, AMD3100. The C-terminal-modified LY2510924 would constitute a versatile scaffold to develop CXCR4-targeting probes or therapeutics for tumor imaging or therapy.
Collapse
|
37
|
Cell-penetrating mechanism of intracellular targeting albumin: Contribution of macropinocytosis induction and endosomal escape. J Control Release 2019; 304:156-163. [DOI: 10.1016/j.jconrel.2019.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 04/16/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
|
38
|
Fazi B, Proserpio C, Galardi S, Annesi F, Cola M, Mangiola A, Michienzi A, Ciafrè SA. The Expression of the Chemokine CXCL14 Correlates with Several Aggressive Aspects of Glioblastoma and Promotes Key Properties of Glioblastoma Cells. Int J Mol Sci 2019; 20:ijms20102496. [PMID: 31117166 PMCID: PMC6566570 DOI: 10.3390/ijms20102496] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/17/2019] [Accepted: 05/19/2019] [Indexed: 02/08/2023] Open
Abstract
Glioblastoma (GBM) is a primary brain tumor whose prognosis is inevitably dismal, leading patients to death in about 15 months from diagnosis. Tumor cells in the mass of the neoplasm are in continuous exchange with cells of the stromal microenvironment, through the production of soluble molecules, among which chemokines play prominent roles. CXCL14 is a chemokine with a pro-tumor role in breast and prostate carcinoma, where it is secreted by cancer associated fibroblasts, and contributes to tumor growth and invasion. We previously observed that CXCL14 expression is higher in GBM tissues than in healthy white matter. Here, we study the effects of exogenously supplemented CXCL14 on key tumorigenic properties of human GBM cell lines. We show that CXCL14 enhances the migration ability and the proliferation of U87MG and LN229 GBM cell lines. None of these effects was affected by the use of AMD3100, an inhibitor of CXCR4 receptor, suggesting that the observed CXCL14 effects are not mediated by this receptor. We also provide evidence that CXCL14 enhances the sphere-forming ability of glioblastoma stem cells, considered the initiating cells, and is responsible for tumor onset, growth and recurrence. In support of our in vitro results, we present data from several GBM expression datasets, demonstrating that CXCL14 expression is inversely correlated with overall survival, that it is enriched at the leading edge of the tumors and in infiltrating tumor areas, and it characterizes mesenchymal and NON G-CIMP tumors, known to have a particularly bad prognosis. Overall, our results point to CXCL14 as a protumorigenic chemokine in GBM.
Collapse
Affiliation(s)
- Barbara Fazi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Carla Proserpio
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Silvia Galardi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Francesca Annesi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Mattia Cola
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Annunziato Mangiola
- Department of Neurosurgery, Università degli Studi "G. D'Annunzio", 65122 Pescara, Italy.
| | - Alessandro Michienzi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.
| | - Silvia Anna Ciafrè
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy.
| |
Collapse
|
39
|
Peters AD, McCallion C, Booth A, Adams JA, Rees-Unwin K, Pluen A, Burthem J, Webb SJ. Synthesis and biological activity of a CXCR4-targeting bis(cyclam) lipid. Org Biomol Chem 2019; 16:6479-6490. [PMID: 30155533 DOI: 10.1039/c8ob01439f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A bis(cyclam)-capped cholesterol lipid designed to bind C-X-C chemokine receptor type 4 (CXCR4) was synthesised in good overall yield from 4-methoxyphenol through a seven step synthetic route, which also provided a bis(cyclam) intermediate bearing an octaethyleneglycol-primary amine that can be easily derivatised. This bis(cyclam)-capped cholesterol lipid was water soluble and self-assembled into micellar and non-micellar aggregates in water at concentrations above 8 μM. The bioactivity of the bis(cyclam)-capped cholesterol lipid was assessed using primary chronic lymphocytic leukaemia (CLL) cells, first with a competition binding assay then with a chemotaxis assay along a C-X-C motif chemokine ligand 12 (CXCL12) concentration gradient. At 20 μM, the bis(cyclam)-capped cholesterol lipid was as effective as the commercial drug AMD3100 for preventing the migration of CLL cells, despite a lower affinity for CXCR4 than AMD3100.
Collapse
Affiliation(s)
- Anna D Peters
- School of Chemistry, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Heon Lee I, Palombo MS, Zhang X, Szekely Z, Sinko PJ. Design and evaluation of a CXCR4 targeting peptide 4DV3 as an HIV entry inhibitor and a ligand for targeted drug delivery. Eur J Pharm Biopharm 2018; 138:11-22. [PMID: 29894816 DOI: 10.1016/j.ejpb.2018.06.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Revised: 04/27/2018] [Accepted: 06/04/2018] [Indexed: 12/09/2022]
Abstract
The feasibility of utilizing the cell surface chemokine receptor CXCR4 for human immunodeficiency virus (HIV) entry inhibition and as an intracellular portal for targeted drug delivery was evaluated. Novel DV3 ligands (1DV3, 2DV3, and 4DV3) were designed, synthesized and conjugated to various probes (fluorescein isothiocyanate (FITC) or biotin) and cargos with sizes ranging from 10 to 50 nm (polyethylene glycol (PEG), streptavidin, and a polymeric nanoparticle). 4DV3 conjugated probes inhibited HIV-1 entry into the CXCR4-expressing reporter cell line TZM-bl (IC50 at 553 nM) whereas 1DV3 and 2DV3 did not. 4DV3 also inhibited binding of anti-CXCR4 antibody 44,708 to TZM-bl cells with nanomolar potency, while the small-molecule CXCR4 antagonist AMD3100 did not. Molecular modeling suggested simultaneous binding of a single 4DV3 molecule to four CXCR4 molecules. Differences in CXCR4-binding sites could explain the discrete inhibitory effects observed for 4DV3, the 44,708 antibody and AMD3100. In the Sup-T1 cell chemotaxis assay, the 4DV3 ligand functioned as a CXCR4 allosteric enhancer. In addition, 4DV3 ligand-conjugated cargos with sizes ranging from 10 to 50 nm were taken up into CXCR4-expressing Sup-T1 and TZM-bl cells, demonstrating that CXCR4 could serve as a drug delivery portal for nanocarriers. The uptake of 4DV3 functionalized nanocarriers combined with the allosteric interaction with CXCR4 suggests enhanced endocytosis occurs when 4DV3 is the targeting ligand. The current results indicate that 4DV3 might serve as a prototype for a new type of dual function ligand, one that acts as a HIV-1 entry inhibitor and a CXCR4 drug delivery targeting ligand.
Collapse
Affiliation(s)
- In Heon Lee
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd, Piscataway, NJ 08854, USA.
| | - Matthew S Palombo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd, Piscataway, NJ 08854, USA.
| | - Xiaoping Zhang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd, Piscataway, NJ 08854, USA.
| | - Zoltan Szekely
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd, Piscataway, NJ 08854, USA.
| | - Patrick J Sinko
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, 160 Frelinghuysen Rd, Piscataway, NJ 08854, USA.
| |
Collapse
|
41
|
Berg C, Daugvilaite V, Steen A, Jørgensen AS, Våbenø J, Rosenkilde MM. Inhibition of HIV Fusion by Small Molecule Agonists through Efficacy-Engineering of CXCR4. ACS Chem Biol 2018; 13:881-886. [PMID: 29461034 DOI: 10.1021/acschembio.8b00061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CXC chemokine receptor 4 (CXCR4) is involved in multiple physiological and pathological processes, notably as a coreceptor for human immunodeficiency virus (HIV) cell entry. Its broad expression pattern and vital biological importance make CXCR4 a troublesome drug target, as disruption of the interaction with its endogenous ligand, CXC chemokine ligand 12 (CXCL12), has severe consequences. In fact, only one CXCR4 drug, the bicyclam antagonist and HIV entry inhibitor AMD3100 (Plerixafor/Mozobil), has been approved for clinical use, however only for stem cell mobilization-a consequence of CXCR4 antagonism. Here, we report the engineering of an efficacy switch mutation in CXCR4-F292A7.43 in the middle of transmembrane helix 7-that converted the antagonists AMD3100 and AMD11070 into partial agonists. As agonists on F292A CXCR4, AMD3100 and AMD11070 were less disruptive to CXCR4 signaling while they remained efficient inhibitors of HIV fusion. This demonstrates that small molecule CXCR4 agonists can have a therapeutic potential as HIV entry inhibitors.
Collapse
Affiliation(s)
- Christian Berg
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
- Infectious Diseases Unit, Department of Medicine, Herlev−Gentofte Hospital, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Viktorija Daugvilaite
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Anne Steen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Astrid Sissel Jørgensen
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Jon Våbenø
- Helgeland Hospital Trust, Prestmarkveien 1, 8800 Sandnessjøen, Norway
| | - Mette Marie Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| |
Collapse
|
42
|
Peng D, Cao B, Zhou YJ, Long YQ. The chemical diversity and structure-based evolution of non-peptide CXCR4 antagonists with diverse therapeutic potential. Eur J Med Chem 2018; 149:148-169. [PMID: 29500940 DOI: 10.1016/j.ejmech.2018.02.043] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/11/2018] [Accepted: 02/13/2018] [Indexed: 12/11/2022]
Abstract
The CXC chemokine receptor 4 (CXCR4) is a highly reserved G-protein coupled 7-transmembrane (TM) chemokine receptor which consists of 352 amino acids. CXCR4 has only one endogenous chemokine ligand of CXCL12, besides several other natural nonchemokine ligands such as extracellular ubiquitin and noncognate ligand of MIF. CXCR4 strongly binds to CXCL12 and the resulting CXCLl2/CXCR4 axis is the molecular basis of their various biological functions, which include: (1) mediating immune and inflammatory response; (2) regulation of hematopoietic stem cell migration and homing; (3) an essential co-receptor for HIV entry into host cells; (4) participation in the process of embryonic development; (5) malignant tumor invasion and metastasis; (6) myocardial infarction, ischemic stroke and acute kidney injury. Correspondingly, CXCR4 antagonists find potential therapeutic applications in HIV infection, as well as hematopoietic stem cell migration, inflammation, immune-related diseases, tumor and ischemic diseases. Recently, great achievements have been made and a number of non-peptide CXCR4 antagonists with diversity scaffolds have been discovered. In this review, the discovery of small molecule CXCR4 antagonists focused on the structures, activities, evolution and development of representative CXCR4 antagonists is comprehensively described. The central role of CXCR4 in diverse cellular signaling pathways and its involvement in several diseases progressions are discussed as well.
Collapse
Affiliation(s)
- Dian Peng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Bin Cao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ying-Jun Zhou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Ya-Qiu Long
- College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou 215123, China.
| |
Collapse
|
43
|
Micallef IN, Stiff PJ, Nademanee AP, Maziarz RT, Horwitz ME, Stadtmauer EA, Kaufman JL, McCarty JM, Vargo R, Cheverton PD, Struijs M, Bolwell B, DiPersio JF. Plerixafor Plus Granulocyte Colony-Stimulating Factor for Patients with Non-Hodgkin Lymphoma and Multiple Myeloma: Long-Term Follow-Up Report. Biol Blood Marrow Transplant 2018; 24:1187-1195. [PMID: 29410180 DOI: 10.1016/j.bbmt.2018.01.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 01/26/2018] [Indexed: 12/22/2022]
Abstract
The purpose of this report is to analyze long-term clinical outcomes of patients exposed to plerixafor plus granulocyte colony-stimulating factor (G-CSF) for stem cell mobilization. This was a study of patients with non-Hodgkin lymphoma (NHL; n = 167) and multiple myeloma (MM; n = 163) who were enrolled in the long-term follow-up of 2 pivotal phase III studies (NCT00741325 and NCT00741780) of 240 µg/kg plerixafor plus 10 µg/kg G-CSF, or placebo plus 10 µg/kg G-CSF to mobilize and collect CD34+ cells for autologous hematopoietic stem cell transplantation. Overall survival (OS) and progression-free survival (PFS) were evaluated over a 5-year period following the first dose of plerixafor or placebo. The probability of OS was not significantly different in patients with NHL or MM treated with plerixafor or placebo (NHL: 64%; 95% confidence interval [CI], 56% to 71% versus 56%; 95% CI, 44% to 67%, respectively; MM: 64%; 95% CI, 54% to 72% versus 64%; 95% CI, 53% to 73%, respectively). In addition, there was no statistically significant difference in the probability of PFS over 5 years between treatment groups in patients with NHL (50%; 95% CI, 44% to 67% for plerixafor versus 43%; 95% CI, 31% to 54% for placebo) or those with MM (17%; 95% CI, 10% to 24% for plerixafor versus 30%; 95% CI, 21% to 40% for placebo). In this long-term follow-up study, the addition of plerixafor to G-CSF for stem cell mobilization did not affect 5-year survival in patients with NHL or patients with MM.
Collapse
Affiliation(s)
| | - Patrick J Stiff
- Division of Hematology/Oncology, Loyola University, Chicago, Illinois
| | | | - Richard T Maziarz
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
| | - Mitchell E Horwitz
- Division of Hematologic Malignancies and Cellular Therapy, Duke University School of Medicine, Durham, North Carolina
| | - Edward A Stadtmauer
- Division of Hematology/Oncology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jonathan L Kaufman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia
| | - John M McCarty
- Department of Internal Medicine, Virginia Commonwealth University Massey Cancer Center, Richmond, Virginia
| | | | | | | | - Brian Bolwell
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - John F DiPersio
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
44
|
Zhou Y, Yu F, Zhang F, Chen G, Wang K, Sun M, Li J, Oupický D. Cyclam-Modified PEI for Combined VEGF siRNA Silencing and CXCR4 Inhibition To Treat Metastatic Breast Cancer. Biomacromolecules 2018; 19:392-401. [PMID: 29350899 DOI: 10.1021/acs.biomac.7b01487] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chemokine receptor CXCR4 plays an important role in cancer cell invasion and metastasis. Recent findings suggest that anti-VEGF therapies upregulate CXCR4 expression, which contributes to resistance to antiangiogenic therapies. Here, we report the development of novel derivatives of polyethylenimine (PEI) that effectively inhibit CXCR4 while delivering anti-VEGF siRNA. PEI was alkylated with different amounts of a CXCR4-binding cyclam derivative to prepare PEI-C. Modification with the cyclam derivatives resulted in a considerable decrease in cytotoxicity when compared with unmodified PEI. All the PEI-C showed significant CXCR4 antagonism and the ability to inhibit cancer cell invasion. Polyplexes of PEI-C prepared with siVEGF showed effective silencing of the VEGF expression in vitro. In vivo testing in a syngeneic breast cancer model showed promising antitumor and antimetastatic activity of the PEI-C/siVEGF polyplexes. Our data demonstrate the feasibility of using PEI-C as a carrier for simultaneous VEGF silencing and CXCR4 inhibition for enhanced antiangiogenic cancer therapies.
Collapse
Affiliation(s)
- Yiwen Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , Nanjing, 210009, China
| | - Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha Nebraska 68198, United States
| | - Feiran Zhang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , Nanjing, 210009, China
| | - Gang Chen
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , Nanjing, 210009, China
| | - Kaikai Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , Nanjing, 210009, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , Nanjing, 210009, China
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha Nebraska 68198, United States
| | - David Oupický
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University , Nanjing, 210009, China.,Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center , Omaha Nebraska 68198, United States
| |
Collapse
|
45
|
Monteiro AR, Hill R, Pilkington GJ, Madureira PA. The Role of Hypoxia in Glioblastoma Invasion. Cells 2017; 6:E45. [PMID: 29165393 PMCID: PMC5755503 DOI: 10.3390/cells6040045] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), a grade IV astrocytoma, is the most common and deadly type of primary malignant brain tumor, with a patient's median survival rate ranging from 15 to 17 months. The current treatment for GBM involves tumor resection surgery based on MRI image analysis, followed by radiotherapy and treatment with temozolomide. However, the gradual development of tumor resistance to temozolomide is frequent in GBM patients leading to subsequent tumor regrowth/relapse. For this reason, the development of more effective therapeutic approaches for GBM is of critical importance. Low tumor oxygenation, also known as hypoxia, constitutes a major concern for GBM patients, since it promotes cancer cell spreading (invasion) into the healthy brain tissue in order to evade this adverse microenvironment. Tumor invasion not only constitutes a major obstacle to surgery, radiotherapy, and chemotherapy, but it is also the main cause of death in GBM patients. Understanding how hypoxia triggers the GBM cells to become invasive is paramount to developing novel and more effective therapies against this devastating disease. In this review, we will present a comprehensive examination of the available literature focused on investigating how GBM hypoxia triggers an invasive cancer cell phenotype and the role of these invasive proteins in GBM progression.
Collapse
Affiliation(s)
- Ana Rita Monteiro
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, Room 3.4, 8005-139 Faro, Portugal.
| | - Richard Hill
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| | - Geoffrey J Pilkington
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| | - Patrícia A Madureira
- Centre for Biomedical Research (CBMR), University of Algarve, Campus of Gambelas, Building 8, Room 3.4, 8005-139 Faro, Portugal.
- Brain Tumour Research Centre of Excellence, Institute of Biomedical and Biomolecular Sciences, University of Portsmouth, Portsmouth PO1 2DT, UK.
| |
Collapse
|
46
|
Alves LG, Pinheiro PF, Feliciano JR, Dâmaso DP, Leitão JH, Martins AM. Synthesis, antimicrobial activity and toxicity to nematodes of cyclam derivatives. Int J Antimicrob Agents 2017; 49:646-649. [PMID: 28315730 DOI: 10.1016/j.ijantimicag.2017.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 12/28/2016] [Accepted: 03/13/2017] [Indexed: 11/19/2022]
Abstract
The antimicrobial activity and toxicity to nematodes of the cyclam salt [H2{H2(4-CF3PhCH2)2Cyclam}](CH3COO)2⋅(CH3COOH)2 were evaluated. Estimated minimum inhibitory concentrations (MICs) of 9, 261 and 15 µg/mL were obtained for Escherichia coli, Pseudomonas aeruginosa and Staphylococcus aureus, respectively. For selected Candida spp., the estimated MICs obtained ranged from 32 µg/mL to 63 µg/mL. Bactericidal activity was demonstrated but the compound was not reliably fungicidal. Concentrations of the cyclam salt up to 32 µg/mL did not significantly affect survival of the nematode Caenorhabditis elegans; however, concentrations equal or above this value significantly affected nematode survival in a dose-dependent manner.
Collapse
Affiliation(s)
- Luis G Alves
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
| | - Pedro F Pinheiro
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal
| | - Joana R Feliciano
- iBB-Instituto de Bioengenharia e Biociências, Departamento de Bioengenharia, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, Torre Sul, Piso 6, 1049-001 Lisbon, Portugal
| | - Diana P Dâmaso
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal; iBB-Instituto de Bioengenharia e Biociências, Departamento de Bioengenharia, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, Torre Sul, Piso 6, 1049-001 Lisbon, Portugal
| | - Jorge H Leitão
- iBB-Instituto de Bioengenharia e Biociências, Departamento de Bioengenharia, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, Torre Sul, Piso 6, 1049-001 Lisbon, Portugal.
| | - Ana M Martins
- Centro de Química Estrutural, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais 1, 1049-001 Lisbon, Portugal.
| |
Collapse
|
47
|
Arimont M, Sun SL, Leurs R, Smit M, de Esch IJP, de Graaf C. Structural Analysis of Chemokine Receptor-Ligand Interactions. J Med Chem 2017; 60:4735-4779. [PMID: 28165741 PMCID: PMC5483895 DOI: 10.1021/acs.jmedchem.6b01309] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
![]()
This
review focuses on the construction and application of structural chemokine
receptor models for the elucidation of molecular determinants of chemokine
receptor modulation and the structure-based discovery and design of
chemokine receptor ligands. A comparative analysis of ligand binding
pockets in chemokine receptors is presented, including a detailed
description of the CXCR4, CCR2, CCR5, CCR9, and US28 X-ray structures,
and their implication for modeling molecular interactions of chemokine
receptors with small-molecule ligands, peptide ligands, and large
antibodies and chemokines. These studies demonstrate how the integration
of new structural information on chemokine receptors with extensive
structure–activity relationship and site-directed mutagenesis
data facilitates the prediction of the structure of chemokine receptor–ligand
complexes that have not been crystallized. Finally, a review of structure-based
ligand discovery and design studies based on chemokine receptor crystal
structures and homology models illustrates the possibilities and challenges
to find novel ligands for chemokine receptors.
Collapse
Affiliation(s)
- Marta Arimont
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Shan-Liang Sun
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Martine Smit
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam , De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
48
|
Peng L, Damschroder MM, Cook KE, Wu H, Dall'Acqua WF. Molecular basis for the antagonistic activity of an anti-CXCR4 antibody. MAbs 2016; 8:163-75. [PMID: 26514996 PMCID: PMC4966504 DOI: 10.1080/19420862.2015.1113359] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Antagonistic antibodies targeting the G-protein C-X-C chemokine receptor 4 (CXCR4) hold promising therapeutic potential in various diseases. We report for the first time the detailed mechanism of action at a molecular level of a potent anti-CXCR4 antagonistic antibody (MEDI3185). We characterized the MEDI3185 paratope using alanine scanning on all 6 complementary-determining regions (CDRs). We also mapped its epitope using CXCR4 mutagenesis to assess the relative importance of the CXCR4 N-terminal peptide, extracellular loops (ECL) and ligand-binding pocket. We show that the interaction between MEDI3185 and CXCR4 is mediated mostly by CDR3H in MEDI3185 and ECL2 in CXCR4. The MEDI3185 epitope comprises the entire ECL2 sequence, lacks any so-called ‘hot-spot’ and is remarkably resistant to mutations. The structure of MEDI3185 variable domains was modeled, and suggested a β-strand/β-strand interaction between MEDI3185 CDR3H and CXCR4 ECL2, resulting in direct steric hindrance with CXCR4 ligand SDF-1. These findings may have important implications for designing antibody therapies against CXCR4.
Collapse
Affiliation(s)
- Li Peng
- a Department of Antibody Discovery and Protein Engineering ; MedImmune LLC; One MedImmune Way ; Gaithersburg , MD 20878 , USA
| | - Melissa M Damschroder
- a Department of Antibody Discovery and Protein Engineering ; MedImmune LLC; One MedImmune Way ; Gaithersburg , MD 20878 , USA
| | - Kimberly E Cook
- a Department of Antibody Discovery and Protein Engineering ; MedImmune LLC; One MedImmune Way ; Gaithersburg , MD 20878 , USA
| | - Herren Wu
- a Department of Antibody Discovery and Protein Engineering ; MedImmune LLC; One MedImmune Way ; Gaithersburg , MD 20878 , USA
| | - William F Dall'Acqua
- a Department of Antibody Discovery and Protein Engineering ; MedImmune LLC; One MedImmune Way ; Gaithersburg , MD 20878 , USA
| |
Collapse
|
49
|
Maples RD, Cain AN, Burke BP, Silversides JD, Mewis RE, D'huys T, Schols D, Linder DP, Archibald SJ, Hubin TJ. Aspartate-Based CXCR4 Chemokine Receptor Binding of Cross-Bridged Tetraazamacrocyclic Copper(II) and Zinc(II) Complexes. Chemistry 2016; 22:12916-30. [PMID: 27458983 PMCID: PMC5224883 DOI: 10.1002/chem.201601468] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Indexed: 12/22/2022]
Abstract
The CXCR4 chemokine receptor is implicated in a number of diseases including HIV infection and cancer development and metastasis. Previous studies have demonstrated that configurationally restricted bis-tetraazamacrocyclic metal complexes are high-affinity CXCR4 antagonists. Here, we present the synthesis of Cu(2+) and Zn(2+) acetate complexes of six cross-bridged tetraazamacrocycles to mimic their coordination interaction with the aspartate side chains known to bind them to CXCR4. X-ray crystal structures for three new Cu(2+) acetate complexes and two new Zn(2+) acetate complexes demonstrate metal-ion-dependent differences in the mode of binding the acetate ligand concomitantly with the requisite cis-V-configured cross-bridged tetraazamacrocyle. Concurrent density functional theory molecular modelling studies produced an energetic rationale for the unexpected [Zn(OAc)(H2 O)](+) coordination motif present in all of the Zn(2+) cross-bridged tetraazamacrocycle crystal structures, which differs from the chelating acetate [Zn(OAc)](+) structures of known unbridged and side-bridged tetraazamacrocyclic Zn(2+) -containing CXCR4 antagonists.
Collapse
Affiliation(s)
- Randall D Maples
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK, 73096, USA
| | - Amy N Cain
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK, 73096, USA
| | - Benjamin P Burke
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Hull, HU6 7RX, UK
| | - Jon D Silversides
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Hull, HU6 7RX, UK
| | - Ryan E Mewis
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Hull, HU6 7RX, UK
| | - Thomas D'huys
- Rega Institute for Medical Research, KU Leuven, 3000, Leuven, Belgium
| | - Dominique Schols
- Rega Institute for Medical Research, KU Leuven, 3000, Leuven, Belgium
| | - Douglas P Linder
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK, 73096, USA
| | - Stephen J Archibald
- Department of Chemistry and Positron Emission Tomography Research Centre, University of Hull, Hull, HU6 7RX, UK.
| | - Timothy J Hubin
- Department of Chemistry and Physics, Southwestern Oklahoma State University, Weatherford, OK, 73096, USA.
| |
Collapse
|
50
|
Yu F, Li J, Xie Y, Sleightholm RL, Oupický D. Polymeric chloroquine as an inhibitor of cancer cell migration and experimental lung metastasis. J Control Release 2016; 244:347-356. [PMID: 27473763 DOI: 10.1016/j.jconrel.2016.07.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 01/16/2023]
Abstract
Chloroquine (CQ) is a widely used antimalarial drug with emerging potential in anticancer therapies due to its apparent inhibitory effects on CXCR4 chemokine receptor, autophagy, and cholesterol metabolism. This study reports on polymeric CQ (pCQ) as a macromolecular drug with antimetastatic activity. The pCQ polymers were synthesized by copolymerization of methacryloylated hydroxy-CQ (HCQ) and N-(2-hydroxypropyl)methacrylamide (HPMA). The results show that pCQ is significantly more effective in inhibiting cancer cell migration and invasion when compared with the parent HCQ. The proposed mechanism of action at least partially relies on the ability of pCQ to inhibit cell migration mediated by the CXCR4/CXCL12 pathway. The pCQ also demonstrates superior inhibitory activity over HCQ when tested in a mouse model of experimental lung metastasis. Lastly, pCQ shows the ability to efficiently translocate to the cytoplasm while exhibiting lower cytotoxicity than HCQ. Overall, this study supports pCQ as a promising polymeric drug platform suitable for use in combination antimetastatic strategies and potential use in cytoplasmic drug delivery.
Collapse
Affiliation(s)
- Fei Yu
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jing Li
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ying Xie
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Richard L Sleightholm
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|