22 results on '"Simon P. Fricker"'
Search Results
2. Chemokine receptor modeling: an interdisciplinary approach to drug design
- Author
-
Markus Metz and Simon P. Fricker
- Subjects
Pharmacology ,CCR1 ,Binding Sites ,biology ,Chemokine receptor CCR5 ,HIV Infections ,C-C chemokine receptor type 7 ,C-C chemokine receptor type 6 ,Protein Structure, Tertiary ,Cell biology ,Molecular Docking Simulation ,Chemokine receptor ,HIV Fusion Inhibitors ,Drug Design ,Drug Discovery ,biology.protein ,Humans ,Molecular Medicine ,Receptors, Chemokine ,CCR10 ,CXC chemokine receptors ,Chemokines ,CCL21 - Abstract
Chemokines and their receptors are integral components of the immune response, regulating lymphocyte development, homing and trafficking, and playing a key role in the pathophysiology of many diseases. Chemokine receptors have, therefore, become the target for both small-molecule, peptide and antibody therapeutics. Chemokine receptors belong to the family of seven transmembrane receptor class A G protein-coupled receptors. The publication of the crystal structure of the archetypal class A seven transmembrane receptor protein rhodopsin, and other G protein-coupled receptors, including C-X-C chemokine receptor 4 and C-C chemokine receptor 5, provided the opportunity to create homology models of chemokine receptors. In this review, we describe an interdisciplinary approach to chemokine receptor modeling and the utility of this approach for structure-based drug design of chemokine receptor inhibitors.
- Published
- 2014
- Full Text
- View/download PDF
3. The molecular pharmacology of AMD11070: An orally bioavailable CXCR4 HIV entry inhibitor
- Author
-
Dana Huskens, Marilyn C. Darkes, Gary Bridger, Jennifer H. Cox, Simon P. Fricker, Zefferino Santucci, Renee Mosi, Jean Labrecque, Kim L. Nelson, Renato Skerlj, Stefan R. Idzan, Virginia Anastassova, Dominique Schols, Ketan Patel, Rebecca S.Y. Wong, and Gloria Lau
- Subjects
CCR1 ,Receptors, CXCR4 ,Anti-HIV Agents ,Allosteric regulation ,Administration, Oral ,Biological Availability ,C-C chemokine receptor type 7 ,Pharmacology ,Biology ,Butylamines ,Virus Replication ,CXCR3 ,Biochemistry ,Cell Line ,Heterocyclic Compounds, 1-Ring ,Dogs ,Calcium flux ,medicine ,Animals ,Humans ,Receptor ,Molecular Structure ,Ligand binding assay ,Virus Internalization ,Chemokine CXCL12 ,Entry inhibitor ,Gene Expression Regulation ,Aminoquinolines ,HIV-1 ,Benzimidazoles ,Protein Binding ,Signal Transduction ,medicine.drug - Abstract
In order to enter and infect human cells HIV must bind to CD4 in addition to either the CXCR4 or the CCR5 chemokine receptor. AMD11070 was the first orally available small molecule antagonist of CXCR4 to enter the clinic. Herein we report the molecular pharmacology of AMD11070 which is a potent inhibitor of X4 HIV-1 replication and the gp120/CXCR4 interaction. Using the CCRF-CEM T cell line that endogenously expresses CXCR4 we have demonstrated that AMD11070 is an antagonist of SDF-1α ligand binding (IC50 = 12.5 ± 1.3 nM), inhibits SDF-1 mediated calcium flux (IC50 = 9.0 ± 2.0 nM) and SDF-1α mediated activation of the CXCR4 receptor as measured by a Eu-GTP binding assay (IC50 =39.8 ± 2.5 nM) or a [(35)S]-GTPγS binding assay (IC50 =19.0 ± 4.1 nM), and inhibits SDF-1α stimulated chemotaxis (IC50 =19.0 ± 4.0 nM). AMD11070 does not inhibit calcium flux of cells expressing CXCR3, CCR1, CCR2b, CCR4, CCR5 or CCR7, or ligand binding to CXCR7 and BLT1, demonstrating selectivity for CXCR4. In addition AMD11070 is able to inhibit the SDF-1β isoform interactions with CXCR4; and N-terminal truncated variants of CXCR4 with equal potency to wild type receptor. Further mechanistic studies indicate that AMD11070 is an allosteric inhibitor of CXCR4.
- Published
- 2012
- Full Text
- View/download PDF
4. Strategies for the Biological Evaluation of Gold Anticancer Agents
- Author
-
Simon P. Fricker
- Subjects
Models, Molecular ,Pharmacology ,Cancer Research ,Auranofin ,business.industry ,Drug Evaluation, Preclinical ,Cancer therapy ,Antineoplastic Agents ,Integrated approach ,Approved drug ,Gold Compounds ,Biological Testing ,In vivo ,Neoplasms ,medicine ,Animals ,Humans ,Molecular Medicine ,business ,Organogold Compounds ,medicine.drug ,Biological evaluation - Abstract
Since the introduction of the monomeric orally bioavailable anti-arthritic gold compound auranofin in 1985, and the success of the platinum-based anti-cancer drugs, there has been a great deal of interest in the use of gold compounds for cancer therapy. However this early promise has not materialized into an approved drug in spite of extensive and innovative efforts in gold chemistry. Therefore, in the light of this lack of success, the strategies for the biological evaluation of potential gold-based anti-cancer drugs are discussed. It is proposed that the biological testing strategy should be multi-faceted incorporating an understanding of the molecular properties of the compounds under investigation related to their behaviour in a biological environment, an evaluation of their comparative in vitro potency against tumor cells, ascertaining the biochemical mechanism of action and target identification to aid in medicinal chemistry design, evaluation of in vivo activity in relevant tumor models, and an understanding of their toxicological and pharmacokinetic properties. This strategy will be exemplified with work on Au(III) cyclometallated complexes in which an integrated approach to the search for new metal-based anticancer drugs was adopted, incorporating in vitro screening, in vivo human tumor xenograft models, and mechanistic studies. The importance of mechanistic studies which have led to the identification of new molecular targets for gold drugs, and in vivo evaluation are emphasized.
- Published
- 2011
- Full Text
- View/download PDF
5. Prospective CCR5 Small Molecule Antagonist Compound Design Using a Combined Mutagenesis/Modeling Approach
- Author
-
Zefferino Santucci, Gary Bridger, Gloria Lau, Dominique Schols, Elyse Bourque, Wen Yang, Simon P. Fricker, Markus Metz, Curtis Harwig, Marilyn C. Darkes, Renato T. Skerlj, Jonathan Langille, Jean Labrecque, and Sanjay Danthi
- Subjects
Models, Molecular ,Anti-HIV Agents ,Stereochemistry ,hERG ,Allosteric regulation ,Mutagenesis (molecular biology technique) ,Microbial Sensitivity Tests ,Computational biology ,CCR5 receptor antagonist ,Biochemistry ,Catalysis ,Structure-Activity Relationship ,Colloid and Surface Chemistry ,medicine ,Humans ,Urea ,Structure–activity relationship ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,Chemistry ,Stereoisomerism ,General Chemistry ,Small molecule ,Ether-A-Go-Go Potassium Channels ,Entry inhibitor ,Molecular Weight ,Mutagenesis ,Drug Design ,CCR5 Receptor Antagonists ,HIV-1 ,Leukocytes, Mononuclear ,biology.protein ,Pharmacophore ,medicine.drug - Abstract
The viral resistance of marketed antiviral drugs including the emergence of new viral resistance of the only marketed CCR5 entry inhibitor, maraviroc, makes it necessary to develop new CCR5 allosteric inhibitors. A mutagenesis/modeling approach was used (a) to remove the potential hERG liability in an otherwise very promising series of compounds and (b) to design a new class of compounds with an unique mutant fingerprint profile depending on residues in the N-terminus and the extracellular loop 2. On the basis of residues, which were identified by mutagenesis as key interaction sites, binding modes of compounds were derived and utilized for compound design in a prospective manner. The compounds were then synthesized, and in vitro evaluation not only showed that they had good antiviral potency but also fulfilled the requirement of low hERG inhibition, a criterion necessary because a potential approved drug would be administered chronically. This work utilized an interdisciplinary approach including medicinal chemistry, molecular biology, and computational chemistry merging the structural requirements for potency with the requirements of an acceptable in vitro profile for allosteric CCR5 inhibitors. The obtained mutant fingerprint profiles of CCR5 inhibitors were used to translate the CCR5 allosteric binding site into a general pharmacophore, which can be used for discovering new inhibitors.
- Published
- 2011
- Full Text
- View/download PDF
6. Inhibition of the cathepsin cysteine proteases B and K by square-planar cycloaurated gold(III) compounds and investigation of their anti-cancer activity
- Author
-
Ling Qin, Renato Skerlj, Yongbao Zhu, Zefferino Santucci, Renee Mosi, Jennifer H. Cox, Simon P. Fricker, Virginia Anastassov, Beth R. Cameron, and Markus Metz
- Subjects
Male ,Cathepsin ,Chemistry ,Stereochemistry ,Cathepsin K ,Antineoplastic Agents ,Biological activity ,Mice, SCID ,Cysteine Proteinase Inhibitors ,Biochemistry ,Cysteine protease ,Cathepsin B ,Inorganic Chemistry ,Mice ,Cathepsin O ,Cysteine Proteases ,Cell Line, Tumor ,Animals ,Humans ,Gold ,Cysteine - Abstract
Gold(III) compounds have been examined for potential anti-cancer activity. It is proposed that the molecular targets of these compounds are thiol-containing biological molecules such as the cathepsin cysteine proteases. These enzymes have been implicated in many diseases including cancer. The catalytic mechanism of the cathepsin cysteine proteases is dependent upon a cysteine at the active site which is accessible to the interaction of thiophilic metals such as gold. The synthesis and biological activity of square-planar six-membered cycloaurated Au(III) compounds with a pyridinyl-phenyl linked backbone and two monodentate or one bidentate leaving group is described. Gold(III) cycloaurated compounds were able to inhibit both cathepsins B and K. Structure/activity was investigated by modifications to the pyridinyl-phenyl backbone, and leaving groups. Optimal activity was seen with substitution at the 6 position of the pyridine ring. The reversibility of inhibition was tested by reactivation in the presence of cysteine with a bidentate thiosalicylate compound being an irreversible inhibitor. Five compounds were evaluated for in vitro cytotoxicity against a panel of human tumor cell lines. The thiosalicylate compound was tested in vivo against the HT29 human colon tumor xenograft model. A modest decrease in tumor growth was observed compared with the untreated control tumor.
- Published
- 2011
- Full Text
- View/download PDF
7. CXCL12 (SDF-1)/CXCR4 Pathway in Cancer
- Author
-
Simon P. Fricker and Beverly A. Teicher
- Subjects
Receptors, CXCR4 ,Cancer Research ,Chemokine ,CXCR4 Inhibitor ,biology ,Chemotaxis ,Chemokine CXCL12 ,Cell biology ,Drug Delivery Systems ,Oncology ,Neoplasms ,Cancer cell ,biology.protein ,Humans ,CXC chemokine receptors ,Signal transduction ,CXCL14 ,Intracellular ,Signal Transduction - Abstract
Chemokines, small proinflammatory chemoattractant cytokines that bind to specific G-protein-coupled seven-span transmembrane receptors, are major regulators of cell trafficking and adhesion. The chemokine CXCL12 [stromal cell-derived factor-1 (SDF-1)] binds primarily to CXC receptor 4 (CXCR4; CD184). The binding of CXCL12 to CXCR4 induces intracellular signaling through several divergent pathways initiating signals related to chemotaxis, cell survival and/or proliferation, increase in intracellular calcium, and gene transcription. CXCR4 is expressed on multiple cell types including lymphocytes, hematopoietic stem cells, endothelial and epithelial cells, and cancer cells. The CXCL12/CXCR4 axis is involved in tumor progression, angiogenesis, metastasis, and survival. This pathway is a target for therapeutics that can block the CXCL12/CXCR4 interaction or inhibit downstream intracellular signaling. Clin Cancer Res; 16(11); 2927–31. ©2010 AACR.
- Published
- 2010
- Full Text
- View/download PDF
8. Metal compounds for the treatment of parasitic diseases
- Author
-
Patricia S. Doyle, Zefferino Santucci, Gloria Lau, Ling Qin, Elizabeth Hansell, Jonathan Langille, Youngbao Zhu, Simon P. Fricker, Virginia Anastassov, Renato Skerlj, Beth R. Cameron, Micki Olsen, Jennifer H. Cox, Ian Baird, James H. McKerrow, Rebecca S.Y. Wong, and Renee Mosi
- Subjects
Trypanosoma ,Proteases ,Stereochemistry ,chemistry.chemical_element ,Cysteine Proteinase Inhibitors ,Biochemistry ,Cathepsin B ,Rhodium ,Inorganic Chemistry ,Metal ,Animals ,Humans ,Chagas Disease ,Osmium ,Leishmaniasis ,Leishmania ,Chemistry ,Trypanocidal Agents ,Cysteine protease ,Cysteine Endopeptidases ,Inorganic Chemicals ,Metals ,visual_art ,visual_art.visual_art_medium ,Cysteine ,Palladium - Abstract
The cysteine proteases of the trypanosomatid parasitic protozoa have been validated as targets for chemotherapy of Chagas' disease and leishmaniasis. Metal complexes of gold, platinum, iridium, palladium, rhodium and osmium have been reported to have activity against a variety of trypanosomatids, but the molecular target of these compounds has not been defined. The activity of gold(III) and palladium(II) cyclometallated complexes, and oxorhenium(V) complexes against mammalian and parasitic cysteine proteases was investigated. All gold(III) complexes (1-6) inhibited cathepsin B with IC(50) values in the range of 0.2-1.4 microM. Of the six palladium compounds, aceto[2,6-bis[(butylthio-kappa S)methyl]phenyl-kappa C]-, (SP-4-3)-palladium(II) (11) was the most potent inhibitor of cathepsin B with an IC(50) of 0.4 microM. A clear structure-activity relationship was observed with the oxorhenium(V) complexes with chloro[2,2'-(thio-kappa S)bis[ethanethiolato-kappa S)]] oxorhenium(V) (16) being the most potent inhibitor of cathepsin B with an IC(50) of 0.009 microM. Six complexes were further tested against the parasite cysteine proteases, cruzain from T. cruzi, and cpB from L. major; the most potent inhibitors were the two rhenium complexes (2(1H)-pyridinethionato-kappa S(2))[2,6-bis[(mercapto-kappa S)methyl]pyridine-kappa N(1)] oxorhenium(V) (15) and chloro[2,2'-(thio-kappa S)bis[ethanethiolato-kappa S)]] oxorhenium(V) (16). The compounds were also evaluated in assays for parasite growth. Two oxorhenium(V) compounds ((p-methoxyphenylthiolato-S)[2,6-bis[(mercapto-kappa S)methyl]pyridine-kappa N(1)] oxorhenium(V) (14) and (methanethiolato)[2,2'-(thio-kappa S)bis[ethanethiolato-kappa S)]] oxorhenium (V) (18)) and the palladium compound 11 inhibited T. cruzi intracellular growth, and compound 11 inhibited promastigote growth in three Leishmania species. In conclusion this preliminary data indicates that metal complexes targeted at parasite cysteine proteases show promise for the treatment of both Chagas' disease and leishmaniasis.
- Published
- 2008
- Full Text
- View/download PDF
9. Characterization of the molecular pharmacology of AMD3100: A specific antagonist of the G-protein coupled chemokine receptor, CXCR4
- Author
-
Ling Qin, Rebecca Wong, Virginia Anastassov, Simon P. Fricker, Jennifer H. Cox, Renee Mosi, Stefan R. Idzan, Jean Labrecque, Kim L. Nelson, Ognjen Grujic, Gloria Lau, Marilyn C. Darkes, and Zeffy Santucci
- Subjects
CCR1 ,Benzylamines ,Receptors, CXCR4 ,Chemokine receptor CCR5 ,C-C chemokine receptor type 7 ,C-C chemokine receptor type 6 ,Cyclams ,Biochemistry ,Cell Line ,Chemokine receptor ,Heterocyclic Compounds ,Humans ,CCL17 ,CXCL14 ,Pharmacology ,biology ,Chemotaxis ,Chemokine CXCL12 ,Cell biology ,biology.protein ,Cancer research ,XCL2 ,Calcium ,Chemokines, CXC ,Protein Binding ,Signal Transduction - Abstract
The chemokine receptor CXCR4 is widely expressed on different cell types, is involved in leukocyte chemotaxis, and is a co-receptor for HIV. AMD3100 has been shown to be a CXCR4 receptor antagonist, and to block HIV infection of T-tropic, X4-using, virus in vitro and in vivo. AMD3100 is an effective mobilizer of hematopoietic stem cells and is being investigated in clinical trials in multiple myeloma and non-Hodgkins lymphoma patients. Using the CCRF-CEM T-cell line that constitutively expresses CXCR4 we confirmed that AMD3100 was an antagonist of SDF-1/CXCL12 ligand binding (IC50=651+/-37 nM). We have also shown that AMD3100 inhibits SDF-1 mediated GTP-binding (IC50=27+/-2.2 nM), SDF-1 mediated calcium flux (IC50=572+/-190 nM), and SDF-1 stimulated chemotaxis (IC50=51+/-17 nM). AMD3100 did not inhibit calcium flux against cells expressing CXCR3, CCR1, CCR2b, CCR4, CCR5 or CCR7 when stimulated with their cognate ligands, nor did it inhibit receptor binding of LTB4. AMD3100 did not, on its own, induce a calcium flux in the CCRF-CEM cells, which express multiple GPCRs including CXCR4, CCR4 and CCR7. Furthermore, AMD3100 neither stimulated GTP-binding, an assay for GPCR activation, in CEM cell membranes; nor chemotaxis of CCRF-CEM cells. These data therefore demonstrate that AMD3100 is a specific antagonist of CXCR4, is not cross-reactive with other chemokine receptors, and is not an agonist of CXCR4.
- Published
- 2006
- Full Text
- View/download PDF
10. Rhenium Inhibitors of Cathepsin B (ReO(SYS)X (Where Y = S, py; X = Cl, Br, SPhOMe-p)): Synthesis and Mechanism of Inhibition
- Author
-
Simon P. Fricker, Virginia Anastassov, Beth R. Cameron, Ian R. Baird, Renee Mosi, Jennifer H. Cox, and Renato T. Skerlj
- Subjects
Time Factors ,Stereochemistry ,Cathepsin K ,chemistry.chemical_element ,Cysteine Proteinase Inhibitors ,Ligands ,Chemical synthesis ,Cathepsin B ,Structure-Activity Relationship ,chemistry.chemical_compound ,Bromide ,Drug Discovery ,Organometallic Compounds ,Humans ,Cathepsin ,Binding Sites ,Molecular Structure ,Ligand ,Leaving group ,Stereoisomerism ,Rhenium ,Cathepsins ,chemistry ,Molecular Medicine ,Selectivity - Abstract
The synthesis of four new oxorhenium(V) complexes containing the “3 + 1” mixed-ligand donor set, ReO(SYS)X (where Y = S, py; X = Cl, Br), is described. All of the complexes tested exhibited selectivity for cathepsin B over K. Most notably, compound 6, ReO(SSS-2,2‘)Br (IC50(cathepsin B) = 1.0 nM), was 260 times more potent against cathepsin B. It was also discovered that complexes containing the same tridentate (SSS) ligand were more potent when the leaving group was bromide versus chloride (e.g., IC50(cathepsin B): ReO(SSS-2,2‘)Cl (4), 8.8 nM; ReO(SSS-2,2‘)Br (6), 1.0 nM). Mechanistic studies with cathepsin B showed that both compounds 2 (ReO(SpyS)(SPhOMe-p)) and 4 were active-site-directed. Compound 2 was determined to be a tight-binding, reversible inhibitor, while compound 4 was a time-dependent, slowly reversible inhibitor. The results described in this paper show that the oxorhenium(V) “3 + 1” complexes are potent, selective inhibitors of cathepsin B and have potential for the treatment of cancer.
- Published
- 2006
- Full Text
- View/download PDF
11. Ruthenium as an Effective Nitric Oxide Scavenger
- Author
-
Clodagh Mulcahy, Beth R. Cameron, Simon P. Fricker, and Celine J. Marmion
- Subjects
Models, Molecular ,chemistry.chemical_classification ,Antineoplastic Agents ,Biological activity ,Endogeny ,Free Radical Scavengers ,General Medicine ,Metabolism ,Biology ,Nitric Oxide ,Models, Biological ,Ruthenium ,Scavenger (chemistry) ,Nitric oxide ,Pathogenesis ,chemistry.chemical_compound ,Enzyme ,Biosynthesis ,chemistry ,Biochemistry ,Drug Discovery ,Organometallic Compounds ,Humans - Abstract
Whilst nitric oxide (NO) has emerged as one of the most versatile and ubiquitous molecules in the human body with a diverse range of physiological functions, dysfunction in NO biosynthesis or metabolism has led to the pathogenesis of a number of disease states. A variety of therapeutic strategies have therefore emerged that either reduce or increase endogenous NO levels depending on the disease pathology. The predominant strategy to date to reduce levels of NO is to utilise specific isoform selective inhibitors of nitric oxide synthases, the enzymes responsible for NO biosynthesis. An alternative line of attack, not r elate d to specificity for a particular enzyme, but rather on compartmental localisation and pharmacokinetics, is to re move or sc ave nge the exce ss NO re sponsible for the dise ase pathology. In this regard, a number of NO scavenger molecules have demonstrated pharmacological activity across a broad spectrum of disease states. This review will highlight the rationale behind the development, and the current state of play, of one such class of NO scavengers, complexes of the d-block transition metal ruthenium. Prior to this, a brief overview of the remarkable diversity of NO, both from a chemical and biological viewpoint, will be provided for perspective.
- Published
- 2004
- Full Text
- View/download PDF
12. Design of substituted imidazolidinylpiperidinylbenzoic acids as chemokine receptor 5 antagonists: potent inhibitors of R5 HIV-1 replication
- Author
-
Curtis Harwig, Dominique Schols, Gang Chen, Wen Yang, Duane Veale, Susan Nan, Simon P. Fricker, Alan Kaller, Danielle Guay, Jason M. Crawford, Michael Satori, Markus Metz, Yongbao Zhu, Helen Yee, Maria Krumpak, Elyse Bourque, Wilson Trevor R, David Bogucki, Bryon Carpenter, Michael Bey, David C. Leitch, Ron MacFarland, David Gauthier, Ian Baird, Rebecca Wong, Renato T. Skerlj, Jonathan Langille, Yuanxi Zhou, Gary Bridger, Renee Mosi, Ernest J. McEachern, Tuya Ba, Tong-Shuang Li, Krystyna Vocadlo, and Veronique Bodart
- Subjects
Receptors, CCR5 ,Anti-HIV Agents ,Cell Survival ,hERG ,Human immunodeficiency virus (HIV) ,CHO Cells ,Pharmacology ,medicine.disease_cause ,Imidazolidines ,Virus Replication ,Benzoates ,Chemokine receptor ,Structure-Activity Relationship ,Cricetulus ,Piperidines ,In vivo ,Cricetinae ,Drug Discovery ,medicine ,Animals ,Humans ,Cells, Cultured ,Cardiotoxicity ,biology ,Molecular Structure ,Chemistry ,Imidazoles ,In vitro ,Receptor selectivity ,HEK293 Cells ,Biochemistry ,Models, Chemical ,Drug Design ,Lipophilicity ,CCR5 Receptor Antagonists ,Host-Pathogen Interactions ,biology.protein ,HIV-1 ,Leukocytes, Mononuclear ,Molecular Medicine - Abstract
The redesign of the previously reported thiophene-3-yl-methyl urea series, as a result of potential cardiotoxicity, was successfully accomplished, resulting in the identification of a novel potent series of CCR5 antagonists containing the imidazolidinylpiperidinyl scaffold. The main redesign criteria were to reduce the number of rotatable bonds and to maintain an acceptable lipophilicity to mitigate hERG inhibition. The structure–activity relationship (SAR) that was developed was used to identify compounds with the best pharmacological profile to inhibit HIV-1. As a result, five advanced compounds, 6d, 6e, 6i, 6h, and 6k, were further evaluated for receptor selectivity, antiviral activity against CCR5 using (R5) HIV-1 clinical isolates, and in vitro and in vivo safety. On the basis of these results, 6d and 6h were selected for further development.
- Published
- 2013
13. Antitumor Properties of Some 2-[(Dimethylamino)methyl]phenylgold(III) Complexes
- Author
-
Lloyd R. Kelland, Simon P. Fricker, Brian P. Howe, Robert G. Buckley, Brian R. C. Theobald, Amanda M. Elsome, Richard V. Parish, and Graham R. Henderson
- Subjects
Benzylamines ,Magnetic Resonance Spectroscopy ,Stereochemistry ,Mice, Nude ,Antineoplastic Agents ,Flow cytometry ,Mice ,In vivo ,Ovarian carcinoma ,Drug Discovery ,Organometallic Compounds ,Tumor Cells, Cultured ,medicine ,Animals ,Humans ,Mode of action ,Cisplatin ,medicine.diagnostic_test ,Chemistry ,Flow Cytometry ,Molecular biology ,In vitro ,Mechanism of action ,Cell culture ,Molecular Medicine ,Drug Screening Assays, Antitumor ,medicine.symptom ,Organogold Compounds ,Cell Division ,Neoplasm Transplantation ,medicine.drug - Abstract
Four analogues of the gold(III) complex [AuCl2(damp)] (1) (damp = 2-[(dimethylamino)methyl]phenyl) have been evaluated for antitumor activity. The compounds have structural features in common with cisplatin which was included as a comparison in the study. In vitro, against a panel of cell lines established from tumors of different tissue types, the gold complexes showed broadly similar growth inhibitory properties with some selectivity to the HT1376 bladder cell line. In a panel of human ovarian carcinoma cell lines, non-cross-resistance to cisplatin was observed, for the complexes, in an acquired cisplatin-resistant line. In vivo, using subcutaneously implanted xenografts derived from the HT1376 bladder and CH1 ovarian cell lines, [Au(acetato)2(damp)] (3) and [Au(malonato)(damp)] (5) (administered intraperitoneally) gave significant tumor inhibition. Mechanistic studies performed with compound 3 showed marked differences to cisplatin. Thus, much higher concentrations of the gold compound were required to affect Col E1 plasmid mobility, and an alkaline elution study showed that 3 did not cause interstrand DNA cross-links in SK-OV-3 cells. Exposure of SK-OV-3 cells to 3 induced only relatively minor changes in cell cycle distribution. Furthermore 3 was only marginally active in vivo against the cisplatin-sensitive murine ADJ/PC6 plasmacytoma. In summary, the gold-(III) complexes 3 and 5 exhibited selective cytotoxicity in vitro and showed in vivo antitumor activity against human carcinoma xenografts. Also, although 3 has some structural similarity to cisplatin, its mode of action appears to be different.
- Published
- 1996
- Full Text
- View/download PDF
14. Cysteine proteases as targets for metal-based drugs
- Author
-
Simon P. Fricker
- Subjects
chemistry.chemical_classification ,Cathepsin ,Proteases ,biology ,Thioredoxin reductase ,Metals and Alloys ,Biophysics ,Active site ,Antineoplastic Agents ,Biochemistry ,Deubiquitinating enzyme ,Biomaterials ,Enzyme ,Drug Delivery Systems ,chemistry ,Chemistry (miscellaneous) ,Coordination Complexes ,Cysteine Proteases ,biology.protein ,Humans ,Cisplatin ,Caspase ,Cysteine - Abstract
The discovery of the platinum anticancer drug cisplatin provided a major stimulus for research into metal-based drugs. The molecular target for the platinum agents is DNA; however recent developments in inorganic medicinal chemistry have identified several alternative novel targets for metal-based drugs. Biological molecules with essential thiol groups are attractive targets. Thiol-containing molecular targets include the redox enzymes thioredoxin reductase and glutathione reductase, transcription factors, and cysteine proteases such as caspases and cathepsins. Inorganic chemistry offers many opportunities for medicinal chemistry, and alternative targets for metal-based drugs are reviewed, with a focus on cysteine proteases. The cathepsin cysteine proteases have numerous physiological functions, and have been implicated in diseases including cancer, autoimmune and inflammatory, and parasitic diseases. The catalytic mechanism of these enzymes is dependent upon a cysteine at the active site. We postulate that metal complexes can inhibit these enzymes via a ligand substitution with the thiol of the active site cysteine. We have investigated several classes of metal complexes including cyclometalated organo gold(III) and Pd(II) complexes, and a series of rhenium(V) mixed ligand oxorhenium complexes as inhibitors of cathepsin cysteine proteases. Mechanistic studies were conducted on the latter supporting the hypothesis of active site-directed inhibition. These data are reviewed below and discussed in the context of possible therapeutic applications including cancer and parasitic disease.
- Published
- 2010
15. A time-resolved fluorescent lanthanide (Eu)-GTP binding assay for chemokine receptors as targets in drug discovery
- Author
-
Jean, Labrecque, Rebecca S Y, Wong, and Simon P, Fricker
- Subjects
Radioligand Assay ,Receptors, CXCR4 ,Leukemia, T-Cell ,Europium ,Cell Membrane ,Drug Discovery ,Tumor Cells, Cultured ,Humans ,Guanosine Triphosphate - Abstract
Chemokines are a family of chemoattractant cytokines involved in leukocyte trafficking, activation, development, and hematopoeisis. Chemokines and their receptors have been implicated in several disease processes, particularly inflammatory and autoimmune disorders and cancer, and are therefore attractive targets for drug development. Chemokine receptors are members of the seven-transmembrane, G protein-coupled receptor (GPCR) family. As such they can be studied using GPCR assays such as ligand binding, G protein activation, and downstream signaling processes such as intracellular calcium flux. In this respect assessing GPCR activation by GTP binding is an important tool to study the early stage of signal transduction. Previously this has been done using the radiolabeled non-hydrolyzable GTP analogue [(35)S]GTPgammaS. In order to avoid the problems involved in working with radioactivity, a new non-radioactive version of the assay has been developed using a europium-labeled GTP analogue in which europium-GTP binding can be assayed using time-resolved fluorescence. We have adapted this assay for chemokine receptors. In this chapter, using the chemokine receptor CXCR4 as an example, we describe the steps for assay optimization. In addition we describe adaptation of this assay for the high-throughput screening of chemokine antagonists.
- Published
- 2009
16. Pharmacology of AMD3465: a small molecule antagonist of the chemokine receptor CXCR4
- Author
-
Melanie Ruzek, Kim L. Nelson, Robert J. Scarborough, Stefan R. Idzan, Gary Bridger, Virginia Anastassov, Kathleen S. Neff, Gloria Lau, Marilyn C. Darkes, Rebecca S.Y. Wong, Jean Labrecque, Zefferino Santucci, Renee Mosi, Veronique Bodart, Ketan Patel, Simon P. Fricker, and Ron MacFarland
- Subjects
Male ,medicine.medical_specialty ,Receptors, CXCR4 ,Maximum Tolerated Dose ,Leukocytosis ,Metabolic Clearance Rate ,Pyridines ,C-C chemokine receptor type 7 ,CHO Cells ,Biology ,Pharmacology ,CXCR3 ,Kidney ,Transfection ,Biochemistry ,CXCR4 ,Absorption ,Cell Line ,Chemokine receptor ,Inhibitory Concentration 50 ,Mice ,Cricetulus ,Dogs ,Heterocyclic Compounds ,Internal medicine ,Cricetinae ,Calcium flux ,medicine ,Animals ,Humans ,Receptor ,Fluorescent Dyes ,Mice, Inbred BALB C ,CXCR4 antagonist ,Dose-Response Relationship, Drug ,Molecular Structure ,Chemotaxis ,Antagonist ,Fluoresceins ,Chemokine CXCL12 ,Mice, Inbred C57BL ,Endocrinology ,Mice, Inbred DBA ,Area Under Curve ,Calcium ,Half-Life ,Protein Binding - Abstract
CXCR4 is widely expressed in multiple cell types, and is involved in neonatal development, hematopoiesis, and lymphocyte trafficking and homing. Disruption of the CXCL12/CXCR4 interaction has been implicated in stem cell mobilization. Additionally CXCR4 is a co-receptor for HIV. Selective small molecule antagonists of CXCR4 therefore have therapeutic potential. AMD3465 is an N-pyridinylmethylene monocyclam CXCR4 antagonist which can block infection of T-tropic, CXCR4-using HIV. Using the CCRF-CEM T-cell line which expresses CXCR4 we have demonstrated that AMD3465 is an antagonist of SDF-1 ligand binding ( K i of 41.7 ± 1.2 nM), and inhibits SDF-1 mediated signaling as shown by inhibition of GTP binding, calcium flux, and inhibition of chemotaxis. AMD3465 is selective for CXCR4 and does not inhibit chemokine-stimulated calcium flux in cells expressing CXCR3, CCR1, CCR2b, CCR4, CCR5 or CCR7, nor does it inhibit binding of LTB 4 to its receptor, BLT1. The pharmacokinetics of AMD3465 was investigated in mice and dogs. Absorption was rapid following subcutaneous administration. AMD3465 was cleared from dog plasma in a biphasic manner with a terminal half-life of 1.56–4.63 h. Comparison of exposure to the intravenous and subcutaneous doses indicated 100% bioavailability following subcutaneous administration. AMD3465 caused leukocytosis when administered subcutaneously in mice and dogs, with peak mobilization occurring between 0.5 and 1.5 h following subcutaneous dosing in mice and with maximum peak plasma concentration of compound preceding peak mobilization in dogs, indicating that AMD3465 has the potential to mobilize hematopoietic stem cells. These data demonstrate the therapeutic potential for the CXCR4 antagonist AMD3465.
- Published
- 2009
17. Comparison of the potential multiple binding modes of bicyclam, monocylam, and noncyclam small-molecule CXC chemokine receptor 4 inhibitors
- Author
-
Markus Metz, Simon P. Fricker, Gary Bridger, Veronique Bodart, Rebecca S.Y. Wong, and Jean Labrecque
- Subjects
Models, Molecular ,Benzylamines ,Receptors, CXCR4 ,CXCR4 Inhibitor ,Chemokine receptor CCR5 ,Anti-HIV Agents ,Pyridines ,C-C chemokine receptor type 7 ,C-C chemokine receptor type 6 ,CXCR3 ,Butylamines ,Cyclams ,Ligands ,Binding, Competitive ,Cell Line ,Cell Fusion ,Chemokine receptor ,Heterocyclic Compounds, 1-Ring ,Radioligand Assay ,Cricetulus ,Heterocyclic Compounds ,Cricetinae ,Animals ,Humans ,CXC chemokine receptors ,Pharmacology ,Binding Sites ,biology ,Biochemistry ,Mutation ,biology.protein ,Aminoquinolines ,Mutagenesis, Site-Directed ,Molecular Medicine ,Benzimidazoles ,CCL21 - Abstract
CXC chemokine receptor (CXCR)4 is an HIV coreceptor and a chemokine receptor that plays an important role in several physiological and pathological processes, including hematopoiesis, leukocyte homing and trafficking, metastasis, and angiogenesis. This receptor belongs to the class A family of G protein-coupled receptors and is a validated target for the development of a new class of antiretroviral therapeutics. This study compares the interactions of three structurally diverse small-molecule CXCR4 inhibitors with the receptor and is the first report of the molecular interactions of the nonmacrocyclic CXCR4 inhibitor (S)-N'-(1H-benzimidazol-2-ylmethyl)-N'-(5,6,7,8-tetrahydroquinolin-8-yl)butene-1,4-diamine (AMD11070). Fourteen CXCR4 single-site mutants representing amino acid residues that span the entire putative ligand binding pocket were used in this study. These mutants were used in binding studies to examine how each single-site mutation affected the ability of the inhibitors to compete with (125)I-stromal-derived factor-1alpha binding. Our data suggest that these CXCR4 inhibitors bind to overlapping but not identical amino acid residues in the transmembrane regions of the receptor. In addition, our results identified amino acid residues that are involved in unique interactions with two of the CXCR4 inhibitors studied. These data suggest an extended binding pocket in the transmembrane regions close to the second extracellular loop of the receptor. Based on site-directed mutagenesis and molecular modeling, several potential binding modes were proposed for each inhibitor. These mechanistic studies might prove to be useful for the development of future generations of CXCR4 inhibitors with improved clinical pharmacology and safety profiles.
- Published
- 2008
18. AMD3100 mobilizes hematopoietic stem cells with long-term repopulating capacity in nonhuman primates
- Author
-
Cynthia E. Dunbar, Donald Orlic, Gary Bridger, Robert E. Donahue, Allen E. Krouse, Mark E. Metzger, Andre Larochelle, Peiman Hematti, and Simon P. Fricker
- Subjects
Benzylamines ,Receptors, CXCR4 ,Immunology ,Genetic Vectors ,CD34 ,Stem cell factor ,Antigens, CD34 ,Biology ,Integrin alpha4beta1 ,Cyclams ,Biochemistry ,CXCR4 ,Transplantation, Autologous ,Mice ,Dogs ,Heterocyclic Compounds ,Granulocyte Colony-Stimulating Factor ,Animals ,Humans ,Hematopoietic Stem Cell Mobilization ,Interleukin 3 ,Transplantation ,Base Sequence ,Hematopoietic Stem Cell Transplantation ,Cell Biology ,Hematology ,DNA ,Hematopoietic Stem Cells ,Macaca mulatta ,Recombinant Proteins ,Cell biology ,Endothelial stem cell ,Stem cell ,Moloney murine leukemia virus ,Adult stem cell - Abstract
AMD3100, a bicyclam antagonist of the chemokine receptor CXCR4, has been shown to induce rapid mobilization of CD34(+) hematopoietic cells in mice, dogs, and humans, offering an alternative to G-CSF mobilization of peripheral-blood hematopoietic stem cells. In this study, AMD3100-mobilized CD34(+) cells were phenotypically analyzed, marked with Neo(R)-containing retroviral vectors, and subsequently transplanted into myeloablated rhesus macaques. We show engraftment of transduced AMD3100-mobilized CD34(+) cells with Neo(R) gene marked myeloid and lymphoid cells up to 32 months after transplantation, demonstrating the ability of AMD3100 to mobilize true long-term repopulating hematopoietic stem cells. More AMD3100-mobilized CD34(+) cells are in the G(1) phase of the cell cycle and more cells express CXCR4 and VLA-4 compared with G-CSF-mobilized CD34(+) cells. In vivo gene marking levels obtained with AMD3100-mobilized CD34(+) cells were better than those obtained using CD34(+) cells mobilized with G-CSF alone. Overall, these results indicate that AMD3100 mobilizes a population of hematopoietic stem cells with intrinsic characteristics different from those of hematopoietic stem cells mobilized with G-CSF, suggesting fundamental differences in the mechanism of AMD3100-mediated and G-CSF-mediated hematopoietic stem cell mobilization. Thus, AMD3100-mobilized CD34(+) cells represent an alternative source of hematopoietic stem cells for clinical stem cell transplantation and genetic manipulation with integrating retroviral vectors.
- Published
- 2006
19. The development of an europium-GTP assay to quantitate chemokine antagonist interactions for CXCR4 and CCR5
- Author
-
Gloria Lau, Jean Labrecque, Marilyn C. Darkes, Simon P. Fricker, Renee Mosi, and Virginia Anastassov
- Subjects
CCR1 ,Chemokine ,Benzylamines ,Receptors, CXCR4 ,Time Factors ,GTP' ,Receptors, CCR5 ,Anti-HIV Agents ,Drug Evaluation, Preclinical ,CCR5 receptor antagonist ,Biology ,Sodium Chloride ,Cyclams ,Guanosine Diphosphate ,Fluorescence ,Cell Line ,Chemokine receptor ,Europium ,Heterocyclic Compounds ,Drug Discovery ,Humans ,G protein-coupled receptor ,CXCR4 antagonist ,Dose-Response Relationship, Drug ,Cell Membrane ,Temperature ,Reproducibility of Results ,Saponins ,Biochemistry ,CCR5 Receptor Antagonists ,biology.protein ,Molecular Medicine ,Calcium ,Guanosine Triphosphate ,Signal transduction ,Chemokines ,Signal Transduction - Abstract
Chemokine receptors have been implicated in several disease processes such as acute and chronic inflammation, cancer, and allograft rejection and are therefore targets for drug development. The chemokine receptors CCR5 and CXCR4 are of particular interest as they serve as entry cofactors for human immunodeficiency virus. These receptors are members of the G protein-coupled receptor (GPCR) family. In this respect, assessing GPCR activation by GTP binding is an important tool to study the early stage of signal transduction. The assay normally utilizes the non-hydrolysable GTP analogue guanosine 5'-gamma-[35S]thiotriphosphate. In order to avoid the problems involved in working with radioactivity, a new non-radioactive version of the assay was developed using a europium-labeled GTP analogue in which europium-GTP binding can be assayed using time-resolved fluorescence. The assay was optimized for CXCR4 and CCR5 and validated for screening of chemokine antagonists using the small molecule CXCR4 antagonist AMD3100 and CCR5 antagonists.
- Published
- 2006
20. Metal complexes as therapeutic agents in nitrogen monoxide modulation
- Author
-
Simon P, Fricker
- Subjects
Disease Models, Animal ,Metals ,Organometallic Compounds ,Animals ,Humans ,Ruthenium Compounds ,Nitric Oxide Donors ,Nitric Oxide ,Ruthenium - Published
- 2004
21. The in vitro antitumour profile of some 1,2-diaminocyclohexane organotin complexes
- Author
-
Josiah J. Bonire and Simon P. Fricker
- Subjects
Cisplatin ,Cyclohexylamines ,Molecular Structure ,Spectrophotometry, Infrared ,Ligand ,Chemistry ,Stereochemistry ,Platinum compounds ,Infrared spectroscopy ,Antineoplastic Agents ,Biochemistry ,In vitro ,Inorganic Chemistry ,Metal ,Spectroscopy, Mossbauer ,visual_art ,medicine ,visual_art.visual_art_medium ,Organotin Compounds ,Tumor Cells, Cultured ,Molecule ,Humans ,Drug Screening Assays, Antitumor ,Cytotoxicity ,medicine.drug - Abstract
Platinum compounds containing the ligand 1,2-diaminocyclohexane (DACH) such as tetraplatin [PtCl 4 (DACH)] have been found to be active in cisplatin-resistant tumour models. In an attempt to develop novel metal-based drugs with a different therapeutic profile to cisplatin, we have synthesised a series of tin compounds containing the DACH ligand, including the Sn analogue of tetraplatin [SnCl 4 (DACH)], and the di- and monoorganotin complexes [Ph 2 Sn(OAc) 2 (DACH)], [Bu 2 Sn(OAc) 2 (DACH)], [PhSnCl 3 (DACH)], [BuSn(OAc) 3 (DACH)], [BuSnCl 3 (DACH)], and [PhSn(OCOCF 3 ) 3 (DACH)]. Mossbauer and IR spectroscopy indicates that the Sn(DACH) complexes are hexacoordinated with a molecular structure similar to that of tetraplatin. These compounds were tested for potential antitumour activity against a panel of human tumour cell lines, (SW620, SW1116 colon carcinoma, ZR-75-1 breast carcinoma, HT1376 bladder carcinoma, SKOV-3, PA-1 ovarian carcinoma). [Ph 2 Sn(penicillinate)], [Ph 2 Sn(OCOCH 2 NCOCH 2 NH 2 )], [Ph 2 Sn(OAc) 2 ] were included for comparison. The results show that whereas [SnCl 4 (DACH)] and the monoorganotin complexes had limited or no activity, the diorganotin DACH complexes were cytotoxic with an associated increase in potency on going from diphenyl to dibutyltin, with mean IC 50 values of 7.26±4.09 μmol ml −1 for [Ph 2 Sn(OAc) 2 (DACH)] and 2.58±0.83 μmol ml −1 for [Bu 2 Sn(OAc) 2 (DACH)] across the cell line panel. Comparison with [Ph 2 Sn(OAc) 2 ] (IC 50 0.69±0.43 μmol ml −1 ) indicated that addition of the DACH ligand resulted in a decrease in cytotoxicity but increased differential toxicity across the cell line panel. These results indicate that the diorganotin DACH complexes merit further investigation as potential metal-based antitumour drugs.
- Published
- 2001
22. The therapeutic application of lanthanides
- Author
-
Simon P. Fricker
- Subjects
Molecular Structure ,Chemistry ,Inorganic chemistry ,Context (language use) ,Nanotechnology ,General Chemistry ,Lanthanoid Series Elements ,Phosphates ,Lanthanum carbonate ,Biological property ,Organometallic Compounds ,medicine ,Humans ,Kidney Diseases ,Biological scientists ,medicine.drug - Abstract
The biological properties of the lanthanides, based on their similarity to calcium, have stimulated research into their therapeutic application. Historical medical uses of the lanthanides and recent advances and successes will be described in the context of the biological chemistry of lanthanides, including a new metal-based drug, lanthanum carbonate, which has recently been approved as a phosphate binder for the treatment of hyperphosphatemia. This tutorial review will be of interest to those working on metal-based drugs, including inorganic chemists, and biological scientists.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.