Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Histochem Cytochem ; 67(4): 257-266, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30562126

RESUMO

Chemokines and their receptors have been implicated in cancer biology. The CXCL12/CXCR4 axis is essential for the homing and retention of hematopoietic stem cells in bone marrow niches, and has a significant role in neonatal development. It is also implicated in multiple facets of cancer biology including metastasis, angiogenesis/neo-vasculogenesis, and immune cell trafficking at the tumor microenvironment (TME). Immunohistochemistry (IHC) is an ideal method for investigating involvement of CXCL12 in the TME. Three antibodies were evaluated here for their suitability to stain CXCL12. Both D8G6H and K15C gave apparent specific staining in both lymphoid and tumor tissue, but with converse staining patterns. D8G6H stained cells in the parafollicular zone whereas K15C showed staining of lymphoid cells in the interfollicular zone of tonsil tissue. Using a cell line with high CXCL12 expression, TOV21G, as a positive control, it was found that D8G6H gave strong staining of TOV21G cells whereas no staining was observed with K15C indicating that D8G6H specifically stains CXCL12. Significant staining of CXCL12 in the ovarian TME using tissue microarray was observed using D8G6H. These data demonstrate the importance of antibody characterization for IHC applications, and provide further evidence for the involvement of CXCL12 in ovarian cancer biology.


Assuntos
Anticorpos/análise , Quimiocina CXCL12/análise , Imuno-Histoquímica/métodos , Células A549 , Animais , Anticorpos Monoclonais/análise , Células CACO-2 , Feminino , Células HT29 , Humanos , Camundongos , Neoplasias Ovarianas/patologia , Coelhos , Microambiente Tumoral
3.
J Biol Chem ; 290(17): 11041-51, 2015 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-25767113

RESUMO

The chemokine receptors CCR5 and CCR2b share 89% amino acid homology. CCR5 is a co-receptor for HIV and CCR5 antagonists have been investigated as inhibitors of HIV infection. We describe the use of two CCR5 antagonists, Schering-C (SCH-C), which is specific for CCR5, and TAK-779, a dual inhibitor of CCR5 and CCR2b, to probe the CCR5 inhibitor binding site using CCR5/CCR2b chimeric receptors. Compound inhibition in the different chimeras was assessed by inhibition of chemokine-induced calcium flux. SCH-C inhibited RANTES (regulated on activation, normal T cell expressed and secreted) (CCL5)-mediated calcium flux on CCR5 with an IC50 of 22.8 nM but was inactive against monocyte chemoattractant protein-1 (CCL2)-mediated calcium flux on CCR2b. However, SCH-C inhibited CCL2-induced calcium flux against a CCR5/CCR2b chimera consisting of transmembrane domains IV-VI of CCR5 with an IC50 of 55 nM. A sequence comparison of CCR5 and CCR2b identified a divergent amino acid sequence located at the junction of transmembrane domain V and second extracellular loop. Transfer of the CCR5 sequence KNFQTLKIV into CCR2b conferred SCH-C inhibition (IC50 of 122 nM) into the predominantly CCR2b chimera. Furthermore, a single substitution, R206I, conferred partial but significant inhibition (IC50 of 1023 nM) by SCH-C. These results show that a limited amino acid sequence is responsible for SCH-C specificity to CCR5, and we propose a model showing the interaction with CCR5 Ile(198).


Assuntos
Amidas/química , Antagonistas dos Receptores CCR5/química , Modelos Moleculares , Compostos de Amônio Quaternário/química , Receptores CCR5/química , Sequência de Aminoácidos , Animais , Sinalização do Cálcio , Células HEK293 , Humanos , Isoleucina/química , Isoleucina/genética , Isoleucina/metabolismo , Macaca , Estrutura Terciária de Proteína , Receptores CCR2/antagonistas & inibidores , Receptores CCR2/química , Receptores CCR2/genética , Receptores CCR2/metabolismo , Receptores CCR5/genética , Receptores CCR5/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
4.
Future Med Chem ; 6(1): 91-114, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24358950

RESUMO

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.


Assuntos
Desenho de Fármacos , Simulação de Acoplamento Molecular , Receptores de Quimiocinas/antagonistas & inibidores , Sítios de Ligação , Quimiocinas/metabolismo , Inibidores da Fusão de HIV/química , Inibidores da Fusão de HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Humanos , Estrutura Terciária de Proteína , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo
5.
Transfus Med Hemother ; 40(4): 237-45, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24179472

RESUMO

Autologous hematopoietic stem cell (HSC) transplantation is an important therapeutic option for patients with non-Hodgkin's lymphoma and multiple myeloma. The primary source of HSC is from the peripheral blood which requires mobilization from the bone marrow. Current mobilization regimens include cytokines such as G-CSF and/or chemotherapy. However not all patients mobilize enough HSC to proceed to transplant. The chemokine receptor CXCR4 and its ligand CXCL12 are an integral part of the mechanism of HSC retention in the bone marrow niche. The discovery of plerixafor, a selective inhibitor of CXCR4, has provided a new additional means of mobilizing HSC for autologous transplantation. Plerixafor consists of two cyclam rings with a phenylenebis(methylene) linker. It inhibits CXCL12 binding to CXCR4 and subsequent downstream events including chemotaxis. The molecular interactions of plerixafor have been defined indicating a unique binding mode to CXCR4. Plerixafor rapidly mobilizes HSC within hours compared with the multi-day treatment required by G-CSF in mouse, dog and non-human primate. The mobilized cells once transplanted are capable of timely and endurable engraftment. Additionally CXCR4 has been implicated in the pathology of HIV, inflammatory disease and cancer and the pharmacology of plerixafor in various disease models is described.

6.
Biochem Pharmacol ; 83(4): 472-9, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22146583

RESUMO

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-1ß 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.


Assuntos
Aminoquinolinas/farmacologia , Aminoquinolinas/farmacocinética , Benzimidazóis/farmacologia , Benzimidazóis/farmacocinética , HIV-1/efeitos dos fármacos , Receptores CXCR4/metabolismo , Internalização do Vírus/efeitos dos fármacos , Administração Oral , Aminoquinolinas/administração & dosagem , Animais , Fármacos Anti-HIV/administração & dosagem , Fármacos Anti-HIV/farmacocinética , Fármacos Anti-HIV/farmacologia , Benzimidazóis/administração & dosagem , Disponibilidade Biológica , Butilaminas , Linhagem Celular , Quimiocina CXCL12/antagonistas & inibidores , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Cães , Regulação da Expressão Gênica/efeitos dos fármacos , HIV-1/fisiologia , Compostos Heterocíclicos com 1 Anel , Humanos , Estrutura Molecular , Ligação Proteica , Receptores CXCR4/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
7.
J Am Chem Soc ; 133(41): 16477-85, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-21942640

RESUMO

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.


Assuntos
Fármacos Anti-HIV/farmacologia , Desenho de Fármacos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , HIV-1/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Ureia/farmacologia , Fármacos Anti-HIV/síntese química , Fármacos Anti-HIV/química , Antagonistas dos Receptores CCR5 , Relação Dose-Resposta a Droga , Humanos , Testes de Sensibilidade Microbiana , Modelos Moleculares , Estrutura Molecular , Peso Molecular , Mutagênese , Estereoisomerismo , Relação Estrutura-Atividade , Ureia/análogos & derivados , Ureia/química
8.
Anticancer Agents Med Chem ; 11(10): 940-52, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21864236

RESUMO

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.


Assuntos
Antineoplásicos/química , Antineoplásicos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos/métodos , Compostos de Ouro/química , Compostos de Ouro/uso terapêutico , Neoplasias/tratamento farmacológico , Compostos Organoáuricos/química , Compostos Organoáuricos/uso terapêutico , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Compostos de Ouro/farmacocinética , Compostos de Ouro/farmacologia , Humanos , Modelos Moleculares , Compostos Organoáuricos/farmacocinética , Compostos Organoáuricos/farmacologia
9.
Mol Cancer Ther ; 10(9): 1571-80, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21712476

RESUMO

The role of endogenous nitric oxide (NO) in the growth and vascularization of a rat carcinosarcoma (P22) has been investigated. Tumor-bearing animals were treated with (i) nitric oxide synthase (NOS) inhibitors, administered via the drinking water, including N(G)-nitro-l-arginine methyl ester (L-NAME), a nonisoform-selective inhibitor, and 2 others that target the inducible (NOS II) enzyme preferentially, namely 1-amino-2-hydroxyguanidine or N-[3-(aminomethyl)benzyl]acetamidine hydrochloride; or (ii) daily injections (intraperitoneally) of 2 Ru(III) polyaminocarboxylates, AMD6221 and AMD6245, both of which are effective NO scavengers. L-NAME, AMD6221, and AMD6245 reduced tumor growth by approximately 60% to 75% of control rates. Tumor sections stained with abs to CD-31/platelet endothelial cell adhesion molecule-1 or NOS III showed that this was associated with a marked reduction (60%-77%) of tumor microvascular densitiy (MVD). Tumors resumed growing promptly when treatment was discontinued, accompanied by partial or complete restoration of MVDs. In contrast, NOS-II selective inhibitors had no effect on tumor growth or vascularization, indicating that both responses require complete blockade of NO production. The results corroborate the view that endogenous NO facilitates tumor development. We suggest that NO deprivation causes tumor feeder vessels to constrict, reducing tumor blood flow. The delivery of oxygen and essential nutrients to the developing tumor is impaired as a consequence, hampering further growth. Normalizing NO levels by withholding treatment causes tumor feeder vessels to dilate, increasing tumor perfusion and reestablishing conditions that allow tumors to begin growing again.


Assuntos
Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico/metabolismo , Rutênio/farmacologia , Sarcoma Experimental/tratamento farmacológico , Animais , Inibidores Enzimáticos/uso terapêutico , Sequestradores de Radicais Livres/metabolismo , Sequestradores de Radicais Livres/uso terapêutico , Masculino , NG-Nitroarginina Metil Éster/farmacologia , Neovascularização Patológica/tratamento farmacológico , Compostos Organometálicos/farmacologia , Ácido Pentético/análogos & derivados , Ácido Pentético/farmacologia , Ratos , Sarcoma Experimental/metabolismo , Transplante Isogênico , Carga Tumoral/efeitos dos fármacos
10.
J Inorg Biochem ; 105(5): 754-62, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21481817

RESUMO

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.


Assuntos
Antineoplásicos/química , Catepsina B/antagonistas & inibidores , Catepsina K/antagonistas & inibidores , Inibidores de Cisteína Proteinase/química , Ouro/química , Animais , Antineoplásicos/farmacologia , Catepsina B/química , Catepsina B/metabolismo , Catepsina K/química , Catepsina K/metabolismo , Linhagem Celular Tumoral , Cisteína Proteases/metabolismo , Inibidores de Cisteína Proteinase/farmacologia , Humanos , Masculino , Camundongos , Camundongos SCID
11.
Virology ; 413(2): 231-43, 2011 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-21388649

RESUMO

Based on the attrition rate of CCR5 small molecule antagonists in the clinic the discovery and development of next generation antagonists with an improved pharmacology and safety profile is necessary. Herein, we describe a combined molecular modeling, CCR5-mediated cell fusion, and receptor site-directed mutagenesis approach to study the molecular interactions of six structurally diverse compounds (aplaviroc, maraviroc, vicriviroc, TAK-779, SCH-C and a benzyloxycarbonyl-aminopiperidin-1-yl-butane derivative) with CCR5, a coreceptor for CCR5-tropic HIV-1 strains. This is the first study using an antifusogenic assay, a model of the interaction of the gp120 envelope protein with CCR5. This assay avoids the use of radioactivity and HIV infection assays, and can be used in a high throughput mode. The assay was validated by comparison with other established CCR5 assays. Given the hydrophobic nature of the binding pocket several binding models are suggested which could prove useful in the rational drug design of new lead compounds.


Assuntos
Antagonistas dos Receptores CCR5 , Inibidores da Fusão de HIV/farmacologia , HIV-1/fisiologia , HIV-1/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Conformação Proteica , Receptores CCR5/genética , Reprodutibilidade dos Testes , Estereoisomerismo , Internalização do Vírus
12.
Metallomics ; 2(6): 366-77, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21072382

RESUMO

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.


Assuntos
Cisplatino/farmacologia , Complexos de Coordenação/farmacologia , Cisteína Proteases/efeitos dos fármacos , Sistemas de Liberação de Medicamentos , Antineoplásicos/química , Antineoplásicos/farmacologia , Cisplatino/química , Complexos de Coordenação/química , Cisteína Proteases/química , Humanos
13.
Clin Cancer Res ; 16(11): 2927-31, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20484021

RESUMO

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.


Assuntos
Quimiocina CXCL12/metabolismo , Neoplasias/metabolismo , Receptores CXCR4/antagonistas & inibidores , Sistemas de Liberação de Medicamentos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Transdução de Sinais/genética
14.
Methods Mol Biol ; 552: 153-69, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19513648

RESUMO

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.


Assuntos
Membrana Celular/metabolismo , Európio/metabolismo , Guanosina Trifosfato/metabolismo , Leucemia de Células T/metabolismo , Ensaio Radioligante/métodos , Receptores CXCR4/metabolismo , Descoberta de Drogas , Humanos , Receptores CXCR4/análise , Células Tumorais Cultivadas
15.
Biochem Pharmacol ; 78(8): 993-1000, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19540208

RESUMO

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+/-1.2nM), 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 1.56-4.63h. 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 1.5h 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.


Assuntos
Compostos Heterocíclicos/farmacologia , Piridinas/farmacologia , Piridinas/farmacocinética , Receptores CXCR4/antagonistas & inibidores , Absorção , Animais , Área Sob a Curva , Células CHO , Cálcio/análise , Cálcio/metabolismo , Linhagem Celular , Quimiocina CXCL12/antagonistas & inibidores , Quimiotaxia/efeitos dos fármacos , Cricetinae , Cricetulus , Cães , Relação Dose-Resposta a Droga , Fluoresceínas/metabolismo , Corantes Fluorescentes/metabolismo , Meia-Vida , Humanos , Concentração Inibidora 50 , Rim/citologia , Leucocitose/etiologia , Masculino , Dose Máxima Tolerável , Taxa de Depuração Metabólica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Estrutura Molecular , Ligação Proteica , Piridinas/efeitos adversos , Piridinas/sangue , Piridinas/química , Transfecção
16.
Expert Opin Investig Drugs ; 17(11): 1749-60, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18922110

RESUMO

BACKGROUND: Hematopoietic stem cell (HSC) transplantation is a treatment option for hematological malignancies. Current mobilization regimes frequently result in inadequate numbers of HSC for transplant therefore alternative methods of mobilization are required. OBJECTIVE: The chemokine receptor CXCR4 and ligand SDF-1 are integrally involved in HSC homing and mobilization. Disruption of the SDF-1/CXCR4 axis by the CXCR4 anatagonist, plerixafor, is shown to improve HSC mobilization. METHODS: The molecular and in vivo pharmacology of plerixafor and subsequent clinical development is reviewed. RESULTS/CONCLUSION: Preclinical studies demonstrate that plerixafor is a selective antagonist of CXCR4 and can rapidly mobilize HSC. Clinical trials demonstrated improved HSC mobilization when plerixafor was included in the mobilization regimen. These data suggest the potential for a significant role for plerixafor in hematological disease.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Receptores CXCR4/antagonistas & inibidores , Animais , Benzilaminas , Ciclamos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Compostos Heterocíclicos/química , Compostos Heterocíclicos/farmacologia , Humanos , Receptores CXCR4/metabolismo
17.
Mol Pharmacol ; 74(6): 1485-95, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18768385

RESUMO

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.


Assuntos
Aminoquinolinas/farmacologia , Fármacos Anti-HIV/farmacologia , Benzimidazóis/farmacologia , Compostos Heterocíclicos/farmacologia , Piridinas/farmacologia , Receptores CXCR4/antagonistas & inibidores , Aminoquinolinas/metabolismo , Animais , Fármacos Anti-HIV/metabolismo , Benzimidazóis/metabolismo , Benzilaminas , Sítios de Ligação , Ligação Competitiva , Butilaminas , Fusão Celular , Linhagem Celular , Cricetinae , Cricetulus , Ciclamos , Compostos Heterocíclicos/metabolismo , Compostos Heterocíclicos com 1 Anel , Humanos , Ligantes , Modelos Moleculares , Mutagênese Sítio-Dirigida , Mutação , Piridinas/metabolismo , Ensaio Radioligante , Receptores CXCR4/genética , Receptores CXCR4/metabolismo
18.
J Inorg Biochem ; 102(10): 1839-45, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18684510

RESUMO

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.


Assuntos
Catepsina B/metabolismo , Doença de Chagas/tratamento farmacológico , Inibidores de Cisteína Proteinase/farmacologia , Leishmania/efeitos dos fármacos , Leishmaniose/tratamento farmacológico , Metais/farmacologia , Tripanossomicidas/farmacologia , Trypanosoma/efeitos dos fármacos , Animais , Catepsina B/antagonistas & inibidores , Cisteína Endopeptidases/metabolismo , Inibidores de Cisteína Proteinase/uso terapêutico , Humanos , Compostos Inorgânicos/química , Compostos Inorgânicos/farmacologia , Metais/uso terapêutico , Tripanossomicidas/uso terapêutico
19.
Am J Pathol ; 169(2): 424-32, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16877345

RESUMO

CXCR4 is a major receptor for CXCL12 and is known to participate in multiple physiological systems. The present study tested a second generation CXCR4 antagonist, AMD3465, for effects on highly defined models of Th1- and Th2-cell-mediated hypersensitivity-type pulmonary granuloma formation. Type-1 and type-2 granulomas were induced, respectively, by intravenous challenge of sensitized CBA/J mice with Mycobacteria bovis purified protein derivative- or Schistosoma mansoni egg antigen-coated beads. Before challenge, mice were implanted with osmotic pumps releasing AMD3465 at 5 microg/hour (6 mg/kg/day). Compared to vehicle, AMD3465 had minimal effect on type-1 inflammation or cytokine responses in draining lymph nodes, but the type-2 inflammation was significantly abrogated with reductions in lesion size and eosinophil content as well as abrogated interleukin (IL)-5, IL-10, and IL-13 cytokine production in draining lymph nodes. The biased effect of AMD3465 correlated with greater CXCR4 ligand expression in the type-2 model. Treatment during a primary response impaired lymph node IL-2 production after both Mycobacteria bovis purified protein derivative and Schistosoma mansoni egg antigen challenge indicating an unbiased effect during immune induction. In summary, CXCR4 blockade inhibited eosinophil recruitment during type-2 granuloma formation and interfered with primary and secondary T-cell activation events in lymphoid tissue, suggesting potential therapeutic application for chronic hypersensitivity diseases.


Assuntos
Antígenos de Helmintos/imunologia , Granuloma do Sistema Respiratório/induzido quimicamente , Pulmão/efeitos dos fármacos , Pulmão/patologia , Piridinas/farmacologia , Receptores CXCR4/antagonistas & inibidores , Schistosoma mansoni/imunologia , Animais , Células Cultivadas , Quimiocina CXCL12 , Quimiocinas CXC/genética , Citocinas/biossíntese , Relação Dose-Resposta a Droga , Eosinófilos/imunologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Granuloma do Sistema Respiratório/patologia , Interleucina-2/biossíntese , Linfonodos/efeitos dos fármacos , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores CXCR4/genética , Células Th2/efeitos dos fármacos
20.
J Med Chem ; 49(17): 5262-72, 2006 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-16913715

RESUMO

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.


Assuntos
Catepsina B/antagonistas & inibidores , Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/farmacologia , Compostos Organometálicos/síntese química , Compostos Organometálicos/farmacologia , Rênio/química , Sítios de Ligação , Catepsina B/química , Catepsina K , Catepsinas/antagonistas & inibidores , Catepsinas/química , Inibidores de Cisteína Proteinase/química , Humanos , Ligantes , Estrutura Molecular , Compostos Organometálicos/química , Estereoisomerismo , Relação Estrutura-Atividade , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...