Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Future Microbiol ; 16: 589-606, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33998266

RESUMEN

Aim: To predict glycosylphosphatidylinositol (GPI)-anchored proteins in the genome of Paracoccidioides brasiliensis and Paracoccidioides lutzii. Materials & methods: Five different bioinformatics tools were used for predicting GPI-anchored proteins; we considered as GPI-anchored proteins those detected by at least two in silico analysis methods. We also performed the proteomic analysis of P. brasiliensis cell wall by mass spectrometry. Results: Hundred GPI-anchored proteins were predicted in P. brasiliensis and P. lutzii genomes. A series of 57 proteins were classified in functional categories and 43 conserved proteins were reported with unknown functions. Four proteins identified by in silico analyses were also identified in the cell wall proteome. Conclusion: The data obtained in this study are important resources for future research of GPI-anchored proteins in Paracoccidioides spp. to identify targets for new diagnostic tools, drugs and immunological tests.


Asunto(s)
Proteínas Fúngicas/genética , Glicosilfosfatidilinositoles/metabolismo , Proteínas de la Membrana/genética , Paracoccidioides/metabolismo , Secuencia de Aminoácidos , Pared Celular/genética , Pared Celular/metabolismo , Biología Computacional , Secuencia Conservada , Proteínas Fúngicas/química , Proteínas Fúngicas/metabolismo , Genoma Fúngico/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Sistemas de Lectura Abierta , Paracoccidioides/genética , Paracoccidioides/patogenicidad , Paracoccidioidomicosis/microbiología , Proteómica , Virulencia
2.
Lancet Oncol ; 19(6): 812-824, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29706375

RESUMEN

BACKGROUND: The C-X-C chemokine receptor type 4 (CXCR4)-stromal cell-derived factor-1α (SDF-1α) axis regulates function and trafficking of immune cells and the tumour microenvironment. CXCR4 antagonists have been shown to enhance the activity of different anticancer treatments in preclinical models. We assessed the safety, tolerability, pharmacokinetics, and preliminary phase 1 activity of the CXCR4 antagonist, balixafortide, in combination with eribulin chemotherapy in patients with heavily pretreated, relapsed metastatic breast cancer. METHODS: This single-arm, dose-escalation, phase 1 trial enrolled patients at 11 sites in Spain and the USA. Eligible patients were women aged 18 years or older who had histologically confirmed HER2-negative metastatic breast cancer, evidence of tumour cell CXCR4 expression, an Eastern Cooperative Oncology Group performance status of 0 or 1, and who had previously received between one and three chemotherapy regimens for metastatic breast cancer, and at least one endocrine therapy if they had hormone receptor-positive disease, unless they were considered unsuitable for endocrine therapy. A standard 3+3 dose-escalation design was used, followed by an expanded cohort at the established maximum tolerated dose or highest dose if no dose-limiting toxicity was observed for the combination. After a treatment-related fatal adverse event in the first cohort who received 21-day cycles of treatment with eribulin and balixafortide, a protocol amendment modified the study design to be done in two parts. Patients enrolled to part 1 received an initial 28-day run-in cycle, with some cohorts receiving de-escalated doses of eribulin plus balixafortide to assess the safety and pharmacokinetics of the combination. The evaluation of part 1 did not confirm any dose-limiting toxicities or eribulin-balixafortide interactions, and therefore part 2 started enrolling patients to receive eribulin at the originally planned dose of 1·4 mg/m2 on days 2 and 9 of a 21-day cycle and balixafortide from a starting dose of 2 mg/kg with dose increments of 0·5 or 1 mg/kg on days 1-3 and 8-10 of the 21-day cycle. Both drugs were administered as intravenous infusions. All patients were to receive treatment until disease progression or unacceptable toxicity. The primary endpoints were dose-limiting toxicities and adverse events, and the establishment of a maximum tolerated dose or recommended phase 2 dose, and pharmacokinetic parameters. Safety analysis was done in all patients who received at least one dose of study treatment. Analysis of antitumour activity was done in all patients who received at least one full cycle of study treatment. The trial is registered at ClinicalTrials.gov, number NCT01837095, and is closed to accrual. FINDINGS: Between Jan 28, 2014, and Oct 4, 2016, 56 patients were enrolled into the trial. No dose-limiting toxicities were confirmed and the maximum tolerated dose was not reached. The highest dose was established as eribulin 1·4 mg/m2 on days 2 and 9, and balixafortide 5·5 mg/kg on days 1-3 and 8-10 of the 21-day cycle. Objective responses (all partial responses) were observed in 16 (30%; 95% CI 18-44) of 54 patients who were evaluable for antitumour activity. The most common treatment-emergent adverse events of any grade were fatigue (44 [79%] of 56 patients), neutropenia (32 [57%]), infusion-related reactions (27 [48%]), alopecia (26 [46%]), constipation (26 [46%]), and nausea (25 [45%]). Serious adverse events occurred in 21 (38%) of 56 patients, including febrile neutropenia in five (9%) of 56 patients, neutrophil count decrease in two (4%) patients, constipation in two (4%) patients, pneumonia in two (4%) patients, and urinary tract infection in three (5%) patients. Two (4%) of 56 patients died while receiving study treatment; one from septic shock and one from pneumonia. INTERPRETATION: The safety and tolerability of balixafortide plus eribulin seems to be similar to that of eribulin or balixafortide monotherapy, and the preliminary activity of the combination seems promising in patients with HER-negative metastatic breast cancer. The results suggest that balixafortide plus eribulin has potential to provide a new therapeutic option in heavily pretreated patients with metastatic breast cancer and warrants further investigation in randomised trials. FUNDING: Polyphor.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/tratamiento farmacológico , Furanos/administración & dosificación , Cetonas/administración & dosificación , Péptidos Cíclicos/administración & dosificación , Receptor ErbB-2/análisis , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Neoplasias de la Mama/química , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Femenino , Furanos/efectos adversos , Furanos/farmacocinética , Humanos , Cetonas/efectos adversos , Cetonas/farmacocinética , Dosis Máxima Tolerada , Persona de Mediana Edad , Metástasis de la Neoplasia , Péptidos Cíclicos/efectos adversos , Péptidos Cíclicos/farmacocinética , Receptores CXCR4/antagonistas & inhibidores , España , Factores de Tiempo , Resultado del Tratamiento , Estados Unidos
3.
Am J Pathol ; 187(9): 2080-2094, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28734730

RESUMEN

Resistance to antiangiogenic therapy in glioblastoma (GBM) patients may involve hypoxia-induced expression of C-X-C motif chemokine receptor 4 (CXCR4) on invading tumor cells, macrophage/microglial cells (MGCs), and glioma stem cells (GSCs). We determined whether antagonizing CXCR4 with POL5551 disrupts anti-vascular endothelial growth factor (VEGF) therapy-induced glioma growth and dissemination. Mice bearing orthotopic CT-2A or GL261 gliomas received POL5551 and/or anti-VEGF antibody B20-4.1.1. Brain tissue was analyzed for tumor volume, invasiveness, hypoxia, vascular density, proliferation, apoptosis, GSCs, and MGCs. Glioma cells were evaluated for CXCR4 expression and polymorphism and POL5551's effects on CXCR4 ligand binding, cell viability, and migration. No CXCR4 mutations were identified. POL5551 inhibited CXCR4 binding to its ligand, stromal cell-derived factor-1α, and reduced hypoxia- and stromal cell-derived factor-1α-mediated migration dose-dependently but minimally affected cell viability. In vivo, B20-4.1.1 increased hypoxic foci and invasiveness, as seen in GBM patients receiving anti-VEGF therapy. Combination of POL5551 and B20-4.1.1 reduced both glioma invasiveness by 16% to 39% and vascular density compared to B20-4.1.1 alone in both glioma models. Reduced populations of GSCs and MGCs were also seen in CT-2A tumors. POL5551 concentrations, evaluated by mass spectrometry, were higher in tumors than in neighboring brain tissues, likely accounting for the results. Inhibition of CXCR4-regulated tumoral, stem cell, and immune mechanisms by adjunctive CXCR4 antagonists may help overcome antiangiogenic therapy resistance, benefiting GBM patients.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Anticuerpos/uso terapéutico , Glioma/tratamiento farmacológico , Receptores CXCR4/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/inmunología , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Movimiento Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Glioma/metabolismo , Glioma/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Proteínas/farmacología , Transducción de Señal/efectos de los fármacos
4.
J Transl Med ; 15(1): 2, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-28049490

RESUMEN

BACKGROUND: Certain disadvantages of the standard hematopoietic stem and progenitor cell (HSPC) mobilizing agent G-CSF fuel the quest for alternatives. We herein report results of a Phase I dose escalation trial comparing mobilization with a peptidic CXCR4 antagonist POL6326 (balixafortide) vs. G-CSF. METHODS: Healthy male volunteer donors with a documented average mobilization response to G-CSF received, following ≥6 weeks wash-out, a 1-2 h infusion of 500-2500 µg/kg of balixafortide. Safety, tolerability, pharmacokinetics and pharmacodynamics were assessed. RESULTS: Balixafortide was well tolerated and rated favorably over G-CSF by subjects. At all doses tested balixafortide mobilized HSPC. In the dose range between 1500 and 2500 µg/kg mobilization was similar, reaching 38.2 ± 2.8 CD34 + cells/µL (mean ± SEM). Balixafortide caused mixed leukocytosis in the mid-20 K/µL range. B-lymphocytosis was more pronounced, whereas neutrophilia and monocytosis were markedly less accentuated with balixafortide compared to G-CSF. At the 24 h time point, leukocytes had largely normalized. CONCLUSIONS: Balixafortide is safe, well tolerated, and induces efficient mobilization of HSPCs in healthy male volunteers. Based on experience with current apheresis technology, the observed mobilization at doses ≥1500 µg/kg of balixafortide is predicted to yield in a single apheresis a standard dose of 4× 10E6 CD34+ cells/kg from most individuals donating for an approximately weight-matched recipient. Exploration of alternative dosing regimens may provide even higher mobilization responses. Trial Registration European Medicines Agency (EudraCT-Nr. 2011-003316-23) and clinicaltrials.gov (NCT01841476).


Asunto(s)
Voluntarios Sanos , Movilización de Célula Madre Hematopoyética , Péptidos Cíclicos/farmacología , Péptidos/farmacología , Receptores CXCR4/antagonistas & inhibidores , Diferenciación Celular/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética/efectos adversos , Humanos , Masculino , Péptidos/farmacocinética , Péptidos Cíclicos/farmacocinética , Receptores CXCR4/metabolismo
5.
Oncotarget ; 6(31): 30902-18, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26360610

RESUMEN

The importance of the cell surface receptor CXCR4 and the chemokine stromal cell-derived factor-1 (SDF-1/CXCL12) is well-established in normal and malignant hematopoiesis. The Protein Epitope Mimetic POL5551 is a novel and potent antagonist of CXCR4. POL5551 efficiently mobilizes hematopoietic stem and progenitor cells, but its effects in acute lymphoblastic leukemia (ALL) have not been reported. Here, we demonstrate that POL5551 is a potent antagonist of CXCR4 in pre-B and T cell ALL cell lines and pediatric ALL primary samples. POL5551 has activity at nanomolar concentrations in decreasing CXCR4 antibody binding, blocking SDF-1α-mediated phosphorylation of ERK1/2, inhibiting SDF-1α-induced chemotaxis, and reversing stromal-mediated protection from chemotherapy. POL5551 is significantly more effective at inhibiting CXCR4 antibody binding than the FDA-approved CXCR4 inhibitor plerixafor in ALL cell lines and primary samples. We also show that treatment with POL5551 in vitro and cytarabine +/- POL5551 in vivo modulates surface expression of adhesion molecules, findings that may guide the optimal clinical use of POL5551. Finally, we demonstrate that POL5551 increases sensitivity to cytarabine in a xenograft model of a high-risk pediatric ALL, infant MLL-rearranged (MLL-R) ALL. Therefore, disruption of the CXCR4/SDF-1 axis with POL5551 may improve outcomes in children with high-risk ALL.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Proteínas/farmacología , Receptores CXCR4/antagonistas & inhibidores , Células del Estroma/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Niño , Citometría de Flujo , Humanos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células del Estroma/metabolismo , Células del Estroma/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cancer Ther ; 14(11): 2473-85, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26269605

RESUMEN

The SDF-1 receptor CXCR4 has been associated with early metastasis and poorer prognosis in breast cancers, especially the most aggressive triple-negative subtype. In line with previous reports, we found that tumoral CXCR4 expression in patients with locally advanced breast cancer was associated with increased metastases and rapid tumor progression. Moreover, high CXCR4 expression identified a group of bone marrow-disseminated tumor cells (DTC)-negative patients at high risk for metastasis and death. The protein epitope mimetic (PEM) POL5551, a novel CXCR4 antagonist, inhibited binding of SDF-1 to CXCR4, had no direct effects on tumor cell viability, but reduced migration of breast cancer cells in vitro. In two orthotopic models of triple-negative breast cancer, POL5551 had little inhibitory effect on primary tumor growth, but significantly reduced distant metastasis. When combined with eribulin, a chemotherapeutic microtubule inhibitor, POL5551 additively reduced metastasis and prolonged survival in mice after resection of the primary tumor compared with single-agent eribulin. Hypothesizing that POL5551 may mobilize tumor cells from their microenvironment and sensitize them to chemotherapy, we used a "chemotherapy framing" dosing strategy. When administered shortly before and after eribulin treatment, three doses of POL5551 with eribulin reduced bone and liver tumor burden more effectively than chemotherapy alone. These data suggest that sequenced administration of CXCR4 antagonists with cytotoxic chemotherapy synergize to reduce distant metastases.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas/farmacología , Receptores CXCR4/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Animales , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Epítopos/metabolismo , Furanos/administración & dosificación , Furanos/farmacología , Humanos , Cetonas/administración & dosificación , Cetonas/farmacología , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Metástasis de la Neoplasia , Unión Proteica/efectos de los fármacos , Proteínas/administración & dosificación , Receptores CXCR4/metabolismo , Transducción de Señal/efectos de los fármacos , Análisis de Supervivencia , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 5(20): 9811-22, 2014 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-25238146

RESUMEN

Glioblastoma recurrence involves the persistence of a subpopulation of cells with enhanced tumor-initiating capacity (TIC) that reside within the perivascular space, or niche (PVN). Anti-angiogenic therapies may prevent the formation of new PVN but have not prevented recurrence in clinical trials, suggesting they cannot abrogate TIC activity. We hypothesized that combining anti-angiogenic therapy with blockade of PVN function would have superior anti-tumor activity. We tested this hypothesis in an established intracranial xenograft model of GBM using a monoclonal antibody specific for murine and human VEGF (mcr84) and a Protein Epitope Mimetic (PEM) CXCR4 antagonist, POL5551. When doses of POL5551 were increased to overcome an mcr84-induced improvement in vascular barrier function, combinatorial therapy significantly inhibited intracranial tumor growth and improved survival. Anti-tumor activity was associated with significant changes in tumor cell proliferation and apoptosis, and a reduction in the numbers of perivascular cells expressing the TIC marker nestin. A direct effect on TICs was demonstrated for POL5551, but not mcr84, in three primary patient-derived GBM isolates. These findings indicate that targeting the structure and function of the PVN has superior anti-tumor effect and provide a strong rationale for clinical evaluation of POL5551 and Avastin in patients with GBM.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Terapia Molecular Dirigida , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas/administración & dosificación , Proteínas/farmacología , Distribución Aleatoria , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Blood ; 124(12): 1905-14, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25079359

RESUMEN

The CXCR4/stromal cell-derived factor-1 (SDF-1) axis is essential for cell trafficking and has been shown to regulate tumor progression and metastasis in many tumors including multiple myeloma (MM). A second chemokine receptor for SDF-1, CXCR7 was discovered recently and found on activated endothelial cells. We examined the role of CXCR7 in angiogenic mononuclear cells (AMCs) trafficking in MM. Our data demonstrate that AMCs are circulating in patients with MM and in vivo studies show that they specifically home to areas of MM tumor growth. CXCR7 expression is important for regulating trafficking and homing of AMCs into areas of MM tumor growth and neoangiogenesis. We demonstrate that the CXCR7 inhibitor, POL6926, abrogated trafficking of AMCs to areas of MM tumor progression leading to a significant inhibition of tumor progression. These effects were through regulation of endothelial cells and not through a direct tumor effect, indicating that targeting a bone marrow microenvironmental cell can lead to a delay in MM tumor progression. In conclusion, our studies demonstrate that CXCR7 may play an important role in the regulation of tumor progression in MM through an indirect effect on the recruitment of AMCs to areas of MM tumor growth in the bone marrow niche.


Asunto(s)
Mieloma Múltiple/etiología , Mieloma Múltiple/inmunología , Receptores CXCR/metabolismo , Animales , Materiales Biomiméticos/farmacología , Línea Celular Tumoral , Progresión de la Enfermedad , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Mieloma Múltiple/patología , Células Neoplásicas Circulantes/inmunología , Células Neoplásicas Circulantes/patología , Neovascularización Patológica , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Receptores CXCR/antagonistas & inhibidores , Nicho de Células Madre/inmunología , Microambiente Tumoral/inmunología
9.
J Mass Spectrom ; 45(9): 1075-80, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20814905

RESUMEN

In this study, the simultaneous determination of deoxynivalenol (DON), zearalenone (ZEA), T-2 and HT-2 toxins in foodstuff was investigated. A new kind of multi-mycotoxin immunoaffinity columns (IACs) available on the market (DZT MS-PREP(®)) was tested. A sensitive, selective and accurate method by high-performance liquid chromatography and tandem mass spectrometry was developed, with electrospray ionization mass spectrometer operating in multiple reaction monitoring (MRM) mode, with negative-positive-negative ion switching. The method was used for the analysis of samples marked in Italy, in the frame of official monitoring plans. The advantages of combining IACs and LC-MS/MS technique are as follows: efficient removal of matrix interferences, simple chromatographic outline, high selectivity, low detection limits (DLs) and separation of a wide range of molecules with different physico-chemical properties in a single run. The method was studied on two different matrices, breakfast cereal and baby food, at contamination levels close to Regulation limits (EC) 1126/2007. The recoveries obtained (60-100%) fulfil the performance criteria required by Regulation (EC) 401/2006. The DL is 60 µg/kg for DON and 10 µg/kg for ZEA, T-2 and HT-2. Linearity range of the calibration curves is suitable for adult and baby food.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Grano Comestible/química , Alimentos Infantiles/análisis , Espectrometría de Masas en Tándem/métodos , Tricotecenos/análisis , Zearalenona/análisis , Calibración , Modelos Lineales , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
10.
Mol Pharmacol ; 73(4): 1264-73, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18182480

RESUMEN

We have studied the mechanism of action of Arg(*)-Arg-Nal(2)-Cys(1x)-Tyr-Gln-Lys-(d-Pro)-Pro-Tyr-Arg-Cit-Cys(1x)-Arg-Gly-(d-Pro)(*) (POL3026), a novel specific beta-hairpin mimetic CXC chemokine receptor (CXCR)4 antagonist. POL3026 specifically blocked the binding of anti-CXCR4 monoclonal antibody 12G5 and the intracellular Ca(2+) signal induced by CXC chemokine ligand 12. POL3026 consistently blocked the replication of human immunodeficiency virus (HIV), including a wide panel of X4 and dualtropic strains and subtypes in several culture models, with 50% effective concentrations (EC(50)) at the subnanomolar range, making POL3026 the most potent CXCR4 antagonist described to date. However, 1-[[4-(1,4,8,11-tetrazacyclotetradec-1-ylmethyl)phenyl]methyl]-1,4,8,11-tetrazacyclotetradecane (AMD3100)-resistant and stromal cell-derived factor-1alpha-resistant HIV-1 strains were cross-resistant to POL3026. Time of addition experiments and a multiparametric evaluation of HIV envelope function in the presence of test compounds confirmed the activity of POL3026 at an early step of virus replication: interaction with the coreceptor. Generation of HIV-1 resistance to POL3026 led to the selection of viruses 12- and 25-fold less sensitive and with mutations in gp120, including the V3 loop region. However, POL3026 prevented the emergence of CXCR4-using variants from an R5 HIV-1 strain that may occur in the presence of anti-HIV agents targeting CC chemokine receptor 5.


Asunto(s)
Fármacos Anti-VIH/farmacología , Farmacorresistencia Viral/efectos de los fármacos , Péptidos Cíclicos/farmacología , Receptores CXCR4/antagonistas & inhibidores , Fármacos Anti-VIH/química , Anticuerpos Monoclonales , Bencilaminas , Señalización del Calcio/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Quimiocina CXCL12/farmacología , Quimiotaxis/efectos de los fármacos , Ciclamas , VIH/efectos de los fármacos , VIH/fisiología , Compuestos Heterocíclicos/farmacología , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Tejido Linfoide/efectos de los fármacos , Tejido Linfoide/virología , Macrófagos/efectos de los fármacos , Macrófagos/virología , Péptidos Cíclicos/química , Factores de Tiempo , Proteínas del Envoltorio Viral/metabolismo , Replicación Viral/efectos de los fármacos
11.
Bioorg Med Chem ; 14(24): 8396-404, 2006 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-17010618

RESUMEN

Novel highly potent CXCR4 inhibitors with good pharmacokinetic properties were designed and optimized starting from the naturally occurring beta-hairpin peptide polyphemusin II. The design involved incorporating important residues from polyphemusin II into a macrocyclic template-bound beta-hairpin mimetic. Using a parallel synthesis approach, the potency and ADME properties of the mimetics were optimized in iterative cycles, resulting in the CXCR4 inhibitors POL2438 and POL3026. The inhibitory potencies of these compounds were confirmed in a series of HIV-1 invasion assays in vitro. POL3026 showed excellent plasma stability, high selectivity for CXCR4, favorable pharmacokinetic properties in the dog, and thus has the potential to become a therapeutic compound for application in the treatment of HIV infections (as an entry inhibitor), cancer (for angiogenesis suppression and inhibition of metastasis), inflammation, and in stem cell transplant therapy.


Asunto(s)
Fármacos Anti-VIH/farmacología , Péptidos Catiónicos Antimicrobianos/química , VIH-1/efectos de los fármacos , Imitación Molecular , Oligopéptidos/farmacología , Péptidos Cíclicos/farmacología , Receptores CXCR4/antagonistas & inhibidores , Animales , Fármacos Anti-VIH/química , Fármacos Anti-VIH/farmacocinética , Calcio/metabolismo , Quimiocina CXCL12 , Quimiocinas CXC/metabolismo , Quimiocinas CXC/farmacología , Quimiotaxis/efectos de los fármacos , Perros , Diseño de Fármacos , VIH-1/fisiología , Humanos , Leucemia/patología , Microsomas/efectos de los fármacos , Oligopéptidos/química , Oligopéptidos/farmacocinética , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacocinética , Unión Proteica , Estructura Secundaria de Proteína , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Células Tumorales Cultivadas
12.
Bioorg Med Chem Lett ; 14(23): 5845-9, 2004 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-15501053

RESUMEN

The synthesis and evaluation of a group of 2,6-, 2,7- and 3,6-bis-aminoalkylamido acridones are reported, which show a similar level of activity against telomerase in vitro compared to their acridine counterparts. Computer modelling and calculations of relative binding energies suggest an equivalent binding mode to human intramolecular G-quadruplex DNA, but with significantly reduced affinity, as a result of the limited delocalisation of the acridone chromophore compared to the acridine system. Thermal melting studies on acridone and acridine quadruplex complexes using a FRET approach support these predictions. Long-term cell proliferation studies at sub-cytotoxic doses with two representative acridones using the SKOV3 cell line, show that neither compound produces growth arrest, in contrast with the effects produced by the tri-substituted acridine compound BRACO-19. It is concluded that telomerase inhibitory activity is a necessary though by itself insufficient property in order for cellular growth arrest to occur at sub-toxic concentrations, and that tight quadruplex binding is also required.


Asunto(s)
Acridinas/química , Acridinas/metabolismo , ADN/metabolismo , Telomerasa/antagonistas & inhibidores , Telomerasa/metabolismo , Acridinas/farmacología , Acridonas , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos , G-Cuádruplex , Humanos , Modelos Moleculares , Unión Proteica/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...