Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
3.
Nat Metab ; 2(12): 1413-1426, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33230295

RESUMEN

Inhibiting glycolysis remains an aspirational approach for the treatment of cancer. We have previously identified a subset of cancers harbouring homozygous deletion of the glycolytic enzyme enolase (ENO1) that have exceptional sensitivity to inhibition of its redundant paralogue, ENO2, through a therapeutic strategy known as collateral lethality. Here, we show that a small-molecule enolase inhibitor, POMHEX, can selectively kill ENO1-deleted glioma cells at low-nanomolar concentrations and eradicate intracranial orthotopic ENO1-deleted tumours in mice at doses well-tolerated in non-human primates. Our data provide an in vivo proof of principle of the power of collateral lethality in precision oncology and demonstrate the utility of POMHEX for glycolysis inhibition with potential use across a range of therapeutic settings.


Asunto(s)
Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/genética , Proteínas de Unión al ADN/genética , Inhibidores Enzimáticos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Fosfopiruvato Hidratasa/antagonistas & inhibidores , Proteínas Supresoras de Tumor/genética , Animales , Línea Celular Tumoral , Femenino , Glioma/tratamiento farmacológico , Glucólisis/efectos de los fármacos , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones SCID , Fosfopiruvato Hidratasa/genética , Medicina de Precisión , Eliminación de Secuencia , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Nat Chem Biol ; 12(12): 1053-1058, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27723749

RESUMEN

Despite being crucial for energy generation in most forms of life, few if any microbial antibiotics specifically inhibit glycolysis. To develop a specific inhibitor of the glycolytic enzyme enolase 2 (ENO2) for the treatment of cancers with deletion of ENO1 (encoding enolase 1), we modeled the synthetic tool compound inhibitor phosphonoacetohydroxamate (PhAH) into the active site of human ENO2. A ring-stabilized analog of PhAH, in which the hydroxamic nitrogen is linked to Cα by an ethylene bridge, was predicted to increase binding affinity by stabilizing the inhibitor in a bound conformation. Unexpectedly, a structure-based search revealed that our hypothesized backbone-stabilized PhAH bears strong similarity to SF2312, a phosphonate antibiotic of unknown mode of action produced by the actinomycete Micromonospora, which is active under anaerobic conditions. Here, we present multiple lines of evidence, including a novel X-ray structure, that SF2312 is a highly potent, low-nanomolar inhibitor of enolase.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Organofosfonatos/farmacología , Fosfopiruvato Hidratasa/antagonistas & inhibidores , Pirrolidinonas/farmacología , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Humanos , Modelos Moleculares , Estructura Molecular , Organofosfonatos/química , Fosfopiruvato Hidratasa/metabolismo , Pirrolidinonas/química , Relación Estructura-Actividad
5.
Oncotarget ; 7(32): 51435-51449, 2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27283492

RESUMEN

Metformin displays antileukemic effects partly due to activation of AMPK and subsequent inhibition of mTOR signaling. Nevertheless, Metformin also inhibits mitochondrial electron transport at complex I in an AMPK-independent manner, Here we report that Metformin and rotenone inhibit mitochondrial electron transport and increase triglyceride levels in leukemia cell lines, suggesting impairment of fatty acid oxidation (FAO). We also report that, like other FAO inhibitors, both agents and the related biguanide, Phenformin, increase sensitivity to apoptosis induction by the bcl-2 inhibitor ABT-737 supporting the notion that electron transport antagonizes activation of the intrinsic apoptosis pathway in leukemia cells. Both biguanides and rotenone induce superoxide generation in leukemia cells, indicating that oxidative damage may sensitize toABT-737 induced apoptosis. In addition, we demonstrate that Metformin sensitizes leukemia cells to the oligomerization of Bak, suggesting that the observed synergy with ABT-737 is mediated, at least in part, by enhanced outer mitochondrial membrane permeabilization. Notably, Phenformin was at least 10-fold more potent than Metformin in abrogating electron transport and increasing sensitivity to ABT-737, suggesting that this agent may be better suited for targeting hematological malignancies. Taken together, our results suggest that inhibition of mitochondrial metabolism by Metformin or Phenformin is associated with increased leukemia cell susceptibility to induction of intrinsic apoptosis, and provide a rationale for clinical studies exploring the efficacy of combining biguanides with the orally bioavailable derivative of ABT-737, Venetoclax.


Asunto(s)
Apoptosis/efectos de los fármacos , Biguanidas/farmacología , Compuestos de Bifenilo/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Transporte de Electrón/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Nitrofenoles/farmacología , Sulfonamidas/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Metformina/farmacología , Mitocondrias/metabolismo , Fenformina/farmacología , Piperazinas/farmacología , Rotenona/farmacología , Células U937
6.
Mol Cancer Ther ; 15(7): 1757-67, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27196777

RESUMEN

UNLABELLED: Ionizing radiation (IR) is a key therapeutic regimen for many head and neck cancers (HNC). However, the 5-year overall survival rate for locally advanced HNCs is approximately 50% and better therapeutic efficacy is needed. NAD(P)H: quinone oxidoreductase 1 (NQO1) is overexpressed in many cancers, and ß-lapachone (ß-lap), a unique NQO1 bioactivatable drug, exploits this enzyme to release massive reactive oxygen species (ROS) that synergize with IR to kill by programmed necrosis. ß-Lap represents a novel therapeutic opportunity in HNC leading to tumor-selective lethality that will enhance the efficacy of IR. Immunohistochemical staining and Western blot assays were used to assess the expression levels of NQO1 in HNC cells and tumors. Forty-five percent of endogenous HNCs expressed elevated NQO1 levels. In addition, multiple HNC cell lines and tumors demonstrated elevated levels of NQO1 expression and activity and were tested for anticancer lethality and radiosensitization by ß-lap using long-term survival assays. The combination of nontoxic ß-lap doses and IR significantly enhanced NQO1-dependent tumor cell lethality, increased ROS, TUNEL-positive cells, DNA damage, NAD(+), and ATP consumption, and resulted in significant antitumor efficacy and prolonged survival in two xenograft murine HNC models, demonstrating ß-lap radiosensitization of HNCs through a NQO1-dependent mechanism. This translational study offers a potential biomarker-driven strategy using NQO1 expression to select tumors susceptible to ß-lap-induced radiosensitization. Mol Cancer Ther; 15(7); 1757-67. ©2016 AACR.


Asunto(s)
Expresión Génica , Neoplasias de Cabeza y Cuello/genética , NAD(P)H Deshidrogenasa (Quinona)/genética , Tolerancia a Radiación/genética , Radiación Ionizante , Adenosina Trifosfato/metabolismo , Animales , Catalasa/genética , Catalasa/metabolismo , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Modelos Animales de Enfermedad , Activación Enzimática , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Especies Reactivas de Oxígeno/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
7.
PLoS Pathog ; 11(3): e1004770, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25822986

RESUMEN

Imatinib mesylate (Gleevec) inhibits Abl1, c-Kit, and related protein tyrosine kinases (PTKs) and serves as a therapeutic for chronic myelogenous leukemia and gastrointestinal stromal tumors. Imatinib also has efficacy against various pathogens, including pathogenic mycobacteria, where it decreases bacterial load in mice, albeit at doses below those used for treating cancer. We report that imatinib at such low doses unexpectedly induces differentiation of hematopoietic stem cells and progenitors in the bone marrow, augments myelopoiesis but not lymphopoiesis, and increases numbers of myeloid cells in blood and spleen. Whereas progenitor differentiation relies on partial inhibition of c-Kit by imatinib, lineage commitment depends upon inhibition of other PTKs. Thus, imatinib mimics "emergency hematopoiesis," a physiological innate immune response to infection. Increasing neutrophil numbers by adoptive transfer sufficed to reduce mycobacterial load, and imatinib reduced bacterial load of Franciscella spp., which do not utilize imatinib-sensitive PTKs for pathogenesis. Thus, potentiation of the immune response by imatinib at low doses may facilitate clearance of diverse microbial pathogens.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Francisella/inmunología , Infecciones por Bacterias Gramnegativas/inmunología , Mesilato de Imatinib/farmacología , Mielopoyesis/efectos de los fármacos , Neutrófilos/inmunología , Animales , Diferenciación Celular/inmunología , Recuento de Leucocitos , Ratones , Mielopoyesis/inmunología
8.
Cell Cycle ; 13(16): 2587-99, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25486199

RESUMEN

Indole-3-carbinol (I3C) is a natural anti-carcinogenic compound found at high concentrations in Brassica vegetables. I3C was recently reported to inhibit neutrophil elastase (NE) activity, while consequently limiting the proteolytic processing of full length cyclin E into pro-tumorigenic low molecular weight cyclin E (LMW-E). In this study, we hypothesized that inhibition of NE activity and resultant LMW-E generation is critical to the anti-tumor effects of I3C. LMW-E was predominately expressed by ERα-negative breast cancer cell lines. However, ERα-positive cell lines demonstrated the greatest sensitivity to the anti-tumor effects of I3C and its more potent N-alkoxy derivatives. We found that I3C was incapable of inhibiting NE activity or the generation of LMW-E. Therefore, this pathway did not contribute to the anti-tumor activity of I3C. Gene expression analyzes identified ligand-activated aryl hydrocarbon receptor (AhR), which mediated sensitivity to the anti-tumor effects of I3C in ERα-positive MCF-7 cells. In this model system, the reactive oxygen species (ROS)-induced upregulation of ATF-3 and pro-apoptotic BH3-only proteins (e.g. NOXA) contributed to the sensitivity of ERα-positive breast cancer cells to the anti-tumor effects of I3C. Overexpression of ERα in MDA-MB-231 cells, which normally lack ERα expression, increased sensitivity to the anti-tumor effects of I3C, demonstrating a direct role for ERα in mediating the sensitivity of breast cancer cell lines to I3C. Our results suggest that ERα signaling amplified the pro-apoptotic effect of I3C-induced AhR signaling in luminal breast cancer cell lines, which was mediated in part through oxidative stress induced upregulation of ATF-3 and downstream BH3-only proteins.


Asunto(s)
Alcoholes/farmacología , Antineoplásicos/farmacología , Neoplasias de la Mama/metabolismo , Receptor alfa de Estrógeno/metabolismo , Indoles/farmacología , Redes y Vías Metabólicas/efectos de los fármacos , Alcoholes/química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina E/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/química , Elastasa de Leucocito/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal/efectos de los fármacos
9.
Tetrahedron ; 70(4): 984-990, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-25110363

RESUMEN

A virtual library of 54 inositol analog mimics of In(1,4,5)P3 has been docked, scored, and ranked within the binding site of human inositol 1,4,5-trisphosphate 3-kinase A (IP3-3KA). Chemical synthesis of the best scoring structure that also met distance criteria for 3'-OH to -P in Phosphate has been attempted along with the synthesis of (1S,2R,3S,4S)-3-fluoro-2,4-dihydroxycyclohexanecarboxylic acid as an inositol analog, useful for non-invasive visualization and quantitation of IP3-3KA enzymatic activity.

10.
Mol Cancer Ther ; 13(7): 1848-59, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24739393

RESUMEN

Aberrant activation of multiple signaling pathways is common in acute myelogenous leukemia (AML) cells, which can be linked to a poor prognosis for patients with this disease. Previous research with mTOR or MEK inhibitors revealed cytostatic, rather than cytotoxic, effects in in vitro and in vivo AML models. We evaluated the combination effect of the mTOR inhibitor AZD8055 and the MEK inhibitor selumetinib on human AML cell lines and primary AML samples. This combination demonstrated synergistic proapoptotic effects in AML cells with high basal activation of MEK and mTOR. We next incorporated the BH3 mimetic ABT-737 into this combination regimen to block Bcl-2, which further enhanced the apoptogenic effect of MEK/mTOR inhibition. The combination treatment also had a striking proapoptotic effect in CD33(+)/CD34(+) AML progenitor cells from primary AML samples with NRAS mutations. Mechanistically, upregulation of the proapoptotic protein Bim, accompanied by the downregulation of the antiapoptotic protein Mcl-1 (mainly via protein degradation), seemed to play critical roles in enhancing the combination drug effect. Furthermore, the modulation of survivin, Bax, Puma, and X-chromosome-linked inhibitor of apoptosis protein (XIAP) expression suggested a role for mitochondria-mediated apoptosis in the cytotoxicity of the drug combination. Consequently, the concomitant blockade of prosurvival MEK/mTOR signaling and the deactivation of Bcl-2 could provide a mechanism-based integrated therapeutic strategy for the eradication of AML cells.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Bencimidazoles/farmacología , Leucemia Mieloide Aguda/tratamiento farmacológico , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Morfolinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Bencimidazoles/administración & dosificación , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Expresión Génica , Humanos , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/patología , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Morfolinas/administración & dosificación , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Transfección , Células U937
11.
Antioxid Redox Signal ; 21(2): 237-50, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24512128

RESUMEN

AIMS: ß-Lapachone (ß-lap), a novel radiosensitizer with potent antitumor efficacy alone, selectively kills solid cancers that over-express NAD(P)H: quinone oxidoreductase 1 (NQO1). Since breast or other solid cancers have heterogeneous NQO1 expression, therapies that reduce the resistance (e.g., NQO1(low)) of tumor cells will have significant clinical advantages. We tested whether NQO1-proficient (NQO1(+)) cells generated sufficient hydrogen peroxide (H2O2) after ß-lap treatment to elicit bystander effects, DNA damage, and cell death in neighboring NQO1(low) cells. RESULTS: ß-Lap showed NQO1-dependent efficacy against two triple-negative breast cancer (TNBC) xenografts. NQO1 expression variations in human breast cancer patient samples were noted, where ~60% cancers over-expressed NQO1, with little or no expression in associated normal tissue. Differential DNA damage and lethality were noted in NQO1(+) versus NQO1-deficient (NQO1(-)) TNBC cells and xenografts after ß-lap treatment. ß-Lap-treated NQO1(+) cells died by programmed necrosis, whereas co-cultured NQO1(-) TNBC cells exhibited DNA damage and caspase-dependent apoptosis. NQO1 inhibition (dicoumarol) or H2O2 scavenging (catalase [CAT]) blocked all responses. Only NQO1(-) cells neighboring NQO1(+) TNBC cells responded to ß-lap in vitro, and bystander effects correlated well with H2O2 diffusion. Bystander effects in NQO1(-) cells in vivo within mixed 50:50 co-cultured xenografts were dramatic and depended on NQO1(+) cells. However, normal human cells in vitro or in vivo did not show bystander effects, due to elevated endogenous CAT levels. Innovation and Conclusions: NQO1-dependent bystander effects elicited by NQO1 bioactivatable drugs (ß-lap or deoxynyboquinone [DNQ]) likely contribute to their efficacies, killing NQO1(+) solid cancer cells and eliminating surrounding heterogeneous NQO1(low) cancer cells. Normal cells/tissue are protected by low NQO1:CAT ratios.


Asunto(s)
Efecto Espectador/efectos de los fármacos , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/farmacología , Quinonas/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Femenino , Humanos , Ratones , Ratones Desnudos , NAD(P)H Deshidrogenasa (Quinona)/deficiencia , NAD(P)H Deshidrogenasa (Quinona)/genética , Oxidación-Reducción/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Bioorg Med Chem ; 22(4): 1450-8, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24457091

RESUMEN

A series of degrasyn-like symmetrical compounds have been designed, synthesized, and screened against B cell malignancy (multiple myeloma, mantle cell lymphoma) cell lines. The lead compounds T5165804 and CP2005 showed higher nanomolar potency against these tumor cells in comparison to degrasyn and inhibited Usp9x activity in vitro and in intact cells. These observations suggest that this new class of compounds holds promise as cancer therapeutic agents.


Asunto(s)
Antineoplásicos/química , Nitrilos/química , Piridinas/química , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cianoacrilatos , Dimerización , Humanos , Modelos Moleculares , Mieloma Múltiple/tratamiento farmacológico , Nitrilos/farmacología , Nitrilos/uso terapéutico , Piridinas/farmacología , Piridinas/uso terapéutico , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/metabolismo
13.
Bioorg Med Chem ; 22(1): 623-32, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24280068

RESUMEN

We synthesized two series of imatinib mesylate (STI-571) analogs to develop a Bcr-Abl and c-KIT receptor-specific labeling agent for positron emission tomography (PET) imaging to measure Bcr-Abl and c-KIT expression levels in a mouse model. The methods of molecular modeling, synthesis of STI-571 and its analogs, in vitro kinase assays, and radiolabeling are described. Molecular modeling revealed that these analogs bind the same Bcr-Abl and c-KIT binding sites as those bound by STI-571. The analogs potently inhibit the tyrosine kinase activity of Bcr-Abl and c-KIT, similarly to STI-571. [(18)F]-labeled STI-571 was prepared with high specific activity (75 GBq/µmol) by nucleophilic displacement and an average radiochemical yield of 12%. [(131)I]-labeled STI-571 was prepared with high purity (>95%) and an average radiochemical yield of 23%. The uptake rates of [(18)F]-STI-571 in K562 cells expressing Abl and in U87WT cells overexpressing c-KIT were significantly higher than those in the U87 cell and could be inhibited by STI-71 (confirming the specificity of uptake). PET scans of K562 and U87WT tumor-bearing mice with [(18)F]-STI-571 as a contrast agent showed visible tumor uptake and tumor-to-non-target contrast.


Asunto(s)
Antineoplásicos/uso terapéutico , Benzamidas/uso terapéutico , Proteínas de Fusión bcr-abl/metabolismo , Piperazinas/uso terapéutico , Tomografía de Emisión de Positrones/métodos , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/uso terapéutico , Animales , Antineoplásicos/química , Benzamidas/química , Modelos Animales de Enfermedad , Humanos , Mesilato de Imatinib , Ratones , Modelos Moleculares , Piperazinas/química , Pirimidinas/química
14.
Bioorg Med Chem ; 22(1): 435-9, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24280069

RESUMEN

A gram scale synthesis of the glucuronide metabolites of curcumin were completed in four steps. The newly synthesized curcumin glucuronide compounds 2 and 3 along with curcumin 1 were tested and their anti-proliferative effects against KBM-5, Jurkat cell, U266, and A549 cell lines were reported. Biological data revealed that as much as 1 µM curcumin 1 exhibited anticancer activity and almost 100% cell kill was noted at 10 µM on two out of four cell lines; while curcumin mono-glucuronide 2 as well as di-glucuronide 3 displayed no suppression of cell proliferation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Curcumina/farmacología , Línea Celular Tumoral , Curcumina/síntesis química , Humanos , Células Jurkat , Relación Estructura-Actividad
15.
PLoS One ; 8(10): e77262, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204784

RESUMEN

OBJECTIVE: The iboga alkaloids are a class of small molecules defined structurally on the basis of a common ibogamine skeleton, some of which modify opioid withdrawal and drug self-administration in humans and preclinical models. These compounds may represent an innovative approach to neurobiological investigation and development of addiction pharmacotherapy. In particular, the use of the prototypic iboga alkaloid ibogaine for opioid detoxification in humans raises the question of whether its effect is mediated by an opioid agonist action, or if it represents alternative and possibly novel mechanism of action. The aim of this study was to independently replicate and extend evidence regarding the activation of µ-opioid receptor (MOR)-related G proteins by iboga alkaloids. METHODS: Ibogaine, its major metabolite noribogaine, and 18-methoxycoronaridine (18-MC), a synthetic congener, were evaluated by agonist-stimulated guanosine-5´-O-(γ-thio)-triphosphate ([(35)S]GTPγS) binding in cells overexpressing the recombinant MOR, in rat thalamic membranes, and autoradiography in rat brain slices. RESULTS AND SIGNIFICANCE: In rat thalamic membranes ibogaine, noribogaine and 18-MC were MOR antagonists with functional Ke values ranging from 3 uM (ibogaine) to 13 uM (noribogaine and 18MC). Noribogaine and 18-MC did not stimulate [(35)S]GTPγS binding in Chinese hamster ovary cells expressing human or rat MORs, and had only limited partial agonist effects in human embryonic kidney cells expressing mouse MORs. Ibogaine did not did not stimulate [(35)S]GTPγS binding in any MOR expressing cells. Noribogaine did not stimulate [(35)S]GTPγS binding in brain slices using autoradiography. An MOR agonist action does not appear to account for the effect of these iboga alkaloids on opioid withdrawal. Taken together with existing evidence that their mechanism of action also differs from that of other non-opioids with clinical effects on opioid tolerance and withdrawal, these findings suggest a novel mechanism of action, and further justify the search for alternative targets of iboga alkaloids.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/farmacología , Ibogaína/análogos & derivados , Ibogaína/farmacología , Receptores Opioides mu/metabolismo , Tálamo/efectos de los fármacos , Animales , Autorradiografía , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Femenino , Expresión Génica , Guanosina 5'-O-(3-Tiotrifosfato)/farmacología , Células HEK293 , Humanos , Especificidad de Órganos , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/agonistas , Receptores Opioides mu/antagonistas & inhibidores , Receptores Opioides mu/genética , Síndrome de Abstinencia a Sustancias/prevención & control , Tálamo/metabolismo
16.
Mol Cancer Ther ; 12(10): 2110-20, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23883585

RESUMEN

Improving patient outcome by personalized therapy involves a thorough understanding of an agent's mechanism of action. ß-Lapachone (clinical forms, Arq501/Arq761) has been developed to exploit dramatic cancer-specific elevations in the phase II detoxifying enzyme NAD(P)H:quinone oxidoreductase (NQO1). NQO1 is dramatically elevated in solid cancers, including primary and metastatic [e.g., triple-negative (ER-, PR-, Her2/Neu-)] breast cancers. To define cellular factors that influence the efficacy of ß-lapachone using knowledge of its mechanism of action, we confirmed that NQO1 was required for lethality and mediated a futile redox cycle where ∼120 moles of superoxide were formed per mole of ß-lapachone in 2 minutes. ß-Lapachone induced reactive oxygen species (ROS), stimulated DNA single-strand break-dependent poly(ADP-ribose) polymerase-1 (PARP1) hyperactivation, caused dramatic loss of essential nucleotides (NAD(+)/ATP), and elicited programmed necrosis in breast cancer cells. Although PARP1 hyperactivation and NQO1 expression were major determinants of ß-lapachone-induced lethality, alterations in catalase expression, including treatment with exogenous enzyme, caused marked cytoprotection. Thus, catalase is an important resistance factor and highlights H2O2 as an obligate ROS for cell death from this agent. Exogenous superoxide dismutase enhanced catalase-induced cytoprotection. ß-Lapachone-induced cell death included apoptosis-inducing factor (AIF) translocation from mitochondria to nuclei, TUNEL+ staining, atypical PARP1 cleavage, and glyceraldehyde 3-phosphate dehydrogenase S-nitrosylation, which were abrogated by catalase. We predict that the ratio of NQO1:catalase activities in breast cancer versus associated normal tissue are likely to be the major determinants affecting the therapeutic window of ß-lapachone and other NQO1 bioactivatable drugs.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/administración & dosificación , Poli(ADP-Ribosa) Polimerasas/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Catalasa/genética , Catalasa/metabolismo , Roturas del ADN de Cadena Simple/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/metabolismo , NAD(P)H Deshidrogenasa (Quinona)/genética , Necrosis/genética , Necrosis/patología , Poli(ADP-Ribosa) Polimerasa-1 , Especies Reactivas de Oxígeno/metabolismo
17.
Bioorg Med Chem ; 21(17): 5182-7, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23859776

RESUMEN

An improved method for the synthesis of 17ß-hydroxy-16α-iodo-wortmannin along with the first synthesis of 17ß-hydroxy-16α-iodoPX866 and [(131)I] radiolabeled 17ß-hydroxy-16α-[(131)I]iodo-wortmannin, as potential PET tracers for PI3K was also described. The differences between wortmannin and its iodo analogue were compared by covalently docking each structure to L833 in PI3K.


Asunto(s)
Androstadienos/química , Androstadienos/síntesis química , Gonanos/síntesis química , Radiofármacos/síntesis química , Sitios de Unión , Gonanos/química , Radioisótopos de Yodo/química , Marcaje Isotópico , Simulación del Acoplamiento Molecular , Fosfatidilinositol 3-Quinasa/química , Fosfatidilinositol 3-Quinasa/metabolismo , Tomografía de Emisión de Positrones , Estructura Terciaria de Proteína , Radiofármacos/química , Wortmanina
18.
J Biol Chem ; 288(27): 19414-28, 2013 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-23703610

RESUMEN

Activation of the integrin family of cell adhesion receptors on progenitor cells may be a viable approach to enhance the effects of stem cell-based therapies by improving cell retention and engraftment. Here, we describe the synthesis and characterization of the first small molecule agonist identified for the integrin α4ß1 (also known as very late antigen-4 or VLA-4). The agonist, THI0019, was generated via two structural modifications to a previously identified α4ß1 antagonist. THI0019 greatly enhanced the adhesion of cultured cell lines and primary progenitor cells to α4ß1 ligands VCAM-1 and CS1 under both static and flow conditions. Furthermore, THI0019 facilitated the rolling and spreading of cells on VCAM-1 and the migration of cells toward SDF-1α. Molecular modeling predicted that the compound binds at the α/ß subunit interface overlapping the ligand-binding site thus indicating that the compound must be displaced upon ligand binding. In support of this model, an analog of THI0019 modified to contain a photoreactive group was used to demonstrate that when cross-linked to the integrin, the compound behaves as an antagonist instead of an agonist. In addition, THI0019 showed cross-reactivity with the related integrin α4ß7 as well as α5ß1 and αLß2. When cross-linked to αLß2, the photoreactive analog of THI0019 remained an agonist, consistent with it binding at the α/ß subunit interface and not at the ligand-binding site in the inserted ("I") domain of the αL subunit. Co-administering progenitor cells with a compound such as THI0019 may provide a mechanism for enhancing stem cell therapy.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Integrina alfa4beta1/agonistas , Modelos Moleculares , Células Madre/metabolismo , Antígeno CD11a/genética , Antígeno CD11a/metabolismo , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Compuestos Heterocíclicos de 4 o más Anillos/química , Células Endoteliales de la Vena Umbilical Humana , Humanos , Integrina alfa4beta1/genética , Integrina alfa4beta1/metabolismo , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Células Jurkat , Células Madre/citología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
19.
Bioorg Med Chem ; 21(4): 932-9, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23294827

RESUMEN

Curcumin (diferuloylmethane) is a potent anti-inflammatory and anti-tumorigenic agent that has shown preclinical activity in diverse cancers. Curcumin up-regulates heat shock protein 70 (hsp70) mRNA in several different cancer cell lines. Hsp70 contributes to an escape from the apoptotic effects of curcumin by several different mechanisms including prevention of the release of apoptosis inducing factor from the mitochondria and inhibition of caspases 3 and 9. Previously we showed that the combination of curcumin plus a heat shock protein inhibitor was synergistic in its down-regulation of the proliferation of a human schwannoma cell line (HEI-193) harboring an NF2 mutation, possibly because curcumin up-regulated hsp70, which also binds merlin, the NF2 gene product. In order to determine if curcumin also interacts directly with hsp70 and to discover other binding partners of curcumin, we synthesized biotinylated curcumin (bio-curcumin) and treated HEI-193 schwannoma cells. Cell lysates were prepared and incubated with avidin-coated beads. Peptides pulled down from this reaction were sequenced and it was determined that biotinylated curcumin bound hsp70, hsp90, 3-phosphoglycerate dehydrogenase, and a ß-actin variant. These binding partners may serve to further elucidate the underlying mechanisms of curcumin's actions.


Asunto(s)
Curcumina/química , Proteínas HSP70 de Choque Térmico/química , Proteínas HSP90 de Choque Térmico/química , Fosfoglicerato-Deshidrogenasa/química , Sitios de Unión , Biotina/química , Línea Celular Tumoral , Curcumina/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Neurilemoma/metabolismo , Neurilemoma/patología , Fosfoglicerato-Deshidrogenasa/metabolismo , Unión Proteica , Estructura Terciaria de Proteína
20.
Mol Cancer Ther ; 12(5): 654-62, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23303403

RESUMEN

We have previously shown that the antiallergic drug cromolyn blocks S100P interaction with its receptor receptor for advanced glycation end product (RAGE) and improves gemcitabine effectiveness in pancreatic ductal adenocarcinoma (PDAC). However, the concentration required to achieve its effectiveness was high (100 µmol/L). In this study, we designed and synthesized analogs of cromolyn and analyzed their effectiveness compared with the parent molecule. An ELISA was used to confirm the binding of S100P with RAGE and to test the effectiveness of the different analogs. Analog 5-methyl cromolyn (C5OH) blocked S100P binding as well as the increases in NF-κB activity, cell growth, and apoptosis normally caused by S100P. In vivo C5OH systemic delivery reduced NF-κB activity to a greater extent than cromolyn and at 10 times lesser dose (50 mg vs. 5 mg). Treatment of mice-bearing syngeneic PDAC tumors showed that C5OH treatment reduced both tumor growth and metastasis. C5OH treatment of nude mice bearing orthotopic highly aggressive pancreatic Mpanc96 cells increased the overall animal survival. Therefore, the cromolyn analog, C5OH, was found to be more efficient and potent than cromolyn as a therapeutic for PDAC.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Unión al Calcio/antagonistas & inhibidores , Carcinoma Ductal Pancreático/metabolismo , Cromolin Sódico/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Neoplasias Pancreáticas/metabolismo , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Proteínas de Unión al Calcio/metabolismo , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/mortalidad , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromolin Sódico/análogos & derivados , Cromolin Sódico/química , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Humanos , Masculino , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Unión Proteica/efectos de los fármacos , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA