Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Alzheimers Dement ; 2022 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715930

RESUMEN

INTRODUCTION: AMYPAD Diagnostic and Patient Management Study (DPMS) aims to investigate the clinical utility and cost-effectiveness of amyloid-PET in Europe. Here we present participants' baseline features and discuss the representativeness of the cohort. METHODS: Participants with subjective cognitive decline plus (SCD+), mild cognitive impairment (MCI), or dementia were recruited in eight European memory clinics from April 16, 2018, to October 30, 2020, and randomized into three arms: ARM1, early amyloid-PET; ARM2, late amyloid-PET; and ARM3, free-choice. RESULTS: A total of 840 participants (244 SCD+, 341 MCI, and 255 dementia) were enrolled. Sociodemographic/clinical features did not differ significantly among recruiting memory clinics or with previously reported cohorts. The randomization assigned 35% of participants to ARM1, 32% to ARM2, and 33% to ARM3; cognitive stages were distributed equally across the arms. DISCUSSION: The features of AMYPAD-DPMS participants are as expected for a memory clinic population. This ensures the generalizability of future study results.

2.
Sci Rep ; 11(1): 21576, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34732782

RESUMEN

Many agents targeting the colchicine binding site in tubulin have been developed as potential anticancer agents. However, none has successfully made it to the clinic, due mainly to dose limiting toxicities and the emergence of multi-drug resistance. Chalcones targeting tubulin have been proposed as a safe and effective alternative. We have shown previously that quinolone chalcones target tubulin and maintain potent anti-proliferative activity vis-à-vis colchicine, while also having high tolerability and low toxicity in mouse models of cancer and refractivity to multi-drug resistance mechanisms. To identify the most effective anticancer chalcone compound, we synthesized 17 quinolone-chalcone derivatives based on our previously published CTR-17 and CTR-20, and then carried out a structure-activity relationship study. We identified two compounds, CTR-21 [((E)-8-Methoxy-3-(3-(2-methoxyphenyl)-3-oxoprop-1-enyl) quinolin-2(1H)-one)] and CTR-32 [((E)-3-(3-(2-ethoxyphenyl)-3-oxoprop-1-enyl) quinolin-2(1H)-one)] as potential leads, which contain independent moieties that play a significant role in their enhanced activities. At the nM range, CTR-21 and CTR-32 effectively kill a panel of different cancer cells originated from a variety of different tissues including breast and skin. Both compounds also effectively kill multi-drug resistant cancer cells. Most importantly, CTR-21 and CTR-32 show a high degree of selectivity against cancer cells. In silico, both of them dock near the colchicine-binding site with similar energies. Whereas both CTR-21 and CTR-32 effectively prevents tubulin polymerization, leading to the cell cycle arrest at G2/M, CTR-21 has more favorable metabolic properties. Perhaps not surprisingly, the combination of CTR-21 and ABT-737, a Bcl-2 inhibitor, showed synergistic effect in killing cancer cells, since we previously found the "parental" CTR-20 also exhibited synergism. Taken together, CTR-21 can potentially be a highly effective and relatively safe anticancer drug.


Asunto(s)
Chalconas/química , Diseño de Fármacos/métodos , Quinolonas/química , Relación Estructura-Actividad , Animales , Apoptosis , Línea Celular Tumoral , Chalconas/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Citometría de Flujo , Células HeLa , Humanos , Enlace de Hidrógeno , Leucocitos Mononucleares/metabolismo , Células MCF-7 , Ratones , Microsomas/química , Paclitaxel/farmacología , Quinolonas/farmacología , Tubulina (Proteína)/química , Moduladores de Tubulina/farmacología
3.
PLoS One ; 16(9): e0257143, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34492076

RESUMEN

Clostridioides difficile infection (CDI) is the primary cause of hospital-acquired diarrhea, and responsible for over 500,000 enteric infections a year in the United States alone. Although most patients with CDI are successfully treated with metronidazole or vancomycin, the high rate of recurrence is still a serious problem, in which case these antibiotics are usually not very effective. The primary objective of this research is to develop a potentially effective therapeutic agent against C. difficile that are resistant to metronidazole or vancomycin. The susceptibility to metronidazole and vancomycin was examined with 194 C. difficile clinical isolates. Sixty of these isolates chosen based on a variety of criteria were examined for their susceptibility against the 4-chloro-1-piperidin-1ylmethyl-1H-indole-2,3-dione compound (Raja 42), a novel isatin-benzothiazole analogue containing a gamma-lactam structure, as we previously found that this novel compound is effective against a variety of different bacteria. Most of the 60 isolates were resistant to ceftriaxone and ciprofloxacin, raising the possibility that they might have been exposed previously to these or structurally similar antibiotics (e.g., ß-lactam and quinolone compounds). Among the isolates, 48 (80%) and 54 (90%) were susceptible to metronidazole and vancomycin, respectively. Raja 42 was found to be effective against most of the isolates, especially so against metronidazole-resistant C. difficile. Most importantly, five isolates that show resistance to metronidazole and vancomycin were sensitive to Raja 42. Thus, Raja 42, a gamma lactam antibiotic, has the potential to effectively control C. difficile strains that are resistant to metronidazole and vancomycin.


Asunto(s)
Clostridioides difficile/efectos de los fármacos , Indoles/farmacología , Clostridioides difficile/aislamiento & purificación , Humanos , Indoles/química , Metronidazol/farmacología , Pruebas de Sensibilidad Microbiana , Vancomicina/farmacología
4.
Int J Pharm ; 606: 120849, 2021 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-34216770

RESUMEN

Cancer treatment remains unsatisfactory with high rates of recurrence and metastasis. Immunomodulatory agents capable of promoting cellular antitumor immunity while inhibiting the local immunosuppressive tumor microenvironment could greatly improve cancer treatment. We have developed a multi-targeted mannosylated cationic liposome delivery system containing muramyl dipeptide (DS) and low doses of the chemotherapeutic agent cytarabine (Ara-C). Immunomodulation of primary immune cells and immortalized cancer cell lines by Ara-C/DS was assessed by measuring cytokine levels and surface marker expression. As a proof of concept, the generation of targeted cellular immunity was investigated in the context of responses to viral antigens. This report is the first demonstrating that Ara-C combined with DS can modulate immune responses and revert immunosuppression as evidenced by increased IFN-γ and IL-12p40 without changes in IL-10 in peripheral blood mononuclear cells, and increased CD80 and decreased CD163 on immunosuppressive macrophages. Furthermore, Ara-C/DS increased MHC class I expression on cancer cells while increasing the production of antigen-specific IFN-γ+ CD8+ T cells in viral peptide-challenged lymphocytes from both humans and vaccinated mice. Taken together, these results are the first to document immunomodulatory properties of Ara-C linked with recognition of antigens and potentially the generation of antitumor immune memory.


Asunto(s)
Citarabina , Liposomas , Animales , Linfocitos T CD8-positivos , Inmunidad Celular , Inmunomodulación , Leucocitos Mononucleares , Ratones
5.
Molecules ; 25(16)2020 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-32824689

RESUMEN

Natural products remain a viable source of novel therapeutics, and as detection and extraction techniques improve, we can identify more molecules from a broader set of plant tissues. The aim of this study was an investigation of the cytotoxic and anti-plasmodial activities of the methanol extract from Stephania dielsiana Y.C. Wu leaves and its isolated compounds. Our study led to the isolation of seven alkaloids, among which oxostephanine (1) is the most active against several cancer cell lines including HeLa, MDA-MB231, MDA-MB-468, MCF-7, and non-cancer cell lines, such as 184B5 and MCF10A, with IC50 values ranging from 1.66 to 4.35 µM. Morever, oxostephanine (1) is on average two-fold more active against cancer cells than stephanine (3), having a similar chemical structure. Cells treated with oxostephanine (1) are arrested at G2/M cell cycle, followed by the formation of aneuploidy and apoptotic cell death. The G2/M arrest appears to be due, at least in part, to the inactivation of Aurora kinases, which is implicated in the onset and progression of many forms of human cancer. An in-silico molecular modeling study suggests that oxostephanine (1) binds to the ATP binding pocket of Aurora kinases to inactivate their activities. Unlike oxostephanine (1), thailandine (2) is highly effective against only the triple-negative MDA-MB-468 breast cancer cells. However, it showed excellent selectivity against the cancer cell line when compared to its effects on non-cancer cells. Furthermore, thailandine (2) showed excellent anti-plasmodial activity against both chloroquine-susceptible 3D7 and chloroquine-resistant W2 Plasmodium falciparum strains. The structure-activity relationship of isolated compound was also discussed in this study. The results of this study support the traditional use of Stephania dielsiana Y.C. Wu and the lead molecules identified can be further optimized for the development of highly effective and safe anti-cancer and anti-plasmodial drugs.


Asunto(s)
Alcaloides/farmacología , Antimaláricos/farmacología , Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Extractos Vegetales/farmacología , Plasmodium falciparum/efectos de los fármacos , Stephania/química , Apoptosis , Humanos , Malaria Falciparum/tratamiento farmacológico , Malaria Falciparum/parasitología , Neoplasias/patología , Pruebas de Sensibilidad Parasitaria , Hojas de la Planta/química , Células Tumorales Cultivadas
6.
J Immunol ; 204(4): 788-795, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31915262

RESUMEN

We previously found that the novel VR23 proteasome inhibitor not only possesses an effective antitumor activity without causing any ill effects to animals but also reduces side effects caused by a partner drug when used in combination. In this article, we report that VR23, unlike other proteasome inhibitors, exhibits potent anti-inflammatory activity. In the LPS-induced THP-1 monocyte model, VR23 downregulates proinflammatory cytokines IL-1ß, TNF-α, IL-6, and IL-8 at a similar efficacy to dexamethasone. In contrast, two well-known proteasome inhibitors, bortezomib and carfilzomib, do not effectively downregulate these proinflammatory cytokines. Data from a study with SW982 synovial cell line and primary human synoviocytes showed that VR23 not only effectively downregulates IL-6 but also inhibits cell migration. Interestingly, the IL-6 downregulation by VR23 was significantly more pronounced in the primary synovial cells from rheumatoid arthritis patients than those from healthy donors, suggesting that VR23 can be selective against rheumatoid arthritis. Finally, VR23 effectively reduces neutrophil migration, TNF-α secretion, and tissue inflammation in mice (female BALB/c strain) with an LPS-induced acute lung injury. Thus, our current data indicate that VR23 can be effective on both acute and chronic inflammatory conditions. Taken together with our previous work, VR23 is not only effective on inflammatory conditions but also applicable to different aspects of cancer control, including the treatment and prevention of tumor development by chronic inflammatory responses.


Asunto(s)
Antiinflamatorios/farmacología , Artritis Reumatoide/inmunología , Citocinas/efectos de los fármacos , Neumonía/inmunología , Quinolinas/farmacología , Sulfonamidas/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Sinoviocitos/efectos de los fármacos
7.
Breast Cancer Res Treat ; 178(2): 327-335, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31392517

RESUMEN

PURPOSE: Chloroquine has demonstrated anti-tumor activities through autophagy inhibition and cell cycle disruption. This study aimed to assess the effect of single-agent chloroquine on breast tumor cellular proliferation in a randomized, phase II, double-blind, placebo-controlled, pre-surgical window of opportunity trial. METHODS: Patients with newly diagnosed breast cancer were randomized 2:1 to chloroquine 500 mg daily or placebo for 2- to 6-weeks prior to their breast surgery. The primary outcome was the relative change in measures of proliferation (Ki67) in primary breast cancer cells pre- and post-treatment. Adverse events and toxicity profiles were also evaluated. RESULTS: From September 2015 to December 2016, 70 patients were randomized [46 (66%) chloroquine and 24 (34%) placebo]. Ten patients who were randomized to chloroquine withdrew from study due to adverse events. Mean duration of drug intake was 15 days (range 14-29 days). There were no significant differences between the chloroquine or placebo arms with respect to either the percentage change (- 0.4 vs. - 1.2, p = 0.088) or absolute change (- 2.0% vs. - 5.2%, p = 0.066) in Ki67 index pre- and post-drug treatment. Although adverse effects were minimal and all classified as grade 1, the effects were significant enough to cause nearly 15% of patients to discontinue therapy. CONCLUSIONS: Treatment with single-agent chloroquine 500 mg daily in the preoperative setting was not associated with any significant effects on breast cancer cellular proliferation. It was, however, associated with toxicity that may affect its broader use in oncology.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Cloroquina/uso terapéutico , Adolescente , Adulto , Antineoplásicos/administración & dosificación , Antineoplásicos/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/mortalidad , Cloroquina/administración & dosificación , Cloroquina/efectos adversos , Femenino , Humanos , Antígeno Ki-67/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Resultado del Tratamiento , Adulto Joven
8.
Sci Rep ; 9(1): 6315, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-31004122

RESUMEN

In an attempt to develop effective and potentially safe anticancer agents, thirty-six 4-aminoquinoline derived sulfonyl analogs were designed and synthesized using a hybrid pharmacophore approach. The cytotoxicity of these compounds was determined using three breast tumor cell lines (MDA-MB231, MDA-MB468 and MCF7) and two matching non-cancer breast epithelial cell lines (184B5 and MCF10A). Although most of the compounds were quite effective on the breast cancer cells, the compound 7-chloro-4-(4-(2,4-dinitrophenylsulfonyl)piperazin-1-yl)quinoline (13; VR23) emerged as potentially the most desirable one in this series of compounds. Data from the NCI-60 cancer panel screening show that compound 13 is effective on a wide range of different cancers. Importantly, compound 13 is needed up to 17.6-fold less doses to achieve the same IC50 against cancer than non-cancer cells (MDA-MB468 vs MCF10A), suggesting that it can potentially be less toxic to normal cells. Cancer cells formed multiple centrosomes in the presence of compound 13, resulting in the cell cycle arrest at prometa-meta phase. This abnormality leads to eventual cell demise with sub-G1 DNA content typically shown with apoptotic cells. In addition, compound 13 also causes an increase in lysosomal volume in cancer but not in non-cancer cells, which may contribute at least in part to its preferential cancer cell-killing. The cancer cell-killing effect of compound 13 is highly potentiated when combined with either bortezomib or monastrol.


Asunto(s)
Aminoquinolinas , Antineoplásicos , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Neoplasias , Aminoquinolinas/síntesis química , Aminoquinolinas/química , Aminoquinolinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Células MCF-7 , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología
9.
Int J Mol Sci ; 20(3)2019 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-30704031

RESUMEN

Cancer immune therapy has recently shown tremendous promise to combat many different cancers. The microtubule is a well-defined and very effective cancer therapeutic target. Interestingly, several lines of evidence now suggest that microtubules are intimately connected to the body's immune responses. This raises the possibility that the combination of microtubule inhibitors and immune therapy can be a highly effective option for cancer treatments. However, our understanding on this potentially important aspect is still very limited, due in part to the multifaceted nature of microtubule functions. Microtubules are not only involved in maintaining cell morphology, but also a variety of cellular processes, including the movement of secretory vesicles and organelles, intracellular macromolecular assembly, signaling pathways, and cell division. Microtubule inhibitors may be subdivided into two classes: Anti-depolymerization agents such as the taxane family, and anti-polymerization agents such as colchicine and vinka alkaloids. These two different classes may have different effects on immune cell subtypes. Anti-depolymerization agents can not only induce NK cells, but also appear to inhibit T regulatory (Treg) cells. However, different inhibitors may have different functions even among the same class. For example, the doxetaxel anti-depolymerization agent up-regulates cytotoxic T cells, while paclitaxel down-regulates them. Certain anti-polymerization agents such as colchicine appear to down-regulate most immune cell types, while inducing dendritic cell maturation and increasing M1 macrophage population. In contrast, the vinblastine anti-polymerization agent activates many of these cell types, albeit down-regulating Treg cells. In this review, we focus on the various effects of tubulin inhibitors on the activities of the body's immune system, in the hope of paving the way to develop an effective cancer therapy by combining tubulin-targeting anticancer agents and immune therapy.


Asunto(s)
Inmunoterapia/métodos , Microtúbulos/metabolismo , Neoplasias/terapia , Tubulina (Proteína)/metabolismo , Animales , Ciclo Celular/inmunología , Humanos , Neoplasias/inmunología , Neoplasias/metabolismo , Linfocitos T/inmunología
10.
J Enzyme Inhib Med Chem ; 34(1): 620-630, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30727782

RESUMEN

In an attempt to improve anti-breast cancer activity, a new series of 4-piperazinylquinoline derivatives based on the urea/thiourea scaffold were designed and synthesised by a pharmacophore hybrid approach. We then examined for their antiproliferative effects on three human breast tumor cell lines, MDA-MB231, MDA-MB468 and MCF7, and two non-cancer breast epithelial cell lines, 184B5 and MCF10A. Among those 26 novel compounds examined, 5, 9, 17, 18, 21, 23 and 29 showed significantly improved antiproliferative activity on breast cancer cells. Compound 23 (4-(7-chloro-quinolin-4-yl)-piperazine-1-carbothioic acid (2-morpholin-4-yl-ethyl)-amide) (RL-15) is especially desirable, since its antigrowth/cell-killing activity is 7-11 fold higher on cancer than non-cancer cells. Data from cell biological studies demonstrated that cancer cells compromised plasma membrane integrity in the presence of compound 23. The cancer cell-specific property of compound 23 shown in cell culture stands in vivo test, this compound can be an excellent lead for effective and safe anticancer drug.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Diseño de Fármacos , Piperazinas/farmacología , Quinolinas/farmacología , Tiourea/farmacología , Urea/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Células MCF-7 , Estructura Molecular , Piperazinas/síntesis química , Piperazinas/química , Quinolinas/síntesis química , Quinolinas/química , Relación Estructura-Actividad , Tiourea/química , Células Tumorales Cultivadas , Urea/química
11.
Eur J Med Chem ; 143: 1028-1038, 2018 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-29232580

RESUMEN

In an attempt to develop effective and safe anticancer agents, we designed, synthesized and examined 23 novel quinacrine (QC) derivatives by combining the 9-aminoacridine scaffold and the [1,3]thiazinan-4-ones group. Most of these hybrids showed strong anticancer activities, among which 3-(3-(6-chloro-2-methoxyacridin-9-ylamino)propyl)-2-(thiophen-2-yl)-1,3-thiazinan-4-one (25; VR151) effectively killed many different cancer cell types, including eight breast cancer cell lines with different genetic background, two prostate cancer and two lung cancer cell lines. In contrast, compound 25 is less effective against non-cancer cells, suggesting it may be less toxic to humans. Our data showed that cancer cells are arrested in S phase for a prolonged period due to the down-regulation of DNA replication, leading to eventual cell death. We have also shown that the S phase arrest may be resulted by the down-regulation of cyclin A coupled with the continued up-regulation of cyclin E, which coincide with the down-regulation of mTor-S6K and mTor-4EBP1 pathways.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Diseño de Fármacos , Quinacrina/análogos & derivados , Tiazinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Estructura Molecular , Quinacrina/síntesis química , Quinacrina/química , Quinacrina/farmacología , Relación Estructura-Actividad , Tiazinas/síntesis química , Tiazinas/química
12.
Sci Rep ; 7(1): 10298, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28860494

RESUMEN

Agents targeting colchicine-binding pocket usually show a minimal drug-resistance issue, albeit often associated with high toxicity. Chalcone-based compounds, which may bind to colchicine-binding site, are found in many edible fruits, suggesting that they can be effective drugs with less toxicity. Therefore, we synthesized and examined 24 quinolone chalcone compounds, from which we identified ((E)-3-(3-(2-Methoxyphenyl)-3-oxoprop-1-enyl) quinolin-2(1H)-one) (CTR-17) and ((E)-6-Methoxy-3-(3-(2-methoxyphenyl)-3-oxoprop-1-enyl) quinolin-2(1H)-one) (CTR-20) as promising leads. In particular, CTR-20 was effective against 65 different cancer cell lines originated from 12 different tissues, largely in a cancer cell-specific manner. We found that both CTR-17 and CTR-20 reversibly bind to the colchicine-binding pocket on ß-tubulin. Interestingly however, both the CTRs were highly effective against multidrug-resistant cancer cells while colchicine, paclitaxel and vinblastine were not. Our study with CTR-20 showed that it overcomes multidrug-resistance through its ability to impede MRP1 function while maintaining strong inhibition against microtubule activity. Data from mice engrafted with the MDA-MB-231 triple-negative breast cancer cells showed that both CTR-17 and CTR-20 possess strong anticancer activity, alone or in combination with paclitaxel, without causing any notable side effects. Together, our data demonstrates that both the CTRs can be effective and safe drugs against many different cancers, especially against multidrug-resistant tumors.


Asunto(s)
Chalconas/química , Colchicina/química , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Tubulina (Proteína)/química , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Segregación Cromosómica/efectos de los fármacos , Colchicina/metabolismo , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Microtúbulos/química , Microtúbulos/metabolismo , Modelos Moleculares , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Paclitaxel/química , Paclitaxel/farmacología , Conformación Proteica , Multimerización de Proteína , Quinolonas/química , Quinolonas/farmacología , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química , Moduladores de Tubulina/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Phytother Res ; 31(9): 1357-1368, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28703314

RESUMEN

Extracts from the tubers of Stephania venosa (Blum) Spreng growing in Vietnam significantly inhibited cell proliferation against a number of cancer cells including HeLa, MDA-MB231 and MCF-7 cells. A bioassay-guided fractionation led to the isolation of four aporphine and one tetrahydroprotoberberine alkaloids: dehydrocrebanine 1, tetrahydropalmatine 2, stephanine 3, crebanine 4 and O-methylbulbocapnine 5. The characterization of these compounds was based on MS, NMR and published data. A study by structure-bioactivity relationship on these isolates showed that stephanine is the most active compound. Cell biological studies showed that stephanine induces the reverse of mitotic exit, eventually leading to cell death by apoptosis. This data suggests that stephanine has a unique mode of cell-killing activity against cancer cells, which is seldom observed with known synthetic compounds. In addition to its anticancer property, our data from an in vitro study showed that S. venosa also possesses effective antiplasmodial activity and stephanine was also the most interesting compound but is the most cytotoxic with the lowest selectivity index. Copyright © 2017 Her Majesty the Queen in Right of Canada Phytotherapy Research StartCopTextCopyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Antimaláricos/farmacología , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Aporfinas/farmacología , Extractos Vegetales/farmacología , Alcaloides/farmacología , Alcaloides de Berberina/química , Alcaloides de Berberina/farmacología , Humanos , Células MCF-7 , Mitosis/efectos de los fármacos , Estructura Molecular , Fitoterapia , Tubérculos de la Planta/química , Plasmodium falciparum/efectos de los fármacos , Stephania/química , Vietnam
14.
Eur J Med Chem ; 137: 156-166, 2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28586716

RESUMEN

Both quinacrine, which contains a 9-aminoacridine scaffold, and thiazolidin-4-one are promising anticancer leads. In an attempt to develop effective and potentially safe anticancer agents, we synthesized 23 novel hybrid compounds by linking the main structural unit of the 9-aminoacridine ring with the thiazolidin-4-one ring system, followed by examination of their anticancer effects against three human breast tumor cell lines and matching non-cancer cells. Most of the hybrid compounds showed good activities, and many of them possessed the preferential killing property against cancer over non-cancer cells. In particular, 3-[3-(6-chloro-2-methoxy-acridin-9-ylamino)-propyl]-2-(2,6-difluoro-phenyl)-thiazolidin-4-one (11; VR118) effectively killed/inhibited proliferation of cancer cells at IC50 values in the range of 1.2-2.4 µM. Furthermore, unlike quinacrine or cisplatin, compound 11 showed strong selectivity for cancer cell killing, as it could kill cancer cells 7.6-fold (MDA-MB231 vs MCF10A) to 14.7-fold (MCF7 vs MCF10A) more effectively than matching non-cancer cells. Data from flow cytometry, TUNEL and Western blot assays showed that compound 11 kills cancer cells by apoptosis through the down-regulation of Bcl-2 (but not Bcl-XL) survival protein and up-regulation of Bad and Bax pro-apoptotic proteins. Thus, compound 11 is a highly promising lead for an effective and potentially anticancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Diseño de Fármacos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Quinacrina/farmacología , Proteína X Asociada a bcl-2/metabolismo , Proteína Letal Asociada a bcl/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Estructura Molecular , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Quinacrina/síntesis química , Quinacrina/química , Relación Estructura-Actividad , Regulación hacia Arriba/efectos de los fármacos
15.
Cell Cycle ; 15(17): 2321-35, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27341472

RESUMEN

The co-regulation of DNA replication and gene transcription is still poorly understood. To gain a better understanding of this important control mechanism, we examined the DNA replication and transcription using the Dbf4 origin-promoter and Dbf4 pseudogene models. We found that origin firing and Dbf4 transcription activity were inversely regulated in a cell cycle-dependent manner. We also found that proteins critical for the regulation of replication (ORC, MCM), transcription (SP1, TFIIB), and cohesin (Smc1, Smc3) and Mediator functions (Med1, Med12) interact with specific sites within and the surrounding regions of the Dbf4 locus in a cell cycle-dependent manner. As expected, replication initiation occurred within a nucleosome-depleted region, and nucleosomes flanked the 2 replication initiation zones. Further, the histone H3 in this region was distinctly acetylated or trimethylated on lysine 9 in a cell cycle-dependent fluctuation pattern: H3K9ac was most prevalent when the Dbf4 transcription level was highest whereas the H3K9me3 level was greatest during and just after replication. The KDM4A histone demethylase, which is responsible for the H3K9me3 modification, was enriched at the Dbf4 origin in a manner coinciding with H3K9me3. Finally, HP1γ, a protein known to interact with H3K9me3 in the heterochromatin was also found enriched at the origin during DNA replication, indicating that H3K9me3 may be required for the regulation of replication at both heterochromatin and euchromatin regions. Taken together, our data show that mammalian cells employ an extremely sophisticated and multilayered co-regulation mechanism for replication and transcription in a highly coordinated manner.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN , Sitios Genéticos , Histonas/metabolismo , Lisina/metabolismo , Regiones Promotoras Genéticas , Transcripción Genética , Acetilación , Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/metabolismo , Regulación hacia Abajo/genética , Células HEK293 , Células HeLa , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Complejo Mediador/metabolismo , Metilación , Nucleosomas/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional/genética , Seudogenes , Origen de Réplica/genética , Cohesinas
16.
Cell Cycle ; 15(11): 1494-505, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27105124

RESUMEN

To maintain genetic stability, the entire mammalian genome must replicate only once per cell cycle. This is largely achieved by strictly regulating the stepwise formation of the pre-replication complex (pre-RC), followed by the activation of individual origins of DNA replication by Cdc7/Dbf4 kinase. However, the mechanism how Cdc7 itself is regulated in the context of cell cycle progression is poorly understood. Here we report that Cdc7 is phosphorylated by a Cdk1-dependent manner during prometaphase on multiple sites, resulting in its dissociation from origins. In contrast, Dbf4 is not removed from origins in prometaphase, nor is it degraded as cells exit mitosis. Our data thus demonstrates that constitutive phosphorylation of Cdc7 at Cdk1 recognition sites, but not the regulation of Dbf4, prevents the initiation of DNA replication in normally cycling cells and under conditions that promote re-replication in G2/M. As cells exit mitosis, PP1α associates with and dephosphorylates Cdc7. Together, our data support a model where Cdc7 (de)phosphorylation is the molecular switch for the activation and inactivation of DNA replication in mitosis, directly connecting Cdc7 and PP1α/Cdk1 to the regulation of once-per-cell cycle DNA replication in mammalian cells.


Asunto(s)
Proteínas de Ciclo Celular/genética , Quinasas Ciclina-Dependientes/genética , Replicación del ADN , Mitosis , Proteína Fosfatasa 1/genética , Proteínas Serina-Treonina Quinasas/genética , Proteína Quinasa CDC2 , Proteínas de Ciclo Celular/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Fosforilación , Proteína Fosfatasa 1/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Fluorescente Roja
17.
Bioorg Med Chem ; 23(24): 7629-40, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26602827

RESUMEN

A series of 25 novel quinolino-stilbene derivatives were designed, synthesized and evaluated for their potential as anticancer agents. Three of them not only displayed quite potent antiproliferative activity with IC50 values<4µM but also showed approximately twofold selectivity against cancer cells, compared to non-cancerous cells. Three other compounds exhibited comparatively good activity with IC50 values in the range of 4-10µM, and the rest was moderately active or inactive. One of these viz. 3-[E-(4-fluorostyryl)]-2-chloroquinoline (compound 7B) caused substantial DNA damage and arrested cell cycle in S phase. Interestingly, 7B was very active against MDA-MB468 (IC50=0.12µM), but not against other cell lines examined. Compound 3-[Z-(3-(trifluoromethyl)styryl)]-2-chloroquinoline (12A), the most effective against all cancer cell lines examined, caused prolonged cell cycle arrest at mitosis and eventually apoptosis. Data from an in vitro study showed that compound 12A inhibited microtubule polymerization in a similar fashion to nocodazole. Further study using in silico molecular modeling revealed that 12A causes the impediment of microtubule polymerization by binding to tubulin at the same cavity where podophyllotoxin binds.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Quinolinas/química , Quinolinas/farmacología , Estilbenos/química , Estilbenos/farmacología , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Modelos Moleculares , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología , Quinolinas/síntesis química , Estilbenos/síntesis química , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacología
18.
Cancer Res ; 75(19): 4164-75, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26238784

RESUMEN

The proteasome is clinically validated as a target for cancer therapeutics. However, proteasome-inhibitory agents that are cancer selective have yet to be developed. In this study, we report the identification of a safe and effective proteasome inhibitor with selective anticancer properties. We screened a chemical library constructed using a hybrid approach that incorporated a 4-piperazinylquinoline scaffold and a sulfonyl phamarcophore. From this library, we identified 7-chloro-4-(4-(2,4-dinitrophenylsulfonyl)piperazin-1-yl)quinoline (VR23) as a small molecule that potently inhibited the activities of trypsin-like proteasomes (IC50 = 1 nmol/L), chymotrypsin-like proteasomes (IC50 = 50-100 nmol/L), and caspase-like proteasomes (IC50 = 3 µmol/L). Data from molecular docking and substrate competition assays established that the primary molecular target of VR23 was ß2 of the 20S proteasome catalytic subunit. Notably, VR23 was structurally distinct from other known proteasome inhibitors and selectively killed cancer cells by apoptosis, with little effect on noncancerous cells. Mechanistic investigations showed that cancer cells exposed to VR23 underwent an abnormal centrosome amplification cycle caused by the accumulation of ubiquitinated cyclin E. In combinations with the clinically approved chymotrypsin-like proteasome inhibitor bortezomib, VR23 produced a synergistic effect in killing multiple myeloma cells, including those that were resistant to bortezomib. VR23 was effective in vivo in controlling multiple myelomas and metastatic breast cancer cells, in the latter case also enhancing the antitumor activity of paclitaxel while reducing its side effects. Overall, our results identify VR23 as a structurally novel proteasome inhibitor with desirable properties as an anticancer agent.


Asunto(s)
Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Centrosoma/efectos de los fármacos , Ciclina E/fisiología , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteasoma/farmacología , Quinolinas/farmacología , Sulfonamidas/farmacología , Proteínas Ubiquitinadas/fisiología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/secundario , Animales , Apoptosis/efectos de los fármacos , Unión Competitiva , Bortezomib/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Centrosoma/metabolismo , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Simulación del Acoplamiento Molecular , Estructura Molecular , Mieloma Múltiple/patología , Proteínas de Neoplasias/fisiología , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Paclitaxel/toxicidad , Unión Proteica , Quinolinas/administración & dosificación , Quinolinas/química , Sulfonamidas/administración & dosificación , Sulfonamidas/química , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Eur J Pharmacol ; 762: 472-86, 2015 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25959387

RESUMEN

Chloroquine (CQ) has a broad spectrum of pharmacological activities including anticancer and anti-inflammatory, in addition to its well-known antimalarial activity. This very useful property of CQ may be rendered through a variety of different molecular and cellular mechanisms, including the induction of apoptosis, necrosis and lysosomal dysfunction. CQ alone may not be as effective as many well-known anticancer drugs; however, it often shows synergisticts when combined with other anticancer agents, without causing substantial ill-effects. To increase its pharmacological activity, scientists synthesized many different chloroquine derivatives by a repositioning approach, some of which show higher activities than the parental CQ. To further improve anticancer activity, medicinal chemists have recently been focusing on generating CQ hybrid molecules by joining, directly or through a linker, 4-aminoquinoline and other pharmacologically active phamarcophore(s). Indeed, some CQ hybrid molecules substantially improved anticancer activity while maintaining desirable CQ property, providing an excellent opportunity of developing effective and safe novel anticancer agents. Since the approach of developing CQ hybrid molecules has advanced much more in the antimalarial drug research, it can provide an excellent template for anticancer drug development. This review provides an overview of CQ-based hybrid molecules by focusing on: (1) the potential advantage of the hybrid approach in developing effective and safe anticancer agents; (2) what we can learn from the CQ hybrid approach used in the development of effective antimalarial agents; and (3) CQ hybrid molecules as potential anticancer agents in different categories classified based on their chemical compositions.


Asunto(s)
Antimaláricos/química , Antimaláricos/farmacología , Antineoplásicos/química , Antineoplásicos/farmacología , Cloroquina/química , Cloroquina/farmacología , Descubrimiento de Drogas/métodos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...