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










Base de dados
Intervalo de ano de publicação
1.
Expert Opin Drug Saf ; 17(12): 1171-1183, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30394114

RESUMO

Introduction: This review provides a guide for the rational use of prescription drugs in patients of reproductive age. Areas covered: A comprehensive retrieval of the labels of FDA-approved drugs was performed to identify drugs where the label recommends contraceptive use during and/or after treatment. The acquired data were analyzed and organized into a table. Contraception was recommended or mandated for 268 single-ingredient drugs. These could be divided into four main categories, with many having effects across several categories: 177 drugs required contraception because they were associated with pregnancy loss or stillbirth, 177 drugs were associated with teratogenesis, 136 were associated with non-teratogenic adverse peri- or postnatal effects on the fetus (e.g. low birth weight), and 44 were associated with decreased efficacy of contraception or a change in ovulatory cycle. We also discuss the period of time contraception is required, as well as the known or hypothesized reasons for the reproductive toxicity of these agents. Expert opinion: We have provided a comprehensive overview of the FDA-approved drugs where the warning labels currently stipulate that contraception should be used. Although other references are available for clinicians, this review provides a useful source of information regarding the single-ingredient prescription drugs that may affect the outcome of pregnancy. This information is particularly relevant for researchers, as it provides an overview of the different drugs with reproductive toxicity, and because it highlights the specific needs for future research. In particular, more work (especially epidemiological studies) is needed to clarify the clinical relevance of these findings, most of which were obtained through animal studies.


Assuntos
Anticoncepção/métodos , Rotulagem de Medicamentos/legislação & jurisprudência , Medicamentos sob Prescrição/efeitos adversos , Anormalidades Induzidas por Medicamentos/prevenção & controle , Aborto Espontâneo/induzido quimicamente , Aborto Espontâneo/prevenção & controle , Animais , Feminino , Humanos , Gravidez , Medicamentos sob Prescrição/administração & dosagem , Fatores de Tempo , Estados Unidos , United States Food and Drug Administration
2.
J Assist Reprod Genet ; 35(2): 191-212, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29063992

RESUMO

PURPOSE: This study reviews FDA-approved drugs that negatively impact spermatozoa in animals, as well as how these findings reflect on observations in human male gametes. METHODS: The FDA drug warning labels included in the DailyMed database and the peer-reviewed literature in the PubMed database were searched for information to identify single-ingredient, FDA-approved prescription drugs with spermatotoxic effects. RESULTS: A total of 235 unique, single-ingredient, FDA-approved drugs reported to be spermatotoxic in animals were identified in the drug labels. Forty-nine of these had documented negative effects on humans in either the drug label or literature, while 31 had no effect or a positive impact on human sperm. For the other 155 drugs that were spermatotoxic in animals, no human data was available. CONCLUSION: The current animal models are not very effective for predicting human spermatotoxicity, and there is limited information available about the impact of many drugs on human spermatozoa. New approaches should be designed that more accurately reflect the findings in men, including more studies on human sperm in vitro and studies using other systems (ex vivo tissue culture, xenograft models, in silico studies, etc.). In addition, the present data is often incomplete or reported in a manner that prevents interpretation of their clinical relevance. Changes should be made to the requirements for pre-clinical testing, drug surveillance, and the warning labels of drugs to ensure that the potential risks to human fertility are clearly indicated.


Assuntos
Espermatozoides/efeitos dos fármacos , Testes de Toxicidade/métodos , Animais , Aprovação de Drogas , Rotulagem de Medicamentos , Humanos , Masculino , PubMed , Espermatogênese/efeitos dos fármacos , Estados Unidos , United States Food and Drug Administration
3.
Crit Rev Clin Lab Sci ; 54(1): 1-17, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27193822

RESUMO

Drug-related laboratory test interference or drug/laboratory test interactions (DLTI) are a major source of laboratory errors. DLTI is of concern with regard to both the clinical diagnosis and the monitoring of patients. Although there have been numerous reports about specific drugs that interfere with laboratory tests, there has not been a recent review on the topic. We herein provide a review of the known DLTI of US FDA-approved drugs based on a systematic search of DailyMed, a website containing the labels of US FDA-approved drugs. The labels for all human single-ingredient prescription drugs included in the database (1368) were searched using stemmed keywords and were manually reviewed for their relevance to DLTI. A total of 134 labels were positive, which indicated that the drug interferes with at least one clinical laboratory test. Antibacterial agents, psychotropic drugs and contrast media are the classes of drugs most likely to lead to DLTI. Urine was the clinical sample most frequently affected by DLTI. The FDA drug label is a source of information for studies of DLTI, although information is still lacking for most drugs, and additional improvements are needed for many of the existing records. Medical professionals, clinicians and laboratory staff should keep these possible interactions in mind when interpreting the results of laboratory tests, and should ensure that they obtain a complete and accurate record of all drugs being used by patients in order to anticipate potential DLTI. The development of a reporting system to address potential DLTI is warranted.


Assuntos
Técnicas de Laboratório Clínico/normas , Interações Medicamentosas , Rotulagem de Medicamentos , Antibacterianos , Meios de Contraste , Humanos , Psicotrópicos , Estados Unidos , United States Food and Drug Administration
4.
Oncotarget ; 8(6): 10714-10725, 2017 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-27801671

RESUMO

We herein provide an overview of the single-ingredient U.S. Food and Drug Administration (FDA)-approved drugs that affect human spermatogenesis, potentially resulting in a negative impact on male fertility. To provide this information, we performed an in-depth search of DailyMed, the official website for FDA-approved drug labels. Not surprisingly, hormone-based agents were found to be the drugs most likely to affect human spermatogenesis. The next category of drugs most likely to have effects on spermatogenesis was the antineoplastic agents. Interestingly, the DailyMed labels indicated that several anti-inflammatory drugs affect spermatogenesis, which is not supported by the peer-reviewed literature. Overall, there were a total of 65 labels for drugs of various classes that showed that they have the potential to affect human sperm production and maturation. We identified several drugs indicated to be spermatotoxic in the drug labels that were not reported in the peer-reviewed literature. However, the details about the effects of these drugs on human spermatogenesis are largely lacking, the mechanisms are often unknown, and the clinical impact of many of the findings is currently unclear. Therefore, additional work is needed at both the basic research level and during clinical trials and post-marketing surveillance to fill the gaps in the current knowledge. The present findings will be of interest to physicians and pharmacists, researchers, and those involved in drug development and health care policy.


Assuntos
Aprovação de Drogas , Fertilidade/efeitos dos fármacos , Infertilidade Masculina/induzido quimicamente , Espermatogênese/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Animais , Rotulagem de Medicamentos , Humanos , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Infertilidade Masculina/fisiopatologia , Masculino , Medição de Risco , Fatores de Risco , Espermatozoides/metabolismo , Espermatozoides/patologia , Estados Unidos , United States Food and Drug Administration
5.
PLoS One ; 7(9): e44883, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028659

RESUMO

The purpose of the present study was to determine the in vitro and in vivo anti-cancer activity and pharmacological properties of 3,4-dimethoxy-N-[(2,2-dimethyl-2H-chromen-6-yl)methyl]-N-phenylbenzenesulfonamide, KCN1. In the present study, we investigated the in vitro activity of KCN1 on cell proliferation and cell cycle distribution of pancreatic cancer cells, using the MTT and BrdUrd assays, and flow cytometry. The in vivo anti-cancer effects of KCN1 were evaluated in two distinct xenograft models of pancreatic cancer. We also developed an HPLC method for the quantitation of the compound, and examined its stability in mouse plasma, plasma protein binding, and degradation by mouse S9 microsomal enzymes. Furthermore, we examined the pharmacokinetics of KCN1 following intravenous or intraperitoneal injection in mice. Results showed that, in a dose-dependent manner, KCN1 inhibited cell growth and induced cell cycle arrest in human pancreatic cancer cells in vitro, and showed in vivo anticancer efficacy in mice bearing Panc-1 or Mia Paca-2 tumor xenografts. The HPLC method provided linear detection of KCN1 in all of the matrices in the range from 0.1 to 100 µM, and had a lower limit of detection of 0.085 µM in mouse plasma. KCN1 was very stable in mouse plasma, extensively plasma bound, and metabolized by S9 microsomal enzymes. The pharmacokinetic studies indicated that KCN1 could be detected in all of the tissues examined, most for at least 24 h. In conclusion, our preclinical data indicate that KCN1 is a potential therapeutic agent for pancreatic cancer, providing a basis for its future development.


Assuntos
Antineoplásicos/farmacologia , Benzopiranos/farmacologia , Neoplasias Pancreáticas/patologia , Sulfonamidas/farmacologia , Animais , Antineoplásicos/sangue , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Benzopiranos/sangue , Benzopiranos/metabolismo , Benzopiranos/farmacocinética , Disponibilidade Biológica , Proteínas Sanguíneas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estabilidade de Medicamentos , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Meia-Vida , Humanos , Camundongos , Reprodutibilidade dos Testes , Sulfonamidas/sangue , Sulfonamidas/metabolismo , Sulfonamidas/farmacocinética , Temperatura , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Biomed Chromatogr ; 25(5): 628-634, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20845374

RESUMO

We herein describe the development and validation of an HPLC method for the quantitation of 7-(benzylamino)-1,3,4,8-tetrahydropyrrolo [4,3,2-de]quinolin-8(1H)-one (BA-TPQ), a newly synthesized iminoquinone anticancer agent. BA-TPQ was extracted from plasma and tissue samples by first precipitating proteins with acetonitrile followed by a liquid-liquid extraction with ethyl acetate. Chromatographic separation was carried out using a gradient flow rate on a Zorbax SB C(18) column, and the effluent was monitored by UV detection at 346 nm. The method was found to be precise, accurate, and specific, with a linear range of 3.91-1955.0 ng/mL in plasma, 19.55-1955.0 ng/mL in spleen, brain, and liver homogenates and 19.55-3910.0 ng/mL in heart, lung and kidney homogenates. The method was stable under all relevant conditions. Using this method, we also carried out an initial study determining plasma pharmacokinetics and tissue distribution of BA-TPQ in mice following intravenous administration. In summary, this simple and sensitive HPLC method can be used in future preclinical and clinical studies of BA-TPQ.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Pirróis/análise , Pirróis/farmacocinética , Quinolonas/análise , Quinolonas/farmacocinética , Animais , Antineoplásicos/análise , Antineoplásicos/sangue , Antineoplásicos/farmacocinética , Análise dos Mínimos Quadrados , Masculino , Camundongos , Pirróis/sangue , Quinolonas/sangue , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual
7.
Mar Drugs ; 8(7): 2129-41, 2010 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-20714427

RESUMO

Marine natural products and their synthetic derivatives represent a major source of novel candidate anti-cancer compounds. We have recently tested the anti-cancer activity of more than forty novel compounds based on an iminoquinone makaluvamine scaffold, and have found that many of the compounds exert potent cytotoxic activity against human cancer cell lines. One of the most potent compounds, BA-TPQ [(11,12),7-(benzylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one], was active against a variety of human cancer cell lines, and inhibited the growth of breast and prostate xenograft tumors in mice. However, there was some toxicity noted in the mice following administration of the compound. In order to further the development of BA-TPQ, and in a search for potential sites of accumulation that might underlie the observed toxicity of the compound, we accomplished preclinical pharmacological studies of the compound. We herein report the in vitro and in vivo pharmacological properties of BA-TPQ, including its stability in plasma, plasma protein binding, metabolism by S9 enzymes, and plasma and tissue distribution. We believe these studies will be useful for further investigations, and may be useful for other investigators examining the use of similar compounds for cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Pirróis/farmacologia , Quinolonas/farmacologia , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Avaliação Pré-Clínica de Medicamentos , Feminino , Humanos , Masculino , Camundongos , Neoplasias/patologia , Ligação Proteica , Pirróis/farmacocinética , Pirróis/toxicidade , Quinolonas/farmacocinética , Quinolonas/toxicidade , Distribuição Tecidual , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Breast Cancer Res Treat ; 123(2): 321-31, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19936915

RESUMO

Herein, we report our examination of the anti-breast cancer activity of a novel synthetic compound, 7-(benzylamino)-1, 3, 4, 8-tetrahydropyrrolo [4, 3, 2-de]quinolin-8(1H)-one (BA-TPQ). This agent is an analog of a naturally occurring marine compound, and was found to be the most active out of more than 40 related compounds. We investigated the in vitro activity of BA-TPQ on the survival, proliferation, and apoptosis of breast cancer cells using the MTT and BrdUrd assays, and Annexin/Annexin-PI staining and flow cytometry. The in vivo anti-cancer effects of BA-TPQ were evaluated in xenograft models of breast cancer. Finally, the mechanisms of action of the compound were also assessed by cDNA microarrays, RT-PCR and Western blotting. In a dose-dependent manner, BA-TPQ inhibited cell growth and induced apoptosis and cell cycle arrest in human MCF-7 and MDA-MB-468 breast cancer cells in vitro, and showed in vivo efficacy in mice bearing MCF-7 or MDA-MB-468 xenograft tumors. We demonstrated that BA-TPQ modifies the expression of numerous molecules involved in cell cycle progression and apoptosis. Similar changes in protein expression were observed in vitro and in vivo, as determined by examination of cells and excised xenograft tumors. Our preclinical data indicate that BA-TPQ is a potential therapeutic agent for breast cancer that has multiple hormone-, Her2-, and p53-independent mechanisms of action, providing a basis for further development of the compound as a novel anticancer agent.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Pirróis/farmacologia , Quinolonas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Camundongos Nus , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Invest New Drugs ; 28(3): 234-41, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19274441

RESUMO

We recently synthesized a series of novel makaluvamine compounds, and found that the most potent was FBA-TPQ. The effects of FBA-TPQ on human (LNCaP and PC3) and murine (TRAMP C1) prostate cancer cells were evaluated. Potential mechanisms of action of the compound were also determined. FBA-TPQ exhibited dose-dependent cytotoxicity in the low micromolar range, inhibited proliferation, caused cell cycle arrest, and induced apoptosis in prostate cancer cell lines. The compound also decreased the expression of the androgen receptor and PSA. The results presented herein support the further development of FBA-TPQ as a novel agent for prostate cancer.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Pirróis/uso terapêutico , Quinolonas/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , Antígeno Prostático Específico/metabolismo , Pirróis/síntese química , Pirróis/farmacologia , Quinolonas/síntese química , Quinolonas/farmacologia , Receptores Androgênicos/metabolismo
10.
Anticancer Agents Med Chem ; 9(8): 882-903, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19538162

RESUMO

The MDM2 oncogene is overexpressed in various human cancers. Its expression correlates with the phenotypes of high-grade, late-stage, and more resistant tumors. The auto-regulatory loop between MDM2 and the tumor suppressor p53 has long been considered the epitome of a rational target for cancer therapy. As such, many novel agents have been generated to interfere with the interaction of the two proteins, which results in the activation of p53. Among these agents are several small molecule inhibitors synthesized based upon the crystal structures of the MDM2-p53 complex. With use of high-throughput screening, several specific and effective agents for inhibition of the protein-protein interaction were discovered. Recent investigations, however, have demonstrated that many proteins regulate the MDM2-p53 interaction, and that MDM2 may have p53-independent oncogenic functions. In order for novel MDM2 inhibitors to be translated to the clinic, it is necessary to obtain a better understanding of the regulation of MDM2 and of the MDM2-p53 interaction. In particular, the implications of various interactions between certain regulator(s) and MDM2/p53 under different circumstances need to be elucidated to determine which pathway(s) represent the best targets for therapy. Targeting both MDM2 itself and regulators of MDM2 and the MDM2-p53 interaction, or use of MDM2 inhibitors in combination with conventional treatments, may improve prospects for tumor eradication.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-mdm2/genética , Ciclo Celular , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Oncogenes , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo
11.
Med Chem ; 5(3): 227-36, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19442212

RESUMO

The high mortality rate and lack of effective therapies make lung cancer an ideal target for novel therapeutic agents. The present study was designed to implement a novel chemical synthesis pathway and to determine the biological activities of synthetic makaluvamine analogs in human lung cancer. Seventeen compounds were synthesized and purified, and their chemical structures were elucidated on the basis of physicochemical constants and NMR spectra. Their in vitro activity was determined in human lung cancer cell lines. Based on initial screens, compound Ic was found to be the most potent, and was therefore used as a model for further studies in lung cancer cells. Ic induced both apoptosis and S-phase cell cycle arrest. Furthermore, it activated p53 and induced cleavage of PARP and caspases 8 and 9. Our preclinical data indicate that the makaluvamine analogs are potential therapeutic agents against lung cancer, providing a basis for further development of Ic (and perhaps other analogs) as a novel anti-cancer agent.


Assuntos
Alcaloides/síntese química , Alcaloides/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Quinolonas/síntese química , Quinolonas/farmacologia , Alcaloides/química , Alcaloides/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/patologia , Quinolonas/química , Quinolonas/uso terapêutico
12.
Clin Cancer Res ; 15(10): 3511-8, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19451594

RESUMO

PURPOSE: The present study was designed to determine biological structure-activity relationships for four newly synthesized analogues of natural compounds (makaluvamines). The compounds, 7-(4-fluorobenzylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one (FBA-TPQ); 7-(phenethylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one (PEA-TPQ); 7-(3,4-methylenedioxyphenethylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one (MPA-TPQ); and 7-(3,4-dimethoxyphenethylamino)-1,3,4,8-tetrahydropyrrolo[4,3,2-de]quinolin-8(1H)-one (DPA-TPQ), were synthesized and purified, and their chemical structures were elucidated on the basis of physicochemical constants and nuclear magnetic resonance spectra. EXPERIMENTAL DESIGN: The structure-activity relationship of the compounds was initially evaluated by comparing their in vitro cytotoxicity against 14 human cell lines. Detailed in vitro and in vivo studies were then done in MCF-7 and MDA-MB-468 breast cancer cell lines. RESULTS: The in vitro cytotoxicity was compound, dose, and cell line dependent. Whereas all of the compounds exerted some activity, FBA-TPQ was the most potent inducer of apoptosis and the most effective inhibitor of cell growth and proliferation, with half maximal inhibitory concentration values for most cell lines in the range of 0.097 to 2.297 mumol/L. In MCF-7 cells, FBA-TPQ exposure led to an increase in p53/p-p53, Bax, ATM/p-ATM, p-chk1 and p-chk2, and p-H2AX; and cleavage of poly(ADP)ribose polymerase, caspase-3, caspase-8, and caspase-9. It also decreased the levels of MDM2, E2F1, Bcl-2, chk1/2, and proteins associated with cell proliferation [cyclin-dependent kinase (Cdk)2, Cdk4, Cdk6, cyclin D1, etc.]. Moreover, FBA-TPQ inhibited the growth of breast cancer xenograft tumors in nude mice in a dose-dependent manner. Western blot analysis ofthe xenograft tumors indicated that similar changes in protein expression also occur in vivo. CONCLUSION: Our preclinical data indicate that FBA-TPQ is a potential therapeutic agent for breast cancer, providing a basis for the development of the compound as a novel anticancer agent.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Mamárias Experimentais/tratamento farmacológico , Pirróis/farmacologia , Quinolonas/farmacologia , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Estrutura Molecular , Pirróis/síntese química , Pirróis/química , Quinolonas/síntese química , Quinolonas/química , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cancer Lett ; 278(2): 241-248, 2009 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-19203832

RESUMO

We recently isolated and characterized two novel ginsenosides, 25-OH-PPD and 25-OCH(3)-PPD. We investigated whether these ginsenosides could represent safe and effective therapeutic agents for pancreatic cancer. In vitro and in vivo studies demonstrated that both compounds inhibited proliferation, caused cell cycle arrest, and induced apoptosis. They also both inhibited the growth of xenograft tumors without any host toxicity. Preliminary investigations into the mechanisms of action of the compounds suggest that their effects may be partially mediated by their inhibition of the MDM2 oncogene and related pathways. The data presented here support further evaluation of the ginsenosides for pancreatic cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Ginsenosídeos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Triterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Lung Cancer ; 65(3): 306-11, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19131140

RESUMO

20(S)-25-methoxyl-dammarane-3beta, 12beta, 20-triol (25-OCH(3)-PPD), a newly identified natural product from Panax notoginseng, exhibits activity against a variety of cancer cells. Herein, we report the effects of this compound on human A549, H358, and H838 lung cancer cells, and compare these effects with a control lung epithelial cell line, BEAS-2B. 25-OCH(3)-PPD decreased survival, inhibited proliferation, and induced apoptosis and G1 cell cycle arrest in the lung cancer cell lines. The P. notoginseng compound also decreased the levels of proteins associated with cell proliferation and cell survival. Moreover, 25-OCH(3)-PPD inhibited the growth of A549 lung cancer xenograft tumors. 25-OCH(3)-PPD demonstrated low toxicity to non-cancer cells, and no observable toxicity was seen when the compound was administered to animals. In conclusion, our preclinical data indicate that 25-OCH(3)-PPD is a potential therapeutic agent in vitro and in vivo, and further preclinical and clinical development of this agent for lung cancer is warranted.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Ginsenosídeos/farmacologia , Neoplasias Pulmonares/tratamento farmacológico , Triterpenos/farmacologia , Adenocarcinoma/patologia , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Neoplasias Pulmonares/patologia , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Panax notoginseng , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/patologia , Carga Tumoral/efeitos dos fármacos
15.
EMBO Rep ; 10(2): 166-72, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19098711

RESUMO

The mouse double minute 2 (MDM2)-p53 interaction regulates the activity of p53 and is a potential target for human cancer therapy. Here, we report that RYBP (RING1- and YY1-binding protein), a member of the polycomb group (PcG), interacts with MDM2 and decreases MDM2-mediated p53 ubiquitination, leading to stabilization of p53 and an increase in p53 activity. RYBP induces cell-cycle arrest and is involved in the p53 response to DNA damage. Expression of RYBP is decreased in human cancer tissues compared with adjacent normal tissues. These results show that RYBP is a new regulator of the MDM2-p53 loop and that it has tumour suppressor activity.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Células COS , Ciclo Celular/fisiologia , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Chlorocebus aethiops , Dano ao DNA , Regulação para Baixo , Etoposídeo/farmacologia , Retroalimentação Fisiológica , Genes Supressores de Tumor , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Hepáticas/química , Neoplasias Pulmonares/química , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Ligação Proteica , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-mdm2/biossíntese , Proteínas Proto-Oncogênicas c-mdm2/genética , Interferência de RNA , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Repressoras , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/genética , Técnicas do Sistema de Duplo-Híbrido , Ubiquitinação
16.
Mol Cell Pharmacol ; 1(1): 29-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-20333321

RESUMO

Inflammation is closely linked to cancer, and many anti-cancer agents are also used to treat inflammatory diseases, such as rheumatoid arthritis. Moreover, chronic inflammation increases the risk for various cancers, indicating that eliminating inflammation may represent a valid strategy for cancer prevention and therapy. This article explores the relationship between inflammation and cancer with an emphasis on epidemiological evidence, summarizes the current use of anti-inflammatory agents for cancer prevention and therapy, and describes the mechanisms underlying the anti-cancer effects of anti-inflammatory agents. Since monotherapy is generally insufficient for treating cancer, the combined use of anti-inflammatory agents and conventional cancer therapy is also a focal point in discussion. In addition, we also briefly describe future directions that should be explored for anti-cancer anti-inflammatory agents.

17.
Drug Discov Today ; 13(11-12): 513-21, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18549978

RESUMO

Antisense oligonucleotides can regulate gene expression in living cells. As such, they regulate cell function and division, and can modulate cellular responses to internal and external stresses and stimuli. Although encouraging results from preclinical and clinical studies have been obtained and significant progress has been made in developing these agents as drugs, they are not yet recognized as effective therapeutics. Several major hurdles remain to be overcome, including problems with efficacy, off-target effects, delivery and side effects. The lessons learned from antisense drug development can help in the development of other oligonucleotide-based therapeutics such as CpG oligonucleotides, RNAi and miRNA.


Assuntos
Inativação Gênica , Oligonucleotídeos Antissenso/farmacologia , Interferência de RNA , Ensaios Clínicos como Assunto , Desenho de Fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética/métodos , Humanos , Oligonucleotídeos Antissenso/metabolismo
18.
Prostate ; 68(8): 809-19, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18324646

RESUMO

BACKGROUND: Ginseng and its components exert various biological effects, including antioxidant, anti-carcinogenic, anti-mutagenic, and anti-tumor activity, and recent research has focused on their value in human cancer prevention and treatment. We recently isolated 25-hydroxyprotopanaxadiol (25-OH-PPD) and 25-hydroxyprotopanaxatriol (25-OH-PPT) from Panax ginseng and evaluated their anti-cancer activity in vitro. METHODS: We compared the effects of the two compounds on human prostate cancer LNCaP and PC3 cells in vitro and in a mouse PC3 xenograft tumor model. We also accomplished a preliminary determination of the mechanisms of action of the compounds. RESULTS: 25-OH-PPD, but not 25-OH-PPT, inhibited prostate cancer cell growth and proliferation, induced apoptosis, and led to arrest in the G1 phase of the cell cycle. In nude mice bearing PC3 xenograft tumors, 25-OH-PPD inhibited tumor growth in a dose-dependent manner and could be safely combined with chemotherapeutic agents (taxotere and gemcitabine) and radiation therapy to improve the anti-tumor effects. Further, in both PC3 and LNCaP cell lines, 25-OH-PPD increased expression of p21, p27, and Bax, induced PARP cleavage and activated caspases. The compound also reduced expression of MDM2, E2F1, Bcl2, cdk2/4/6, and cyclin D1, which correlated with the cell cycle arrest in G1 and the decrease in proliferation. Moreover, 25-OH-PPD demonstrated low toxicity to non-cancer cells and no observable host toxicity in animals either alone or in combination with conventional therapies. CONCLUSIONS: The newly identified ginsenoside 25-OH-PPD may have potential as a novel prostate cancer therapeutic agent.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Ginsenosídeos/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/química , Apoptose , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Terapia Combinada , Fibroblastos/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Ginsenosídeos/química , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/patologia , Neoplasias da Próstata/radioterapia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Int J Oncol ; 30(6): 1511-9, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17487373

RESUMO

The present study demonstrates the efficacy of utilizing TLR9 (toll-like receptor 9) agonism as a potential therapy for colon cancer. We examined the effects of two types of TLR9 agonists: a traditional CpG oligonucleotide and a novel immunomodulatory oligonucleotide in models of colon cancer, both alone and in combination with conventional cancer therapies. Because the tumor suppressor p53 is involved in many anti-cancer pathways, and is mutated in more than 50% of cancers, we determined whether p53 is necessary for the anti-tumor effects observed following treatment with TLR9 agonists. We also established that colon cancer cells express TLR9, which has not been demonstrated previously. The effects of TLR9 agonism on the growth, proliferation and apoptosis of colon cancer cells in vitro was then examined. We report five major discoveries: i) TLR9 agonism results in significant activity in models of colon cancer, ii) TLR9 agonists increase the anti-tumor effects of radiation and chemotherapy, iii) p53 is not required for the anti-cancer effects of TLR9 agonism, iv) human colon cancer cells express TLR9, and v) TLR9 agonism leads to decreased cell survival and proliferation and induces apoptosis of colon cancer cells in vitro. These results provide a basis for future studies determining the potential of utilizing TLR9 agonists for human colon cancer therapy.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias do Colo/tratamento farmacológico , Oligonucleotídeos/uso terapêutico , Receptor Toll-Like 9/agonistas , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Ilhas de CpG , Feminino , Humanos , Camundongos , Receptor Toll-Like 9/biossíntese
20.
Cancer Epidemiol Biomarkers Prev ; 16(4): 789-95, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17416772

RESUMO

The overexpression of cyclic AMP (cAMP)-dependent protein kinase (PKA) has been reported in patients with cancer, and PKA inhibitors have been tested in clinical trials as a novel cancer therapy. The present study was designed to characterize the population distribution of extracellular activity of cAMP-dependent protein kinase (ECPKA) and its potential value as a biomarker for cancer detection and monitoring of cancer therapy. The population distribution of ECPKA activity was determined in serum samples from a Chinese population consisting of a total of 603 subjects (374 normal healthy volunteers and 229 cancer patients). The serum ECPKA was determined by a validated sensitive radioassay, and its diagnostic values (including positive and negative predictive values) were analyzed. The majority of normal subjects (>70%) have undetectable or very low levels of serum ECPKA. In contrast, the majority of cancer patients (>85%) have high levels of ECPKA. The mean ECPKA activity in the sera of cancer patients was 10.98 units/mL, 5-fold higher than that of the healthy controls (2.15 units/mL; P < 0.001). In both normal subjects and cancer patients, gender and age had no significant influence on the serum ECPKA. Among factors considered, logistic analysis revealed that the disease (cancer) is the only factor contributing to the elevation of ECPKA activity in cancer patients. In conclusion, ECPKA may function as a cancer marker for various human cancers and can be used in cancer detection and for monitoring response to therapy with other screening or diagnostic techniques.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/sangue , Neoplasias/enzimologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Análise de Variância , Biomarcadores Tumorais/sangue , Estudos de Casos e Controles , Distribuição de Qui-Quadrado , China/epidemiologia , Feminino , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Neoplasias/epidemiologia , Valor Preditivo dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...