Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Int J Oncol ; 30(4): 947-53, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17332934

RESUMO

Dexamethasone (DEX) is mainly used as an anti-emetic agent in cancer therapy. We have recently demonstrated that DEX pretreatment increases the antitumor activity of the cancer chemotherapeutic agents carboplatin and gemcitabine, and decreases host toxicity in nude mouse xenograft models of human cancer. However, the underlying mechanisms are not fully understood. The present study was designed to determine the effects of DEX pretreatment on the anticancer activity of adriamycin (ADR) in a syngeneic model of breast cancer (4T1), emphasizing the effects of DEX on cytokine expression and modulation of ADR pharmacokinetics. We have demonstrated five major new findings about DEX pretreatment: a) it enhances the therapeutic effect of ADR, inducing almost complete inhibition of tumor growth; b) it increases tumor ADR accumulation; c) it modulates the expression of cytokines produced by the tumor, increasing TNFalpha and decreasing IL-1beta and VEGF expression; d) it enhances the effects of ADR on induction of apoptosis and inhibition of cell proliferation; and e) it suppresses nuclear NFkappaB activation and inhibits ADR-induced NFkappaB activation, possibly via IkappaB up-regulation. These findings suggest that DEX can be used as a chemosensitizer and chemoprotectant. These results provide a rationale for the expanded clinical use of DEX for cancer therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Citocinas/metabolismo , Dexametasona/uso terapêutico , Doxorrubicina/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Proliferação de Células/efeitos dos fármacos , Citocinas/genética , Dexametasona/administração & dosagem , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Sinergismo Farmacológico , Feminino , Camundongos , NF-kappa B/metabolismo , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Cancer Chemother Pharmacol ; 59(5): 589-601, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16924497

RESUMO

PURPOSE: Panax ginseng and its extracts have long been used for medical purposes; there is increasing interest in developing ginseng products as cancer preventive or therapeutic agents. The present study was designed to determine biological structure-activity relationships (SAR) for saponins present in Panax ginseng fruits. METHODS: Eleven saponins were extracted from P. ginseng fruits and purified by use of D(101) resin and ordinary and reverse-phase silica gel column chromatography. Their chemical structures were elucidated on the basis of physicochemical constants and NMR spectra. Compounds were then evaluated for SAR with their in vitro cytotoxicity against several human cancer cell lines. RESULTS: The 11 compounds were identified as 20(R)-dammarane-3beta,12beta,20,25-tetrol (25-OH-PPD, 1); 20(R)-dammarane-3beta,6alpha,12beta,20,25-pentol (25-OH-PPT, 2); 20(S)-protopanaxadiol (PPD, 3); daucosterine 4, 20(S)-ginsenoside-Rh(2) (Rh(2), 5); 20(S)-ginsenoside-Rg(3) (Rg(3,) 6); 20(S)-ginsenoside-Rg(2) (Rg(2), 7); 20(S)-ginsenoside-Rg(1) (Rg(1), 8); 20(S)-ginsenoside-Rd (Rd, 9); 20(S)-ginsenoside-Re (Re, 10); and 20(S)-ginsenoside-Rb(1) (Rb(1), 11). Among the eleven compounds, 1, 3 and 5 were the most effective inhibitors of cell growth and proliferation and inducers of apoptosis and cell cycle arrest. For 1, the IC(50) values for most cell lines were in the range of 10-60 microM, at least twofold lower than for any of the other compounds. Compounds 1 and 3 had significant, dose-dependent effects on apoptosis, proliferation, and cell cycle progression. CONCLUSIONS: The results suggest that the type of dammarane, the number of sugar moieties, and differences in the substituent groups affect their anti-cancer activity. This information may be useful for evaluating the structure/function relationship of other ginsenosides and their aglycones and for development of novel anticancer agents.


Assuntos
Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Frutas/química , Ginsenosídeos/química , Ginsenosídeos/farmacologia , Panax/química , Antineoplásicos Fitogênicos/isolamento & purificação , Apoptose/efeitos dos fármacos , Produtos Biológicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ginsenosídeos/isolamento & purificação , Humanos , Indicadores e Reagentes , Espectroscopia de Ressonância Magnética , Relação Estrutura-Atividade
15.
Mol Cancer Ther ; 5(8): 2106-14, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16928832

RESUMO

Oligonucleotides containing CpG motifs and immunomodulatory oligonucleotides (IMO) containing a synthetic immunostimulatory dinucleotide and a novel DNA structure have been suggested to have potential for the treatment of various human diseases. In the present study, a newly designed IMO was evaluated in several models of human (MCF-7 and BT474 xenograft) and murine (4T1 syngeneic) breast cancer. Pharmacokinetics studies of the IMO administered by s.c., i.v., p.o., or i.p. routes were also accomplished. The IMO was widely distributed to various tissues by all four routes, with s.c. administration yielding the highest concentration in tumor tissue. The IMO inhibited the growth of tumors in all three models of breast cancer, with the lowest dose of the IMO inhibiting MCF-7 xenograft tumor growth by >40%. Combining the IMO with the anticancer antibody, Herceptin, led to potent antitumor effects, resulting in >96% inhibition of tumor growth. The IMO also exerted in vitro antitumor activity, as measured by cell growth, apoptosis, and proliferation assays in the presence of Lipofectin. This is the first report of the pharmacokinetics of this agent in normal and tumor-bearing mice. Based on the present results, we believe that the IMO is a good candidate for clinical development for breast cancer therapy used either alone or in combination with conventional cancer therapeutic agents.


Assuntos
Neoplasias da Mama/terapia , Fatores Imunológicos/farmacologia , Imunoterapia/métodos , Oligonucleotídeos/imunologia , Oligonucleotídeos/farmacologia , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Fatores Imunológicos/farmacocinética , Camundongos , Camundongos Nus , Oligonucleotídeos/farmacocinética , Distribuição Tecidual , Trastuzumab , Células Tumorais Cultivadas
16.
Mol Cancer Ther ; 5(6): 1585-92, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16818518

RESUMO

Lung cancer is a leading cause of death world-wide and the long-term survival rate for lung cancer patients is one of the lowest for any cancer. New therapies are urgently needed. The present study was designed to evaluate an immunomodulatory oligonucleotide as a novel type of therapy for lung cancer. The in vivo effects of the immunomodulatory oligonucleotides were determined in four tumor models derived from human non-small cell lung cancer (NSCLC) cell lines (A549, H1299, H358, and H520), administered alone or in combination with conventional chemotherapeutic agents used to treat lung cancer. The in vitro effects of the immunomodulatory oligonucleotide on the growth, apoptosis, and proliferation of NSCLC cells were also determined. We also examined NSCLC cells for expression of Toll-like receptor 9 (TLR9), the receptor for the immunomodulatory oligonucleotide. We showed several important findings: (a) treatment with the immunomodulatory oligonucleotide led to potent antitumor effects, inhibiting tumor growth by at least 60% in all four in vivo models; (b) combination with the immunomodulatory oligonucleotide led to enhanced effects following treatment with gemcitabine or Alimta; (c) the immunomodulatory oligonucleotide increased apoptosis, decreased proliferation, and decreased survival in A549 cells in vitro; and (d) both TLR9 mRNA and protein were expressed in NSCLC cells. The immunomodulatory oligonucleotide has potent antitumor effects as monotherapy and in combination with conventional chemotherapeutic agents, and may act directly on NSCLC cells via TLR9. The present study provides a rationale for developing the immunomodulatory oligonucleotide for lung cancer therapy.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Oligonucleotídeos/uso terapêutico , Receptor Toll-Like 9/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Tolerância a Medicamentos , Feminino , Glutamatos/administração & dosagem , Guanina/administração & dosagem , Guanina/análogos & derivados , Humanos , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Nus , Pemetrexede , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Toll-Like 9/imunologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
17.
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
18.
Oncogene ; 24(48): 7238-47, 2005 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-16170383

RESUMO

Although previous studies suggested that the tumorigenicity of mouse double minute 2 (MDM2) was due to its negative regulation of p53, the p53-independent interactions may be equally as important. During recent studies utilizing MDM2 inhibitors, we noted that E2F transcription factor 1 (E2F1) was down regulated upon inhibition of MDM2, regardless of the p53 status of the cancer. The present study investigated the mechanisms responsible for the MDM2-mediated increase in E2F1 expression. MDM2 prolongs the half-life of the E2F1 protein by inhibiting its ubiquitination. MDM2 displaces SCF(SKP2), the E2F1 E3 ligase. Direct binding between MDM2 and E2F1 is necessary for the negative effects of MDM2 on E2F1 ubiquitination, and deletion of the MDM2 nuclear localization signal does not result in loss of the ability to increase the E2F1 protein level. The downregulation of E2F1 upon MDM2 inhibition was not due to either pRB or p14(Arf). In addition, E2F1 was responsible for at least part of the inhibition of cell proliferation induced by MDM2 knockdown. In conclusion, the present study provides evidence that stabilization of the E2F1 protein is likely another p53-independent component of MDM2-mediated tumorigenesis. More knowledge about the MDM2-E2F1 interaction may be helpful in developing novel anticancer therapies.


Assuntos
Fator de Transcrição E2F1/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ubiquitina/metabolismo , Animais , Neoplasias da Mama/patologia , Células COS , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Regulação para Baixo , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Deleção de Genes , Meia-Vida , Humanos , Masculino , Microscopia Confocal , Oligonucleotídeos Antissenso/farmacologia , Testes de Precipitina , Neoplasias da Próstata/patologia , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/genética , Regulação para Cima
19.
Am J Pharmacogenomics ; 5(3): 173-90, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15952871

RESUMO

In the past 10 years, progress made in cancer biology, genetics, and biotechnology has led to a major transition in cancer drug design and development. There has been a change from an emphasis on non-specific, cytotoxic agents to specific, molecular-based therapeutics. Mechanism-based therapy is designed to act on cellular and molecular targets that are causally involved in the formation, growth, and progression of human cancers. These agents, which may have greater selectivity for cancer versus normal cells, and which may produce better anti-tumor efficacy and lower host toxicity, can be small molecules, natural or engineered peptides, proteins, antibodies, or synthetic nucleic acids (e.g. antisense oligonucleotides, ribozymes, and siRNAs). Novel targets are identified and validated by state-of-the-art approaches, including high-throughput screening, combinatorial chemistry, and gene expression arrays, which increase the speed and efficiency of drug discovery and development. Examples of oncogene-based, molecular therapeutics that show promising clinical activity include trastuzumab (Herceptin), imatinib (Gleevec), and gefitinib (Iressa). However, the full potential of oncogenes as novel targets for cancer therapy has not been realized and many challenges remain, from the validation of novel targets, to the design of specific agents, to the evaluation of these agents in both preclinical and clinical settings. In maximizing the benefits of molecular therapeutics in monotherapy or combination therapy of cancer, it is necessary to have an understanding of the underlying molecular abnormalities and mechanisms involved. This is the first part of a four-part review in which we discuss progress made in the last decade as it relates to the discovery of novel oncogenes and signal transduction pathways, in the context of their potential as targets for cancer therapy. This part delineates the latest discoveries about the potential use of growth factors and protein tyrosine kinases as targets for therapy. Later parts focus on intermediate signaling pathways, transcription factors, and proteins involved in cell cycle, DNA damage, and apoptotic pathways.


Assuntos
Neoplasias/genética , Neoplasias/terapia , Oncogenes , Animais , Inibidores do Crescimento/uso terapêutico , Substâncias de Crescimento/genética , Substâncias de Crescimento/metabolismo , Humanos , Neoplasias/metabolismo , Oncogenes/efeitos dos fármacos , Farmacogenética , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais
20.
Am J Pharmacogenomics ; 5(4): 247-57, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16078861

RESUMO

This is the second part of a four-part review on potential therapeutic targeting of oncogenes. The previous part introduced the new technologies responsible for the advancement of oncogene identification, target validation, and drug design. Because of such advances, new specific and more efficient therapeutic agents can be developed for cancer. This part of the review continues the exploration of various oncogenes, which we have grouped within seven categories: growth factors, tyrosine kinases, intermediate signaling molecules, transcription factors, cell cycle regulators, DNA damage repair genes, and genes involved in apoptosis. Part I included a discussion of growth factors and tyrosine kinases. This portion of the review covers intermediate signaling molecules and the various strategies used to inhibit their expression or decrease their activities.


Assuntos
Terapia Genética , Neoplasias/genética , Neoplasias/terapia , Oncogenes/genética , Animais , DNA de Neoplasias/genética , Humanos , Neoplasias/fisiopatologia , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA