Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
2.
Mol Pharmacol ; 87(6): 996-1005, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25814515

RESUMO

COH29 [N-(4-(3,4-dihydroxyphenyl)-5-phenylthiazol-2-yl)-3,4-dihydroxybenzamide], a novel antimetabolite drug developed at City of Hope Cancer Center, has anticancer activity that stems primarily from the inhibition of human ribonucleotide reductase (RNR). This key enzyme in deoxyribonucleotide biosynthesis is the target of established clinical agents such as hydroxyurea and gemcitabine because of its critical role in DNA replication and repair. Herein we report that BRCA-1-defective human breast cancer cells are more sensitive than wild-type BRCA-1 counterparts to COH29 in vitro and in vivo. Microarray gene expression profiling showed that COH29 reduces the expression of DNA repair pathway genes, suggesting that COH29 interferes with these pathways. It is well established that BRCA1 plays a role in DNA damage repair, especially homologous recombination (HR) repair, to maintain genome integrity. In BRCA1-defective HCC1937 breast cancer cells, COH29 induced more double-strand breaks (DSBs) and DNA-damage response than in HCC1937 + BRCA1 cells. By EJ5- and DR-green fluorescent protein (GFP) reporter assay, we found that COH29 could inhibit nonhomologous end joining (NHEJ) efficiency and that no HR activity was detected in HCC1937 cells, suggesting that repression of the NHEJ repair pathway may be involved in COH29-induced DSBs in BRCA1-deficient HCC1937 cells. Furthermore, we observed an accumulation of nuclear Rad51 foci in COH29-treated HCC1937 + BRCA1 cells, suggesting that BRCA1 plays a crucial role in repairing and recovering drug-induced DNA damage by recruiting Rad51 to damage sites. In summary, we describe here additional biologic effects of the RNR inhibitor COH29 that potentially strengthen its use as an anticancer agent.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Benzamidas/farmacologia , Reparo do DNA/efeitos dos fármacos , Ribonucleotídeo Redutases/antagonistas & inibidores , Tiazóis/farmacologia , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Proteína BRCA1/genética , Benzamidas/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA por Junção de Extremidades/efeitos dos fármacos , Feminino , Xenoenxertos , Humanos , Camundongos Endogâmicos NOD , Testes de Mutagenicidade , Transplante de Neoplasias , Tiazóis/uso terapêutico , Peixe-Zebra
3.
Cancers (Basel) ; 16(4)2024 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-38398204

RESUMO

From the perspective of health economics, the evaluation of drug-related cost effectiveness and clinical utility is crucial. We conducted a cost-utility analysis of two first-line drugs, tenofovir alafenamide (TAF) and entecavir (ETV), in the treatment of chronic hepatitis B (CHB) patients. We performed inverse probability of treatment weighting (IPTW) to match the independent variables between the two treatment groups. The incremental cost effectiveness ratio (ICER) of the two treatment groups was simulated using a decision tree with the Markov annual-cycle model. A total of 54 patients treated with TAF and 98 with ETV from January 2016 to December 2020 were enrolled. The total medical cost in the TAF group was NT$76,098 less than that in the ETV group, and TAF demonstrated more effectiveness than ETV by 3.19 quality-adjusted life years (QALYs). When the time horizon was set at 30 years, the ICER of the TAF group compared with the ETV group was -NT$23,878 per QALY, suggesting more cost savings for TAF. Additionally, with the application of TAF, over NT$366 million (approximately US$12 million) can be saved annually. TAF demonstrates cheaper medical costs and more favorable clinical QALYs than ETV. To balance health insurance benefits and cost effectiveness, TAF is the optimal treatment for CHB.

4.
Cancer Res ; 67(1): 16-21, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17210678

RESUMO

p53R2, which is one of the two known ribonucleotide reductase small subunits (the other being M2), is suggested to play an important role in supplying deoxynucleotide triphosphates (dNTP) for DNA repair during the G(1) or G(2) phase of the cell cycle. The ability of p53R2 to supply dNTPs for repairing DNA damages requires the presence of a functional p53 tumor suppressor. Here, we report in vivo physical interaction and colocalization of p53R2 and p21 before DNA damage. Mammalian two-hybrid assay further indicates that the amino acids 1 to 113 of p53R2 are critical for interacting with the NH(2)-terminal region (amino acids 1-93) of p21. The binding between p21 and p53R2 decreases inside the nucleus in response to UV, the time point of which corresponds to the increased binding of p21 with cyclin-dependent kinase-2 (Cdk2), and the decreased Cdk2 activity in the nucleus at G(1). Interestingly, p53R2 dissociates from p21 but facilitates the accumulation of p21 in the nucleus in response to UV. On the other hand, the ribonucleotide reductase activity increases at the corresponding time in response to UV. These data suggest a new function of p53R2 of cooperating with p21 during DNA repair at G(1) arrest.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Dano ao DNA/fisiologia , Fase G1/efeitos da radiação , Ribonucleotídeo Redutases/metabolismo , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Núcleo Celular/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética , DNA de Neoplasias/efeitos da radiação , Fase G1/fisiologia , Humanos , Células KB , Ligação Proteica , Ribonucleotídeo Redutases/biossíntese , Ribonucleotídeo Redutases/genética , Relação Estrutura-Atividade , Transfecção
7.
Cancer Genomics Proteomics ; 5(3-4): 151-60, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18820369

RESUMO

BACKGROUND: The expression and activity of ribonucleotide reductase (RR) has been associated with resistance to multiple drugs in human cancer. The use of antisense oligonucleotide drug, GTI-2040, a 20-mer phosphorothioate oligonucleotide complemented to the human RR M2 subunit mRNA, represents an effective strategy for inhibiting RR. The increased specificity due to the anti-resistance effect of GTI-2040 may also lead to a more favorable therapeutic outcome. MATERIALS AND METHODS: To understand the molecular mechanism underlying RR inhibition, patients' blood samples were analyzed using multiple dimensional proteomics technology via matrix-assisted laser desorption and ionization time-of-flight (MALDI-TOF) mass spectrometry. RESULTS: A major difference occurred at 5k m/z in the MALDI profile, which appeared only in the non-responsive group and diminished after GTI-2040 treatment. This specific peptide peak remained at the basal level in responsive patients. The peak was identified to represent the F-box/LLR-repeat protein 17 (FBXL17) through nanoelectrospray ionization liquid chromatography-tandem mass spectrometry (nanoESI LC-MS/MS). Further characterization revealed that FBXL17 [corrected] directly interacts with the human RR M2 (RRM2) subunit to promote hRRM2 overexpression in the breast cancer cell line MCF-7. CONCLUSION: Validation of this protein using real-time RT-PCR indicates the F-box protein 17 (FBXL17) can serve as a therapeutic target and surrogate marker for breast cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Proteínas F-Box/metabolismo , Oligodesoxirribonucleotídeos/uso terapêutico , Sequência de Bases , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida , Primers do DNA , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
14.
15.
Biochem Pharmacol ; 73(12): 1927-38, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17428446

RESUMO

Mechanistic aberrations leading to Gemcitabine (2',2'-dFdCyd,2,2-difluorodeoxycytidine, Gem) resistance may include alteration in its transport, metabolism and incorporation into DNA. To explore the mechanism of Gem resistance, the restriction fragment differential display PCR (RFDD-PCR) was employed to compare the mRNA expression patterns of KBGem (Gem resistant), KBHURs (hydroxyurea resistant) and KBwt (parental KB cell). Nine gene fragments were overexpressed specifically in the KBGem clone. Sequencing and BLAST results showed that three fragments represent cytochrome C oxidase (CCOX, respiration complex IV) subunit III (CCOX3). The cDNA microarray confirmed that the mRNAs of CCOX and ATP synthase subunits were upregulated in KBGem as compared to KBwt and KBHURs. The increase in CCOX1 protein and activity led to the increase of free ATP concentration, which is consistent with the gene expression profile of KBGem. Furthermore, the sensitivity to Gem could be reversed by sodium azide, a CCOX inhibitor. Following the treatment of sodium azide, the cellular accumulation of [3H]-Gem increased in a dose (of azide)-dependent manner, which is associated with increase of [3H]-Gem incorporation into DNA in KBGem. In summary, an increase of CCOX activity and free ATP level may reduce the transport, metabolism and DNA incorporation of Gem, resulting in Gem resistance.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , DNA/metabolismo , Desoxicitidina/análogos & derivados , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Trifosfato de Adenosina/análise , Western Blotting , Células Clonais , DNA/genética , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Complexo IV da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Complexo IV da Cadeia de Transporte de Elétrons/genética , Inibidores Enzimáticos/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Células KB , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , RNA Mensageiro/metabolismo , Azida Sódica/farmacologia , Gencitabina
16.
Clin Colorectal Cancer ; 6(5): 374-81, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17311703

RESUMO

BACKGROUND: Ribonucleoside diphosphate reductase plays a key role in converting ribonucleoside diphosphate to 2'-deoxyribonucleoside diphosphate, which is necessary for DNA repair and replication. To determine if human ribonucleotide reductase small subunit M2 (hRRM2) and p53-dependent human ribonucleotide reductase small subunit R2 (p53R2) play roles on invasion ability of cancer cells, the gene transferring technique was used to construct stable hRRM2 and p53R2 overexpression transfectants. Increase of hRRM2 dramatically enhanced the cell migration in KB and PC-3 cells, but p53R2 overexpression reduced cellular invasion potential to 50% and 40% in KB and PC-3 cells, respectively. Furthermore, hRRM2 enhanced cancer cells to induce the cell migration of Human Umbilical Vein Endothelial Cells, but p53R2 reduced this ability in transfectants. PATIENTS AND METHODS: To further determine the role of human ribonucleotide reductase subunits on cancer metastasis, a tissue array, including 59 primary and 49 metastatic colon adenocarcinoma samples, was used. Immunohistochemistry was used to evaluate the relationship between human ribonucleotide reductase subunits and metastasis. RESULTS: Univariate and multivariate analysis revealed that p53R2 is negatively related to the metastasis of colon adenocarcinoma samples (odds ratio, 0.23; P < 0.05); hRRM2 increases the risk of metastasis in colon cancer, but did not show significantly. Thus, opposing regulation of hRRM2 and p53R2 in invasion potential might play a critical role in determining the invasion and metastasis phenotype in cancer cells. CONCLUSION: The expression level of ribonucleotide reductase small subunits could serve as biomarkers to predict the malignancy potential of human cancers in the future.


Assuntos
Biomarcadores Tumorais , Neoplasias do Colo/fisiopatologia , Ribonucleotídeo Redutases/metabolismo , Proteína Supressora de Tumor p53 , Adulto , Idoso , Proteínas de Ciclo Celular , Movimento Celular , Sobrevivência Celular , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Prognóstico , Proteínas de Ligação a RNA , Ribonucleosídeo Difosfato Redutase/metabolismo , Risco , Proteínas Supressoras de Tumor
17.
Clin Cancer Res ; 12(21): 6337-44, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17085643

RESUMO

PURPOSE: Previous gene transfection studies have shown that the accumulation of human ribonucleotide reductase small subunit M2 (hRRM2) enhances cellular transformation, tumorigenesis, and malignancy potential. The latest identified small subunit p53R2 has 80% homology to hRRM2. Here, we investigate the role of p53R2 in cancer invasion and metastasis. EXPERIMENTAL DESIGN: The immunohistochemistry was conducted on a tissue array including 49 primary and 59 metastatic colon adenocarcinoma samples to determine the relationship between p53R2 expression and metastasis. A Matrigel invasive chamber was used to sort the highly invasive cells and to evaluate the invasion potential of p53R2. RESULTS: Univariate and multivariate analyses revealed that p53R2 is negatively related to the metastasis of colon adenocarcinoma samples (odds ratio, 0.23; P<0.05). The decrease of p53R2 is associated with cell invasion potential, which was observed in both p53 wild-type (KB) and mutant (PC-3 and Mia PaCa-2) cell lines. An increase in p53R2 expression by gene transfection significantly reduced the cellular invasion potential to 54% and 30% in KB and PC-3 cells, respectively, whereas inhibition of p53R2 by short interfering RNA resulted in a 3-fold increase in cell migration. CONCLUSIONS: Opposite regulation of hRRM2 and p53R2 in invasion potential might play a critical role in determining the invasion and metastasis phenotype in cancer cells. The expression level of ribonucleotide reductase small subunits may serve as a biomarker to predict the malignancy potential of human cancers in the future.


Assuntos
Adenocarcinoma/metabolismo , Biomarcadores Tumorais/análise , Proteínas de Ciclo Celular/metabolismo , Neoplasias do Colo/metabolismo , Invasividade Neoplásica , Ribonucleotídeo Redutases/metabolismo , Western Blotting , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Humanos , Imuno-Histoquímica , RNA Interferente Pequeno , Ribonucleosídeo Difosfato Redutase/metabolismo , Transfecção , Proteína Supressora de Tumor p53/metabolismo
18.
Anticancer Res ; 26(4B): 2761-7, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16886595

RESUMO

BACKGROUND: Ribonucleotide reductase (RR) inhibition by hydroxyurea (HU) causes deoxyribonucleotide (dNTP) depletion, which activates the replication checkpoint, a part of the S-phase checkpoint that responds to DNA damage by inhibiting late origin firing. It also transactivates RR and other genes involved in DNA replication and repair. ICBP90 (overexpressed in breast cancer) is a novel Rb-associating transactivator for the human topoisomerase IIalpha gene and responds to DNA damage-induced checkpoint signaling. MATERIALS AND METHODS: ICBP90 expression was monitored by Western blot. Promoter activity was detected via the luciferase assay and gene silencing via siRNA. Cell death was monitored by the MTT assay. RESULTS: dNTP depletion by HU induced ICBP90, ICBP90 transactivated RR's M2 subunit gene, and ICBP90 induction was necessary for HU-induced M2 accumulation. Blocking the M2 accumulation via anti-ICBP90 siRNA caused greater sensitivity in HU-resistant human cancer. CONCLUSION: A transcriptional intervention strategy is presented through which HU-resistant cancers may be eradicated without dose escalation.


Assuntos
Proteínas Estimuladoras de Ligação a CCAAT/biossíntese , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/metabolismo , Hidroxiureia/farmacologia , Ribonucleosídeo Difosfato Redutase/antagonistas & inibidores , Antineoplásicos/farmacologia , Proteínas Estimuladoras de Ligação a CCAAT/antagonistas & inibidores , Proteínas Estimuladoras de Ligação a CCAAT/genética , Desoxirribonucleotídeos/metabolismo , Indução Enzimática , Inativação Gênica , Neoplasias de Cabeça e Pescoço/enzimologia , Neoplasias de Cabeça e Pescoço/genética , Humanos , Células KB , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Ribonucleosídeo Difosfato Redutase/biossíntese , Ribonucleosídeo Difosfato Redutase/metabolismo , Ativação Transcricional , Ubiquitina-Proteína Ligases
19.
Cancer Res ; 73(4): 1298-307, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23393200

RESUMO

Fibroblast growth factor (FGF) receptor (FGFR) substrate 2 (FRS2) is an adaptor protein that plays a critical role in FGFR signaling. FRS2 is located on chromosome 12q13-15 that is frequently amplified in liposarcomas. The significance of FRS2 and FGFR signaling in high-grade liposarcomas is unknown. Herein, we first comparatively examined the amplification and expression of FRS2 with CDK4 and MDM2 in dedifferentiated liposarcoma (DDLS) and undifferentiated high-grade pleomorphic sarcoma (UHGPS). Amplification and expression of the three genes were identified in 90% to 100% (9-11 of 11) of DDLS, whereas that of FRS2, CDK4, and MDM2 were observed in 55% (41 of 75), 48% (36 of 75), and 44% (33/75) of clinically diagnosed UHGPS, suggesting that these "UHGPS" may represent DDLS despite lacking histologic evidence of lipoblasts. Immunohistochemical analysis of phosphorylated FRS2 protein indicated that the FGFR/FRS2 signaling axis was generally activated in about 75% of FRS2-positive high-grade liposarcomas. Moreover, we found that FRS2 and FGFRs proteins are highly expressed and functional in three high-grade liposarcoma cell lines: FU-DDLS-1, LiSa-2, and SW872. Importantly, the FGFR selective inhibitor NVP-BGJ-398 significantly inhibited the growth of FU-DDLS-1 and LiSa-2 cells with a concomitant suppression of FGFR signal transduction. Attenuation of FRS2 protein in FU-DDLS-1 and LiSa-2 cell lines decreased the phosphorylated extracellular signal-regulated kinase 1/2 and AKT and repressed cell proliferation. These findings indicate that analysis of FRS2 in combination with CDK4 and MDM2 will more accurately characterize pathologic features of high-grade liposarcomas. Activated FGFR/FRS2 signaling may play a functional role in the development of high-grade liposarcomas, therefore, serve as a potential therapeutic target.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Lipossarcoma/genética , Proteínas de Membrana/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Arginina/análogos & derivados , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Amplificação de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Imuno-Histoquímica , Lipossarcoma/metabolismo , Lipossarcoma/patologia , Masculino , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Gradação de Tumores , Compostos de Fenilureia/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Pirimidinas/farmacologia , Interferência de RNA , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
20.
Anticancer Res ; 32(11): 4685-90, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23155230

RESUMO

BACKGROUND: Ribonucleotide reductase composed of the hRRM1 and hRRM2 subunits catalyzes the conversion of ribonucleotides to their corresponding deoxy forms for DNA replication. Anti-hRRM2 siRNA degrades hRRM2's mRNA and suppresses tumorigenesis. A Phase I clinical trial demonstrated its therapy potential. HN-1 represents a tumor-specifically internalizing peptide for targeted-drug delivery into human head and neck squamous cell carcinoma. MATERIALS AND METHODS: Internalization of peptide was monitored by fluorescence microscopy. The peptide-siRNA conjugate was chemically synthesized. The hRRM2 expression was monitored by western blot analysis. RESULTS: HN-1(TYR) (HN-1 with two N-terminally added tyrosines) was internalized by human head and neck or breast cancer cells. Anti-hRRM2 siRNA(R) (resistant to RNase degradation) was conjugated to HN-1(TYR) without compromising their properties. The treatment with HN-1(TYR)-anti-hRRM2 siRNA(R) partly suppressed the endogenously expressed hRRM2 in human breast cancer cells. CONCLUSION: Our results establish the utility of tumor-specifically internalizing peptides for targeted siRNA delivery into human cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Terapia Genética/métodos , Neoplasias de Cabeça e Pescoço/metabolismo , Oligopeptídeos/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Western Blotting , Neoplasias da Mama/genética , Neoplasias da Mama/terapia , Linhagem Celular Tumoral , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/terapia , Humanos , Microscopia de Fluorescência , Oligopeptídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ribonucleotídeo Redutases/biossíntese , Ribonucleotídeo Redutases/deficiência , Ribonucleotídeo Redutases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA