Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS One ; 17(12): e0278152, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36455056

RESUMO

BACKGROUND: This study characterized undernutrition among children (0-24 months) by age groups specified for Infant and Young Child-feeding (IYCF) and determined the association between child malnutrition and IYCF. METHODS: This cross-sectional survey recruited mother-children dyads (N = 1443). WHO standards were used to assess nutritional status and IYCF indicators. Multivariate analyses were performed to assess the association between IYCF and nutritional indicators. RESULTS: Stunting, underweight, wasting, overweight, and obesity were prevalent in 33.1%, 26%, 20.2%, 4.6%, and 2.9% of the children, respectively. Age-wise distribution of undernutrition identified severity of stunting and underweight at 10-24 months (median < -1.6 SD; < -1.2 SD; 25th percentile at -2.6 & -2.2 SD respectively) and wasting highest at 0-6 months (25th percentile close to -2SD). Boys manifested higher stunting (lower value -5.2 SD) and were more wasted (lower value -4.7 SD). IYCF prevalence recorded early initiation at 45.2%, exclusive breastfeeding at 23.1%, and prelacteal and bottle-feeding at 37.5 and 22.5% respectively. Child minimum diet diversity (MDD) ≥4 was not achieved by 84%. Minimum meal frequency and minimum acceptable diet were achieved by 75% and 14% respectively. Bottle-feeding increased the odds of wasting [AOR: 1.501 (95% CI: 1.062-2.121)], severe stunting [AOR: 1.595 (95% CI: 1.079-2.358)] and underweight [AOR: 1.519 (95% CI 1.102-2.094)]. Wasting according to BAZ scores was associated with delayed initiation of breastfeeding [AOR: 1.387 (95% CI: 1.018-1.889)] and bottle feeding [AOR: 1.538 (95% CI: 1.087-2.175)]. Delayed introduction of complementary feeding increased the odds of severe stunting [AOR: 2.189 (95% CI: 1.090-4.399)]. Formula feeding increased the odds of underweight [AOR: 1.738 (95% CI: 1.046-2.888)] and obesity [AOR: 4.664 (95% CI: 1.351-16.10)]. Prelacteal feeding increased the odds of severe forms of stunting and underweight by 56% and 79% respectively, and overweight by 96%. CONCLUSION: Setting and age-specific interventions to improve age-appropriate child-feeding practices are vital to address the double burden of malnutrition in the critical age group.


Assuntos
Desnutrição , Áreas de Pobreza , Masculino , Criança , Lactente , Humanos , Pré-Escolar , Magreza/epidemiologia , Sobrepeso/epidemiologia , Estudos Transversais , Índia/epidemiologia , Caquexia , Transtornos do Crescimento/epidemiologia , Transtornos do Crescimento/prevenção & controle , Obesidade
2.
Front Oncol ; 12: 881902, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35814366

RESUMO

Objective: To determine the efficacy, safety, and durability of the use of AHCC supplementation for 6 months to support the host immune system to clear high-risk human papillomavirus (HPV) infections. The AHCC supplement is a proprietary, standardized extract of cultured lentinula edodes mycelia (AHCC®, Amino Up, Ltd., Sapporo, Japan) that has been shown to have unique immune modulatory benefits. Study Design: This was a randomized, double-blind, placebo-controlled study (CTN: NCT02405533) in 50 women over 30 years of age with confirmed persistent high-risk HPV infections for greater than 2 years. Patients were randomized to placebo once daily for 12 months (N = 25) or AHCC 3-g supplementation by mouth once daily on empty stomach for 6 months followed by 6 months of placebo (N = 25). Every 3 months, patients were evaluated with HPV DNA and HPV RNA testing as well as a blood sample collected to evaluate a panel of immune markers including interferon-alpha, interferon-beta (IFN-ß), interferon-gamma (IFN-γ), IgG1, T lymphocytes, and natural killer (NK) cell levels. At the completion of the 12-month study period, patients on the placebo arm were given the option to continue on the study to receive AHCC supplementation unblinded for 6 months with the same follow-up appointments and testing as the intervention arm. Results: Fifty women with high-risk HPV were enrolled, and 41 completed the study. Fourteen (63.6%) of the 22 patients in the AHCC supplementation arm were HPV RNA/HPV DNA negative after 6 months, with 64.3% (9/14) achieving a durable response defined as being HPV RNA/HPV DNA negative 6 months off supplementation. On the placebo arm, two (10.5%) of 19 patients were HPV negative at 12 months. In the twelve placebo arm patients who elected to continue on the unblinded study, 50% (n = 6) were HPV RNA/HPV DNA negative after 6 months of AHCC supplementation. At the time of completion of the study, there were a total of 34 patients (22 blinded and 12 unblinded) who had received AHCC supplementation with an overall response rate of 58.8% that cleared HPV persistent infections. At the time of enrollment, the mean IFN-ß level was 60.5 ± 37.6 pg/ml in women with confirmed persistent HPV infections. Suppression of IFN-ß to less than 20 pg/ml correlated with an increase in T lymphocytes and IFN-γ and durable clearance of HPV infections in women who received AHCC supplementation. Conclusion: Results from this phase II study demonstrated that AHCC 3 g once daily was effective to support the host immune system to eliminate persistent HPV infections and was well tolerated with no significant adverse side effects reported. The duration of AHCC supplementation required beyond the first negative result needs more evaluation to optimize success for durable outcomes. The suppression of the IFN-ß level to less than 20 pg/ml correlated with clearance of HPV infections and merits further evaluation as a clinical tool for monitoring patients with HPV infections. Clinical Trial Registration: clinicaltrials.gov/ct2/, identifier NCT02405533.

3.
Front Oncol ; 9: 173, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30949451

RESUMO

Objective: There is currently no effective medicine or supplement for clearance of high risk- human papillomavirus (HR-HPV) infections. We have taken a systematic approach evaluating the potential use of AHCC supplementation to support clearance of HR-HPV infections. The primary objective of this research was to evaluate AHCC supplementation to modulation of the host immune system to clear HR-HPV infections from bench to bedside. Methods: Cervical cancer cells, CaSki (HPV16+), HeLa(HPV18+), SiHa(HPV16/18+), and C-33A(HPV-), were treated in vitro with AHCC 0.42 mg/mL daily x7 days then observed x7 days with daily sample collection. A confirmatory study in cervical cancer mouse models, SiHa(HPV16/18+) and C-33A(HPV-), was conducted: mice were divided into three groups per cell line then dosed with AHCC 50 mg/kg/d (N = 10), or vehicle alone (N = 10), or no supplementation (N = 10) for a total of 90 days followed by 30 days of observation. Tumors were measured 3x/week and blood samples collected bi-weekly to evaluate interferon (IFN) alpha(α), beta(ß), and gamma(γ) and immunoglobulin G(IgG) by immunoassays. Tumors were evaluated for HR-HPV expression by PCR. Two pilot studies of 10 patients each were conducted in women with confirmed persistent HR-HPV+ infections. The 1st study evaluated AHCC 3g from 5 weeks up to 6 months and 2nd study evaluated AHCC 1g < 8 months. HR-HPV DNA status and the immune panel were monitored at each visit. Results: HR-HPV clearance was observed in vitro and confirmed in the animal studies as a durable response. Four of six (66.7%) patients had confirmed HR-HPV clearance after 3-6 months of AHCC 3g. Similarly, 4 of 9 (44%) patients had confirmed HR-HPV clearance after 7 months of AHCC 1g. Suppression of IFNß <25 pg/mL was observed in those clearing the HR-HPV infection. Conclusion: Pre-clinical in vitro and in vivo studies demonstrated durable clearance of HR-HPV infections. The preliminary data from the two pilot studies suggested that AHCC supplementation supports the host immune system for successful clearance of HR-HPV infections. A confirmatory phase II randomized, double-blinded, placebo-controlled study is ongoing.

4.
Gynecol Oncol ; 152(2): 265-269, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30466807

RESUMO

OBJECTIVE: Niraparib is a poly (ADP-ribose) polymerase inhibitor (PARP) approved for use in maintenance therapy for ovarian cancer that is associated with the unpredictable grade 3/4 thrombocytopenia. This study was conducted to refine patient dosing recommendations for niraparib based upon clinical practice observations of grade 3/4 thrombocytopenia. METHODS AND MATERIALS: Six patient cases were reviewed to identify similarities in patient factors. An in vitro study was conducted using healthy volunteer blood spiked with Niraparib concentrations ranging from 0 ng/mL to 5000 ng/mL. Manual platelet counts were evaluated at different time intervals for each concentration and compared to untreated controls. Data was then analyzed based on percent change in platelet count versus untreated control for each concentration/time point. RESULTS: In three patients with body weight > 80 kg and platelet count >200 × 109/L, decreased creatinine clearance (CrCl) <60 mL/min was identified as potential signal. An additional three patients with weights below 77 kg and/or baseline platelet counts <150 × 109/L were re-evaluated, and it was observed that all had decreased CrCl of <60 mL/min. Albumin <3.5 g/dL was also observed in some patients with thrombocytopenia. The in vitro study, observed a direct concentration-dependent relationship between niraparib and thrombocytopenia. CONCLUSION: The data suggests that renal insufficiency and hypoalbuminemia may be associated with the development of niraparib-induced thrombocytopenia. Moreover, the preliminary in vitro studies also demonstrated a concentration-dependent relationship between niraparib and direct toxicity to platelets.


Assuntos
Indazóis/efeitos adversos , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/tratamento farmacológico , Piperidinas/efeitos adversos , Trombocitopenia/induzido quimicamente , Idoso , Plaquetas/efeitos dos fármacos , Feminino , Humanos , Indazóis/administração & dosagem , Indazóis/sangue , Pessoa de Meia-Idade , Piperidinas/administração & dosagem , Piperidinas/sangue , Inibidores de Poli(ADP-Ribose) Polimerases/administração & dosagem , Inibidores de Poli(ADP-Ribose) Polimerases/efeitos adversos , Inibidores de Poli(ADP-Ribose) Polimerases/sangue , Fatores de Risco , Trombocitopenia/sangue
5.
Integr Cancer Ther ; 17(3): 755-761, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29156992

RESUMO

OBJECTIVE: To determine the activity of fucoidan from Undaria pinnatifida (UPF) and Fucus vesiculosus (FVF) when given in combination of chemotherapy drugs using selected human breast or ovarian cancer orthotopic mouse models. METHODS: Mice were inoculated with 1 × 106 cells of TOV-112d, MCF-7, or ZR-75 subcutaneously or SKOV3-GFP-Luc intraperitoneally on day 0. MCF-7 and ZR-75 mice were administered with estradiol valerate 2 mg/kg in 0.2 mL castor oil subcutaneously two days prior to cell inoculation. Mice were randomized to one of six arms (N = 10/arm) paclitaxel, UPF/paclitaxel, FVF/paclitaxel, tamoxifen, UPF/tamoxifen, or FVF/tamoxifen. Tumors were measured three times per week for 28 days. RESULTS: Improved activity was observed with UPF or FVF in combination with tamoxifen in both the MCF-7 and ZR-75D breast cancer mouse models. Decreased activity of paclitaxel was observed when given in combination with UPF or FVF in both breast cancer mouse models. The combination of FVF/tamoxifen in the TOV-112d ovarian cancer mouse model had improved activity but no there was difference observed with the UPF/tamoxifen in either ovarian cancer mouse model. No difference was observed with combination of UPF or FVF with paclitaxel in human ovarian cancer SKOV3 or TOV-112d orthotopic mouse models. CONCLUSION: This study did confirm that UPF/FVF in combination with tamoxifen did not decrease tamoxifen activity in both breast and ovarian cancer, with some potential to improve activity compared to tamoxifen alone in breast cancers. Previous in vitro studies had suggested UPF and FVF had overall synergistic activity with paclitaxel; however, in the current in vivo human cancer mouse model studies there was no change in paclitaxel activity when given in combination with UPF or FVF in either of the two human ovarian cancer models. Furthermore, this study demonstrated that UPF or FVF given in combination with paclitaxel had a potential antagonistic effect in breast cancer models. Additional studies are warranted to delineate mechanisms contributing to variation in the in vivo activity when given in combination with paclitaxel. As a first step, a clinical pharmacokinetic study evaluating impact of FVF/UPF given in combination with chemotherapy in patients with solid tumors is underway.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Fucus/química , Neoplasias Ovarianas/tratamento farmacológico , Polissacarídeos/farmacologia , Undaria/química , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , Paclitaxel/farmacologia , Tamoxifeno/farmacologia
6.
Integr Cancer Ther ; 16(4): 572-584, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29096568

RESUMO

OBJECTIVES: To evaluate potential hepatic metabolism-mediated drug interactions with fucoidan from Undaria pinnatifida (UPF) or Fucus vesiculosus (FVF) and potential growth inhibition activity with either fucoidan alone or with chemotherapy. In vivo studies were done to confirm safety and investigate fucoidan-mediated immune modulation. METHODS: Cytochrome P450 (CYP450) 3A4, 2C8, 2C9, and 2D6 inhibition experiments were conducted in vitro followed by an ex vivo human hepatocytes model to evaluate the CYP450 induction potential of each fucoidan at highest theoretical concentrations. Four hepatic metabolism phase II pathways-glutathione S transferase (GST), quinone oxidoreductase (QOR), catechol-O-methyltransferases (COMT), and uridine di-phosphate (UDP)-glucuronosyltransferase (UGT)-were evaluated with validated immunoassays. Growth inhibition assays were performed with each fucoidan alone and in combination with chemotherapy agents in a panel of human cancer cell lines. In vivo studies evaluated safety and immune modualtion. RESULTS: CYP450 inhibition was observed with FVF. The GST, QOR, and UGT pathways had no changes. UPF and FVF both interacted with COMT. No growth inhibitory activity in cancer cell lines was observed. UPF and FVF had synergistic activity with paclitaxel or tamoxifen and additive activity with topotecan. In vivo, FVF decreased HeLa human cervical tumor growth and both FVF and UPF decreased TOV-112D human ovarian tumor growth. Otherwise, no significant change in tumor growth was observed. FVF immune modulation of IgG and IL-6 was observed (p<0.03). CONCLUSION: At higher doses, UPF and FVF may have limited potential for drug-supplement interactions, with either CYP450 or COMT hepatic metabolism pathways. Additional studies are warranted to evaluate to confirm findings of fucoidans in combination with chemotherapy.


Assuntos
Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Fucus/química , Polissacarídeos/efeitos adversos , Polissacarídeos/farmacologia , Undaria/química , Animais , Catecol O-Metiltransferase/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores das Enzimas do Citocromo P-450/farmacologia , Sistema Enzimático do Citocromo P-450/metabolismo , Avaliação Pré-Clínica de Medicamentos , Interações Medicamentosas , Feminino , Glucuronosiltransferase/metabolismo , Glutationa Transferase/metabolismo , Células HeLa , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Células MCF-7 , Camundongos , Camundongos Nus , NAD(P)H Desidrogenase (Quinona)/metabolismo
7.
Integr Cancer Ther ; 16(3): 300-307, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28438054

RESUMO

OBJECTIVE: To determine the impact on antitumor activity when active hexose correlated compound (AHCC) in combination with anticancer hormonal agents in orthotopic mouse models of human estrogen receptor positive breast cancer and evaluate impact of AHCC on aromatase activity. METHODS: The study consisted of 7 treatment arms (n=10) conducted in 2 breast cancer mouse models: MCF-7 and ZR-75. Treatment groups included untreated, vehicle, AHCC 50 mg/kg, AHCC 50 mg/kg + tamoxifen 10 mg/kg, tamoxifen 10 mg/kg, AHCC 50 mg/kg + letrozole 10 µg/mouse, or letrozole 10 µg/mouse. All treatments were administered daily by oral gavage for 12 weeks. Tumors were measured 3 times a week. In vitro estrone and 17ß-estradiol enzyme immunoassay was used to evaluate aromatase activity. RESULTS: There was no difference in the activity with the combination of AHCC + tamoxifen compared with tamoxifen ( P = 0.29). In the ZR-75 model (catechol- O-methyltransferase [COMT] wild-type), there was no difference in activity with the letrozole + AHCC compared with letrozole. However, in the MCF-7 model (COMT variant), AHCC + letrozole resulted in a decrease in activity compared with letrozole ( P < 0.01). Immunoassay data suggested that AHCC is a potential inducer of aromatase activity. In both tumor models, there was cytotoxicity observed with AHCC compared with untreated ( P < 0.02). CONCLUSION: AHCC did not change the activity of tamoxifen. AHCC may have some interaction with letrozole in patients with COMT variant genotype. AHCC had cytotoxicity that warrents additional studies to evaluate its potential role for consolidation/prevention of breast cancer.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Polissacarídeos/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Inibidores da Aromatase/farmacologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Estradiol/metabolismo , Antagonistas de Estrogênios/farmacologia , Feminino , Humanos , Letrozol , Células MCF-7 , Camundongos , Nitrilas/farmacologia , Tamoxifeno/farmacologia , Triazóis/farmacologia
8.
Reprod Toxicol ; 53: 92-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25813292

RESUMO

The objective of this study was to elucidate the possible toxic effects on the fetal tissues after exposure to two clinically relevant concentrations of granisetron. Primary cells were isolated from human fetal organs of 16-19 weeks gestational age and treated with 3 ng/mL or 30 ng/mL of granisetron. Cell cycle progression was evaluated by flow cytometry. ELISA was used to detect alterations in major apoptotic proteins. Up to 10% apoptosis in cardiac tissue was observed following treatment with 30 ng/mL granisetron. Neither concentration of granisetron caused alteration in cell cycle progression or alterations in apoptotic proteins in any of the other tissues. At 30 ng/mL granisetron concentration had the potential to induce up to 10% apoptosis in cardiac tissue; clinical significance needs further evaluation. At granisetron 3 ng/mL there was no detectable toxicity or on any fetal tissue in this study. Further research is needed to confirm these preliminary findings and determine if clinically significant.


Assuntos
Antieméticos/toxicidade , Feto/efeitos dos fármacos , Granisetron/toxicidade , Proteínas Reguladoras de Apoptose/metabolismo , Encéfalo/anatomia & histologia , Encéfalo/efeitos dos fármacos , Feminino , Feto/anatomia & histologia , Feto/metabolismo , Coração/anatomia & histologia , Coração/efeitos dos fármacos , Humanos , Intestino Delgado/efeitos dos fármacos , Rim/anatomia & histologia , Rim/efeitos dos fármacos , Pulmão/anatomia & histologia , Pulmão/efeitos dos fármacos , Troca Materno-Fetal , Miocárdio/citologia , Miocárdio/metabolismo , Gravidez
9.
Am J Obstet Gynecol ; 210(3): 275.e1-9, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24333234

RESUMO

OBJECTIVE: The objective of this study was to determine the fetal drug compartment concentrations when various concentrations of carboplatin cross the placental-trophoblastic barrier and the effect on the fetal kidneys. STUDY DESIGN: An ex vivo human placenta perfusion model was utilized. Term human placentae (n = 9) were collected immediately after delivery and then reperfused with plasma concentrations achieved with carboplatin an area under the curve of 5 (1000 ng/mL), 7.5 (5000 ng/mL), or 11 (11,000 ng/mL). Antipyrine was used as a reference compound. Samples were collected over 2 hours. Placental transfer was evaluated by computation of transport fraction and clearance index. Primary cells isolated by explant culture of 16-18 week old fetal organ tissues were incubated with carboplatin for up to 48 hours with untreated cell as controls. Immunohistochemical, flow cytometry analysis, and immunoblotting were applied for the expression of apoptosis-related proteins. RESULTS: Mean transport fractions for carboplatin at low, middle, and high concentrations were 0.05 ± 0.02, 0.04 ± 0.01, and 0.10 ± 0.01, respectively, with clearance indexes of 0.22 ± 0.01, 0.14 ± 0.08, and 0.50 ± 0.07, respectively. The fetal peak concentrations of carboplatin achieved were 61 ± 39 ng/mL (low), 375 ± 248 ng/mL (middle), and 2081 ± 529 ng/mL (high). Fetal kidney cells exposed to carboplatin showed a concentration-dependent increased expression of apoptosis-inducing factor and p53 apoptosis proteins and a time-dependent increase in expression Bax apoptosis protein expression. Apoptosis was confirmed at the high concentration by flow cytometry. CONCLUSION: Doses of carboplatin up to an area under the curve of 7.5 were not associated with significant placental transfer, fetal exposure, or fetal toxic effects. This suggests it might not be necessary to empirically reduce carboplatin doses in pregnant women.


Assuntos
Antineoplásicos/farmacologia , Carboplatina/farmacologia , Troca Materno-Fetal/efeitos dos fármacos , Placenta/efeitos dos fármacos , Adulto , Antineoplásicos/farmacocinética , Carboplatina/farmacocinética , Feminino , Humanos , Troca Materno-Fetal/fisiologia , Placenta/metabolismo , Gravidez
10.
J Antimicrob Chemother ; 67(2): 422-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22129592

RESUMO

OBJECTIVES: To determine whether the catechol functional group on echinocandins decreases the catechol-O-methyltransferase (COMT) metabolism of catechol oestrogens (CEs) and the potential role of this functional group in the development of hepatocellular cancer. METHODS: Human COMT expression was measured by RT-PCR in a panel of selected human cancer cell lines and human hepatocytes. An ex vivo human hepatocyte model was employed to evaluate the metabolism of 17-ß-oestradiol to CEs in the presence of a catechol (B(0)C) versus a non-catechol echinocandin (B(0)) compound. COMT inhibition assays were conducted to evaluate the metabolism of CEs in the presence of B(0)C or B(0). Oestrogen receptor expression in human hepatic carcinoma cells was evaluated by RT-PCR and western blotting. Cell proliferation assays were used to evaluate the impact of B(0) or B(0)C on cancer cell growth. RESULTS: MCF-7 and Hep-G2 cells and human hepatocytes expressed variant Met/Met COMT. At clinically relevant concentrations, only B(0)C significantly increased CE levels in the COMT inhibition assays, to 90.0 µM compared with 79.8 µM in the untreated controls (P = 0.032). A high concentration (500 µg/mL) of B(0)C decreased COMT expression to 79%, 94% and 90% of untreated, baseline control levels in the three cell lines, respectively. B(0)C and B(0) did not increase cell growth in the cancer cell lines evaluated. CONCLUSIONS: At clinically achievable concentrations only B(0)C significantly inhibited COMT activity and increased CE concentrations. Short-term exposure did not alter the rate of cancer cell growth. Confirmation is needed to determine the clinical impact of long-term exposure to and the use of echinocandins with catechol functional groups.


Assuntos
Carcinógenos/toxicidade , Carcinoma Hepatocelular/induzido quimicamente , Carcinoma Hepatocelular/secundário , Inibidores de Catecol O-Metiltransferase , Catecóis/toxicidade , Equinocandinas/toxicidade , Western Blotting , Carcinógenos/química , Catecol O-Metiltransferase/metabolismo , Catecóis/química , Linhagem Celular Tumoral , Equinocandinas/química , Estradiol/metabolismo , Perfilação da Expressão Gênica , Humanos , Vírus da Leucemia Murina , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Relação Estrutura-Atividade
11.
J Oncol Pharm Pract ; 15(1): 35-44, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18753181

RESUMO

PURPOSE: This study was conducted to determine the in vitro optimal combination of selected anticancer agents with gefitinib and evaluate its effect on the expression of correlative biological targets in the cell-signaling pathway. In addition, the effect of gefitinib on the expression of ATP-binding cassette (ABC) transport proteins was evaluated. METHODS: Growth inhibition assays were conducted in six human endometrial cancer cell lines to evaluate the activity of selected anticancer agents with gefitinib compared to each alone. Enzyme linked immunosorbant assay (ELISA) assessed the presence of pEGFR in treated and untreated cells. Evaluation of the suppression of correlative biological targets in the cell-signaling pathway was completed by immunoblotting. RT-PCR was used to characterize the expression of MRP and ABC transport proteins. RESULTS: This in vitro study gefitinib did not observe cytotoxic activity as a single agent. However, the activity of gefitinib as EGFR inhibitor was confirmed. The combination of gefitinib with paclitaxel and docetaxel exhibited improved in vitro cytotoxic activity compared to each antineoplastic agent alone. Suppression of pAKT and p27 in the human endometrial cancer cells treated with selected combinations of chemotherapeutic drugs and gefitinib was observed. CONCLUSION: These data suggest that EGFRinhibitors, such as gefitinib, have the potential to modulate common mechanisms of drug resistance and may have a role in optimizing antineoplastic regimens for the treatment of recurrent endometrial cancer. This may represent a promising option for this class of agents in the treatment of endometrial cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias do Endométrio/tratamento farmacológico , Receptores ErbB/antagonistas & inibidores , Transportadores de Cassetes de Ligação de ATP/efeitos dos fármacos , Transportadores de Cassetes de Ligação de ATP/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Ensaio de Imunoadsorção Enzimática , Feminino , Gefitinibe , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quinazolinas/administração & dosagem , Reação em Cadeia da Polimerase Via Transcriptase Reversa
12.
Cancer Chemother Pharmacol ; 62(3): 483-9, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18004570

RESUMO

PURPOSE: To determine the growth inhibitory effects of mifepristone on endometrial cancer cell growth and evaluate its effect on apoptosis using HEC-1-A and Ishikawa human endometrial cancer cell lines. METHODS: The human endometrial cancer cell lines, HEC-1-A and Ishikawa, were cultured in vitro. MTT assays were completed in order to estimate the IC(50) of mifepristone. Both cell lines were then treated with the respective IC(50) values. Immunohistochemistry assays were performed to investigate the expression of estrogen receptors alpha and beta (ERalpha/beta), progesterone receptor alpha and beta (PR alpha/beta), cyclooxygenase-2 (COX-2), bax, p53, and bcl-2. Flow cytometry analysis was performed to study cell cycle arrest and apoptosis. RESULTS: The estimated IC(50) of mifepristone for HEC-1-A and Ishikawa was found to be 16 and 19 mug/ml respectively. At this concentration, there was no change in either ERalpha/beta or PR alpha/beta in Ishikawa. However, PR beta expression increased with time of treatment in HEC-1-A. Expression of p53 was increased with duration of treatment in both cell lines. Consequently a decrease in bcl-2 and an increase in COX-2 expression were seen in HEC-1-A and Ishikawa cells, respectively. Lastly, flow cytometry analysis confirmed an accumulation of cells in G0 phase after 72 h of treatment in both cell lines. CONCLUSIONS: Mifepristone demonstrates activity in both HEC-1-A and Ishikawa cells at clinically relevant concentrations based on an oral human dose of about 200 mg/day. While its mechanism of action remains unknown, this data supports an increase in apoptosis that may be due to p53 activation rather than hormone receptor mediation. Additional studies are needed to help further identify mifepristone mechanism of action.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias do Endométrio , Antagonistas de Hormônios/farmacologia , Mifepristona/farmacologia , Linhagem Celular Tumoral , Ciclo-Oxigenase 2/biossíntese , Neoplasias do Endométrio/tratamento farmacológico , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Concentração Inibidora 50 , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Receptores de Esteroides/biossíntese , Proteína Supressora de Tumor p53/biossíntese
13.
Cancer Chemother Pharmacol ; 62(1): 51-8, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17849119

RESUMO

PURPOSE: This study was conducted to determine the in vitro optimal combination of selected anticancer agents with gefitinib and evaluate its effect on the expression of correlative biological targets in the cell-signaling pathway. In addition, the effect of gefitinib on the expression of ATP-binding cassette (ABC) transport proteins was evaluated. METHODS: Growth inhibition assays were conducted in five human ovarian cancer cell lines to evaluate the activity of selected anticancer agents in combination with gefitinib compared to each alone. Enzyme linked immunosorbant assay (ELISA) assessed the presence of pEGFR in treated and untreated cells. Expression of correlative biological targets in the cell-signaling pathway was completed by immunoblotting. RT-PCR was used to characterize the expression ABC transport proteins. RESULTS: This in vitro study confirmed gefitinib did not have significant cytotoxic activity, the combination of gefitinib with other chemotherapy drugs demonstrated improved in vitro cytotoxic activity in platinum sensitive ovarian cancer cell lines. Suppression of pAKT and p-erk activation in cells treated with combination of cisplatin and gefitinib was observed and suggests the role of gefitinib inhibition of proliferative cell signaling pathway. CONCLUSION: This data suggests that EGFR-inhibitors, such as gefitinib, have the potential to modulate common mechanisms of drug resistance and may have a role in optimizing chemotherapy regimens for the treatment of ovarian cancer.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Ovarianas/tratamento farmacológico , Quinazolinas/farmacologia , Transportadores de Cassetes de Ligação de ATP/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Interações Medicamentosas , Ensaio de Imunoadsorção Enzimática , Feminino , Gefitinibe , Humanos , Indicadores e Reagentes , Neoplasias Ovarianas/patologia , RNA , RNA Neoplásico/biossíntese , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Soluções
14.
J Oncol Pharm Pract ; 13(1): 39-45, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17621566

RESUMO

OBJECTIVE: The primary objective of this study was to evaluate the potential to increase the in vivo activity of liposomal doxorubicin when administered in combination with other chemotherapeutic agents such as topotecan, docetaxel, gemcitabine, capecitabine, or celecoxib in an ovarian cancer xenograft mouse model to identify new treatment options for recurrent platinum-sensitive/resistant ovarian cancer. METHODS: This was a five-arm study in two xenograft ovarian cancer mouse models, ES-2 (platinum-sensitive), and OVCAR3 (platinumresistant), to evaluate the combination of liposomal doxorubicin with the common chemotherapeutic agents. Each cell line had five mice for each treatment arm, five vehicle control mice, and five liposomal doxorubicin alone control mice. Experiments were done in duplicate. RESULTS: The percentage tumor reduction ranged from 52% to 74.1% for the single-agent treatment arms. Tumor growth inhibition and regression (response) was improved on the combination treatment arms ranging from 76.1% to 100%. We observed increased activity in the liposomal doxorubicin plus topotecan arm, with a 27.3% improvement in response, compared with either agent alone. CONCLUSIONS: The addition of liposomal doxorubicin demonstrated increased antitumor activity compared with either agent used alone. The most active combination treatment arm was liposomal doxorubicin with topotecan which is consistent with recent clinical study reports of enhanced activity with the combination of topoisomerase I and topoisomerase II agents. Additional studies are warranted to evaluate the efficacy and safety to optimize the combination of liposomal doxorubicin and topotecan for the treatment of recurrent or refractory ovarian cancer.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Doxorrubicina/uso terapêutico , Neoplasias Ovarianas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Lipossomos , Camundongos , Camundongos Nus , Compostos de Platina/farmacologia , Topotecan/uso terapêutico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Gynecol Oncol ; 103(2): 518-22, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16690105

RESUMO

OBJECTIVES: The primary objective of this study was to determine the mechanism(s) of cisplatin drug resistance in endometrial cancer cell lines. To evaluate the mechanism that gemcitabine modulates cisplatin drug resistance in endometrial cancer cell lines. METHODS: Combination treatment was completed in panel of four human endometrial cancer cell lines. Growth inhibition assays were conducted in each cell line evaluating combinations of the Ic25, Ic50, and Ic90 to determine optimal dosing for the combination of gemcitabine plus cisplatin. Evaluation of the correlative biological targets for modulation of platinum drug resistance was completed by the respective immunohistochemistry assays. RESULTS: Downregulation of glutathione-S-transferase (GST) activity by 11% to 100% was observed with an associated 78.6% to 100% decrease in intracellular glutathione (GSH) concentrations. In the gemcitabine plus cisplatin treatment arm compared to either alone, there was also downregulation of MSH2, p53, and ERCC1 expression. No changes observed in the pro-apoptotic proteins, BAX or BAD, expression, AKT activation, or MDR1/PGP expression regardless of treatment with combination of gemcitabine plus cisplatin or either agent alone. CONCLUSIONS: There is likely more than one mechanism contributing to the increase synergistic in vitro platinum-resistant cell lines and increase clinical activity that has been observed in patients with platinum-resistant tumors. In this in vitro study, we determined the downregulation of intracellular GST activity and GSH concentration were the predominant mechanisms involved in the modulation of platinum resistance. Downregulation of MSH2, p53 and ERCC1 expression may also contribute to increase cytotoxic activity compared to cisplatin alone.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cisplatino/farmacologia , Desoxicitidina/análogos & derivados , Neoplasias do Endométrio/tratamento farmacológico , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Neoplasias do Endométrio/metabolismo , Neoplasias do Endométrio/patologia , Feminino , Glutationa/metabolismo , Humanos , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...