Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 177
Filtrar
1.
Pharm Res ; 38(2): 335-346, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33604784

RESUMO

PURPOSE: Melanoma is an invasive and very aggressive skin cancer due to its multi-drug resistance that results in poor patient survival. There is a need to test new treatment approaches to improve therapeutic efficacy and reduce side effects of conventional treatments. METHODS: PLA/PVA nanoparticles carrying both Dacarbazine and zinc phthalocyanine was produced by double emulsion technique. The characterization was performed by dynamic light scattering and atomic force microscopy. In vitro photodynamic therapy test assay using MV3 melanoma cells as a model has been performed. In vitro cell viability (MTT) was performed to measure cell toxicity of of nanoparticles with and without drugs using human endothelial cells as a model. The in vivo assay (biodistribution/tissue deposition) has been performed using radiolabeled PLA/PVA NPs. RESULTS: The nanoparticles produced showed a mean diameter of about 259 nm with a spherical shape. The in-vitro photodynamic therapy tests demonstrated that the combination is critical to enhance the therapeutic efficacy and it is dose dependent. The in vitro cell toxicity assay using endothelial cells demonstrated that the drug encapsulated into nanoparticles had no significant toxicity compared to control samples. In-vivo results demonstrated that the drug loading affects the biodistribution of the nanoparticle formulations (NPs). Low accumulation of the NPs into the stomach, heart, brain, and kidneys suggested that common side effects of Dacarbazine could be reduced. CONCLUSION: This work reports a robust nanoparticle formulation with the objective to leveraging the synergistic effects of chemo and photodynamic therapies to potentially suppressing the drug resistance and reducing side effects associated with Dacarbazine. The data corroborates that the dual encapsulated NPs showed better in-vitro efficacy when compared with the both compounds alone. The results support the need to have a dual modality NP formulation for melanoma therapy by combining chemotherapy and photodynamic therapy.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Portadores de Fármacos/química , Melanoma/tratamento farmacológico , Fármacos Fotossensibilizantes/administração & dosagem , Neoplasias Cutâneas/tratamento farmacológico , Animais , Antineoplásicos Alquilantes/efeitos adversos , Antineoplásicos Alquilantes/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Composição de Medicamentos/métodos , Células Endoteliais , Humanos , Isoindóis/administração & dosagem , Isoindóis/farmacocinética , Masculino , Melanoma/patologia , Camundongos , Nanopartículas/química , Compostos Organometálicos/administração & dosagem , Compostos Organometálicos/farmacocinética , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacocinética , Poliésteres/química , Álcool de Polivinil/química , Neoplasias Cutâneas/patologia , Distribuição Tecidual , Compostos de Zinco/administração & dosagem , Compostos de Zinco/farmacocinética
2.
BMC Pregnancy Childbirth ; 20(1): 231, 2020 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-32306909

RESUMO

BACKGROUND: Hodgkin lymphoma (HL) is the most common hematological malignancy during pregnancy. The first-line treatment for HL in pregnancy is the standard ABVD regimen without any drug and/or dose adjustment. However, data on chemotherapy during twin pregnancies are sparse, and a better understanding of the mechanisms involved in exposure to and the toxic effects of anticancer drugs in the fetuses is needed. CASE PRESENTATION: A 41-year-old dichorionic diamniotic pregnant patient was given ABVD treatment for HL at a gestational age of 28 weeks and 3 days. The patient received 2 cycles of chemotherapy with a 15-day therapeutic window including an actual 25 mg/m2 dose of doxorubicin per cycle. Unlike the female twin, the male twin presented four days after birth a left cardiac dysfunction. Doxorubicin cardiotoxicity in the male newborn was also supported by high blood levels of troponin. At one month of age, echocardiography findings were normal. We investigated literature data on physiological aspects of pregnancy that may influence doxorubicin pharmacokinetics, and pharmacodynamic and pharmacokinetic data on the use of doxorubicin in pregnancy. We detailed the role of the transporters in doxorubicin placenta distribution, and tried to understand why only one fetus was affected. CONCLUSIONS: Fetal safety depends at least on maternal doxorubicin pharmacokinetics.Because of drug interactions (i.e. drug metabolism and drug transport), care should always be taken to avoid maternal pharmacokinetic variability. The toxic effects were discrepant between the dizygotic twins, suggesting additional fetus-specific pharmacokinetic/pharmacodynamic factors in doxorubicin toxicity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Doença de Hodgkin/tratamento farmacológico , Complicações Neoplásicas na Gravidez/tratamento farmacológico , Gravidez de Gêmeos/efeitos dos fármacos , Adulto , Bleomicina/farmacocinética , Dacarbazina/farmacocinética , Doxorrubicina/farmacocinética , Feminino , Humanos , Recém-Nascido , Masculino , Gravidez , Vimblastina/farmacocinética
3.
Int J Pharm ; 564: 492-498, 2019 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-31022505

RESUMO

The present study aimed to develop a photochemically stabilized formulation of dacarbazine [5-(3,3-dimethyl-1-triazeno)imidazole-4-carboxamide; DTIC] for reducing the production of algogenic photodegradant (5-diazoimidazole-4-carboxamide; Diazo-IC). Photochemical properties of DTIC were characterized by UV-visible light spectral analysis, reactive oxygen species (ROS) assay, and photostability testing. A pharmacokinetic study was conducted after intravenous administration of DTIC formulations (1 mg-DTIC/kg) to rats. DTIC exhibited strong absorption in the UVA range, and photoirradiated DTIC exhibited marked ROS generation. Thus, DTIC had high photoreactive potential. After exposure of DTIC (1 mM) to simulated sunlight (250 W/m2) for 3 min, remaining DTIC and yielded Diazo-IC were estimated to be ca. 230 µM and 600 µM, respectively. The addition of radical scavenger (1 mM), including l-ascorbic acid, l-cysteine (Cys), l-histidine, D-mannitol, l-tryptophan, or l-tyrosine, to DTIC (1 mM) could attenuate DTIC photoreactions, and in particular, the addition of Cys to DTIC brought ca. 34% and 86% inhibition of DTIC photodegradation and Diazo-IC photogeneration, respectively. There were no significant differences in the calculated pharmacokinetic parameters of DTIC between DTIC and DTIC with Cys (0.67 mg/kg). From these findings, the supplementary use of Cys would be an effective approach to improve the photostability of DTIC with less production of Diazo-IC.


Assuntos
Antineoplásicos Alquilantes , Compostos Azo/química , Cisteína/química , Dacarbazina , Sequestradores de Radicais Livres/química , Imidazóis/química , Luz , Animais , Antineoplásicos Alquilantes/sangue , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/efeitos da radiação , Dacarbazina/sangue , Dacarbazina/química , Dacarbazina/farmacocinética , Dacarbazina/efeitos da radiação , Estabilidade de Medicamentos , Masculino , Fotólise , Ratos Sprague-Dawley
4.
Mater Sci Eng C Mater Biol Appl ; 97: 96-102, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30678984

RESUMO

Layered metal hydroxides have exhibited remarkable benefits in drug delivery, days or even weeks of continuous drug release with improved bioavailability and minimized adverse effects. Here, we report synthesis of a new M+ (Na+) and M2+ (Ca2+) layered double metal hydroxide-based phases with the general formula of [Na0.2Ca0.8(OH)1.4] (NO3)0.4, and 3D pseudo-hexagonal morphology. NaCa layered double metal hydroxide (NaCa-LDH), which is biodegradable, biocompatible, and pH-sensitive, could have broad applicability in drug release and other biomedical applications. Dacarbazine (DAC) is one of the most commonly used chemotherapy drugs for treating various cancers. However, its poor water solubility, short half-life in blood circulation, low response rate and high side effects limit its application. This study aimed to increase its half-life and anticancer activity; minimize its side effects; and prolong its drug release by intercalating of DAC in biodegradable NaCa-LDH (DAC-LDH). Results from the intercalation process show that NaCa-LDH is able to intercalate DAC with a simple procedure and with a good drug loading (38% w/w) through a one pot reaction. The DAC shows a sustained and pH-sensitive release, and the release rate of DAC from DAC-LDH at pH 7.4 is remarkably lower than that at pH 6.0 due to its different release mechanisms. In the latter case, the release was not complete at 24 h. We show that DAC-LDH anticancer efficacy on malignant (A-375) melanoma and breast cancer (MCF-7) cell lines is higher than that of free DAC. These nanoparticles may open a significant way toward the development of a pH-sensitive drug release system that minimizes drug side effect for a wide range of applications.


Assuntos
Materiais Biocompatíveis/química , Dacarbazina/farmacologia , Sistemas de Liberação de Medicamentos/métodos , Hidróxidos/química , Melanoma/tratamento farmacológico , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacologia , Materiais Biocompatíveis/síntese química , Materiais Biocompatíveis/farmacologia , Cálcio/química , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Preparações de Ação Retardada , Liberação Controlada de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Meia-Vida , Humanos , Concentração de Íons de Hidrogênio , Hidróxidos/síntese química , Células MCF-7 , Melanoma/patologia , Sódio/química
5.
Neoplasia ; 20(7): 710-720, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29852323

RESUMO

The anticancer drug temozolomide is the only drug with proven activity against high-grade gliomas and has therefore become a part of the standard treatment of these tumors. P-glycoprotein (P-gp; ABCB1) and breast cancer resistance protein (BCRP; ABCG2) are transport proteins, which are present at the blood-brain barrier and limit the brain uptake of substrate drugs. We have studied the effect of P-gp and BCRP on the pharmacokinetics and pharmacodynamics of temozolomide, making use of a comprehensive set of in vitro transport experiments and in vivo pharmacokinetic and antitumor efficacy experiments using wild-type, Abcg2-/-, Abcb1a/b-/-, and Abcb1a/b;Abcg2-/- mice. We here show that the combined deletion of Abcb1a/b and Abcg2 increases the brain penetration of temozolomide by 1.5-fold compared to wild-type controls (P < .001) without changing the systemic drug exposure. Moreover, the same increase was achieved when temozolomide was given to wild-type mice in combination with the dual P-gp/BCRP inhibitor elacridar (GF120918). The antitumor efficacy of temozolomide against three different intracranial tumor models was significantly enhanced when Abcb1a/b and Abcg2 were genetically deficient or pharmacologically inhibited in recipient mice. These findings call for further clinical testing of temozolomide in combination with elacridar for the treatment of gliomas, as this offers the perspective of further improving the antitumor efficacy of this already active agent.


Assuntos
Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Antineoplásicos Alquilantes/farmacologia , Barreira Hematoencefálica/metabolismo , Dacarbazina/análogos & derivados , Proteínas de Neoplasias/antagonistas & inibidores , Subfamília B de Transportador de Cassetes de Ligação de ATP/antagonistas & inibidores , Animais , Antineoplásicos Alquilantes/farmacocinética , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Modelos Animais de Doenças , Humanos , Imageamento por Ressonância Magnética , Masculino , Camundongos , Suínos , Temozolomida
6.
Int J Nanomedicine ; 13: 3039-3051, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29861635

RESUMO

PURPOSE: Glioblastoma multiforme (GBM) is the most common malignant brain tumor originating in the central nervous system in adults. Based on nanotechnology such as liposomes, polymeric nanoparticles, and lipid nanoparticles, recent research efforts have been aimed to target drugs to the brain. METHODS: In this study, lactoferrin- and arginine-glycine-aspartic acid (RGD) dual- ligand-comodified, temozolomide and vincristine-coloaded nanostructured lipid carriers (L/RT/V-NLCs) were introduced for GBM combination therapy. The physicochemical properties of L/R-T/V-NLCs such as particle size, zeta potential, and encapsulated efficiency are measured. The drug release profile, cellular uptake, cytotoxicity, tissue distribution, and antitumor activity of L/R-T/V-NLCs are further investigated in vitro and in vivo. RESULTS: L/R-T/V-NLCs were stable with nanosize and high drug encapsulation efficiency. L/R-T/V-NLCs exhibited sustained-release behavior, high cellular uptake, high cytotoxicity and synergy effects, increased drug accumulation in the tumor tissue, and obvious tumor inhibition efficiency with low systemic toxicity. CONCLUSION: L/R-T/V-NLCs could be a promising drug delivery system for glioblastoma chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Portadores de Fármacos/química , Neoplasias Neuroepiteliomatosas/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Dacarbazina/farmacocinética , Portadores de Fármacos/administração & dosagem , Sistemas de Liberação de Medicamentos/métodos , Liberação Controlada de Fármacos , Humanos , Lactoferrina/administração & dosagem , Lactoferrina/química , Lipídeos/química , Camundongos Endogâmicos BALB C , Nanoestruturas/química , Oligopeptídeos/química , Temozolomida , Distribuição Tecidual , Vincristina/administração & dosagem , Vincristina/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Int J Biol Macromol ; 116: 1260-1267, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29775717

RESUMO

The present study was designed to develop and optimize Temozolomide nano lipid chitosan hydrogel formulations (TMZNLCHG) to target the brain through nasal route. The formulation was developed using chitosan as a gelling agent and Vit E: gelucire 44/14 blend as lipid. The formulations were evaluated for particle size, encapsulation efficiency (%EE), drug loading (DL), morphology, drug release, nasal diffusion, cell line study, and histopathology study. The particle size, PDI, %EE, %DL, and drug release were found to be 134 nm, 0.177, 88.45% ±â€¯4.45%, 9.12% ±â€¯0.78%, and 84.23% ±â€¯2.78%, respectively. The enhancement ratio was more than two folds higher than TMZCHG formulation (control) suggesting the superiority of chitosan with lipid as permeability enhancer. The microscopic image of lyophilized TMZNLCHGopt displayed the spherical and rough surface morphology. IC50 was found to be 3.34 µg/ml for TMZNLCHGopt and was 160 µg/ml for pure TMZ. Further, No structural damage was observed with TMZNLCHGopt treated nasal mucosa upon histopathological examination. Overall, the present study produces encouraging findings in the formulation of a non-invasive intranasal route for brain targeting as an alternate to other route for TMZ.


Assuntos
Encéfalo/metabolismo , Dacarbazina/análogos & derivados , Sistemas de Liberação de Medicamentos/métodos , Hidrogéis , Absorção Nasal , Mucosa Nasal/metabolismo , Administração Intranasal , Animais , Encéfalo/patologia , Linhagem Celular , Linhagem Celular Tumoral , Dacarbazina/química , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Cabras , Hidrogéis/química , Hidrogéis/farmacocinética , Hidrogéis/farmacologia , Mucosa Nasal/patologia , Ratos , Ratos Wistar , Temozolomida
8.
Fluids Barriers CNS ; 15(1): 2, 2018 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-29332604

RESUMO

BACKGROUND: The blood-brain barrier (BBB) severely limits the entry of systemically administered drugs including chemotherapy to the brain. In rodents, regadenoson activation of adenosine A2A receptors causes transient BBB disruption and increased drug concentrations in normal brain. This study was conducted to evaluate if activation of A2A receptors would increase intra-tumoral temozolomide concentrations in patients with glioblastoma. METHODS: Patients scheduled for a clinically indicated surgery for recurrent glioblastoma were eligible. Microdialysis catheters (MDC) were placed intraoperatively, and the positions were documented radiographically. On post-operative day #1, patients received oral temozolomide (150 mg/m2). On day #2, 60 min after oral temozolomide, patients received one intravenous dose of regadenoson (0.4 mg). Blood and MDC samples were collected to determine temozolomide concentrations. RESULTS: Six patients were enrolled. Five patients had no complications from the MDC placement or regadenoson and had successful collection of blood and dialysate samples. The mean plasma AUC was 16.4 ± 1.4 h µg/ml for temozolomide alone and 16.6 ± 2.87 h µg/ml with addition of regadenoson. The mean dialysate AUC was 2.9 ± 1.2 h µg/ml with temozolomide alone and 3.0 ± 1.7 h µg/ml with regadenoson. The mean brain:plasma AUC ratio was 18.0 ± 7.8 and 19.1 ± 10.7% for temozolomide alone and with regadenoson respectively. Peak concentration and Tmax in brain were not significantly different. CONCLUSIONS: Although previously shown to be efficacious in rodents to increase varied size agents to cross the BBB, our data suggest that regadenoson does not increase temozolomide concentrations in brain. Further studies exploring alternative doses and schedules are needed; as transiently disrupting the BBB to facilitate drug entry is of critical importance in neuro-oncology.


Assuntos
Agonistas do Receptor A2 de Adenosina/uso terapêutico , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Purinas/uso terapêutico , Pirazóis/uso terapêutico , Administração Intravenosa , Administração Oral , Adulto , Idoso , Antineoplásicos Alquilantes/farmacocinética , Encéfalo/diagnóstico por imagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/cirurgia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/cirurgia , Dacarbazina/farmacocinética , Dacarbazina/uso terapêutico , Estudos de Viabilidade , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/metabolismo , Glioblastoma/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/cirurgia , Receptor A2A de Adenosina/metabolismo , Temozolomida
9.
Pharm Res ; 35(1): 9, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29294212

RESUMO

PURPOSE: To establish a platform for the possibility of effective and safe delivery of Temozolomide (TMZ) to brain via surface engineered (polyamidoamine) PAMAM dendrimer for the treatment of glioblastoma. METHODS: The present study aims to investigate the efficacy of PAMAM-chitosan conjugate based TMZ nanoformulation (PCT) against gliomas in vitro as well as in vivo. The prepared nanoconjugated formulation was characterized by 1H NMR, FT-IR spectroscopy and for surface morphological parameters. The reported approach was also designed in such a way to ensure toxicity before in vivo delivery through conducting the hemolytic study. RESULT: Surface morphology was found as per nanoformulation via size, pdi and zeta potential measurement. PCT was more efficacious in terms of IC50 values compared to pure TMZ against U-251 and T-98G glioma cell lines. The in vivo pharmacokinetic parameters proved sustained release fashion such as half-life (t1/2) of 22.74 h (PCT) rather than15.35 h (TMZ) only. Higher concentration was found in heart than brain in bio-distribution studies. This study exhibits the potential applicability of dendrimer and CS in improving the anticancer activity and delivery of TMZ to brain. CONCLUSION: The attractive ex vivo cytotoxicity against two glioma cell lines; U-251 and T-98G and phase solubility studies of TMZ revealed remarkable results. In vivo studies of prepared nanoformulation were significant and promising that explored the double concentration of TMZ in brain due to surface functionality of dendrimer. The reported work is novel and non- obvious as none of such approaches using chitosan anchored dendrimer for TMZ delivery has been reported earlier.


Assuntos
Quitosana/síntese química , Dacarbazina/análogos & derivados , Dendrímeros/química , Glioma/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Transporte Biológico , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Química Farmacêutica/métodos , Dacarbazina/administração & dosagem , Dacarbazina/química , Dacarbazina/farmacocinética , Dacarbazina/uso terapêutico , Dendrímeros/síntese química , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Meia-Vida , Humanos , Microscopia de Força Atômica/métodos , Microscopia Eletrônica de Varredura/métodos , Tamanho da Partícula , Ratos , Ratos Wistar , Solubilidade , Espectroscopia de Infravermelho com Transformada de Fourier/métodos , Propriedades de Superfície , Temozolomida , Distribuição Tecidual/efeitos dos fármacos
10.
Cancer Chemother Pharmacol ; 81(3): 455-460, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29305638

RESUMO

PURPOSE: The purpose of this report is to describe, for the first time, the pharmacokinetics of dacarbazine (DTIC) and its metabolites [5-[3-methyl-triazen-1-yl]-imidazole-4-carboxamide (MTIC), 5-[3-hydroxymethyl-3-methyl-triazen-1-yl]-imidazole-4-carboxamide (HMMTIC) and 5-aminoimidazole-4-carboxamide (AIC)] during pregnancy (n = 2) and postpartum (n = 1). METHODS: Non-compartmental DTIC, MTIC, HMMTIC, and AIC pharmacokinetics (PK) were estimated in one case at 29 week gestation and 18 day postpartum and a second case at 32 week gestation, in women receiving DTIC in combination with doxorubicin, bleomycin, and vinblastine for treatment of Hodgkin's lymphoma. Drug concentrations were measured by HPLC. RESULTS: In the subject who completed both pregnancy and postpartum study days, DTIC area under the concentration-time curve (AUC) was 27% higher and metabolite AUCs were lower by 27% for HMMTIC, 38% for MTIC, and 83% of AIC during pregnancy compared to postpartum. At 7 and 9 year follow-up, both subjects were in remission of their Hodgkin's lymphoma. CONCLUSIONS: Based on these two case reports, pregnancy appears to decrease the metabolism of the pro-drug dacarbazine, likely through inhibition of CYP1A2 activity. Lower concentrations of active metabolites and decreased efficacy may result, although both these subjects experienced long-term remission of their Hodgkin's lymphoma.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Dacarbazina , Doença de Hodgkin , Complicações Neoplásicas na Gravidez , Administração Intravenosa , Adulto , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/efeitos adversos , Antineoplásicos Alquilantes/sangue , Antineoplásicos Alquilantes/farmacocinética , Área Sob a Curva , Bleomicina/farmacologia , Dacarbazina/administração & dosagem , Dacarbazina/efeitos adversos , Dacarbazina/análogos & derivados , Dacarbazina/sangue , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Doxorrubicina/farmacologia , Monitoramento de Medicamentos/métodos , Feminino , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/patologia , Humanos , Recém-Nascido , Estadiamento de Neoplasias , Gravidez , Complicações Neoplásicas na Gravidez/tratamento farmacológico , Complicações Neoplásicas na Gravidez/patologia , Resultado da Gravidez , Resultado do Tratamento , Vimblastina/farmacologia
11.
Pharm Res ; 34(12): 2922-2930, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29063342

RESUMO

PURPOSE: The purpose of this article was to develop, characterize and test (in vivo) dacarbazine microparticles that may be labeled with 99mTc and Ra-223 for both use: diagnostic and therapy of metastatic melanoma. METHODS: We developed by double emulsion solvent evaporation methodology the microparticle. The characterization has been done using, Dynamic Light Scattering (DLS) and Scanning Electron Microscopy (SEM). The labeling with 99mTc and Ra-223 has been done by the direct labeling process. Also the formulation has been tested pre-clinically using Balb/c mice inducted with melanoma, performing the the biodistribution and planar imaging. Cytotoxicity evaluation was also done in M3 V cell line. In order to understand the safety aspects of the microparticles, microbiological study (endotoxin and sterility) has been done. Finally, planar imaging was performed to evaluate the diagnosing aspect. RESULTS: The results showed that a 559 nm microparticles was obtained with a spherical shape. The labeling process with 99mTc reached over 90% of efficacy. On the other hand, the labeling process with Ra-223 showed a 70% efficacy. The results in inducted animals demonstrated that the microparticles were able to reach the tumor with a high rate (20%). Also demonstrated a low recognition by the Mononuclear Phagocytic System. The cytotoxicity and the microbiological control, corroborates the safety aspect of these microparticles. CONCLUSION: The planar image and the possible labeling with Ra-223, corroborates the use as a theragnostic agent for imaging and therapy of Metastatic Melanoma.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Dacarbazina/uso terapêutico , Melanoma/diagnóstico , Melanoma/tratamento farmacológico , Rádio (Elemento)/uso terapêutico , Tecnécio/uso terapêutico , Animais , Antineoplásicos Alquilantes/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dacarbazina/farmacocinética , Sistemas de Liberação de Medicamentos , Feminino , Camundongos Endogâmicos BALB C , Camundongos Nus , Rádio (Elemento)/farmacocinética , Tecnécio/farmacocinética , Distribuição Tecidual
12.
Actas dermo-sifiliogr. (Ed. impr.) ; 108(8): 708-720, oct. 2017. ilus, tab
Artigo em Espanhol | IBECS | ID: ibc-167176

RESUMO

Desde la introducción de la inmunoterapia el tratamiento del melanoma ha sufrido una revolución, consiguiéndose cifras de supervivencia superiores a las que se alcanzaban con los tratamientos previos. Los fármacos inmunomoduladores disponibles actualmente van dirigidos principalmente frente a las moléculas de superficie CTLA-4 y PD-1, que ejercen un efecto inhibidor sobre la respuesta inmune antitumoral. Se trata de un tratamiento en pleno proceso de expansión, y la investigación se dirige hacia el descubrimiento de nuevas moléculas, las combinaciones de los fármacos disponibles o la identificación de biomarcadores que permitan seleccionar a los pacientes idóneos para cada terapia (AU)


Approaches to treating melanoma have changed radically since the introduction of immunotherapy, and survival figures are now higher than possible with earlier therapies. The immunomodulators currently available mainly block CTLA-4 (cytotoxicT lymphocyte-associated molecule-4) and PD-1 (programed cell death protein 1) translocated to the cell surface, where they inhibit the antitumor immune response. Treatments blocking these molecules are being more widely used. Research now seeks new molecular targets, the best combinations of available drugs, and biomarkers that can identify ideal candidates for each one (AU)


Assuntos
Humanos , Neoplasias Cutâneas/imunologia , Melanoma/imunologia , Imunoterapia/métodos , Antígeno CTLA-4/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Fatores Imunológicos/farmacocinética , Dacarbazina/farmacocinética , Anticorpos Monoclonais/farmacocinética
13.
Drugs R D ; 17(3): 427-434, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28756607

RESUMO

BACKGROUND: Temozolomide is an antineoplastic agent of proven efficacy against high-grade gliomas. PURPOSE: The objective of this crossover, single-dose, bioequivalence study was to compare the rate and extent of absorption of oral temozolomide after administration of the study product (Dralitem®, Monte Verde Sociedad Anónima) and the reference product (Temodal®, originator product manufactured by Schering Plough Laboratories) in patients with primary central nervous system (CNS) tumors under fasting conditions. METHODS: Sixteen male and female subjects with primary CNS tumors (excluding CNS lymphoma) were recruited, and were administered temozolomide 200 mg/m2 (Dralitem®) on days 1, 2 and 5 of a 5-day treatment. On days 3 and 4, subjects received the same dose of the test product (Dralitem®), or the reference product (Temodal®) on alternate days. The single dose of 200 mg/m2 was reached with three different temozolomide capsule strengths: 20, 100 and 250 mg. On days 3 and 4, blood samples were obtained for pharmacokinetic (PK) evaluation after drug administration. RESULTS: Bioequivalence assessment was made for the 90% confidence interval (CI) for the ratio of log-transformed means (µT/µR) of the area under the concentration-time curve (AUC from time zero to the final quantifiable sample [AUCt] and AUC from time zero to infinity [AUC∞]) and maximum concentration (C max) of both the test (Dralitem®) and reference (Temodal®) products. The point estimate and 90% CI of the ratios of C max, AUCt and AUC∞ values were 94.37 (82.69-107.69), 100.99 (97.81-104.28) and 101.53 (98.60-104.54), respectively. The ratio met the predefined bioequivalence criteria (i.e. 90% CI between 80.00 and 125.00) for C max and AUC. The most commonly reported adverse events (AE) on this study were vomiting, abdominal pain, asthenia and weakness. One subject experienced expressive aphasia, possibly unrelated to the study drug and with no significant sequelae upon recovery. No serious AEs or unexpected AEs were reported. CONCLUSIONS: Temozolomide Dralitem® capsules, 20, 100 and 250 mg, were bioequivalent to Temodal® capsules under fasting conditions in patients with CNS primary tumors, supporting that they are therapeutic equivalents. ClinicalTrials.gov Identifier: NCT02343081.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Neoplasias do Sistema Nervoso Central/tratamento farmacológico , Dacarbazina/análogos & derivados , Adulto , Idoso , Antineoplásicos Alquilantes/efeitos adversos , Antineoplásicos Alquilantes/farmacocinética , Área Sob a Curva , Cápsulas , Estudos Cross-Over , Dacarbazina/administração & dosagem , Dacarbazina/efeitos adversos , Dacarbazina/farmacocinética , Relação Dose-Resposta a Droga , Jejum , Feminino , Glioma/tratamento farmacológico , Humanos , Masculino , Pessoa de Meia-Idade , Temozolomida , Equivalência Terapêutica , Adulto Jovem
14.
J Control Release ; 261: 138-146, 2017 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-28666727

RESUMO

Even though some progress in diagnosis and treatment has been made over the years, there is still no definitive treatment available for Glioblastoma multiforme (GBM). Convection-enhanced delivery (CED), a continuous infusion-mediated pressure gradient via intracranial catheters, studied in clinical trials, enables in situ drug concentrations several orders of magnitude greater than those achieved by systemic administration. We hypothesized that the currently limited efficacy of CED could be enhanced by a liposomal formulation, thus achieving enhanced drug localization to the tumor site with minimal toxicity. We hereby describe a novel approach for treating GBM by CED of liposomes containing the known chemotherapeutic agent, temozolomide (TMZ). A new technique for encapsulating TMZ in hydrophilic (PEGylated) liposomes, characterized by nano-size (121nm), low polydispersity index (<0.13) and with near-neutral charge (-ʒ,0.2mV), has been developed. Co-infusion of PEGylated Gd-DTPA liposomes and TMZ-liposomes by CED in GBM bearing rats, resulted in enhanced tumor detection with longer residence time than free Gd-DTPA. Treatment of GBM-bearing rats with either TMZ solution or TMZ-liposomes resulted in greater tumor inhibition and significantly higher survival. However, the longer survival and smaller tumor volumes exhibited by TMZ liposomal treatment in comparison to TMZ in solution were insignificant (p<0.053); and only significantly lower edema volumes were observed. Thus, there are no clear-cut advantages to use a liposomal delivery system of TMZ via CED over a drug solution.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Sistemas de Liberação de Medicamentos , Glioblastoma/tratamento farmacológico , Animais , Antineoplásicos Alquilantes/administração & dosagem , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/farmacologia , Convecção , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Gadolínio DTPA/administração & dosagem , Lipossomos , Masculino , Nanopartículas , Tamanho da Partícula , Polietilenoglicóis/química , Ratos , Ratos Endogâmicos Lew , Ratos Sprague-Dawley , Taxa de Sobrevida , Temozolomida , Carga Tumoral
15.
Clin Cancer Res ; 23(20): 6215-6226, 2017 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-28679777

RESUMO

Purpose: HSP90, a highly conserved molecular chaperone that regulates the function of several oncogenic client proteins, is altered in glioblastoma. However, HSP90 inhibitors currently in clinical trials are short-acting, have unacceptable toxicities, or are unable to cross the blood-brain barrier (BBB). We examined the efficacy of onalespib, a potent, long-acting novel HSP90 inhibitor as a single agent and in combination with temozolomide (TMZ) against gliomas in vitro and in vivoExperimental Design: The effect of onalespib on HSP90, its client proteins, and on the biology of glioma cell lines and patient-derived glioma-initiating cells (GSC) was determined. Brain and plasma pharmacokinetics of onalespib and its ability to inhibit HSP90 in vivo were assessed in non-tumor-bearing mice. Its efficacy as a single agent or in combination with TMZ was assessed in vitro and in vivo using zebrafish and patient-derived GSC xenograft mouse glioma models.Results: Onalespib-mediated HSP90 inhibition depleted several survival-promoting client proteins such as EGFR, EGFRvIII, and AKT, disrupted their downstream signaling, and decreased the proliferation, migration, angiogenesis, and survival of glioma cell lines and GSCs. Onalespib effectively crossed the BBB to inhibit HSP90 in vivo and extended survival as a single agent in zebrafish xenografts and in combination with TMZ in both zebrafish and GSC mouse xenografts.Conclusions: Our results demonstrate the long-acting effects of onalespib against gliomas in vitro and in vivo, which combined with its ability to cross the BBB support its development as a potential therapeutic agent in combination with TMZ against gliomas. Clin Cancer Res; 23(20); 6215-26. ©2017 AACR.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Dacarbazina/análogos & derivados , Glioma/metabolismo , Glioma/patologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Isoindóis/farmacologia , Animais , Antineoplásicos/farmacocinética , Benzamidas/farmacocinética , Barreira Hematoencefálica/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Modelos Animais de Doenças , Sinergismo Farmacológico , Quimioterapia Combinada , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Expressão Gênica , Glioma/tratamento farmacológico , Glioma/mortalidade , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Isoindóis/farmacocinética , Camundongos , Células-Tronco Neoplásicas/metabolismo , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Taxa de Sobrevida , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
16.
Mater Sci Eng C Mater Biol Appl ; 75: 165-174, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28415451

RESUMO

In the present study, the PCL-Diol-b-PU/Au nanocompsite nanofibers were fabricated via electrospinning process during two different stages to load an anticancer temozolomide (TMZ) drug into the nanofibers. The first stage was the incorporation of Au nanoparticles into the nanofibers and the second stage was coating the gold nanoparticles on the surface of PCL-Diol-b-PU/Au composite nanofibers. The prepared nanofibrous formulations were characterized using FTIR, SEM and TEM analysis. Box-Behnken-design was used to investigate the influence of electrospinning parameters including solution concentration, applied voltage to tip-collector distance ratio and collector speed on the morphology and fiber diameter of PCL-Diol-b-PU/Au nanofibers. Drug loading efficiency, in vitro release profiles of TMZ from PCL-Diol-b-PU/Au and gold-coated PCL-Diol-b-PU/Au composite nanofibers as well as in vitro antitumor efficacy against U-87 MG human glioblastoma cells were carried out. The TMZ release data were well described using Korsmayer-Peppas kinetic model in which results indicated Fickian diffusion of TMZ from nanofibers. The obtained results revealed the higher efficiency of PCL-Diol-b-PU/Au@TMZ nanofibrous implants for treatment of glioblastoma tumors.


Assuntos
Dacarbazina/análogos & derivados , Implantes de Medicamento , Glioblastoma/tratamento farmacológico , Nanocompostos/química , Nanofibras/química , Linhagem Celular Tumoral , Dacarbazina/química , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Implantes de Medicamento/química , Implantes de Medicamento/farmacocinética , Implantes de Medicamento/farmacologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Temozolomida
17.
Biomater Sci ; 5(5): 1041-1050, 2017 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-28378865

RESUMO

Effective penetration through the blood-brain barrier (BBB) remains a challenge for the treatment of many brain diseases. In this study, a small molecule, sinapic acid (SA), extracted from mustard, was selected as a novel bioinspired BBB-permeable ligand for efficient drug delivery in glioma treatment. SA was conjugated on the surface of zwitterionic polymer poly(2-methacryloyloxyethyl phosphorylcholine) (PMPC)-encapsulated bovine serum albumin (BSA)-based nanoparticles, yielding nBSA-SA. The PMPC shell serves as a protective layer to prolong the in vivo blood circulation time with a better chance to cross the BBB. Furthermore, temozolomide (TMZ), which can be loaded onto the nanoparticles via electrostatic interactions with acrylic acid (AA) to generate AA-nBSA-SA-TMZ, was applied as an excellent chemotherapeutic drug for glioma therapy. The obtained nanoparticles with a distinct size show great BBB permeability. Through the mechanism study, it was found that the cell internalization of the SA-conjugated nanoparticles is an energy-dependent process with only transient disruption of the BBB. The biological evaluation results unambiguously suggest that drug-loaded nanoparticles can lead to strong apoptosis on the tumor site and increase the median survival time of glioma-bearing mice. Overall, this novel BBB-permeable ligand SA paves the way for the delivery of cargo into the brain and provides a powerful nanoplatform for glioma therapy via intravenous administration.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Barreira Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamento farmacológico , Ácidos Cumáricos/química , Dacarbazina/análogos & derivados , Glioma/tratamento farmacológico , Nanopartículas/química , Fosforilcolina/análogos & derivados , Ácidos Polimetacrílicos/química , Animais , Antineoplásicos Alquilantes/farmacocinética , Antineoplásicos Alquilantes/uso terapêutico , Transporte Biológico , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/metabolismo , Bovinos , Linhagem Celular Tumoral , Ácidos Cumáricos/metabolismo , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Dacarbazina/uso terapêutico , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Sistemas de Liberação de Medicamentos , Glioma/metabolismo , Humanos , Camundongos , Modelos Moleculares , Mostardeira/química , Nanopartículas/metabolismo , Fosforilcolina/química , Fosforilcolina/metabolismo , Ácidos Polimetacrílicos/metabolismo , Ratos , Soroalbumina Bovina/química , Soroalbumina Bovina/metabolismo , Temozolomida , Distribuição Tecidual
18.
BMC Cancer ; 17(1): 173, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28264648

RESUMO

BACKGROUND: The RAS/RAF/MEK/ERK pathway is constitutively activated in many cancers. Selumetinib (AZD6244, ARRY-142886) is an oral, potent and highly selective, allosteric MEK1/2 inhibitor with a short half-life that has shown clinical activity as monotherapy in phase I and II studies of advanced cancer. Preclinical data suggest that selumetinib may enhance the activity of chemotherapeutic agents. We assessed the safety, tolerability, and pharmacokinetics (PK) of selumetinib (AZD6244, ARRY-142886) in combination with docetaxel or dacarbazine in patients with advanced solid tumors. METHODS: This study was a phase I, open-label, multicenter study in patients aged ≥18 years with advanced solid tumors who were candidates for docetaxel or dacarbazine treatment. Part A of the study (dose escalation) evaluated safety, tolerability, PK, and maximum tolerated dose (MTD) of selumetinib twice daily (BID) with docetaxel 75 mg/m2 or dacarbazine 1000 mg/m2 administered every 21 days. Patients receiving docetaxel could be administered primary prophylactic granulocyte-colony stimulating factor according to standard guidelines. Part B of the study (dose expansion) further evaluated safety, tolerability, and PK in 12 additional patients at the MTD combinations determined in part A. RESULTS: A total of 35 patients received selumetinib plus docetaxel, and 25 received selumetinib plus dacarbazine. The MTD of selumetinib was 75 mg BID in combination with either docetaxel (two dose-limiting toxicity [DLT] events: neutropenia with fever, and thrombocytopenia) or dacarbazine (one DLT event: thrombocytopenia). Common adverse events occurring with each treatment combination were diarrhea, peripheral/periorbital edema, fatigue, and nausea. PK parameters for selumetinib and docetaxel or dacarbazine were similar when administered alone or in combination. Partial responses were reported in 6/35 patients receiving selumetinib plus docetaxel and 4/25 patients receiving selumetinib plus dacarbazine. CONCLUSIONS: The combinations of selumetinib plus docetaxel and selumetinib plus dacarbazine demonstrated manageable safety and tolerability profiles and preliminary signs of clinical activity in patients with advanced solid tumors. TRIAL REGISTRATION: ClinicalTrials.gov NCT00600496; registered 8 July 2009.


Assuntos
Benzimidazóis/efeitos adversos , Dacarbazina/efeitos adversos , Neoplasias/tratamento farmacológico , Taxoides/efeitos adversos , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Benzimidazóis/farmacocinética , Benzimidazóis/uso terapêutico , Dacarbazina/farmacocinética , Dacarbazina/uso terapêutico , Docetaxel , Feminino , Humanos , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Segurança do Paciente , Taxoides/farmacocinética , Taxoides/uso terapêutico
19.
J Neurooncol ; 132(3): 401-407, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28290002

RESUMO

The blood-brain barrier (BBB) limits entry of most chemotherapeutic agents into the CNS, resulting in inadequate exposure within CNS tumor tissue. Intranasal administration is a proposed means of delivery that can bypass the BBB, potentially resulting in more effective chemotherapeutic exposure at the tumor site. The objective of this study was to evaluate the feasibility and pharmacokinetics (plasma and CSF) of intranasal delivery using select chemotherapeutic agents in a non-human primate (NHP) model. Three chemotherapeutic agents with known differences in CNS penetration were selected for intranasal administration in a NHP model to determine proof of principle of CNS delivery, assess tolerability and feasibility, and to evaluate whether certain drug characteristics were associated with increased CNS exposure. Intravenous (IV) temozolomide (TMZ), oral (PO) valproic acid, and PO perifosine were administered to adult male rhesus macaques. The animals received a single dose of each agent systemically and intranasally in separate experiments, with each animal acting as his own control. The dose of the agents administered systemically was the human equivalent of a clinically appropriate dose, while the intranasal dose was the maximum achievable dose based on the volume limitation of 1 mL. Multiple serial paired plasma and CSF samples were collected and quantified using a validated uHPLC/tandem mass spectrometry assay after each drug administration. Pharmacokinetic parameters were estimated using non-compartmental analysis. CSF penetration was calculated from the ratio of areas under the concentration-time curves for CSF and plasma (AUCCSF:plasma). Intranasal administration was feasible and tolerable for all agents with no significant toxicities observed. For TMZ, the degrees of CSF drug penetration after intranasal and IV administration were 36 (32-57) and 22 (20-41)%, respectively. Although maximum TMZ drug concentration in the CSF (Cmax) was lower after intranasal delivery compared to IV administration due to the lower dose administered, clinically significant exposure was achieved in the CSF after intranasal administration with the lower doses. This was associated with lower systemic exposure, suggesting increased efficiency and potentially lower toxicities of TMZ after intranasal delivery. For valproic acid and perifosine, CSF penetration after intranasal delivery was similar to systemic administration. Although this study demonstrates feasibility and safety of intranasal drug administration, further agent-specific studies are necessary to optimize agent selection and dosing to achieve clinically-relevant CSF exposures.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Administração Intranasal , Animais , Antineoplásicos/metabolismo , Barreira Hematoencefálica , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Dacarbazina/metabolismo , Dacarbazina/farmacocinética , Modelos Animais de Doenças , Macaca mulatta , Masculino , Absorção Nasal , Fosforilcolina/administração & dosagem , Fosforilcolina/análogos & derivados , Fosforilcolina/metabolismo , Fosforilcolina/farmacocinética , Temozolomida , Ácido Valproico/administração & dosagem , Ácido Valproico/metabolismo , Ácido Valproico/farmacocinética
20.
Sci Rep ; 7: 43271, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28262735

RESUMO

Localized and controlled delivery of chemotherapeutics directly in brain-tumor for prolonged periods may radically improve the prognosis of recurrent glioblastoma. Here, we report a unique method of nanofiber by fiber controlled delivery of anti-cancer drug, Temozolomide, in orthotopic brain-tumor for one month using flexible polymeric nano-implant. A library of drug loaded (20 wt%) electrospun nanofiber of PLGA-PLA-PCL blends with distinct in vivo brain-release kinetics (hours to months) were numerically selected and a single nano-implant was formed by co-electrospinning of nano-fiber such that different set of fibres releases the drug for a specific periods from days to months by fiber-by-fiber switching. Orthotopic rat glioma implanted wafers showed constant drug release (116.6 µg/day) with negligible leakage into the peripheral blood (<100 ng) rendering ~1000 fold differential drug dosage in tumor versus peripheral blood. Most importantly, implant with one month release profile resulted in long-term (>4 month) survival of 85.7% animals whereas 07 day releasing implant showed tumor recurrence in 54.6% animals, rendering a median survival of only 74 days. In effect, we show that highly controlled drug delivery is possible for prolonged periods in orthotopic brain-tumor using combinatorial nanofibre libraries of bulk-eroding polymers, thereby controlling glioma recurrence.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Preparações de Ação Retardada/administração & dosagem , Glioma/tratamento farmacológico , Recidiva Local de Neoplasia/tratamento farmacológico , Nanomedicina Teranóstica/métodos , Animais , Antineoplásicos/farmacocinética , Neoplasias Encefálicas/diagnóstico , Linhagem Celular , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Modelos Animais de Doenças , Portadores de Fármacos/administração & dosagem , Glioma/diagnóstico , Humanos , Nanofibras/administração & dosagem , Recidiva Local de Neoplasia/diagnóstico , Ratos Wistar , Análise de Sobrevida , Temozolomida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...