Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
J Med Chem ; 67(4): 2619-2630, 2024 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-38294341

RESUMO

Targeting microtubules is the most effective wide-spectrum pharmacological strategy in antitumoral chemotherapy, and current research focuses on reducing main drawbacks: neurotoxicity and resistance. PM534 is a novel synthetic compound derived from the Structure-Activity-Relationship study on the natural molecule PM742, isolated from the sponge of the order Lithistida, family Theonellidae, genus Discodermia (du Bocage 1869). PM534 targets the entire colchicine binding domain of tubulin, covering four of the five centers of the pharmacophore model. Its nanomolar affinity and high retention time modulate a strikingly high antitumor activity that efficiently overrides two resistance mechanisms in cells (detoxification pumps and tubulin ßIII isotype overexpression). Furthermore, PM534 induces significant inhibition of tumor growth in mouse xenograft models of human non-small cell lung cancer. Our results present PM534, a highly effective new compound in the preclinical evaluation that is currently in its first human Phase I clinical trial.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Animais , Camundongos , Colchicina/metabolismo , Tubulina (Proteína)/metabolismo , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Microtúbulos , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/uso terapêutico , Moduladores de Tubulina/química , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Antineoplásicos/química , Sítios de Ligação , Linhagem Celular Tumoral , Proliferação de Células
2.
Brain Res Bull ; 192: 12-20, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36328144

RESUMO

Chemotherapy-induced neuropathy (CIN) is one of the most common complications of cancer treatment with sensory dysfunctions which frequently include pain. The mechanisms underlying pain during CIN are starting to be uncovered. Neuroimaging allows the identification of brain circuitry involved in pain processing and modulation and has recently been used to unravel the disruptions of that circuitry by neuropathic pain. The present study evaluates the effects of paclitaxel, a cytostatic drug frequently used in cancer treatment, at the neuronal function in the anterior cingulate cortex (ACC), hypothalamus and periaqueductal gray (PAG) using manganese-enhanced magnetic resonance imaging (MEMRI). We also studied the metabolic profile at the prefrontal cortex (PFC) and hypothalamus using ex vivo spectroscopy. Wistar male rats were intraperitoneal injected with paclitaxel or vehicle solution (DMSO). The evaluation of mechanical sensitivity using von Frey test at baseline (BL), 21 (T21), 28 (T28), 49 (T49) and 56 days (T56) after CIN induction showed that paclitaxel-injected rats presented mechanical hypersensitivity from T21 until T56 after CIN induction. The evaluation of the locomotor activity and exploratory behaviors using open-field test at T28 and T56 after the first injection of paclitaxel revealed that paclitaxel-injected rats walked higher distance with higher velocity at late point of CIN accompanied with a sustained exhibition of anxiety-like behaviors. Imaging studies performed using MEMRI at T28 and T56 showed that paclitaxel treatment increased the neuronal activation in the hypothalamus and PAG at T56 in comparison with the control group. The analysis of data from ex vivo spectroscopy demonstrated that at T28 paclitaxel-injected rats presented an increase of N-acetyl aspartate (NAA) levels in the PFC and an increase of NAA and decrease of lactate (Lac) concentration in the hypothalamus compared to the control group. Furthermore, at T56 the paclitaxel-injected rats presented lower NAA and higher taurine (Tau) levels in the PFC. Together, MEMRI and metabolomic data indicate that CIN is associated with neuroplastic changes in brain areas involved in pain modulation and suggests that other events involving glial cells may be happening.


Assuntos
Antineoplásicos , Neuralgia , Animais , Ratos , Masculino , Ratos Wistar , Neuralgia/induzido quimicamente , Neuralgia/diagnóstico por imagem , Neuralgia/tratamento farmacológico , Imageamento por Ressonância Magnética/métodos , Encéfalo/metabolismo , Paclitaxel/toxicidade , Paclitaxel/uso terapêutico , Antineoplásicos/toxicidade , Análise Espectral
3.
Science ; 371(6532): 926-931, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33495306

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viral proteins interact with the eukaryotic translation machinery, and inhibitors of translation have potent antiviral effects. We found that the drug plitidepsin (aplidin), which has limited clinical approval, possesses antiviral activity (90% inhibitory concentration = 0.88 nM) that is more potent than remdesivir against SARS-CoV-2 in vitro by a factor of 27.5, with limited toxicity in cell culture. Through the use of a drug-resistant mutant, we show that the antiviral activity of plitidepsin against SARS-CoV-2 is mediated through inhibition of the known target eEF1A (eukaryotic translation elongation factor 1A). We demonstrate the in vivo efficacy of plitidepsin treatment in two mouse models of SARS-CoV-2 infection with a reduction of viral replication in the lungs by two orders of magnitude using prophylactic treatment. Our results indicate that plitidepsin is a promising therapeutic candidate for COVID-19.


Assuntos
Antivirais/farmacologia , Tratamento Farmacológico da COVID-19 , Depsipeptídeos/farmacologia , Fator 1 de Elongação de Peptídeos/antagonistas & inibidores , SARS-CoV-2/efeitos dos fármacos , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/farmacologia , Monofosfato de Adenosina/uso terapêutico , Alanina/análogos & derivados , Alanina/farmacologia , Alanina/uso terapêutico , Animais , Antivirais/uso terapêutico , COVID-19/prevenção & controle , COVID-19/virologia , Proteínas do Nucleocapsídeo de Coronavírus/biossíntese , Proteínas do Nucleocapsídeo de Coronavírus/genética , Depsipeptídeos/administração & dosagem , Depsipeptídeos/uso terapêutico , Avaliação Pré-Clínica de Medicamentos , Feminino , Células HEK293 , Humanos , Pulmão/virologia , Camundongos Endogâmicos C57BL , Mutação , Peptídeos Cíclicos , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , RNA Viral/biossíntese , RNA Viral/genética , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , Replicação Viral/efeitos dos fármacos
4.
Transl Oncol ; 13(11): 100832, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32711367

RESUMO

The majority of patients with gastrointestinal stromal tumors (GIST) eventually become resistant with time due to secondary mutations in the driver receptor tyrosine kinase. Novel treatments that do not target these receptors may therefore be preferable. For the first time, we evaluated a tubulin inhibitor, plocabulin, in patient-derived xenograft (PDX) models of GIST, a disease generally considered to be resistant to cytotoxic agents. Three PDX models of GIST with different KIT genotype were generated by implanting tumor fragments from patients directly into nude mice. We then used these well characterized models with distinct sensitivity to imatinib to evaluate the efficacy of the novel tubulin inhibitor. The efficacy of the drug was assessed by volumetric analysis of the tumors, histopathology, immunohistochemistry and Western blotting. Plocabulin treatment led to extensive necrosis in all three models and significant tumor shrinkage in two models. This histological response can be explained by the drug's vascular-disruptive properties, which resulted in a shutdown of tumor vasculature, reflected by a decreased total vascular area in the tumor tissue. Our results demonstrated the in vivo efficacy of the novel tubulin inhibitor plocabulin in PDX models of GIST and challenge the established view that GIST are resistant to cytotoxic agents in general and to tubulin inhibitors in particular. Our findings provide a convincing rationale for early clinical exploration of plocabulin in GIST and warrant further exploration of this class of drugs in the management of this common sarcoma subtype.

5.
J Hematol Oncol ; 13(1): 32, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32264921

RESUMO

BACKGROUND: In the search for novel antibody-drug conjugates (ADCs) with therapeutic potential, it is imperative to identify novel targets to direct the antibody moiety. CD13 seems an attractive ADC target as it shows a differential pattern of expression in a variety of tumors and cell lines and it is internalized upon engagement with a suitable monoclonal antibody. PM050489 is a marine cytotoxic compound tightly binding tubulin and impairing microtubule dynamics which is currently undergoing clinical trials for solid tumors. METHODS: Anti-CD13 monoclonal antibody (mAb) TEA1/8 has been used to prepare a novel ADC, MI130110, by conjugation to the marine compound PM050489. In vitro and in vivo experiments have been carried out to demonstrate the activity and specificity of MI130110. RESULTS: CD13 is readily internalized upon TEA1/8 mAb binding, and the conjugation with PM050489 did not have any effect on the binding or the internalization of the antibody. MI130110 showed remarkable activity and selectivity in vitro on CD13-expressing tumor cells causing the same effects than those described for PM050489, including cell cycle arrest at G2, mitosis with disarrayed and often multipolar spindles consistent with an arrest at metaphase, and induction of cell death. In contrast, none of these toxic effects were observed in CD13-null cell lines incubated with MI130110. Furthermore, in vivo studies showed that MI130110 exhibited excellent antitumor activity in a CD13-positive fibrosarcoma xenograft murine model, with total remissions in a significant number of the treated animals. Mitotic catastrophes, typical of the payload mechanism of action, were also observed in the tumor cells isolated from mice treated with MI130110. In contrast, MI130110 failed to show any activity in a xenograft mouse model of myeloma cells not expressing CD13, thereby corroborating the selectivity of the ADC to its target and its stability in circulation. CONCLUSION: Our results show that MI130110 ADC combines the antitumor potential of the PM050489 payload with the selectivity of the TEA1/8 monoclonal anti-CD13 antibody and confirm the correct intracellular processing of the ADC. These results demonstrate the suitability of CD13 as a novel ADC target and the effectiveness of MI130110 as a promising antitumor therapeutic agent.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Antígenos CD13/imunologia , Imunoconjugados/farmacologia , Neoplasias/tratamento farmacológico , Policetídeos/farmacologia , Pironas/farmacologia , Animais , Antineoplásicos Imunológicos/química , Antineoplásicos Imunológicos/uso terapêutico , Linhagem Celular Tumoral , Feminino , Humanos , Imunoconjugados/química , Imunoconjugados/uso terapêutico , Camundongos , Camundongos Nus , Neoplasias/imunologia , Policetídeos/química , Policetídeos/uso terapêutico , Pironas/química , Pironas/uso terapêutico
6.
Mater Sci Eng C Mater Biol Appl ; 107: 110262, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31761230

RESUMO

Magnetic resonance imaging (MRI) is the most powerful technique for non-invasive diagnosis of human diseases and disorders. Properly designed contrast agents can be accumulated in the damaged zone and be internalized by cells, becoming interesting cellular MRI probes for disease tracking and monitoring. However, this approach is sometimes limited by the relaxation rates of contrast agents currently in clinical use, which show neither optimal pharmacokinetic parameters nor toxicity. In this work, a suitable contrast agent candidate, based on iron oxide nanoparticles (IONPs) coated with polyethyleneglycol, was finely designed, prepared and fully characterized under a physical, chemical and biological point of view. To stand out the real potential of our study, all the experiments were performed in comparison with Ferumoxytol, a FDA approved IONPs. IONPs with a core size of 15 nm and coated with polyethyleneglycol of 5 kDa (OD15-P5) resulted the best ones, being able to be uptaken by both tumoral cells and macrophages and showing no toxicity for in vitro and in vivo experiments. In vitro and in vivo MRI results for OD15-P5 showed r2 relaxivity values higher than Ferumoxitol. Furthermore, the injected OD15-P5 were completely retained at the tumor site for up to 24 h showing high potential as MRI contrast agents for real time long-lasting monitoring of the tumor evolution.


Assuntos
Meios de Contraste/química , Compostos Férricos/química , Imageamento por Ressonância Magnética/métodos , Nanopartículas de Magnetita/química , Polietilenoglicóis/química , Animais , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Nanopartículas de Magnetita/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/diagnóstico por imagem , Tamanho da Partícula , Siloxanas/química
7.
Mol Cancer Ther ; 17(4): 786-794, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29440297

RESUMO

In the search for novel payloads to design new antibody-drug conjugates (ADC), marine compounds represent an interesting opportunity given their unique chemical features. PM050489 is a marine compound that binds ß-tubulin at a new site and disrupts the microtubule network, hence leading to mitotic aberrations and cell death. PM050489 has been conjugated to trastuzumab via Cys residues through a noncleavable linker, and the resulting ADC, named MI130004, has been studied. Analysis of MI130004 delivered data consistent with the presence of two molecules of PM050489 per antibody molecule, likely bound to both sides of the intermolecular disulfide bond connecting the antibody light and heavy chains. The antitumor activity of MI130004 was analyzed in vitro and in vivo in different cell lines of diverse tumor origin (breast, ovary, and gastric cancer) expressing different levels of HER2. MI130004 showed very high in vitro potency and good selectivity for tumor cells that overexpressed HER2. At the cellular level, MI130004 impaired tubulin polymerization, causing disorganization and disintegration of the microtubule network, which ultimately led to mitotic failure, mirroring the effect of its payload. Treatment with MI130004 in mice carrying histologically diverse tumors expressing HER2 induced a long-lasting antitumor effect with statistically significant inhibition of tumor growth coupled with increases in median survival time compared with vehicle or trastuzumab. These results strongly suggest that MI130004 is endowed with remarkable anticancer activity and confirm the extraordinary potential of marine compounds for the design of new ADCs. Mol Cancer Ther; 17(4); 786-94. ©2018 AACR.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Imunoconjugados/farmacologia , Neoplasias/tratamento farmacológico , Policetídeos/farmacologia , Pironas/farmacologia , Receptor ErbB-2/imunologia , Trastuzumab/farmacologia , Animais , Anticorpos Monoclonais Humanizados/química , Apoptose , Proliferação de Células , Feminino , Humanos , Imunoconjugados/química , Camundongos , Camundongos Nus , Camundongos SCID , Neoplasias/enzimologia , Neoplasias/patologia , Policetídeos/química , Pironas/química , Trastuzumab/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Nanomedicine ; 14(2): 257-267, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29127040

RESUMO

Glutathione degradable polyurethane-polyurea nanoparticles (PUUa NP) with a disulfide-rich multiwalled structure and a cyclic RGD peptide as a targeting moiety were synthesized, incorporating a very lipophilic chemotherapeutic drug named Plitidepsin. In vitro studies indicated that encapsulated drug maintained and even improved its cytotoxic activity while in vivo toxicity studies revealed that the maximum tolerated dose (MTD) of Plitidepsin could be increased three-fold after encapsulation. We also found that pharmacokinetic parameters such as maximum concentration (Cmax), area under the curve (AUC) and plasma half-life were significantly improved for Plitidepsin loaded in PUUa NP. Moreover, biodistribution assays in mice showed that RGD-decorated PUUa NP accumulate less in spleen and liver than non-targeted conjugates, suggesting that RGD-decorated nanoparticles avoid sequestration by macrophages from the reticuloendothelial system. Overall, our results indicate that polyurethane-polyurea nanoparticles represent a very valuable nanoplatform for the delivery of lipophilic drugs by improving their toxicological, pharmacokinetic and whole-body biodistribution profiles.


Assuntos
Antineoplásicos/farmacocinética , Depsipeptídeos/farmacocinética , Sistemas de Liberação de Medicamentos , Integrina alfaVbeta3/antagonistas & inibidores , Nanopartículas/administração & dosagem , Polímeros/química , Poliuretanos/química , Animais , Antineoplásicos/administração & dosagem , Depsipeptídeos/administração & dosagem , Portadores de Fármacos , Feminino , Camundongos , Nanopartículas/química , Peptídeos Cíclicos , Distribuição Tecidual
9.
Dis Model Mech ; 9(12): 1461-1471, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27780828

RESUMO

We explored whether the combination of lurbinectedin (PM01183) with the antimetabolite gemcitabine could result in a synergistic antitumor effect in pancreatic ductal adenocarcinoma (PDA) mouse models. We also studied the contribution of lurbinectedin to this synergism. This drug presents a dual pharmacological effect that contributes to its in vivo antitumor activity: (i) specific binding to DNA minor grooves, inhibiting active transcription and DNA repair; and (ii) specific depletion of tumor-associated macrophages (TAMs). We evaluated the in vivo antitumor activity of lurbinectedin and gemcitabine as single agents and in combination in SW-1990 and MIA PaCa-2 cell-line xenografts and in patient-derived PDA models (AVATAR). Lurbinectedin-gemcitabine combination induced a synergistic effect on both MIA PaCa-2 [combination index (CI)=0.66] and SW-1990 (CI=0.80) tumor xenografts. It also induced complete tumor remissions in four out of six patient-derived PDA xenografts. This synergism was associated with enhanced DNA damage (anti-γ-H2AX), cell cycle blockage, caspase-3 activation and apoptosis. In addition to the enhanced DNA damage, which is a consequence of the interaction of the two drugs with the DNA, lurbinectedin induced TAM depletion leading to cytidine deaminase (CDA) downregulation in PDA tumors. This effect could, in turn, induce an increase of gemcitabine-mediated DNA damage that was especially relevant in high-density TAM tumors. These results show that lurbinectedin can be used to develop 'molecularly targeted' combination strategies.


Assuntos
Adenocarcinoma/tratamento farmacológico , Carbolinas/uso terapêutico , Desoxicitidina/análogos & derivados , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Macrófagos/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Adenocarcinoma/patologia , Animais , Apoptose/efeitos dos fármacos , Carbolinas/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citidina Desaminase/metabolismo , Dano ao DNA , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Nus , Neoplasias Pancreáticas/patologia , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina , Neoplasias Pancreáticas
10.
J Pharm Biomed Anal ; 123: 37-41, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-26871278

RESUMO

Lurbinectedin (PM01183) is a new synthetic tetrahydroisoquinoline alkaloid that binds to selected sequences in the minor groove of DNA, inducing PM01183-DNA adducts that stall replication, DNA repair and transcription and gives rise to double-strand breaks and finally, caspase-dependent apoptotic cell death. PM01183 has demonstrated clinical antitumor activity in platinum resistant/refractory ovarian cancer patients. A rapid and sensitive liquid chromatography/tandem mass spectrometry assay was developed and validated to quantify PM01183 in plasma from nonclinical species. The bioanalysis consisted of a supported liquid extraction, followed by a gradient phase chromatography and, detection by positive ion electrospray tandem mass spectrometry. The calibration range for PM01183 was established using PM01183 standards from 0.1 to 100 ng/mL in blank plasma. The multiple reaction monitoring, based on the transition m/z 767.3→273.0, was specific for PM01183, and that based on the transition m/z 771.4→277.0 was specific for the internal standard (deuterated PM01183). No endogenous material interfered with the analysis of PM01183 and the internal standard from blank plasma. The limit of detection (LOD) of the assay was calculated as 0.025 ng/mL. The correlation coefficients for the calibration curves ranged from 0.9937 to 0.9987. The mean inter-day accuracies for all calibration standards ranged from 92 to 108% (≤8% bias), and the mean inter-day precision for calibration standards was always less than 12%. The mean intra and inter-day assay accuracy for all quality control replicates remained between 91 and 109%. The mean intra and inter-day assay precision was less than 10% for all QC levels. The method was validated to demonstrate the specificity, recovery, limit of quantification, accuracy and precision of measurements. The assay has been used to support preclinical pharmacokinetic and toxicokinetic studies of PM01183 in nonclinical species. The main PK parameters in dogs (3 male and 3 female, respectively) were calculated as follows: maximum concentration (Cmax, 12.9±0.6 and 10.2±3.0 ng/mL) and the area under the plasma concentration-time curve (AUC, 24.9±0.7 and 22.6±6.1 ng h/mL). The results showed that plasma samples could be monitored for PM01183 for long enough to accurately estimate pharmacokinetics information.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacocinética , Carbolinas/química , Carbolinas/farmacocinética , Compostos Heterocíclicos de 4 ou mais Anéis/química , Compostos Heterocíclicos de 4 ou mais Anéis/farmacocinética , Plasma/química , Espectrometria de Massas em Tandem/métodos , Animais , Área Sob a Curva , Calibragem , Cães , Feminino , Limite de Detecção , Macaca fascicularis , Masculino , Camundongos , Ratos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Suínos , Porco Miniatura , Tetra-Hidroisoquinolinas/química
11.
Enferm. glob ; 14(38): 220-234, abr. 2015. tab, graf
Artigo em Espanhol | IBECS | ID: ibc-135460

RESUMO

Introducción. Con el advenimiento de la informatización de la documentación médica, incluida la Prescripción Electrónica Asistida, la comunicación entre Médicos y Enfermeras es cada vez más electrónica y menos 'cara a cara' produciéndose efectos no deseados como cambios en los patrones de comunicación entre profesionales. Objetivos. Definir cuáll es el grado de comunicación verbal existente entre Médicos y personal de Enfermería en la unidad de Medicina Interna y establecer propuestas de mejora. Material y método: Revisión bibliográfica en bases de datos Pubmed, Medline, CINAHL, Scielo España y en base de datos de editorial Elseiver utilizando los descriptores DeCS o palabras clave (Relaciones Médico-Enfermero, Comunicación, Prescripción Electrónica, Pase de Guardia, Grupo de Atención al Paciente). Estudio observacional descriptivo, con una muestra de 19 Médicos y 19 Enfermeros, mediante cuestionario con 10 preguntas cerradas y varias opciones de respuesta. Resultados. Los resultados de la encuesta muestran que ambos colectivos consideran de gran importancia la necesidad de comunicación entre ambos tanto a nivel global como individual así como de trabajo en equipo. Conclusiones. Entre los aspectos más relevantes y que apoyan nuestro planteamiento inicial es que 78% (IC 95%; 85,6%-70,39%) de los encuestados piensa que tras la implantación del programa de PEA ha cambiado el patrón de comunicación entre ambos colectivos disminuyendo considerablemente la comunicación verbal directa (AU)


Introduction: With the advent of computerization of medical records, including electronic prescribing, communication between doctors and nurses is becoming more electronic and less 'face to face', thus producing unwished effects such as changes in the communication patterns. Objectives: Establishing the level of verbal communication between physicians and nurses in an internal medicine unit and suggesting improvement proposals. Material and method: Literature review in databases Pubmed, Medline, CINAHL, SciELO Spain and publishing database Elseiver using MeSH descriptors or keywords (Physician-Nurse Relations, Communication, Electronic Prescribing, Patient Handoff, Patient Care Team). Descriptive observational study with a sample of 19 physicians and 19 nurses, using a questionnaire with 10 closed questions with several answer options. Results: Both groups considered of great importance the need for communication, globally and individually, as well as teamwork. Conclusions: Among the most relevant aspects supporting our initial approach we have to point out that 78% (CI 95%; 85,6%-70,39%) of people being interviewed thought that after the implementation of computerized physician medication order entry the communication behaviour between both groups has changed significantly by reducing direct verbal communication (AU)


Assuntos
Humanos , Masculino , Feminino , Pneumologia/educação , Cardiologia/educação , Prescrição Eletrônica/classificação , Prescrição Eletrônica/enfermagem , Sistemas de Comunicação no Hospital/classificação , Sistemas de Comunicação no Hospital/ética , Bases de Dados Bibliográficas/classificação , Pneumologia , Cardiologia/métodos , Prescrição Eletrônica/economia , Prescrição Eletrônica/história , Sistemas de Comunicação no Hospital/organização & administração , Bases de Dados Bibliográficas/economia , Bases de Dados Bibliográficas , 34002
12.
Int J Pharm ; 483(1-2): 212-9, 2015 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-25681727

RESUMO

The focus of this study is to disclose a new delivery carrier intended to improve the pharmacokinetic characteristics of the anticancer drug plitidepsin and to favor its accumulation within the tumor. These nanocarriers named as nanocapsules, consist of an oily core surrounded by a highly PEGylated polyglutamic acid (PGA-PEG) shell loaded with plitidepsin. They showed a size of around 190 nm, a zeta potential of -24 mV and were able to encapsulate a high percentage (85%) of plitidepsin. In vivo studies, following intravenous injection in healthy mice, indicated that the encapsulation of the drug within PGA-PEG nanocapsules led to an important increase in its area under the curve (AUC) which is related to the important decrease of the clearance, as compared to the values observed for the drug dissolved in a Cremophor(®) EL solution. This improvement of the pharmacokinetic profile of the encapsulated plitidepsin was accompanied by a high increase (2.5-fold) of the maximum tolerated dose (MTD) in comparison to that of plitidepsin Cremophor(®) EL solution. The efficacy study performed in a xenograft tumor mice model evidenced the capacity of PGA-PEG nanocapsules to significantly reduce tumor growth. These promising results highlight the potential of PGA-PEG nanocapsules as an effective drug delivery system for cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Depsipeptídeos/farmacologia , Portadores de Fármacos/química , Nanocápsulas/química , Neoplasias Experimentais/tratamento farmacológico , Polietilenoglicóis/química , Ácido Poliglutâmico/química , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Depsipeptídeos/administração & dosagem , Depsipeptídeos/química , Portadores de Fármacos/administração & dosagem , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Injeções Intravenosas , Masculino , Dose Máxima Tolerável , Camundongos , Camundongos Nus , Nanocápsulas/administração & dosagem , Neoplasias Experimentais/patologia , Tamanho da Partícula , Peptídeos Cíclicos , Polietilenoglicóis/administração & dosagem , Ácido Poliglutâmico/administração & dosagem , Propriedades de Superfície , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Pharm Res ; 31(4): 983-91, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24287622

RESUMO

PURPOSE: Plitidepsin is an antineoplasic currently in clinical evaluation in a phase III trial in multiple myeloma (ADMYRE). Presently, the hydrophobic drug plitidepsin is formulated using Cremophor®, an adjuvant associated with unwanted hypersensitivity reactions. In search of alternatives, we developed and tested two nanoparticle-based formulations of plitidepsin, aiming to modify/improve drug biodistribution and efficacy. METHODS: Using nanoprecipitation, plitidepsin was loaded in polymer nanoparticles made of amphiphilic block copolymers (i.e. PEG-b-PBLG or PTMC-b-PGA). The pharmacokinetics, biodistribution and therapeutic efficacy was assessed using a xenograft renal cancer mouse model (MRI-H-121 xenograft) upon administration of the different plitidepsin formulations at maximum tolerated multiple doses (0.20 and 0.25 mg/kg for Cremophor® and copolymer formulations, respectively). RESULTS: High plitidepsin loading efficiencies were obtained for both copolymer formulations. Considering pharmacokinetics, PEG-b-PBLG formulation showed lower plasma clearance, associated with higher AUC and Cmax than Cremophor® or PTMC-b-PGA formulations. Additionally, the PEG-b-PBLG formulation presented lower liver and kidney accumulation compared with the other two formulations, associated with an equivalent tumor distribution. Regarding the anticancer activity, all formulations elicited similar efficacy profiles, as compared to the Cremophor® formulation, successfully reducing tumor growth rate. CONCLUSIONS: Although the nanoparticle formulations present equivalent anticancer activity, compared to the Cremophor® formulation, they show improved biodistribution profiles, presenting novel tools for future plitidepsin-based therapies.


Assuntos
Depsipeptídeos/farmacocinética , Portadores de Fármacos/farmacocinética , Neoplasias Renais/metabolismo , Nanopartículas/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Animais , Depsipeptídeos/administração & dosagem , Portadores de Fármacos/administração & dosagem , Feminino , Neoplasias Renais/tratamento farmacológico , Camundongos , Camundongos Nus , Nanopartículas/administração & dosagem , Peptídeos Cíclicos , Distribuição Tecidual/efeitos dos fármacos , Distribuição Tecidual/fisiologia , Resultado do Tratamento
14.
J Control Release ; 169(1-2): 10-6, 2013 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-23591661

RESUMO

A critical objective in cancer therapy is to reduce the systemic toxicity through the modification of the biodistribution of anticancer drugs. Herein, we disclose a new biodegradable nanocarrier, polyglutamic acid (PGA) nanocapsules, and present the in vivo pharmacokinetics/toxicity proof-of-concept for the anticancer drug plitidepsin. These novel nanocapsules were prepared using a modified solvent displacement technique where the polyamino acid was electrostatically deposited onto the lipid core. The nanocapsules exhibited an average size of 200 nm, a negative zeta potential and a great capacity for the encapsulation of plitidepsin (encapsulation efficiency above 90%). In addition, the nanocapsules could be freeze-dried and showed an adequate stability profile upon storage. Finally, the in vivo proof-of-concept studies performed in mice indicated that the encapsulation provided the drug with a prolonged blood circulation and a significantly reduced toxicity. In fact, the maximum tolerated dose of the nanoencapsulated drug was more than 3 times that of the reference formulation (Cremophor® EL plitidepsin solution). Overall, beyond the value of this specific formulation, the work reported here represents the evidence of the potential of polyamino acid nanocapsules in nano-oncological therapy.


Assuntos
Antineoplásicos/administração & dosagem , Depsipeptídeos/administração & dosagem , Nanocápsulas/química , Ácido Poliglutâmico/química , Animais , Antineoplásicos/farmacocinética , Depsipeptídeos/farmacocinética , Masculino , Camundongos , Modelos Moleculares , Nanocápsulas/toxicidade , Peptídeos Cíclicos , Ácido Poliglutâmico/toxicidade
15.
Clin Cancer Res ; 18(19): 5399-411, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22896654

RESUMO

PURPOSE: Epithelial ovarian cancer (EOC) is the fifth leading cause of death in women diagnosed with gynecologic malignancies. The low survival rate is because of its advanced-stage diagnosis and either intrinsic or acquired resistance to standard platinum-based chemotherapy. So, the development of effective innovative therapeutic strategies to overcome cisplatin resistance remains a high priority. EXPERIMENTAL DESIGN: To investigate new treatments in in vivo models reproducing EOCs tumor growth, we generated a preclinical model of ovarian cancer after orthotopic implantation of a primary serous tumor in nude mice. Further, matched model of acquired cisplatin-resistant tumor version was successfully derived in mice. Effectiveness of lurbinectedin (PM01183) treatment, a novel marine-derived DNA minor groove covalent binder, was assessed in both preclinical models as a single and a combined-cisplatin agent. RESULTS: Orthotopically perpetuated tumor grafts mimic the histopathological characteristics of primary patients' tumors and they also recapitulate in mice characteristic features of tumor response to cisplatin treatments. We showed that single lurbinectedin or cisplatin-combined therapies were effective in treating cisplatin-sensitive and cisplatin-resistant preclinical ovarian tumor models. Furthermore, the strongest in vivo synergistic effect was observed for combined treatments, especially in cisplatin-resistant tumors. Lurbinectedin tumor growth inhibition was associated with reduced proliferation, increased rate of aberrant mitosis, and subsequent induced apoptosis. CONCLUSIONS: Taken together, preclinical orthotopic ovarian tumor grafts are useful tools for drug development, providing hard evidence that lurbinectedin might be a useful therapy in the treatment of EOC by overcoming cisplatin resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Carbolinas/administração & dosagem , Sinergismo Farmacológico , Compostos Heterocíclicos de 4 ou mais Anéis/administração & dosagem , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Neoplasias Ovarianas/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Cisplatino/administração & dosagem , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Ovarianas/patologia , Transplante Heterólogo
16.
Rapid Commun Mass Spectrom ; 20(18): 2735-40, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16921565

RESUMO

A rapid and sensitive liquid chromatography/tandem mass spectrometry (LC/MS/MS) assay was developed and validated to quantify a novel antineoplastic agent, PM02734, in dog plasma. The method was validated to demonstrate the specificity, limit of quantification (LOQ), accuracy, and precision of measurements. The calibration range for PM02734 was established using PM02734 standards from 0.05 to 100 ng/mL in blank plasma. The dominating ions were doubly charged molecular ions [M+2H]2+ at m/z 740.0 instead of singly charged ones at m/z 1478.4. The selected reaction monitoring (SRM), based on the m/z 740.0 --> 212.2 transition, was specific for PM02734, and that based on the m/z 743.8 --> 212.2 transition was specific for deuterated PM02734 (the internal standard, IS); no endogenous materials interfered with the analysis of PM02734 and IS from blank plasma. The assay was linear over the concentration range 0.05-100 ng/mL. In terms of sensitivity of assay 0.05 ng/mL is a very low LLOQ, especially considering PM02734 is a peptide. The correlation coefficients for the calibration curves ranged from 0.9990 to 0.9999. The mean intraday and interday accuracies for all calibration standards (n = 9) ranged from 93 to 111% (< or =11% bias) in dog plasma, and the mean interday precision for all calibration standards was less than 6.4%. The mean intra- and interday assay accuracy for all quality control replicates in dog plasma (n = 9), determined at each QC level throughout the validated runs, ranged from 85-111% (< or =15% bias) and from 99-109% (< or =9% bias), respectively. The mean intra- and interday assay precision was less than 12.1 and 13.3% for all QC levels, respectively. The assay has been used to support preclinical pharmacokinetic (PK) and toxicokinetic studies. The results showed that preclinical samples could be monitored for PM02734 up to 168 h after dosing, which allowed us to identify multiple elimination phases and accurately estimate PK information.


Assuntos
Antineoplásicos/sangue , Cromatografia Líquida de Alta Pressão , Depsipeptídeos/sangue , Espectrometria de Massas por Ionização por Electrospray/métodos , Espectrometria de Massas em Tandem/métodos , Animais , Antineoplásicos/farmacocinética , Área Sob a Curva , Depsipeptídeos/farmacocinética , Cães , Feminino , Meia-Vida , Injeções Intravenosas , Masculino , Moluscos/química , Reprodutibilidade dos Testes
17.
Artigo em Inglês | MEDLINE | ID: mdl-16497567

RESUMO

PM01218 is a novel marine-derived alkaloid and has shown potent growth inhibitory activity against several human cancer cell lines. A rapid and sensitive high performance liquid chromatography/tandem mass spectrometry (HPLC-MS/MS) method was developed and validated to quantify PM01218 in mouse and rat plasma. The lower limit of quantitation (LLOQ) was 0.05 ng/mL. The calibration curve was linear from 0.05 to 100 ng/mL (R(2)>0.999). The assay was specifically based on the multiple reaction monitoring (MRM) transitions at m/z 278.4-->184.2, no endogenous material interfaced with the analysis of PM01218 and its internal standard from blank mouse and rat plasma. The mean intra- and inter-day assay accuracy remained below 15 and 8%, respectively, for all calibration standards and QC samples. The intra- and inter-day assay precision was less than 12.8 and 8.5% for all QC levels, respectively. The utility of the assay was demonstrated by pharmacokinetics studies of i.v. (bolus) PM01218 on SD rats.


Assuntos
Compostos Aza/sangue , Cromatografia Líquida de Alta Pressão/métodos , Pirimidinas/sangue , Espectrometria de Massas por Ionização por Electrospray/métodos , Animais , Compostos Aza/farmacocinética , Calibragem , Feminino , Masculino , Camundongos , Pirimidinas/farmacocinética , Ratos , Ratos Sprague-Dawley , Padrões de Referência , Sensibilidade e Especificidade
18.
Rapid Commun Mass Spectrom ; 19(5): 689-95, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15702485

RESUMO

A rapid and sensitive liquid chromatography/tandem mass spectrometry (LC/MS/MS) assay was developed and validated to quantify a novel antineoplastic agent, PM00104, in mouse, rat, dog, and human plasma. The method was validated to demonstrate the specificity, limit of quantification (LOQ), accuracy, and precision of measurements. The calibration range for PM00104 was established using PM00104 standards from 0.01-5.0 ng/mL in blank plasma. The selected reaction monitoring (SRM), based on the m/z 692.2 --> 218.2 transition, was specific for PM00104, and that based on the m/z 697.2 --> 218.2 transition was specific for PM00104 ((13)C(2),(2)H(3)) (the internal standard, IS); no endogenous materials interfered with the analysis of PM00104 and IS from blank plasma. The assay was linear over the concentration range 0.01-5.0 ng/mL. The correlation coefficients for the calibration curves ranged from 0.9981-0.9999. The mean intra-day and inter-day accuracies for all calibration standards (n = 8) ranged from 97-105% (< or =5% bias) in human plasma, and the mean inter-day precision for all calibration standards was less than 8.5%. The mean intra- and inter-day assay accuracy for all quality control (QC) replicates in human plasma (n = 9), determined at each QC level throughout the validated runs, ranged from 96-112% (< or =12% bias) and from 102-105% (< or =5% bias), respectively. The mean intra- and inter-day assay precision was less than 15.0 and 11.8% for all QC levels, respectively. For the QC samples prepared in animal species plasma, the %CV values of the assays ranged from 1.8-8.8% in mouse plasma, from 3.7-13.8% in rat plasma, and from 3.0-7.2% in dog plasma. The assay accuracies ranged from 92-102% (< or =8% bias) for all QC levels prepared in mouse plasma; ranged from 93-106% (< or =7% bias) in rat plasma; and ranged from 95-114% (< or =14% bias) in dog plasma. The assay has been used to support preclinical pharmacokinetic and toxicokinetic studies and is currently used to measure PM00104 plasma concentrations to support clinical trials.


Assuntos
Antineoplásicos/sangue , Análise Química do Sangue/métodos , Espectrometria de Massas por Ionização por Electrospray/métodos , Tetra-Hidroisoquinolinas/sangue , Animais , Cromatografia Líquida de Alta Pressão , Cães , Humanos , Camundongos , Ratos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Especificidade da Espécie
19.
Artigo em Inglês | MEDLINE | ID: mdl-12888201

RESUMO

A sensitive high-performance liquid chromatography-tandem mass spectrometry assay for thiocoraline, an anti-tumor depsipeptide, in mouse plasma is described. Echinomycin, a quinoxaline peptide, was used as an internal standard. Thiocoraline was recovered from the mouse plasma using protein precipitation with acetonitrile and followed by solid-phase extraction of the supernatant. The mobile phase consisted of methanol (0.1% formic acid)-water (0.1% formic acid) (90:10, v/v). The analytical column was a YMC C(18). The standard curve was linear from 0.1 to 50 ng/ml (R(2)>0.99). The lower limit of quantitation was 0.1 ng/ml. The assay was specific based on the multiple reaction monitoring transitions at m/z 1157-->215 and m/z 1101-->243 for thiocoraline and the internal standard, echinomycin, respectively. The mean intra- and inter-day assay accuracies remained below 5 and 12%, respectively, for all calibration standards and quality control (QC) samples. The intra- and inter-day assay precisions were less than 11.4 and 9.5% for all QC levels, respectively. The utility of the assay was demonstrated by a pharmacokinetic study of i.v. (bolus) thiocoraline on CD-1 mice. Thiocoraline was stable in mouse plasma in an ice-water bath for 6 h and for three freeze-thaw cycles. The reconstituted thiocoraline after extraction and drying sample process was stable in the autosampler for over 24 h. The assay was able to quantify thiocoraline in plasma up to 48 h following dose. Pharmacokinetic analysis showed that thiocoraline has distinct pharmacokinetic profiling when dosed in different formulation solutions. The assay is currently used to measure thiocoraline plasma concentrations in support of a project to develop a suitable formulation with a desirable pharmacokinetic profile.


Assuntos
Depsipeptídeos , Espectrometria de Massas/métodos , Peptídeos/sangue , Animais , Equinomicina/sangue , Feminino , Masculino , Camundongos , Peptídeos/farmacocinética , Controle de Qualidade , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...