Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Respir Res ; 23(1): 272, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36183104

RESUMEN

BACKGROUND: Oxidative stress associated with severe cardiopulmonary diseases leads to impairment in the nitric oxide/soluble guanylate cyclase signaling pathway, shifting native soluble guanylate cyclase toward heme-free apo-soluble guanylate cyclase. Here we describe a new inhaled soluble guanylate cyclase activator to target apo-soluble guanylate cyclase and outline its therapeutic potential. METHODS: We aimed to generate a novel soluble guanylate cyclase activator, specifically designed for local inhaled application in the lung. We report the discovery and in vitro and in vivo characterization of the soluble guanylate cyclase activator mosliciguat (BAY 1237592). RESULTS: Mosliciguat specifically activates apo-soluble guanylate cyclase leading to improved cardiopulmonary circulation. Lung-selective effects, e.g., reduced pulmonary artery pressure without reduced systemic artery pressure, were seen after inhaled but not after intravenous administration in a thromboxane-induced pulmonary hypertension minipig model. These effects were observed over a broad dose range with a long duration of action and were further enhanced under experimental oxidative stress conditions. In a unilateral broncho-occlusion minipig model, inhaled mosliciguat decreased pulmonary arterial pressure without ventilation/perfusion mismatch. With respect to airway resistance, mosliciguat showed additional beneficial bronchodilatory effects in an acetylcholine-induced rat model. CONCLUSION: Inhaled mosliciguat may overcome treatment limitations in patients with pulmonary hypertension by improving pulmonary circulation and airway resistance without systemic exposure or ventilation/perfusion mismatch. Mosliciguat has the potential to become a new therapeutic paradigm, exhibiting a unique mode of action and route of application, and is currently under clinical development in phase Ib for pulmonary hypertension.


Asunto(s)
Hipertensión Pulmonar , Acetilcolina , Animales , Guanilato Ciclasa/metabolismo , Guanilato Ciclasa/uso terapéutico , Óxido Nítrico/metabolismo , Ratas , Guanilil Ciclasa Soluble/metabolismo , Guanilil Ciclasa Soluble/uso terapéutico , Porcinos , Porcinos Enanos/metabolismo , Tromboxanos/uso terapéutico , Vasodilatadores
2.
Clin Cancer Res ; 25(15): 4723-4734, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31064781

RESUMEN

PURPOSE: Targeted thorium-227 conjugates (TTC) represent a new class of molecules for targeted alpha therapy (TAT). Covalent attachment of a 3,2-HOPO chelator to an antibody enables specific complexation and delivery of the alpha particle emitter thorium-227 to tumor cells. Because of the high energy and short penetration range, TAT efficiently induces double-strand DNA breaks (DSB) preferentially in the tumor cell with limited damage to the surrounding tissue. We present herein the preclinical evaluation of a mesothelin (MSLN)-targeted thorium-227 conjugate, BAY 2287411. MSLN is a GPI-anchored membrane glycoprotein overexpressed in mesothelioma, ovarian, pancreatic, lung, and breast cancers with limited expression in healthy tissue. EXPERIMENTAL DESIGN: The binding activity and radiostability of BAY 2287411 were confirmed bioanalytically. The mode-of-action and antitumor potency of BAY 2287411 were investigated in vitro and in vivo in cell line and patient-derived xenograft models of breast, colorectal, lung, ovarian, and pancreatic cancer. RESULTS: BAY 2287411 induced DSBs, apoptotic markers, and oxidative stress, leading to reduced cellular viability. Furthermore, upregulation of immunogenic cell death markers was observed. BAY 2287411 was well-tolerated and demonstrated significant antitumor efficacy when administered via single or multiple dosing regimens in vivo. In addition, significant survival benefit was observed in a disseminated lung cancer model. Biodistribution studies showed specific uptake and retention of BAY 2287411 in tumors and enabled the development of a mechanistic pharmacokinetic/pharmacodynamic model to describe the preclinical data. CONCLUSIONS: These promising preclinical results supported the transition of BAY 2287411 into a clinical phase I program in mesothelioma and ovarian cancer patients (NCT03507452).


Asunto(s)
Partículas alfa/uso terapéutico , Evaluación Preclínica de Medicamentos/métodos , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/farmacología , Neoplasias/tratamiento farmacológico , Radiofármacos/farmacología , Torio/farmacología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/química , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia Celular , Femenino , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/farmacocinética , Humanos , Inmunoconjugados/administración & dosificación , Inmunoconjugados/química , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mesotelina , Mesotelioma/tratamiento farmacológico , Mesotelioma/metabolismo , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Ratones Desnudos , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Radiofármacos/farmacocinética , Torio/administración & dosificación , Torio/química , Torio/farmacocinética , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
3.
AAPS J ; 19(4): 1186-1195, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28516358

RESUMEN

BAY 1093884 is a fully human monoclonal antibody against the tissue factor pathway inhibitor (TFPI) in development as prophylaxis in patients with hemophilia with or without inhibitors. In vitro, BAY 1093884 binds to human, mouse, and monkey TFPI. The objective of this study was to find a pharmacodynamic (PD) biomarker after administration of BAY 1093884 to normal monkeys. In monkey plasma, BAY 1093884 exhibited an IC50 (concentration that inhibits 50%) of 4.65 and 6.19 nM for free TFPI and diluted prothrombin time (dPT), respectively. The BAY 1093884 pharmacokinetic (PK) profile and its PD effects on dPT and free TFPI levels were assessed after intravenous and subcutaneous administration of BAY 1093884 (5 and 20 mg/kg) to female cynomolgus monkeys. Free TFPI concentrations in plasma decreased rapidly and increased to baseline in a dose-dependent manner. dPT clotting time was shortened and correlated with free TFPI levels and drug concentration in plasma, demonstrating the relationship between PD activities (dPT clotting time and free TFPI levels) and drug concentration. BAY 1093884 exhibited nonlinear PK, and a target-mediated drug disposition model was used to characterize the BAY 1093884 versus TFPI concentration-response relationship. We concluded that a mechanism-based PK/PD binding model could be useful for predicting human response to BAY 1093884. For the first-in-human study, measurement of free TFPI will be included as part of the dose-escalation design.


Asunto(s)
Anticuerpos Neutralizantes/farmacología , Lipoproteínas/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Macaca fascicularis
4.
Mol Cancer Ther ; 16(5): 893-904, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28292941

RESUMEN

C4.4A (LYPD3) has been identified as a cancer- and metastasis-associated internalizing cell surface protein that is expressed in non-small cell lung cancer (NSCLC), with particularly high prevalence in the squamous cell carcinoma (SCC) subtype. With the exception of skin keratinocytes and esophageal endothelial cells, C4.4A expression is scarce in normal tissues, presenting an opportunity to selectively treat cancers with a C4.4A-directed antibody-drug conjugate (ADC). We have generated BAY 1129980 (C4.4A-ADC), an ADC consisting of a fully human C4.4A-targeting mAb conjugated to a novel, highly potent derivative of the microtubule-disrupting cytotoxic drug auristatin via a noncleavable alkyl hydrazide linker. In vitro, C4.4A-ADC demonstrated potent antiproliferative efficacy in cell lines endogenously expressing C4.4A and inhibited proliferation of C4.4A-transfected A549 lung cancer cells showing selectivity compared with a nontargeted control ADC. In vivo, C4.4A-ADC was efficacious in human NSCLC cell line (NCI-H292 and NCI-H322) and patient-derived xenograft (PDX) models (Lu7064, Lu7126, Lu7433, and Lu7466). C4.4A expression level correlated with in vivo efficacy, the most responsive being the models with C4.4A expression in over 50% of the cells. In the NCI-H292 NSCLC model, C4.4A-ADC demonstrated equal or superior efficacy compared to cisplatin, paclitaxel, and vinorelbine. Furthermore, an additive antitumor efficacy in combination with cisplatin was observed. Finally, a repeated dosing with C4.4A-ADC was well tolerated without changing the sensitivity to the treatment. Taken together, C4.4A-ADC is a promising therapeutic candidate for the treatment of NSCLC and other cancers expressing C4.4A. A phase I study (NCT02134197) with the C4.4A-ADC BAY 1129980 is currently ongoing. Mol Cancer Ther; 16(5); 893-904. ©2017 AACR.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Moléculas de Adhesión Celular/inmunología , Inmunoconjugados/administración & dosificación , Aminobenzoatos/química , Aminobenzoatos/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Moléculas de Adhesión Celular/antagonistas & inhibidores , Línea Celular Tumoral , Cisplatino/administración & dosificación , Cisplatino/inmunología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunoconjugados/química , Inmunoconjugados/inmunología , Ratones , Oligopéptidos/química , Oligopéptidos/inmunología , Paclitaxel/administración & dosificación , Paclitaxel/inmunología , Vinblastina/administración & dosificación , Vinblastina/análogos & derivados , Vinblastina/inmunología , Vinorelbina , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 76(21): 6331-6339, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-27543601

RESUMEN

The fibroblast growth factor receptor FGFR2 is overexpressed in a variety of solid tumors, including breast, gastric, and ovarian tumors, where it offers a potential therapeutic target. In this study, we present evidence of the preclinical efficacy of BAY 1187982, a novel antibody-drug conjugate (ADC). It consists of a fully human FGFR2 monoclonal antibody (mAb BAY 1179470), which binds to the FGFR2 isoforms FGFR2-IIIb and FGFR2-IIIc, conjugated through a noncleavable linker to a novel derivative of the microtubule-disrupting cytotoxic drug auristatin (FGFR2-ADC). In FGFR2-expressing cancer cell lines, this FGFR2-ADC exhibited potency in the low nanomolar to subnanomolar range and was more than 100-fold selective against FGFR2-negative cell lines. High expression levels of FGFR2 in cells correlated with efficient internalization, efficacy, and cytotoxic effects in vitro Pharmacokinetic analyses in mice bearing FGFR2-positive NCI-H716 tumors indicated that the toxophore metabolite of FGFR2-ADC was enriched more than 30-fold in tumors compared with healthy tissues. Efficacy studies demonstrated that FGFR2-ADC treatment leads to a significant tumor growth inhibition or tumor regression of cell line-based or patient-derived xenograft models of human gastric or breast cancer. Furthermore, FGFR2 amplification or mRNA overexpression predicted high efficacy in both of these types of in vivo model systems. Taken together, our results strongly support the clinical evaluation of BAY 1187982 in cancer patients and a phase I study (NCT02368951) has been initiated. Cancer Res; 76(21); 6331-9. ©2016 AACR.


Asunto(s)
Aminobenzoatos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Inmunoconjugados/uso terapéutico , Neoplasias/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/análisis , Animales , Anticuerpos Monoclonales Humanizados , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cancer Ther ; 11(12): 2664-73, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23041545

RESUMEN

For treatment of patients with prostate cancer (PCa), we developed a novel T cell-engaging (BiTE) antibody designated AMG 212 or BAY2010112 that is bispecific for prostate-specific membrane antigen (PSMA) and the CD3 epsilon subunit of the T cell receptor complex. AMG 212/BAY2010112 induced target cell-dependent activation and cytokine release of T cells, and efficiently redirected T cells for lysis of target cells. In addition to Chinese hamster ovary cells stably expressing human or cynomolgus monkey PSMA, T cells redirected by AMG 212/BAY2010112 also lysed human PCa cell lines VCaP, 22Rv1, MDA PCa 2b, C4-2, PC-3-huPSMA, and LnCaP at half maximal BiTE concentrations between 0.1 and 4 ng/mL (1.8-72 pmol/L). No lysis of PSMA-negative human PCa cell lines PC-3 and DU145 was observed. The subcutaneous (s.c.) formation of tumors from PC-3-huPSMA cells in NOD/SCID mice was significantly prevented by once daily intravenous (i.v.) injection of AMG 212/BAY2010112 at a dose level as low as 0.005 mg/kg/d. Rapid tumor shrinkage with complete remissions were observed in NOD/SCID mice bearing established s.c. 22Rv1 xenografts after repeated daily treatment with AMG 212/BAY2010112 by either the i.v. or s.c. route. Of note, 22Rv1 tumors were grown in the absence of human T cells followed by intraperitoneal injection of T cells 3 days before BiTE treatment. No effects on tumor growth were observed in the absence of human T cells or AMG 212/BAY2010112. On the basis of these preclinical results, AMG 212/BAY2010112 appears as a promising new BiTE antibody for the treatment of patients with PSMA-expressing PCa.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Antígenos de Superficie/inmunología , Complejo CD3/inmunología , Glutamato Carboxipeptidasa II/inmunología , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Animales , Anticuerpos Biespecíficos/genética , Anticuerpos Biespecíficos/inmunología , Complejo CD3/genética , Células CHO , Cricetinae , Reacciones Cruzadas , Femenino , Haplorrinos , Humanos , Inmunización Pasiva/métodos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Neoplasias de la Próstata/patología , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biopharm Drug Dispos ; 26(6): 259-68, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15966026

RESUMEN

Repinotan hydrochloride (repinotan) is a highly potent and selective 5-HT(1A) full receptor agonist. The ability of repinotan to cross the blood-brain barrier (BBB) and penetrate into rat brain tissue was investigated, because rapid penetration into brain tissue is thought to be essential for neuroprotective efficacy. Intravenous (i.v.) repinotan was rapidly distributed into brain, and the distribution equilibrium between blood and brain was reached immediately after the start of infusion. Free concentrations of repinotan were identical in brain and plasma, indicating that repinotan crosses the BBB freely in both directions with diffusion as a driving force. The brain concentration of repinotan was determined by the free plasma concentration. Thus, the total plasma concentration of repinotan (sum of bound and unbound compound) is only indicative for the brain concentration as long as the unbound fraction remains constant. Metabolites of repinotan do not penetrate the BBB and are retained in the perfusing blood due to their increased polarity. The penetration of [14C] repinotan into ischemic areas of the brain was dependent on time. In studies using injured animals (pMCAO), high levels of [14C] repinotan could be detected in ischemic areas when the compound was administered up to 5 h post injury. [14C] repinotan radioactivity could no longer be detected in ischemic areas when administered 18 h after pMCA-O. After the end of infusion, repinotan was rapidly and completely eliminated from rat brains. Elimination occurred in parallel from plasma and brain with half-lives of about 1 h. In conclusion, repinotan rapidly and to a considerable extent penetrates into brain tissue of healthy and injured animals.


Asunto(s)
Benzopiranos/farmacocinética , Lesiones Encefálicas/metabolismo , Encéfalo/metabolismo , Agonistas de Receptores de Serotonina/farmacocinética , Tiazoles/farmacocinética , Animales , Barrera Hematoencefálica , Masculino , Ratas , Ratas Wistar
8.
Bioorg Med Chem Lett ; 15(2): 401-4, 2005 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-15603962

RESUMEN

Novel cyclohexadienes have been identified as potent and specific IK(Ca)-channel blockers. In this communication we describe their synthesis as well as their chemical and biological properties. A selected derivative is being enriched in rat brain and reduces the infarct volume, intracranial pressure as well as the water content in a rat subdural hematoma model of traumatic brain injury after iv administration.


Asunto(s)
Agua Corporal/efectos de los fármacos , Ciclohexanos/farmacología , Presión Intracraneal/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/metabolismo , Animales , Agua Corporal/metabolismo , Infarto Encefálico/tratamiento farmacológico , Lesiones Encefálicas/tratamiento farmacológico , Ciclohexanos/síntesis química , Ciclohexenos , Modelos Animales de Enfermedad , Hematoma Subdural/tratamiento farmacológico , Canales de Potasio de Conductancia Intermedia Activados por el Calcio , Bloqueadores de los Canales de Potasio/síntesis química , Ratas , Relación Estructura-Actividad
9.
Eur J Neurosci ; 20(7): 1761-8, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15379997

RESUMEN

Early deterioration and death after brain injury is often the result of oedema in the injured and peri-lesional tissue. So far, no pharmacotherapy is available that exhibits significant brain oedema-reducing efficacy in patients. We selected two low molecular weight compounds from different chemical classes, a triazole (1-[(2-chlorophenyl)diphenylmethyl]-1,2,3-triazole) and a cyclohexadiene (methyl 4-[4-chloro-3-(trifluoromethyl)phenyl]-6-methyl-3-oxo-1,4,7-tetrahydroisobenzofuran-5-carboxylate) to characterize their pharmacological properties on KCNN4 channels (intermediate/small conductance calcium-activated potassium channel, subfamily N, member 4) in vitro as well as in vivo. In vitro we replaced potassium by rubidium (Rb+) and determined Rb+ fluxes evoked by 10 micro m of the calcium ionophore A23187 on C6BU1 rat glioma cells. Compared with known KCNN4 blockers, such as clotrimazole (IC50=360 +/- 12 nm) and charybdotoxin (IC50=3.3 +/- 1.9 nm), the triazole and cyclohexadiene were considerably more potent than clotrimazole and displayed similar potencies (IC50=12.1 +/- 8.8 and 13.3 +/- 4.7 nm, respectively). In the rat acute subdural haematoma model, both the triazole and cyclohexadiene displayed reduction of brain water content (-26% at 0.3 mg/kg and -24% at 0.01 mg/kg) and reduction of the intracranial pressure (-46% at 0.1 mg/kg and -60% at 0.003 mg/kg) after 24 h when administered as a 4-h infusion immediately after brain injury. When infarct volumes were determined after 7 days, the triazole as well as the cyclohexadiene displayed strong neuroprotective efficacy (-52% infarct volume reduction at 1.2 mg/kg and -43% at 0.04 mg/kg, respectively). It is concluded that blockade of KCNN4 channels is a new pharmacological approach to attenuate acute brain damage caused by traumatic brain injury.


Asunto(s)
Edema Encefálico/terapia , Lesiones Encefálicas/terapia , Clotrimazol/uso terapéutico , Hematoma Subdural/terapia , Bloqueadores de los Canales de Potasio/uso terapéutico , Animales , Química Encefálica , Calcimicina/farmacología , Línea Celular Tumoral , Infarto Cerebral/patología , Caribdotoxina/uso terapéutico , Cartilla de ADN , Eritrocitos/fisiología , Glioma/genética , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio , Canales de Potasio Calcio-Activados , Ratas , Ratas Wistar , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rubidio/sangre , Agua/análisis
10.
J Pharm Sci ; 93(4): 816-30, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-14999720

RESUMEN

Quantification of protein binding of new chemical entities is an important early screening step during drug discovery and is of fundamental interest for the estimation of safety margins during drug development. In this publication, we describe the development of a new high-throughput assay for the determination of the free drug fraction in plasma (fu). The new technique is an enhancement of the previously published erythrocytes partition method. It is based on the distribution of drugs between plasma water, plasma proteins, and solid-supported lipid membranes (Transil). The execution of protein binding studies by partitioning is dramatically simplified by substituting erythrocytes with commercially available Transil beads, and makes the method particularly suitable for high-throughput studies. Eight Bayer compounds from different compound classes covering a wide range of lipophilicities (log P = 1.9-5.6) and fu values (0.018-35%) were selected for validation of the assay. The results obtained by the new method and by either the erythrocytes partitioning technique or more conventional methods (ultrafiltration and equilibrium dialysis) are identical, confirming that the new method produces valid results even for drugs that are strongly bound to plasma proteins. Precision and accuracy of the data in the cases of very low and high fu values are comparable, indicating that the method is especially suited for highly lipophilic drugs that tend to adsorb to surfaces compared with other methods, like ultrafiltration or equilibrium dialysis, that may produce biased data. The method is also useful for the determination of binding parameters and the pH dependence of fu. In summary, this assay is well suited for high-throughput determination of protein binding during drug discovery and for extended protein binding studies during drug development.


Asunto(s)
Proteínas Sanguíneas/análisis , Preparaciones Farmacéuticas/análisis , Animales , Tampones (Química) , Fenómenos Químicos , Química Física , Perros , Eritrocitos/química , Femenino , Humanos , Indicadores y Reactivos , Macaca mulatta , Masculino , Membranas Artificiales , Fosfatidilcolinas/química , Unión Proteica , Ratas , Ratas Wistar , Dióxido de Silicio/química
11.
Rapid Commun Mass Spectrom ; 17(17): 1950-7, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12913858

RESUMEN

Some cases of occurrence of matrix effects (mostly ion suppression) in protein-precipitated plasma samples, and their influence on the validity of plasma concentrations and pharmacokinetic parameters, are discussed. The comparison of matrix effects using either electrospray (TurboIonspray, TISP) or atmospheric pressure chemical ionization (APCI) indicated that APCI is less prone to matrix effects. Nevertheless, TISP is usually the first choice of ionization technique since unknown thermally labile metabolites might be present in the plasma samples causing erroneous results. A high impact of ion suppression on the plasma concentrations after intravenous (i.v.) administration was found, depending on the drug formulation (vehicle). Since ion suppression caused significantly lower plasma concentrations (by a factor of up to 5.5) after i.v. dosing, the area under the curve (AUC) was underestimated and the plasma clearance was consequently erroneously high, with an impact on drug candidate selection. By simple stepwise dilution (e.g. 10-fold and 50-fold) of the supernatant of protein-precipitated plasma samples, including all calibration and quality control samples, the matrix effects were recognized and eliminated.


Asunto(s)
Plasma/química , Animales , Cromatografía Líquida de Alta Presión , Etanol , Concentración de Iones de Hidrógeno , Indicadores y Reactivos , Inyecciones Intravenosas , Masculino , Espectrometría de Masas , Preparaciones Farmacéuticas/análisis , Vehículos Farmacéuticos , Polietilenglicoles , Ratas , Ratas Wistar , Solventes , Espectrometría de Masa por Ionización de Electrospray , Agua
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...