Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Ther Drug Monit ; 45(4): 428-430, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37253456

RESUMO

ABSTRACT: The authors present a case of a 57-year-old patient with chronic myeloid leukemia who was treated with ponatinib and subsequently treated with dasatinib. The patient showed a major molecular response; however, the BCR-ABL1 signal increased with low ponatinib and dasatinib trough concentrations. Cobicistat was used as a pharmacokinetic booster to increase ponatinib and dasatinib exposure, as opposed to increasing the dose. However, ponatinib exposure was not sufficiently increased by cobicistat. The peak dasatinib concentration was successfully increased with cobicistat treatment. Dasatinib and cobicistat cotreatment induced a response in BCR-ABL1 PCR signal, was well tolerated, and led to a substantial reduction in drug costs.


Assuntos
Antineoplásicos , Piridazinas , Humanos , Pessoa de Meia-Idade , Dasatinibe/efeitos adversos , Cobicistat , Proteínas de Fusão bcr-abl/genética , Imidazóis/efeitos adversos , Piridazinas/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Antineoplásicos/efeitos adversos
2.
Ther Drug Monit ; 40(2): 159-161, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29470228

RESUMO

The authors present a case of a 69-year-old man with arrhythmogenic right ventricular cardiomyopathy controlled with amiodarone and an infected orthopedic prosthesis requiring treatment with rifampicin. This combination involves a pharmacokinetic drug-drug interaction leading to subtherapeutic drug concentrations of amiodarone and its active metabolite. The long half-life of amiodarone and its active metabolite in combination with the late onset and offset of cytochrome P4503A (CYP3A4) induction by rifampicin makes this a challenging drug-drug interaction to cope with in clinical practice. Before, during, and after rifampicin treatment, the serum concentrations of amiodarone and its active metabolite were measured and the amiodarone dose was adjusted accordingly. The amiodarone dose required to maintain effective concentrations was 450% of the initial dose. The drug-drug interaction between amiodarone and rifampicin is relevant, both clinically and pharmacokinetically, and can be managed by dose adjustments of amiodarone based on serum concentrations.


Assuntos
Amiodarona/farmacocinética , Amiodarona/uso terapêutico , Rifampina/efeitos adversos , Rifampina/uso terapêutico , Idoso , Cardiomiopatias/tratamento farmacológico , Citocromo P-450 CYP3A/metabolismo , Interações Medicamentosas , Monitoramento de Medicamentos/métodos , Humanos , Masculino , Próteses e Implantes/microbiologia , Disfunção Ventricular Direita/tratamento farmacológico
3.
Mol Imaging ; 13(5)2014.
Artigo em Inglês | MEDLINE | ID: mdl-24825298

RESUMO

Human epidermal growth factor receptor (HER)2 imaging with radiolabeled trastuzumab might support HER2-targeted therapy. It is, however, frequently questioned whether HER2 imaging is also possible during trastuzumab treatment as the receptor might be saturated. We studied the effect of trastuzumab treatment on 111In-trastuzumab uptake. Patients received trastuzumab weekly and paclitaxel once every 3 weeks. 111In-trastuzumab was injected on day 1 of cycle 1 and day 15 of cycle 4. Whole-body planar scintigraphy was acquired at different time points postinjection. Tumor uptake and organ distribution between the first and repeated scan series were calculated via residence times. Twenty-five tumor lesions in 12 patients were visualized on both scintigraphy series. Tumor uptake decreased (19.6%; p  =  .03). The residence times of normal organs remained similar except for the cardiac blood pool (+ 16.3%; p  =  .014). Trastuzumab treatment decreases tumor 111In-trastuzumab uptake around 20%. HER2 imaging is feasible during trastuzumab treatment.


Assuntos
Anticorpos Monoclonais Humanizados/administração & dosagem , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Radioisótopos de Índio , Anticorpos Monoclonais Humanizados/química , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica , Esquema de Medicação , Feminino , Humanos , Metástase Neoplásica/diagnóstico por imagem , Metástase Neoplásica/tratamento farmacológico , Paclitaxel/administração & dosagem , Cintilografia , Receptor ErbB-2/metabolismo , Trastuzumab
4.
Pharmaceutics ; 16(4)2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38675137

RESUMO

Vesicular hand eczema (VHE), a clinical subtype of hand eczema (HE), showed limited responsiveness to alitretinoin, the only approved systemic treatment for severe chronic HE. This emphasizes the need for alternative treatment approaches. Therefore, our study aimed to identify drug repurposing opportunities for VHE using transcriptomics and genomics data. We constructed a gene network by combining 52 differentially expressed genes (DEGs) from a VHE transcriptomics study with 3 quantitative trait locus (QTL) genes associated with HE. Through network analysis, clustering, and functional enrichment analyses, we investigated the underlying biological mechanisms of this network. Next, we leveraged drug-gene interactions and retrieved pharmaco-transcriptomics data from the DrugBank database to identify drug repurposing opportunities for (V)HE. We developed a drug ranking system, primarily based on efficacy, safety, and practical and pricing factors, to select the most promising drug repurposing candidates. Our results revealed that the (V)HE network comprised 78 genes that yielded several biological pathways underlying the disease. The drug-gene interaction search together with pharmaco-transcriptomics lookups revealed 123 unique drug repurposing opportunities. Based on our drug ranking system, our study identified the most promising drug repurposing opportunities (e.g., vitamin D analogues, retinoids, and immunomodulating drugs) that might be effective in treating (V)HE.

5.
Eur J Drug Metab Pharmacokinet ; 49(4): 517-526, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38878145

RESUMO

BACKGROUND AND OBJECTIVE: Several population pharmacokinetic (popPK) studies have been reported that can guide the prediction of osimertinib plasma concentrations in individual patients. It is currently unclear which popPK model offers the best predictive performance and which popPK models are most suitable for nonadherence management and model-informed precision dosing. Therefore, the objective of this study was to externally validate all osimertinib popPK models available in the current literature. METHODS: Published popPK models for osimertinib were constructed using NONMEM version 7.4.4. The predictive quality of the identified models was assessed with goodness-of-fit (GoF) plots, conditional weighted residuals (CWRES) plots and a prediction-corrected visual predictive check (pcVPC) for osimertinib and its active metabolite AZ5104. A subset from the Dutch OSIBOOST trial, where 11 patients with low osimertinib exposure were included, was used as evaluation cohort. RESULTS: The population GoF plots for all four models poorly followed the line of identity. For the individual GoF plots, all models performed comparable and were closely distributed among the line of identity. CWRES of the four models were skewed. The pcVPCs of all four models showed a similar trend, where all observed concentrations fell in the simulated shaded areas, but in the lower region of the simulated areas. CONCLUSION: All four popPK models can be used to individually predict osimertinib concentrations in patients with low osimertinib exposure. For population predictions, all four popPK models performed poorly in patients with low osimertinib exposure. A novel popPK model with good predictive performance should be developed for patients with low osimertinib exposure. Ideally, the cause for the relatively low osimertinib exposure in our evaluation cohort should be known. CLINICAL TRIALS REGISTRATION: NCT03858491.


Assuntos
Acrilamidas , Compostos de Anilina , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Modelos Biológicos , Humanos , Acrilamidas/farmacocinética , Neoplasias Pulmonares/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Compostos de Anilina/farmacocinética , Masculino , Pessoa de Meia-Idade , Feminino , Idoso , Países Baixos , Antineoplásicos/farmacocinética , Antineoplásicos/administração & dosagem , Estudos de Coortes , Adulto , Idoso de 80 Anos ou mais , Indóis , Pirimidinas
7.
J Geriatr Oncol ; 14(8): 101636, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37806290

RESUMO

INTRODUCTION: Older patients (≥65 years old) make up the majority of the cancer population. Older patients seem to experience more adverse events (AEs) from protein kinase inhibitors (PKIs) in clinical practice. Yet they are underrepresented in clinical trials. We aimed to evaluate whether age-related safety differences were described at authorization of PKIs. Representation of older patients in registration studies was also evaluated. MATERIALS AND METHODS: European Public Assessment Reports (EPARs) of PKIs authorized between 2010 and 2015 were evaluated for the description of age-related safety- and pharmacokinetic differences. The International Council for Harmonization of Technical Requirement for Pharmaceuticals for Human Use (ICH) E7 guideline was applied to EPARs to assess the representation of older patients. Study results were presented descriptively. RESULTS: Eighteen PKIs with 19 EPARs were analyzed. Age-related safety differences were described in 14 out of 19 EPARs, and age-related pharmacokinetic differences in 1 out of 19 EPARs. More than 100 older patients were included in half of the studies. Older patients were not excluded solely by age, although other inclusion and exclusion criteria negatively influenced enrollment of older patients. None of the PKIs met all criteria from the ICH E7 guideline. DISCUSSION: Age-related safety differences are described for most PKIs. Older patients were underrepresented in PKI registration studies. Adequate representation of older patients in clinical trials for PKIs is vital, since they make up most of the cancer population.


Assuntos
Neoplasias , Inibidores de Proteínas Quinases , Humanos , Idoso , Inibidores de Proteínas Quinases/efeitos adversos , Neoplasias/tratamento farmacológico
8.
Target Oncol ; 18(3): 441-450, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37081309

RESUMO

BACKGROUND: Expensive novel anticancer drugs put a serious strain on healthcare budgets, and the associated drug expenses limit access to life-saving treatments worldwide. OBJECTIVE: We aimed to develop alternative dosing regimens to reduce drug expenses. METHODS: We developed alternative dosing regimens for the following monoclonal antibodies used for the treatment of lung cancer: amivantamab, atezolizumab, bevacizumab, durvalumab, ipilimumab, nivolumab, pembrolizumab, and ramucirumab; and for the antibody-drug conjugate trastuzumab deruxtecan. The alternative dosing regimens were developed by means of modeling and simulation based on the population pharmacokinetic models developed by the license holders. They were based on weight bands and the administration of complete vials to limit drug wastage. The resulting dosing regimens were developed to comply with criteria used by regulatory authorities for in silico dose development. RESULTS: We found that alternative dosing regimens could result in cost savings that range from 11 to 28%, and lead to equivalent pharmacokinetic exposure with no relevant increases in variability in exposure. CONCLUSIONS: Dosing regimens based on weight bands and the use of complete vials to reduce drug wastage result in less expenses while maintaining equivalent exposure. The level of evidence of our proposal is the same as accepted by regulatory authorities for the approval of alternative dosing regimens of other monoclonal antibodies in oncology. The proposed alternative dosing regimens can, therefore, be directly implemented in clinical practice.


Assuntos
Antineoplásicos , Imunoconjugados , Neoplasias Pulmonares , Humanos , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Nivolumabe , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico
9.
Mol Pharm ; 9(11): 2995-3002, 2012 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-23003202

RESUMO

Human epidermal growth factor receptor-2 (HER2) directed therapy potentially can be improved by insight in drug effects on HER2 expression. This study evaluates the effects of the EGFR/HER2 tyrosine kinase inhibitor lapatinib, the heat shock protein-90 inhibitor 17AAG, and their combination, on HER2 expression with in vivo HER2-PET imaging. Lapatinib and 17AAG effects on EGFR and HER2 membrane expression were determined in vitro using flow cytometry of human SKBR3 tumor cells. Effect of lapatinib on HER2 internalization was studied in vitro by (89)Zr-trastuzumab-F(ab')(2) internalization. For in vivo evaluation, (89)Zr-trastuzumab-F(ab')(2) µPET imaging was performed two times with a 7 day interval. Lapatinib was administered for 6 days, starting 1 day after the baseline scan. 17AAG was given 1 day before the second (89)Zr-trastuzumab-F(ab')(2) injection. Imaging data were compared with ex vivo biodistribution analysis and HER2 immunohistochemical staining. 17AAG treatment lowered EGFR expression by 41% (P = 0.016) and HER2 by 76% (P = 0.022). EGFR/HER2 downregulation by 17AAG was inhibited by lapatinib pretreatment. Lapatinib reduced internalization of (89)Zr-trastuzumab-F(ab')(2) with 25% (P = 0.0022). (89)Zr-trastuzumab-F(ab')(2) tumor to blood ratio was lowered 32% by lapatinib (P = 0.00004), 34% by 17AAG (P = 0.0022) and even 53% by the combination (P = 0.011). Lapatinib inhibits HER2 internalization and 17AAG lowers HER2 membrane expression. Both drugs reduce (89)Zr-trastuzumab-F(ab')(2) tumor uptake. Based on our findings, supported by previous preclinical data indicating the antitumor potency of lapatinib in combination with HSP90 inhibition, combination of these drugs deserves further investigation.


Assuntos
Anticorpos Monoclonais Humanizados/metabolismo , Antineoplásicos/uso terapêutico , Benzoquinonas/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Fragmentos Fab das Imunoglobulinas/metabolismo , Lactamas Macrocíclicas/uso terapêutico , Quinazolinas/uso terapêutico , Receptor ErbB-2/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Western Blotting , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proliferação de Células , Sinergismo Farmacológico , Feminino , Citometria de Fluxo , Imunofluorescência , Genes erbB-1 , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Técnicas Imunoenzimáticas , Lapatinib , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Receptor ErbB-2/antagonistas & inibidores , Distribuição Tecidual , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
11.
J Nucl Med ; 56(1): 63-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25476536

RESUMO

UNLABELLED: No validated predictive biomarkers for antiangiogenic treatment of metastatic renal cell carcinoma (mRCC) exist. Tumor vascular endothelial growth factor A (VEGF-A) level may be useful. We determined tumor uptake of (89)Zr-bevacizumab, a VEGF-A-binding PET tracer, in mRCC patients before and during antiangiogenic treatment in a pilot study. METHODS: Patients underwent (89)Zr-bevacizumab PET scans at baseline and 2 and 6 wk after initiating either bevacizumab (10 mg/kg every 2 wk) with interferon-α (3-9 million IU 3 times/wk) (n = 11) or sunitinib (50 mg daily, 4 of every 6 wk) (n = 11). Standardized uptake values were compared with plasma VEGF-A and time to disease progression. RESULTS: (89)Zr-bevacizumab PET scans visualized 125 evaluable tumor lesions in 22 patients, with a median SUV(max) (maximum standardized uptake value) of 6.9 (range, 2.3-46.9). Bevacizumab/interferon-α induced a mean change in tumor SUV(max) of -47.0% (range, -84.7 to +20.0%; P < 0.0001) at 2 wk and an additional -9.7% (range, -44.8 to +38.9%; P = 0.015) at 6 wk. In the sunitinib group, the mean change in tumor SUV(max) was -14.3% at 2 wk (range, -80.4 to +269.9; P = 0.006), but at 6 wk the mean change in tumor SUV(max) was +72.6% (range, -46.4 to +236%; P < 0.0001) above baseline. SUV(max) was not related to plasma VEGF-A at all scan moments. A baseline mean tumor SUV(max) greater than 10.0 in the 3 most intense lesions corresponded with longer time to disease progression (89.7 vs. 23.0 wk; hazard ratio, 0.22; 95% confidence interval, 0.05-1.00). CONCLUSION: Tumor uptake of (89)Zr-bevacizumab is high in mRCC, with remarkable interpatient and intrapatient heterogeneity. Bevacizumab/interferon-α strongly decreases tumor uptake whereas sunitinib results in a modest reduction with an overshoot after 2 drug-free weeks. High baseline tumor SUV(max) was associated with longer time to progression.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados/metabolismo , Carcinoma de Células Renais/diagnóstico por imagem , Carcinoma de Células Renais/tratamento farmacológico , Neoplasias Renais/diagnóstico por imagem , Neoplasias Renais/tratamento farmacológico , Idoso , Anticorpos Monoclonais Humanizados/uso terapêutico , Bevacizumab , Transporte Biológico , Carcinoma de Células Renais/sangue , Carcinoma de Células Renais/metabolismo , Feminino , Humanos , Indóis/uso terapêutico , Interferon-alfa/uso terapêutico , Neoplasias Renais/sangue , Neoplasias Renais/metabolismo , Masculino , Pessoa de Meia-Idade , Pirróis/uso terapêutico , Traçadores Radioativos , Radioisótopos , Cintilografia , Sunitinibe , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/sangue , Zircônio
12.
J Nucl Med ; 56(9): 1310-4, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26135113

RESUMO

UNLABELLED: Transforming growth factor-ß (TGF-ß) signaling is involved in glioma development. The monoclonal antibody fresolimumab (GC1008) can neutralize all mammalian isoforms of TGF-ß, and tumor uptake can be visualized and quantified with (89)Zr-fresolimumab PET in mice. The aim of this study was to investigate the fresolimumab uptake in recurrent high-grade gliomas using (89)Zr-fresolimumab PET and to assess treatment outcome in patients with recurrent high-grade glioma treated with fresolimumab. METHODS: Patients with recurrent glioma were eligible. After intravenous administration of 37 MBq (5 mg) of (89)Zr-fresolimumab, PET scans were acquired on day 2 or day 4 after tracer injection. Thereafter, patients were treated with 5 mg of fresolimumab per kilogram intravenously every 3 wk. (89)Zr-fresolimumab tumor uptake was quantified as maximum standardized uptake value (SUVmax). MR imaging for response evaluation was performed after 3 infusions or as clinically indicated. RESULTS: Twelve patients with recurrent high-grade glioma were included: 10 glioblastomas, 1 anaplastic oligodendroglioma, and 1 anaplastic astrocytoma. All patients underwent (89)Zr-fresolimumab PET 4 d after injection. In 4 patients, an additional PET scan was obtained on day 2 after injection. SUVmax on day 4 in tumor lesions was 4.6 (range, 1.5-13.9) versus a median SUVmean of 0.3 (range, 0.2-0.5) in normal brain tissue. All patients showed clinical or radiologic progression after 1-3 infusions of fresolimumab. Median progression-free survival was 61 d (range, 25-80 d), and median overall survival was 106 d (range, 37-417 d). CONCLUSION: (89)Zr-fresolimumab penetrated recurrent high-grade gliomas very well but did not result in clinical benefit.


Assuntos
Anticorpos Monoclonais/farmacocinética , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Glioma/radioterapia , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/radioterapia , Adulto , Idoso , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/radioterapia , Linhagem Celular Tumoral , Feminino , Glioma/diagnóstico por imagem , Humanos , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Radioisótopos/farmacocinética , Compostos Radiofarmacêuticos/farmacocinética , Distribuição Tecidual , Fator de Crescimento Transformador beta/imunologia , Zircônio/farmacocinética
13.
Clin Cancer Res ; 20(15): 3945-54, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-25085789

RESUMO

PURPOSE: HSP90 chaperones have key client proteins that are involved in all hallmarks of breast cancer growth and progression. The primary aim of this clinical trial was to evaluate the feasibility of using (89)Zr-trastuzumab PET (for HER2-positive breast cancer) or (89)Zr-bevacizumab PET [for estrogen receptor (ER)-positive breast cancer] to determine in vivo degradation of client proteins caused by the novel HSP90 inhibitor NVP-AUY922. EXPERIMENTAL DESIGN: Of note, 70 mg/m(2) NVP-AUY922 was administered intravenously in a weekly schedule to patients with advanced HER2 or ER-positive breast cancer. Biomarker analysis consisted of serial PET imaging with 2[18F]fluoro-2-deoxy-D-glucose (FDG), (89)Zr-trastuzumab, or (89)Zr-bevacizumab. Response evaluation was performed according to RECIST1.0. FDG, (89)Zr-trastuzumab, and (89)Zr-bevacizumab distributions were scored visually and quantitatively by calculating the maximum standardized uptake values (SUVmax). In blood samples, serial HSP70 levels, extracellular form of HER2 (HER2-ECD), and pharmacokinetic and pharmacodynamic parameters were measured. RESULTS: Sixteen patients (ten HER2-positive and six ER-positive tumors) were included. One partial response was observed; seven patients showed stable disease. SUVmax change in individual tumor lesions on baseline versus 3 weeks (89)Zr-trastuzumab PET was heterogeneous and related to size change on CT after 8 weeks treatment (r(2) = 0.69; P = 0.006). Tumor response on (89)Zr-bevacizumab PET and FDG-PET was not correlated with CT response. CONCLUSIONS: NVP-AUY922 showed proof-of-concept clinical response in HER2-amplified metastatic breast cancer. Early change on (89)Zr-trastuzumab PET was positively associated with change in size of individual lesions assessed by CT.


Assuntos
Anticorpos Monoclonais Humanizados , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Isoxazóis/farmacologia , Tomografia por Emissão de Pósitrons/métodos , Resorcinóis/farmacologia , Anticorpos Monoclonais Humanizados/farmacocinética , Bevacizumab , Neoplasias da Mama/metabolismo , Neoplasias da Mama/secundário , Ensaios Clínicos Fase II como Assunto , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Radioisótopos , Zircônio
14.
J Nucl Med ; 55(7): 1087-92, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24790218

RESUMO

UNLABELLED: Everolimus increases progression-free survival in patients with advanced neuroendocrine tumors (NETs). Currently, no biomarkers are available for early selection of patients who will benefit from everolimus. Everolimus can reduce vascular endothelial growth factor A (VEGF-A) production by tumor cells. Therefore, we aimed to investigate the effect of everolimus on tumor uptake of the radioactive-labeled VEGF-A antibody bevacizumab with PET in NET patients. METHODS: Patients with advanced progressive well-differentiated NETs underwent (89)Zr-bevacizumab PET scans before and at 2 and 12 wk during everolimus treatment. (89)Zr-bevacizumab uptake was quantified by the maximum standardized uptake value (SUVmax). Tumor response and the percentage change in the sum of target lesion diameters were determined according to Response Evaluation Criteria in Solid Tumors 1.1 on CT (3 monthly). RESULTS: In 4 of the 14 patients entered, no tumor lesions were visualized with (89)Zr-bevacizumab PET. In the remaining patients, 19% of tumor lesions 1 cm or greater known by CT were visualized. Tumor SUVmax decreased during everolimus treatment, with a median of -7% at 2 wk (P = 0.09) and a median of -35% at 12 wk (P < 0.001). The difference in SUVmax at 2 and 12 wk with respect to SUVmax at baseline correlated with percentage change on CT at 6 mo (r(2) = 0.51, P < 0.05, and r(2) = 0.61, P < 0.01, respectively). CONCLUSION: This study demonstrates variable (89)Zr-bevacizumab PET tumor uptake in NET patients. (89)Zr-bevacizumab tumor uptake diminished during everolimus treatment. Serial (89)Zr-bevacizumab PET might be useful as an early predictive biomarker of anti-VEGF-directed treatment in NET patients.


Assuntos
Anticorpos Monoclonais Humanizados/metabolismo , Tumores Neuroendócrinos/tratamento farmacológico , Tumores Neuroendócrinos/metabolismo , Sirolimo/análogos & derivados , Adulto , Idoso , Anticorpos Monoclonais Humanizados/imunologia , Bevacizumab , Transporte Biológico/efeitos dos fármacos , Cromogranina A/sangue , Everolimo , Estudos de Viabilidade , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/sangue , Tumores Neuroendócrinos/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Radioisótopos , Sirolimo/sangue , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Resultado do Tratamento , Fator A de Crescimento do Endotélio Vascular/imunologia , Zircônio
15.
J Nucl Med ; 54(6): 929-35, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23625582

RESUMO

UNLABELLED: Placental growth factor (PlGF) is a member of the proangiogenic vascular endothelial growth factor family, which is upregulated in many tumors. RO5323441, a humanized monoclonal antibody against PlGF, showed antitumor activity in human tumor xenografts. We therefore aimed to radiolabel RO5323441 and preclinically validate this tracer to study drug tumor uptake and organ distribution by PET imaging. (89)Zr-RO5323441 was tested for stability and immunoreactivity in vitro. METHODS: The tumor uptake and organ distribution for 10, 50, and 500 µg of (89)Zr-RO5323441 was assessed in mice bearing human PlGF-expressing hepatocellular cancer (Huh7) xenografts or human renal cell carcinoma (ACHN) xenografts without detectable human PlGF expression. The effect of pretreatment with RO5323441 (20 mg/kg) on (89)Zr-RO5323441 tumor uptake was analyzed in Huh7 xenografts. (111)In-IgG served as a control for nonspecific tumor uptake and organ distribution. Cy5-RO5323441 was injected to study the intratumor distribution of RO5323441 with fluorescence microscopy. RESULTS: (89)Zr-RO5323441 showed a time- and dose-dependent tumor accumulation. Uptake in Huh7 xenografts at 10 µg of (89)Zr-RO5323441 was 8.2% ± 1.7% injected dose (ID)/cm(3) at 144 h after injection, and in ACHN xenografts it was 5.5 ± 0.3 %ID/cm(3) (P = 0.03). RO5323441 pretreatment of Huh7 xenograft-bearing mice reduced (89)Zr-RO5323441 tumor uptake to the level of nonspecific (111)In-IgG uptake. Cy5-RO5323441 was present in the tumors mainly in the microenvironment. CONCLUSION: The findings show that RO5323441 tumor uptake is PlGF-specific and time- and dose-dependent.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacocinética , Proteínas da Gravidez/imunologia , Proteínas da Gravidez/metabolismo , Radioisótopos , Microambiente Tumoral , Zircônio , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados , Transporte Biológico/efeitos dos fármacos , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Marcação por Isótopo , Neoplasias Renais/metabolismo , Neoplasias Renais/patologia , Macrófagos/metabolismo , Masculino , Camundongos , Fator de Crescimento Placentário , Microambiente Tumoral/efeitos dos fármacos
16.
Cancer Res ; 73(11): 3347-55, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23580572

RESUMO

In solid tumors, angiogenesis occurs in the setting of a defective vasculature and impaired lymphatic drainage that is associated with increased vascular permeability and enhanced tumor permeability. These universal aspects of the tumor microenvironment can have a marked influence on intratumoral drug delivery that may often be underappreciated. In this study, we investigated the effect of blood vessel normalization in tumors by the antiangiogenic drug bevacizumab on antibody uptake by tumors. In mouse xenograft models of human ovarian and esophageal cancer (SKOV-3 and OE19), we evaluated antibody uptake in tumors by positron emission tomographic imaging 24 and 144 hours after injection of (89)Zr-trastuzumab (SKOV-3 and OE19), (89)Zr-bevacizumab (SKOV-3), or (89)Zr-IgG (SKOV-3) before or after treatment with bevacizumab. Intratumor distribution was assessed by fluorescence microscopy along with mean vessel density (MVD) and vessel normalization. Notably, bevacizumab treatment decreased tumor uptake and intratumoral accumulation compared with baseline in the tumor models relative to controls. Bevacizumab treatment also reduced MVD in tumors and increased vessel pericyte coverage. These findings are clinically important, suggesting caution in designing combinatorial trials with therapeutic antibodies due to a possible reduction in tumoral accumulation that may be caused by bevacizumab cotreatment.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/farmacocinética , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Imunoglobulina G/metabolismo , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/metabolismo , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/farmacocinética , Bevacizumab , Linhagem Celular Tumoral , Interações Medicamentosas , Neoplasias Esofágicas/irrigação sanguínea , Neoplasias Esofágicas/diagnóstico por imagem , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/diagnóstico por imagem , Tomografia por Emissão de Pósitrons , Radioisótopos , Compostos Radiofarmacêuticos/farmacocinética , Trastuzumab , Ensaios Antitumorais Modelo de Xenoenxerto , Zircônio/farmacocinética
17.
Pharmacol Ther ; 135(2): 123-32, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22587883

RESUMO

Cancer remains one of the leading causes of death in the developed countries and cancer mortality is expected to rise globally. Despite encouraging developments regarding targeted drugs, the most prevalent cancer mortality remains metastatic disease. Therefore, drugs that target cancer progression, invasion and metastasis are clearly needed. One of the most interesting targets in this setting is transforming growth factor ß (TGF-ß). TGF-ß can promote tumor growth, invasion and metastasis. However, TGF-ß also has a physiological, opposing role: maintaining tissue homeostasis and suppression of tumor progression. The window of effective TGF-ß targeting is therefore evidently small, which poses a clear challenge in selecting patients at the right time. Despite this complexity, several TGF-ß inhibitors are currently in clinical development, modulating TGF-ß production, activation or signaling. Still, specificity and long term toxicity remain unclear, emphasizing the importance of careful monitoring of clinical trials. Development and application of these drugs in the clinic require adequate insight and evaluation methods for the role of TGF-ß during tumor invasion and metastasis. In this review, presently available methods for clinical evaluation will be discussed, such as an ex vivo stimulation assay, TGF-ß response signature and molecular imaging techniques. Future clinical trials incorporating the validation of these evaluation methods will show which method will be most predictive and suitable for clinical application.


Assuntos
Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diagnóstico por Imagem , Humanos , Neoplasias/diagnóstico , Neoplasias/tratamento farmacológico , Fator de Crescimento Transformador beta/antagonistas & inibidores
18.
Neoplasia ; 14(8): 709-18, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22952424

RESUMO

Several in vitro and in vivo models have revealed the key role of CXCR4/CXCL12 axis in tumor-stroma interactions. Stromal cells present in the tumor microenvironment express high levels of CXCL12 protein, directly stimulating proliferation and migration of CXCR4-expressing cancer cells. This specific prosurvival influence of stromal cells on tumor cells is thought to protect them from cytotoxic chemotherapy and is postulated as a possible explanation for the minimal residual disease in hematological and solid cancers. Therefore, CXCR4/CXCL12 signaling is an attractive therapeutic target in cancer, as proven in preclinical leukemia mouse models, where CXCR4 inhibition sensitized cancer cells to conventional chemotherapy. This study investigates whether inhibition of CXCR4 with the specific inhibitor AMD3100 sensitizes human prostate cancer cells to docetaxel. We showed that both mouse and human stromal cell lines have a protective effect on PC3-luc cells by promoting their survival after chemotherapy. Furthermore, we demonstrated that AMD3100 sensitizes PC3-luc cells to docetaxel. In a subcutaneous xenograft mouse model of human prostate carcinoma, we showed that a combination of docetaxel and AMD3100 exerts increased antitumor effect compared with docetaxel alone. We concluded that CXCR4 inhibition chemosensitizes prostate cancer cells, both in vitro and in vivo. To explore the relevance of these findings, we analyzed CXCR4 expression levels in human prostate cancer samples. We found that cancer cells present in bone metastatic lesions express higher CXCR4 levels relative to the cells present in primary tumors and lymph node metastatic lesions. These findings underscore the potential of CXCR4 inhibitors as chemosensitizing agents.


Assuntos
Compostos Heterocíclicos/farmacologia , Neoplasias da Próstata/tratamento farmacológico , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Taxoides/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Benzilaminas , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/secundário , Adesão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL12/metabolismo , Ciclamos , Docetaxel , Humanos , Masculino , Camundongos , Neoplasias da Próstata/metabolismo , Distribuição Aleatória , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Cancer Discov ; 1(1): 25-8, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22586317

RESUMO

With current testing strategies, the number of novel targeted anticancer agents will exceed our drug selection capacity. Molecular imaging is a powerful additional tool that can assist us in selecting effective drugs and help patients benefit from targeted agents. Moreover, measurement of the functional effects of such targeted agents could permit dynamic tuning of treatment selection at the earliest time point at which loss of functional effects is observed.


Assuntos
Aprovação de Drogas/métodos , Descoberta de Drogas/métodos , Imagem Molecular/métodos , Neoplasias/tratamento farmacológico , Animais , Ensaios Clínicos como Assunto , Aprovação de Drogas/economia , Descoberta de Drogas/economia , Avaliação Pré-Clínica de Medicamentos , Humanos , Terapia de Alvo Molecular/economia , Terapia de Alvo Molecular/métodos , Neoplasias/economia , Medicina de Precisão/economia
20.
J Nucl Med ; 52(12): 2001-8, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22072706

RESUMO

UNLABELLED: Transforming growth factor-ß (TGF-ß) promotes cancer invasion and metastasis and is therefore a potential drug target for cancer treatment. Fresolimumab, which neutralizes all mammalian active isoforms of TGF-ß, was radiolabeled with (89)Zr for PET to analyze TGF-ß expression, antibody tumor uptake, and organ distribution. METHODS: (89)Zr was conjugated to fresolimumab using the chelator N-succinyldesferrioxamine-B-tetrafluorphenol. (89)Zr-fresolimumab was analyzed for conjugation ratio, aggregation, radiochemical purity, stability, and immunoreactivity. (89)Zr-fresolimumab tumor uptake and organ distribution were assessed using 3 protein doses (10, 50, and 100 µg) and compared with (111)In-IgG in a human TGF-ß-transfected Chinese hamster ovary xenograft model, human breast cancer MDA-MB-231 xenograft, and metastatic model. Latent and active TGF-ß1 expression was analyzed in tissue homogenates with enzyme-linked immunosorbent assay. RESULTS: (89)Zr was labeled to fresolimumab with high specific activity (>1 GBq/mg), high yield, and high purity. In vitro validation of (89)Zr-fresolimumab showed a fully preserved immunoreactivity and long (>1 wk) stability in solution and in human serum. In vivo validation showed an (89)Zr-fresolimumab distribution similar to IgG in most organs, except for a higher uptake in the liver in all mice and higher kidney uptake in the 10-µg group. (89)Zr-fresolimumab induced no toxicity in mice; it accumulated in primary tumors and metastases in a manner similar to IgG. Both latent and active TGF-ß was detected in tumor homogenates, whereas only latent TGF-ß could be detected in liver homogenates. Remarkably high (89)Zr-fresolimumab uptake was seen in sites of tumor ulceration and in scar tissue, processes in which TGF-ß is known to be highly active. CONCLUSION: Fresolimumab tumor uptake and organ distribution can be visualized and quantified with (89)Zr-fresolimumab PET. This technique will be used to guide further clinical development of fresolimumab and could possibly identify patients most likely to benefit.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias da Mama/diagnóstico por imagem , Tomografia por Emissão de Pósitrons/métodos , Radioisótopos , Fator de Crescimento Transformador beta/imunologia , Zircônio , Animais , Anticorpos Monoclonais/farmacocinética , Anticorpos Monoclonais Humanizados , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Células CHO , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Cricetinae , Cricetulus , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Marcação por Isótopo , Fígado/diagnóstico por imagem , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos , Metástase Neoplásica , Transfecção , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA