Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Nucl Med ; 65(2): 221-223, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38071554

RESUMEN

The estrogen receptor (ER), a steroid hormone receptor important in female physiology, is a significant contributor to breast carcinogenesis and progression and, as such, is an important therapeutic target. Approximately 70% of breast cancers will express ER at presentation, and the determination of ER expression by tissue assay, usually by immunohistochemistry, is part of the standard of care for newly diagnosed breast cancer. ER expression is important in guiding the approach to treatment, especially with the increase in relevant systemic therapies. The ER-targeting imaging agent 16α-[18F]fluoro-17ß-estradiol ([18F]FES) is approved for clinical use by regulatory agencies in France and the United States. Multiple studies suggest the advantages of [18F]FES PET in assessing tumor ER expression, the ability of both qualitative and quantitative [18F]FES PET measures to predict response to ER-targeted therapy, and the ability of [18F]FES PET to clarify equivocal staging and restaging results in patients with ER-expressing cancers. [18F]FES PET/CT may also be helpful in staging invasive lobular breast cancer and low-grade ER-expressing invasive ductal cancers and, in some cases, may be a substitute for biopsy. The Society of Nuclear Medicine and Molecular Imaging and the European Association of Nuclear Medicine in June 2023 released a procedure standard/practice guideline for [18F]FES PET ER imaging of patients with breast cancer. The goal of the standard/guideline is to assist physicians in recommending, performing, interpreting, and reporting the results of [18F]FES PET studies for patients with breast cancer and to provide clinicians with the best available evidence, inform them about areas where robust evidence is lacking, and help them deliver the best possible diagnostic efficacy and study quality for their patients. Also reviewed are standardized quality control, quality assurance, and imaging procedures for [18F]FES PET. The authors emphasize the importance of precision, accuracy, repeatability, and reproducibility for both clinical management of patients and for use of [18F]FES PET in multicenter trials. A standardized imaging procedure, in combination with already published appropriate-use criteria, will help promote the use of [18F]FES PET and enhance subsequent research. This brief summary article reviews the content of the joint standard/guideline, which is available in its entirety at https://www.snmmi.org/ClinicalPractice/content.aspx?ItemNumber=6414&navItemNumbe=10790.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Receptores de Estrógenos/metabolismo , Reproducibilidad de los Resultados , Estradiol/metabolismo , Tomografía de Emisión de Positrones/métodos
2.
Ther Adv Med Oncol ; 15: 17588359231170738, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37223262

RESUMEN

Molecular imaging, such as positron emission tomography (PET), is increasingly used as biomarker to predict and assess treatment response in breast cancer. The number of biomarkers is expanding with specific tracers for tumour characteristics throughout the body and this information can be used to aid the decision-making process. These measurements include metabolic activity using [18F]fluorodeoxyglucose PET ([18F]FDG-PET), oestrogen receptor (ER) expression using 16α-[18F]Fluoro-17ß-oestradiol ([18F]FES)-PET and human epidermal growth factor receptor 2 (HER2) expression using PET with radiolabelled trastuzumab (HER2-PET). In early breast cancer, baseline [18F]FDG-PET is frequently used for staging, but limited subtype-specific data reduce its usefulness as biomarker for treatment response or outcome. Early metabolic change on serial [18F]FDG-PET is increasingly used in the neo-adjuvant setting as dynamic biomarker to predict pathological complete response to systemic therapy, potentially allowing de-intensification or step-up intensification of treatment. In the metastatic setting, baseline [18F]FDG-PET and [18F]FES-PET can be used as biomarker to predict treatment response, in triple-negative and ER-positive breast cancer, respectively. Metabolic progression on repeated [18F]FDG-PET appears to precede progressive disease on standard evaluation imaging; however, subtype-specific studies are limited and more prospective data are needed before implementation in clinical practice. Even though (repeated) [18F]FDG-PET, [18F]FES-PET and HER2-PEt all show promising results as biomarkers to predict therapy response and outcome, for eventual integration into clinical practice, future studies will have to clarify at what timepoint this integration has to optimally take place.

3.
Eur Urol Open Sci ; 49: 23-31, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36874601

RESUMEN

Background: Germline and tumour genetic testing in prostate cancer (PCa) is becoming more broadly accepted, but testing indications and clinical consequences for carriers in each disease stage are not yet well defined. Objective: To determine the consensus of a Dutch multidisciplinary expert panel on the indication and application of germline and tumour genetic testing in PCa. Design setting and participants: The panel consisted of 39 specialists involved in PCa management. We used a modified Delphi method consisting of two voting rounds and a virtual consensus meeting. Outcome measurements and statistical analysis: Consensus was reached if ≥75% of the panellists chose the same option. Appropriateness was assessed by the RAND/UCLA appropriateness method. Results and limitations: Of the multiple-choice questions, 44% reached consensus. For men without PCa having a relevant family history (familial PCa/BRCA-related hereditary cancer), follow-up by prostate-specific antigen was considered appropriate. For patients with low-risk localised PCa and a family history of PCa, active surveillance was considered appropriate, except in case of the patient being a BRCA2 germline pathogenic variant carrier. Germline and tumour genetic testing should not be done for nonmetastatic hormone-sensitive PCa in the absence of a relevant family history of cancer. Tumour genetic testing was deemed most appropriate for the identification of actionable variants, with uncertainty for germline testing. For tumour genetic testing in metastatic castration-resistant PCa, consensus was not reached for the timing and panel composition. The principal limitations are as follows: (1) a number of topics discussed lack scientific evidence, and therefore the recommendations are partly opinion based, and (2) there was a small number of experts per discipline. Conclusions: The outcomes of this Dutch consensus meeting may provide further guidance on genetic counselling and molecular testing related to PCa. Patient summary: A group of Dutch specialists discussed the use of germline and tumour genetic testing in prostate cancer (PCa) patients, indication of these tests (which patients and when), and impact of these tests on the management and treatment of PCa.

4.
J Clin Oncol ; 40(31): 3642-3652, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35584346

RESUMEN

PURPOSE: Determining the estrogen receptor (ER) status is essential in metastatic breast cancer (MBC) management. Whole-body ER imaging with 16α-[18F]fluoro-17ß-estradiol positron emission tomography ([18F]FES-PET) is increasingly used for this purpose. To establish the clinical validity of the [18F]FES-PET, we studied the diagnostic accuracy of qualitative and quantitative [18F]FES-PET assessment to predict ER expression by immunohistochemistry in a metastasis. METHODS: In a prospective multicenter trial, 200 patients with newly diagnosed MBC underwent extensive workup including molecular imaging. For this subanalysis, ER expression in the biopsied metastasis was related to qualitative whole-body [18F]FES-PET evaluation and quantitative [18F]FES uptake in the corresponding metastasis. A review and meta-analysis regarding [18F]FES-PET diagnostic performance were performed. RESULTS: Whole-body [18F]FES-PET assessment predicted ER expression in the biopsied metastasis with good accuracy: a sensitivity of 95% (95% CI, 89 to 97), a specificity of 80% (66 to 89), a positive predictive value (PPV) of 93% (87 to 96), and a negative predictive value (NPV) of 85% (72 to 92) in 181 of 200 evaluable patients. Quantitative [18F]FES uptake predicted ER immunohistochemistry in the corresponding metastasis with a sensitivity/specificity of 91%/69% and a PPV/NPV of 90%/71% in 156 of 200 evaluable patients. For bone metastases, PPV/NPV was 92%/81%. Meta-analysis with addition of our data has increased diagnostic performance and narrowed the 95% CIs compared with previous studies with a sensitivity/specificity of both 86% (81 to 90 and 73 to 93, respectively). CONCLUSION: In this largest prospective series so far, we established the clinical validity of [18F]FES-PET to determine tumor ER status in MBC. In view of the high diagnostic accuracy of qualitatively assessed whole-body [18F]FES-PET, this noninvasive imaging modality can be considered a valid alternative to a biopsy of a metastasis to determine ER status in newly MBC (ClinicalTrials.gov identifier: NCT01957332).


Asunto(s)
Neoplasias de la Mama , Estradiol , Humanos , Femenino , Neoplasias de la Mama/patología , Receptores de Estrógenos/metabolismo , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/métodos , Estudios Multicéntricos como Asunto
5.
Clin Lung Cancer ; 23(2): e104-e115, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34325996

RESUMEN

INTRODUCTION: Non-small cell lung cancer (NSCLC) patients with Anaplastic Lymphoma Kinase (ALK) gene fusions respond well to ALK inhibitors but commonly develop on-target resistance mutations. The objective of this study is to collect clinical evidence for subsequent treatment with ALK inhibitors. PATIENTS AND METHODS: Local experience with on-target ALK resistance mutations and review of the literature identified 387 patients with ALK inhibitor resistance mutations. Clinical benefit of mutation-inhibitor combinations was assessed based on reported response, progression-free survival and duration of treatment. Furthermore, this clinical evidence was compared to previously reported in vitro sensitivity of mutations to the inhibitors. RESULTS: Of the pooled population of 387 patients in this analysis, 239 (62%) received at least 1 additional line of ALK inhibition after developing on-target resistance to ALK inhibitor therapy. Clinical benefit was reported for 177 (68%) patients, but differed for each mutation-inhibitor combination. Agreement between in vitro predicted sensitivity of 6 published models and observed clinical benefit ranged from 69% to 89%. The observed clinical evidence for highest probability of response in the context of specific on-target ALK inhibitor resistance mutations is presented. CONCLUSION: Molecular diagnostics performed on tissue samples that are refractive to ALK inhibitor therapy can reveal new options for targeted therapy for NSCLC patients. Our comprehensive overview of clinical evidence of drug actionability of ALK on-target resistance mechanisms may serve as a practical guide to select the most optimal drug for individual patients.


Asunto(s)
Quinasa de Linfoma Anaplásico/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Quinasa de Linfoma Anaplásico/metabolismo , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Mutación , Supervivencia sin Progresión
6.
Cancer Metastasis Rev ; 39(3): 999-1013, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32367253

RESUMEN

Response evaluation for cancer treatment consists primarily of clinical and radiological assessments. In addition, a limited number of serum biomarkers that assess treatment response are available for a small subset of malignancies. Through recent technological innovations, new methods for measuring tumor burden and treatment response are becoming available. By utilization of highly sensitive techniques, tumor-specific mutations in circulating DNA can be detected and circulating tumor DNA (ctDNA) can be quantified. These so-called liquid biopsies provide both molecular information about the genomic composition of the tumor and opportunities to evaluate tumor response during therapy. Quantification of tumor-specific mutations in plasma correlates well with tumor burden. Moreover, with liquid biopsies, it is also possible to detect mutations causing secondary resistance during treatment. This review focuses on the clinical utility of ctDNA as a response and follow-up marker in patients with non-small cell lung cancer, melanoma, colorectal cancer, and breast cancer. Relevant studies were retrieved from a literature search using PubMed database. An overview of the available literature is provided and the relevance of ctDNA as a response marker in anti-cancer therapy for clinical practice is discussed. We conclude that the use of plasma-derived ctDNA is a promising tool for treatment decision-making based on predictive testing, detection of resistance mechanisms, and monitoring tumor response. Necessary steps for translation to daily practice and future perspectives are discussed.


Asunto(s)
ADN Tumoral Circulante/sangre , Neoplasias/genética , Neoplasias/terapia , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , ADN Tumoral Circulante/genética , Humanos , Biopsia Líquida , Mutación , Neoplasias/sangre , Neoplasias/patología , Valor Predictivo de las Pruebas
7.
EJNMMI Res ; 10(1): 40, 2020 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-32307594

RESUMEN

PURPOSE: Correct identification of tumour receptor status is important for treatment decisions in breast cancer. [18F]FES PET and [18F]FDHT PET allow non-invasive assessment of the oestrogen (ER) and androgen receptor (AR) status of individual lesions within a patient. Despite standardised analysis techniques, interobserver variability can significantly affect the interpretation of PET results and thus clinical applicability. The purpose of this study was to determine visual and quantitative interobserver variability of [18F]FES PET and [18F]FDHT PET interpretation in patients with metastatic breast cancer. METHODS: In this prospective, two-centre study, patients with ER-positive metastatic breast cancer underwent both [18F]FES and [18F]FDHT PET/CT. In total, 120 lesions were identified in 10 patients with either conventional imaging (bone scan or lesions > 1 cm on high-resolution CT, n = 69) or only with [18F]FES and [18F]FDHT PET (n = 51). All lesions were scored visually and quantitatively by two independent observers. A visually PET-positive lesion was defined as uptake above background. For quantification, we used standardised uptake values (SUV): SUVmax, SUVpeak and SUVmean. RESULTS: Visual analysis showed an absolute positive and negative interobserver agreement for [18F]FES PET of 84% and 83%, respectively (kappa = 0.67, 95% CI 0.48-0.87), and 49% and 74% for [18F]FDHT PET, respectively (kappa = 0.23, 95% CI - 0.04-0.49). Intraclass correlation coefficients (ICC) for quantification of SUVmax, SUVpeak and SUVmean were 0.98 (95% CI 0.96-0.98), 0.97 (95% CI 0.96-0.98) and 0.89 (95% CI 0.83-0.92) for [18F]FES, and 0.78 (95% CI 0.66-0.85), 0.76 (95% CI 0.63-0.84) and 0.75 (95% CI 0.62-0.84) for [18F]FDHT, respectively. CONCLUSION: Visual and quantitative evaluation of [18F]FES PET showed high interobserver agreement. These results support the use of [18F]FES PET in clinical practice. In contrast, visual agreement for [18F]FDHT PET was relatively low due to low tumour-background ratios, but quantitative agreement was good. This underscores the relevance of quantitative analysis of [18F]FDHT PET in breast cancer. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01988324. Registered 20 November 2013, https://clinicaltrials.gov/ct2/show/NCT01988324?term=FDHT+PET&draw=1&rank=2.

8.
Ther Drug Monit ; 41(6): 693-695, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31169759

RESUMEN

The authors present a case of an 18-year-old man with metastasized osteosarcoma, admitted for methotrexate (MTX) treatment combined with cisplatin and doxorubicin. During the first cycle, severe MTX toxicity was observed with increased MTX serum levels and delayed MTX clearance requiring rescue treatment with intensified leucovorin. In the following cycles, cisplatin and doxorubicin were discontinued, and MTX dose was reduced. The elimination half-life slowly improved over the following cycles suggesting a reversible cause responsible for reduced MTX clearance and toxicity during the first cycle. Cisplatin is well-known for its nephrotoxic effects and can induce reversible tubular injury. Previous treatment with cisplatin may well have been responsible for decreased MTX clearance, and combination treatment should be used with adequate monitoring of MTX levels. Other factors that may have contributed, such as urine alkalization, gene polymorphisms, and other drug-drug interactions are discussed.


Asunto(s)
Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Cisplatino/farmacocinética , Metotrexato/farmacocinética , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Adolescente , Antibacterianos/uso terapéutico , Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Cisplatino/uso terapéutico , Doxorrubicina/efectos adversos , Doxorrubicina/uso terapéutico , Interacciones Farmacológicas , Femenino , Humanos , Enfermedades Renales/inducido químicamente , Leucovorina/farmacología , Metotrexato/sangre , Metotrexato/metabolismo , Metotrexato/uso terapéutico , Morfina/uso terapéutico , Mucositis/inducido químicamente , Mucositis/tratamiento farmacológico , Osteosarcoma/metabolismo
9.
Pharmacol Ther ; 200: 135-147, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31077689

RESUMEN

The androgen receptor (AR) is a drug target in breast cancer, and AR-targeted therapies have induced tumor responses in breast cancer patients. In this review, we summarized the role of AR in breast cancer based on preclinical and clinical data. Response to AR-targeted therapies in unselected breast cancer populations is relatively low. Preclinical and clinical data show that AR antagonists might have a role in estrogen receptor (ER)-negative/AR-positive tumors. The prognostic value of AR for patients remains uncertain due to the use of various antibodies and cut-off values for immunohistochemical assessment. To get more insight into the role of AR in breast cancer, we additionally performed a retrospective pooled analysis to determine the prognostic value of the AR using mRNA profiles of 7270 primary breast tumors. Our analysis shows that a higher AR mRNA level is associated with improved disease outcome in patients with ER-positive/human epidermal growth factor receptor 2 (HER2)-negative tumors, but with worse disease outcome in HER2-positive subgroups. In conclusion, next to AR expression, incorporation of additional tumor characteristics will potentially make AR targeting a more valuable therapeutic strategy in breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Receptores Androgénicos/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Femenino , Humanos , Masculino , Terapia Molecular Dirigida , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , ARN Mensajero/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/fisiología
10.
J Nucl Med ; 59(8): 1212-1218, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29602822

RESUMEN

Heterogeneity of estrogen receptor (ER) expression in breast cancer is recognized. However, knowledge about varying expression across metastases and surrounding normal tissue in patients is scarce. We therefore analyzed 16α-18F-fluoro-17ß-estradiol (18F-FES) PET to assess ER expression heterogeneity. Methods:18F-FES PET on accredited PET/CT camera systems performed in patients with ER-positive metastatic breast cancer November 2009-December 2014 was analyzed. Lesions with an SUVmax 1.5 or more were considered ER-positive, but liver lesions were excluded given high background liver signal. CT lesions with a diameter 10 mm or more were included. We used multilevel linear-mixed models to evaluate determinants of 18F-FES uptake. Cluster analysis was performed with different imaging features per patient as input variables. Results: In 91 patients, 1,617 metastases in bone (78%), lymph node (15%), lung (4%), or liver (2%) were identified by CT (11.2%), PET (56.6%), or both (32.2%). Median tumor uptake varied greatly between patients (SUVmax, 0.54-14.21). 18F-FES uptake in bone metastases was higher than in lymph node and lung metastases (geometric mean SUVmax, 2.61 [95% confidence interval (CI), 2.31-2.94] vs. 2.29 [95% CI, 2.00-2.61; P < 0.001] vs. 2.23 [95% CI, 1.88-2.61; P = 0.021]), respectively. Cluster analysis identified 3 subgroups of patients characterized by particular metastatic sites and 18F-FES PET/CT features. SUVmax in surrounding normal tissue, highest in the bones, varied per patient (range, 0.7-3.3). Conclusion:18F-FES uptake is heterogeneous in tumor and normal tissue and influenced by anatomic site. Different patterns can be distinguished, possibly identifying biologically relevant ER-positive metastatic breast cancer patient subgroups.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Estradiol/análogos & derivados , Adulto , Anciano , Anciano de 80 o más Años , Transporte Biológico , Neoplasias de la Mama/diagnóstico por imagen , Estradiol/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Tomografía Computarizada por Tomografía de Emisión de Positrones , Receptores de Estrógenos/metabolismo
11.
J Nucl Med ; 58(12): 1906-1912, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28912144

RESUMEN

In addition to the well-known estrogen receptor (ER) and human epidermal growth factor receptor 2, the androgen receptor (AR) is also a potential drug target in breast cancer treatment. Whole-body imaging can provide information across lesions within a patient. ER expression in tumor lesions can be visualized by 18F-fluoroestradiol (18F-FES) PET, and AR expression has been visualized in prostate cancer patients with 18F-fluorodihydrotestosterone (18F-FDHT) PET. Our aim was to assess the concordance between 18F-FDHT and 18F-FES PET and tumor AR and ER expression measured immunohistochemically in patients with metastatic breast cancer. Methods: Patients with ER-positive metastatic breast cancer were eligible for the study, irrespective of tumor AR status. The concordance of 18F-FDHT and 18F-FES uptake on PET with immunohistochemical expression of AR and ER in biopsies of corresponding metastases was analyzed. Patients underwent 18F-FDHT PET and 18F-FES PET. A metastasis was biopsied within 8 wk of the PET procedures. Tumor samples with more than 10% and 1% nuclear tumor cell staining were considered, respectively, AR- and ER-positive. Correlations between PET uptake and semiquantitative immunohistochemical scoring (percentage positive cells × intensity) were calculated. The optimum threshold of SUV to discriminate positive and negative lesions for both AR and ER was determined by receiver-operating-characteristic analysis. Results: In the 13 evaluable patients, correlation (R2 ) between semiquantitative AR expression and 18F-FDHT uptake was 0.47 (P = 0.01) and between semiquantitative ER expression and 18F-FES uptake 0.78 (P = 0.01). The optimal cutoff for AR-positive lesions was an SUVmax of 1.94 for 18F-FDHT PET, yielding a sensitivity of 91% and a specificity of 100%; the optimal cutoff was an SUVmax of 1.54 for 18F-FES PET, resulting in a sensitivity and specificity of 100% for ER. Conclusion:18F-FDHT and 18F-FES uptake correlate well with AR and ER expression levels in representative biopsies. These results show the potential use of whole-body imaging for receptor status assessment, particularly in view of biopsy-associated sampling errors and heterogeneous receptor expression in breast cancer metastases.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/metabolismo , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo , Biopsia , Núcleo Celular/patología , Estudios de Factibilidad , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia/diagnóstico por imagen , Tomografía de Emisión de Positrones , Radiofármacos , Tomografía Computarizada de Emisión , Imagen de Cuerpo Entero
12.
Eur J Nucl Med Mol Imaging ; 42(11): 1674-1681, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26091705

RESUMEN

PURPOSE: Whereas anti-oestrogen therapy is widely applied to treat oestrogen receptor (ER) positive breast cancer, paradoxically, oestrogens can also induce tumour regression. Up-regulation of ER expression is a marker for oestrogen hypersensitivity. We, therefore, performed an exploratory study to evaluate positron emission tomography (PET) with the tracer 16α-[(18)F]fluoro-17ß-oestradiol ((18)F-FES) as potential marker to select breast cancer patients for oestradiol therapy. METHODS: Eligible patients had acquired endocrine-resistant metastatic breast cancer that progressed after ≥2 lines of endocrine therapy. All patients had prior ER-positive histology. Treatment consisted of oestradiol 2 mg, three times daily, orally. Patients underwent (18)F-FES-PET/CT imaging at baseline. Tumour (18)F-FES-uptake was quantified for a maximum of 20 lesions and expressed as maximum standardised uptake value (SUVmax). CT-scan was repeated every 3 months to evaluate treatment response. Clinical benefit was defined as time to radiologic or clinical progression ≥24 weeks. RESULTS: (18)F-FES uptake, quantified for 255 lesions in 19 patients, varied greatly between lesions (median 2.8; range 0.6-24.3) and between patients (median 2.5; range 1.1-15.5). Seven (37%) patients experienced clinical benefit of oestrogen therapy, eight progressed (PD), and four were non-evaluable due to side effects. The positive and negative predictive value (PPV/NPV) of (18)F-FES-PET for response to treatment were 60% (95% CI: 31-83%) and 80% (95% CI: 38-96%), respectively, using SUVmax >1.5. CONCLUSION: (18)F-FES-PET may aid identification of patients with acquired antihormone resistant breast cancer that are unlikely to benefit from oestradiol therapy.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Estradiol/análogos & derivados , Estradiol/uso terapéutico , Tomografía de Emisión de Positrones , Adulto , Anciano , Transporte Biológico , Biomarcadores de Tumor/metabolismo , Huesos/metabolismo , Neoplasias de la Mama/metabolismo , Estradiol/efectos adversos , Estradiol/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Resultado del Tratamiento
13.
Gynecol Oncol ; 138(3): 634-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26115976

RESUMEN

OBJECTIVE: Androgen receptor (AR), estrogen receptor α and ß (ERα, ERß), and progesterone receptor (PR) are potential therapeutic targets in epithelial ovarian cancer. In this study we evaluate the prognostic value of these hormone receptors in ovarian cancer patients. METHODS: In a prospective multicenter randomized controlled phase II trial 196 ovarian cancer patients were randomized to carboplatin/docetaxel±celecoxib. Of 121 patients sufficient tumor tissue was available for hormone receptor analysis. Tissue micro-arrays were stained for AR, ERα, ERß, and PR. Cluster analysis was performed to identify subgroups based on hormone receptor expression profile. Receptor expression was correlated to progression-free survival (PFS) and overall survival (OS) in uni- and multivariate analysis. RESULTS: AR, ERα, ERß, and PR were expressed in respectively 10%, 31%, 73%, and 19%. In patients with synchronous metastasis tissue available (n=69 patients), discordant receptor expression was observed in 9-32%. ERß-expression was associated with poor PFS and OS (hazard ratios 1.88 and 1.92). Clustering analysis revealed a subgroup with hormone receptor negative disease that had a favorable PFS and OS. CONCLUSION: Hormone receptors are expressed in the majority of ovarian cancer tumors and may serve as therapeutic targets. Clustering analysis can reveal subgroups with different outcome, which may prove valuable in selecting patients for endocrine therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Glandulares y Epiteliales/sangre , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Ováricas/sangre , Neoplasias Ováricas/tratamiento farmacológico , Receptores Androgénicos/sangre , Receptores de Estrógenos/sangre , Receptores de Progesterona/sangre , Adulto , Anciano , Anciano de 80 o más Años , Carboplatino/administración & dosificación , Carcinoma Epitelial de Ovario , Celecoxib/administración & dosificación , Docetaxel , Femenino , Humanos , Persona de Mediana Edad , Pronóstico , Estudios Prospectivos , Receptores Androgénicos/biosíntesis , Receptores de Estrógenos/biosíntesis , Receptores de Progesterona/biosíntesis , Análisis de Supervivencia , Taxoides/administración & dosificación , Análisis de Matrices Tisulares
14.
Cancer Discov ; 5(1): 72-81, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25380844

RESUMEN

UNLABELLED: It is unknown whether the current dose of fulvestrant, an estrogen receptor (ER) antagonist, is sufficient for maximal ER downregulation in patients with metastatic breast cancer. We performed a feasibility study to assess ER availability before and during fulvestrant. Sixteen patients with ER-positive metastatic breast cancer underwent positron emission tomography/computed tomography (PET/CT) at baseline (scan 1), day 28 (scan 2), and day 84 (scan 3) to monitor tumor [(18)F]fluoroestradiol (FES) uptake. Incomplete reduction in ER availability was predefined as <75% decrease in median tumor FES uptake and a residual standardized uptake value (SUVmax) of ≥1.5. In total, 131 FES-positive lesions were identified (median SUVmax of 2.9; range, 1.7-6.5). The median change in patients during fulvestrant treatment was -85% at scan 2, but varied widely (-99% to +60%). Fulvestrant reduced tumor FES uptake incompletely at scan 2 in 6 (38%) of the 16 patients, which was associated with early progression. SIGNIFICANCE: Serial imaging of tumor estrogen uptake by FES-PET can give insight into the dose needed for ER antagonists to completely abolish ER. FES-PET showed significant residual ER availability in tumors during fulvestrant therapy in 38% of patients, which was associated with early progression.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Estradiol/análogos & derivados , Tomografía de Emisión de Positrones , Receptores de Estrógenos/antagonistas & inhibidores , Tomografía Computarizada por Rayos X , Anciano , Antineoplásicos Hormonales/administración & dosificación , Antineoplásicos Hormonales/farmacocinética , Neoplasias de la Mama/patología , Progresión de la Enfermedad , Estradiol/administración & dosificación , Estradiol/farmacocinética , Estradiol/uso terapéutico , Femenino , Fulvestrant , Humanos , Persona de Mediana Edad , Metástasis de la Neoplasia , Tamoxifeno/administración & dosificación , Tamoxifeno/uso terapéutico , Resultado del Tratamiento
15.
J Nucl Med ; 56(1): 50-5, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25476534

RESUMEN

UNLABELLED: The estrogen receptor α (ERα) is expressed in approximately 70% of ovarian cancer tumors. PET of tumor ERα expression with the tracer 16α-(18)F-fluoro-17ß-estradiol ((18)F-FES) may be valuable to select ovarian cancer patients for endocrine therapy. The aim of this study was to evaluate the feasibility of (18)F-FES PET to determine tumor ERα expression noninvasively in epithelial ovarian cancer patients. METHODS: (18)F-FES PET/CT was performed shortly before cytoreductive surgery. Tumor (18)F-FES uptake was quantified for all lesions 10 mm or greater on CT and expressed as maximum standardized uptake value. (18)F-FES PET/CT findings were compared with histology and immunohistochemistry for ERα, ERß, and progesterone receptor. Receptor expression was scored semiquantitatively using H-scores (percentage of positive tumor cells × staining intensity). The optimum threshold to discriminate ER-positive and -negative lesions was determined by receiver-operating-characteristic analysis. RESULTS: In the 15 included patients with suspected ovarian cancer, 32 measurable lesions greater than 10 mm were present on CT. Tumor (18)F-FES uptake could be quantified for 28 lesions (88%), and 4 lesions were visible but nonquantifiable because of high uptake in adjacent tissue. During surgery, histology was obtained of 23 of 28 quantified lesions (82%). Quantitative (18)F-FES uptake correlated with the semiquantitative immunoscore for ERα (ρ = 0.65, P < 0.01) and weakly with progesterone receptor expression (ρ = 0.46, P = 0.03) and was not associated with ERß expression (ρ = 0.21, P = 0.33). The optimum threshold to discriminate ERα-positive and ERα-negative lesions was a maximum standardized uptake value greater than 1.8, which provided a 79% sensitivity, 100% specificity, and area under the curve of 0.86 (95% confidence interval, 0.70-1.00). In 2 of 7 patients with cytology/histology available at primary diagnosis and at debulking surgery, immunohistochemical ERα expression had changed over time. (18)F-FES PET was in accordance with histology at debulking surgery but not at primary diagnosis, indicating that (18)F-FES PET could provide reliable information about current tumor ERα status. CONCLUSION: (18)F-FES PET/CT can reliably assess ERα status in epithelial ovarian cancer tumors and metastases noninvasively. Evaluation of the predictive value of (18)F-FES PET/CT for endocrine therapy in epithelial ovarian cancer patients is warranted.


Asunto(s)
Estradiol/análogos & derivados , Regulación Neoplásica de la Expresión Génica , Neoplasias Glandulares y Epiteliales/diagnóstico , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/metabolismo , Tomografía de Emisión de Positrones , Receptores de Estrógenos/metabolismo , Tomografía Computarizada por Rayos X , Anciano , Anciano de 80 o más Años , Carcinoma Epitelial de Ovario , Estudios de Factibilidad , Femenino , Humanos , Persona de Mediana Edad , Imagen Multimodal , Neoplasias Glandulares y Epiteliales/sangre , Neoplasias Glandulares y Epiteliales/patología , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Globulina de Unión a Hormona Sexual/metabolismo
16.
Breast Cancer Res ; 16(4): 412, 2014 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-25410383

RESUMEN

DNA methylation and histone modifications are important epigenetic modifications associated with gene (dys)regulation. The epigenetic modifications are balanced by epigenetic enzymes, so-called writers and erasers, such as DNA (de)methylases and histone (de)acetylases. Aberrant epigenetic alterations have been associated with various diseases, including breast cancer. Since aberrant epigenetic modifications are potentially reversible, they might represent targets for breast cancer therapy. Indeed, several drugs have been designed to inhibit epigenetic enzymes (epi-drugs), thereby reversing epigenetic modifications. US Food and Drug Administration approval has been obtained for some epi-drugs for hematological malignancies. However, these drugs have had very modest anti-tumor efficacy in phase I and II clinical trials in breast cancer patients as monotherapy. Therefore, current clinical trials focus on the combination of epi-drugs with other therapies to enhance or restore the sensitivity to such therapies. This approach has yielded some promising results in early phase II trials. The disadvantage of epi-drugs, however, is genome-wide effects, which may cause unwanted upregulation of, for example, pro-metastatic genes. Development of gene-targeted epigenetic modifications (epigenetic editing) in breast cancer can provide a novel approach to prevent such unwanted events. In this context, identification of crucial epigenetic modifications regulating key genes in breast cancer is of critical importance. In this review, we first describe aberrant DNA methylation and histone modifications as two important classes of epigenetic mutations in breast cancer. Then we focus on the preclinical and clinical epigenetic-based therapies currently being explored for breast cancer. Finally, we describe epigenetic editing as a promising new approach for possible applications towards more targeted breast cancer treatment.


Asunto(s)
Neoplasias de la Mama/genética , Epigénesis Genética , Epigenómica , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Ensayos Clínicos como Asunto , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Histona Desacetilasas/uso terapéutico , Histonas/metabolismo , Humanos , Resultado del Tratamiento
18.
Lancet Oncol ; 14(11): e465-e475, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24079874

RESUMEN

Oestrogen receptors are overexpressed in around 70% of all breast cancers, and are a target for endocrine therapy. These receptors can be visualised on PET with use of 16α-[(18)F]-fluoro-17ß-oestradiol ((18)F-FES) as a tracer. Compared with biopsy, which enables assessment of individual sites, whole-body (18)F-FES-PET enables quantification of oestrogen-receptor expression in all metastases. In several studies, measurement of tumour protein expression in oestrogen receptors by (18)F-FES-PET, concurrent with biopsy, detected oestrogen-receptor-positive tumour lesions with a sensitivity of 84% and specificity of 98%. Roughly 45% of patients with metastatic breast cancer have discordant oestrogen-receptor expression across lesions (ie, (18)F-FES-positive and (18)F-FES-negative metastases). Low tumour (18)F-FES uptake in metastases can predict failure of hormonal therapy in patients with oestrogen-receptor-positive primary tumours. Finally, (18)F-FES-PET has shown that oestrogen-receptor binding capacity changes after intervention with hormonal drugs, but findings need to be confirmed. Factors other than oestrogen-receptor expression, including menopausal status and concomitant therapies, that can affect tumour (18)F-FES uptake must be taken into account.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Tomografía de Emisión de Positrones , Receptores de Estrógenos/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos
19.
Eur J Cancer ; 49(18): 3850-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24007820

RESUMEN

Although the majority of endometrial stromal sarcomas (ESSs) express oestrogen receptor (ER), data on the efficacy of ER-targeted therapies are scarce. Using PubMed search engine we identified nine case reports and small series in a total of 25 patients reporting on the efficacy of palliative ER-targeted therapies. Literature supports the efficacy of aromatase inhibitors after the failure of progestins, but not of the partial ER-antagonist tamoxifen. Fulvestrant is a pure ER-antagonist with a distinct mechanism, of which efficacy has not yet been reported in ESS. We present a patient that underwent positron emission tomography and computed tomography (PET/CT) of ER-expression with the tracer (18)F-fluoroestradiol (FES). High levels of ER-expression provided a rationale for fulvestrant therapy. FES-PET/CT was repeated after 6 months and indicated a strong decrease in tumour FES-uptake, and 15% reduction in tumour diameters according to Response Evaluation Criteria in Solid Tumours (RECIST) criteria.


Asunto(s)
Neoplasias Endometriales/metabolismo , Tomografía de Emisión de Positrones/métodos , Receptores de Estrógenos/metabolismo , Sarcoma Estromático Endometrial/metabolismo , Adulto , Neoplasias Endometriales/diagnóstico por imagen , Neoplasias Endometriales/tratamiento farmacológico , Estradiol/análogos & derivados , Estradiol/uso terapéutico , Antagonistas de Estrógenos/uso terapéutico , Femenino , Fulvestrant , Humanos , Receptores de Estrógenos/antagonistas & inhibidores , Sarcoma Estromático Endometrial/diagnóstico por imagen , Sarcoma Estromático Endometrial/tratamiento farmacológico , Factores de Tiempo , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos
20.
Eur J Pharmacol ; 717(1-3): 2-11, 2013 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-23545359

RESUMEN

Currently, tumour response following drug treatment is based on measurement of anatomical size changes. This is often done according to Response Evaluation Criteria in Solid Tumours (RECIST) and is generally performed every 2-3 cycles. Bone metastases, being the most common site of distant metastases in breast cancer, are not measurable by RECIST. The standard response measurement provides no insight in changes of molecular characteristics. In the era of targeted medicine, knowledge of specific molecular tumour characteristics becomes more important. A potential way to assess this is by means of molecular imaging. Molecular imaging can visualise general tumour processes, such as glucose metabolism with (18)F-fluorodeoxyglucose ((18)F-FDG) and DNA synthesis with (18)F-fluorodeoxythymidine ((18)F-FLT). In addition, an increasing number of more specific targets, such as hormone receptors, growth factor receptors, and growth factors can be visualised. In the future molecular imaging may thus be of value for personalised treatment-selection by providing insight in the expression of these drug targets. Additionally, when molecular changes can be detected early during therapy, this may serve as early predictor of response. However, in order to define clinical utility of this approach results from (ongoing) clinical trials is required. In this review we summarise the potential role of molecular imaging of general tumour processes as well as hormone receptors, growth factor receptors, and tumour micro-environment for predicting and monitoring treatment response in breast cancer patients.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/terapia , Imagen Molecular/métodos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Humanos , Receptores de Factores de Crecimiento/metabolismo , Receptores de Esteroides/metabolismo , Resultado del Tratamiento , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...