Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Eur J Cancer ; 177: 25-29, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36323049

RESUMEN

INTRODUCTION: Regulatory decisions on paediatric investigation plans (PIPs) aim at making effective and safe medicines timely available for children with high unmet medical need. At the same time, scientific knowledge progresses continuously leading frequently to the identification of new molecular targets in the therapeutic area of oncology. This, together with further efforts to optimise next generation medicines, results in novel innovative products in development pipelines. In the context of global regulatory development requirements for these growing pipelines of innovative products (e.g. US RACE for children Act), it is an increasing challenge to complete development efforts in paediatric oncology, a therapeutic area of rare and life-threatening diseases with high unmet needs. OBJECTIVE: Regulators recognise feasibility challenges of the regulatory obligations in this context. Here, we explain the EU regulatory decision making strategy applied to paediatric oncology, which aims fostering evidence generation to support developments based on needs and robust science. Because there is a plethora of products under development within given classes of or within cancer types, priorities need to be identified and updated as evidence evolves. This also includes identifying the need for third or fourth generation products to secure focused and accelerated drug development. CONCLUSION: An agreed PIP, as a plan, is a living document which can be modified in light of new evidence. For this to be successful, input from the various relevant stakeholders, i.e. patients/parents, clinicians and investigators is required. To efficiently obtain this input, the EMA is co-organising with ACCELERATE oncology stakeholder engagement platform meetings.


Asunto(s)
Neoplasias , Niño , Humanos , Neoplasias/tratamiento farmacológico , Oncología Médica/métodos , Desarrollo de Medicamentos
2.
Melanoma Res ; 32(6): 460-468, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-35703270

RESUMEN

Recent results of patients with advanced melanoma treated with first-line BRAF-MEK inhibitors in clinical trials showed 5-year survival in one-third of patients with a median overall survival (OS) of more than 2 years. This study aimed to investigate these patients' real-world survival and identify the characteristics of long-term survivors. The study population consisted of patients with advanced cutaneous melanoma with a BRAF-V600 mutated tumor who were treated with first-line BRAF-MEK inhibitors between 2013 and 2017. Long-term survival was defined as a minimum OS of 2 years from start therapy. The median progression-free survival (mPFS) and median OS (mOS) of real-world patients ( n = 435) were respectively 8.0 (95% CI, 6.8-9.4) and 11.7 (95% CI, 10.3-13.5) months. Two-year survival was reached by 28% of the patients, 22% reached 3-year survival and 19% reached 4-year survival. Real-world patients often had brain metastases (41%), stage IV M1c disease (87%), ECOG PS ≥2 (21%), ≥3 organ sites (62%) and elevated LDH of ≥250 U/I (49%). Trial-eligible real-world patients had an mOS of 17.9 months. Patients surviving more than 2 years ( n = 116) more often had an ECOG PS ≤1 (83%), normal LDH (60%), no brain metastases (60%), no liver metastases (63%) and <3 organ sites (60%). Long-term survival of real-world patients treated with first-line BRAF-MEK inhibitors is significantly lower than that of trial patients, which is probably explained by poorer baseline characteristics of patients treated in daily practice. Long-term survivors generally had more favorable characteristics with regard to age, LDH level and metastatic sites, compared to patients not reaching long-term survival.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Encefálicas/secundario , Melanoma/tratamiento farmacológico , Melanoma/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología
3.
J Med Internet Res ; 24(6): e33446, 2022 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-35737449

RESUMEN

BACKGROUND: The Dutch Institute for Clinical Auditing (DICA) Medicines Program was set up in September 2018 to evaluate expensive medicine use in daily practice in terms of real-world effectiveness using only existing data sources. OBJECTIVE: The aim of this study is to describe the potential of the addition of declaration data to quality registries to provide participating centers with benchmark information about the use of medicines and outcomes among patients. METHODS: A total of 3 national population-based registries were linked to clinical and financial data from the hospital pharmacy, the Dutch diagnosis treatment combinations information system including in-hospital activities, and survival data from health care insurers. The first results of the real-world data (RWD) linkage are presented using descriptive statistics to assess patient, tumor, and treatment characteristics. Time-to-next-treatment (TTNT) and overall survival (OS) were estimated using the Kaplan-Meier method. RESULTS: A total of 21 Dutch hospitals participated in the DICA Medicines Program, which included 7412 patients with colorectal cancer, 1981 patients with metastasized colon cancer, 3860 patients with lung cancer, 1253 patients with metastasized breast cancer, and 7564 patients with rheumatic disease. The data were used for hospital benchmarking to gain insights into medication use in specific patient populations, treatment information, clinical outcomes, and costs. Detailed treatment information (duration and treatment steps) led to insights into differences between hospitals in daily clinical practices. Furthermore, exploratory analyses on clinical outcomes (TTNT and OS) were possible. CONCLUSIONS: The DICA Medicines Program shows that it is possible to gather and link RWD about medicines to 4 disease-specific population-based registries. Since these RWD became available with minimal registration burden and effort for hospitals, this method can be explored in other population-based registries to evaluate real-world efficacy.


Asunto(s)
Neoplasias , Benchmarking , Servicios de Salud , Hospitales , Humanos , Neoplasias/tratamiento farmacológico , Sistema de Registros
4.
Eur J Cancer ; 158: 234-245, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34600790

RESUMEN

BACKGROUND: Little is known about outcomes of adjuvant-treated melanoma patients beyond the clinical trial setting. Since 2019, adjuvant-treated melanoma patients have been registered in the DMTR, a population-based registry to monitor the quality and safety of melanoma care in the Netherlands. This study aims to describe treatment patterns, relapse, and toxicity rates of adjuvant-treated melanoma patients beyond the clinical trial setting. METHODS: Analyses were performed on adjuvant-treated melanoma patients included in the DMTR. Descriptive statistics were used to analyse patient-, and treatment characteristics. A baseline registration completeness analysis was performed, and an analysis on trial eligibility in clinical practice patients. Recurrence-free survival (RFS) at 12-months was estimated with the Kaplan-Meier method. RESULTS: A total of 641 patients were treated with adjuvant anti-PD-1 therapy. RFS at 12-months was 70.6% (95% CI, 66.9-74.6) with a median follow-up of 12.8 months. Sex, stage of disease and Breslow thickness were associated with a higher hazard for RFS. Eighteen per cent of the anti-PD-1-treated patients developed grade ≥3 toxicity. Sixty-one per cent of patients prematurely discontinued anti-PD-1 therapy. CONCLUSION: Adjuvant anti-PD-1 treatment of resected stage III/IV melanoma in daily practice showed slightly higher toxicity rates and more frequent premature discontinuation but similar RFS rates compared to trials.


Asunto(s)
Adyuvantes Inmunológicos/uso terapéutico , Antineoplásicos/uso terapéutico , Melanoma/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Femenino , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias/métodos , Países Bajos , Estudios Prospectivos , Adulto Joven
5.
Melanoma Res ; 31(1): 58-66, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33351553

RESUMEN

Postapproval trials and patient registries have their pros and cons in the generation of postapproval data. No direct comparison between clinical outcomes of these data sources currently exists for advanced melanoma patients. We aimed to investigate whether a patient registry can complement or even replace postapproval trials. Postapproval single-arm clinical trial data from the Medicines Evaluation Board and real-world data from the Dutch Melanoma Treatment Registry were used. The study population consisted of advanced melanoma patients with brain metastases treated with targeted therapies (BRAF- or BRAF-MEK inhibitors) in the first line. A Cox hazard regression model and a propensity score matching (PSM) model were used to compare the two patient populations. Compared to patients treated in postapproval trials (n = 467), real-world patients (n = 602) had significantly higher age, higher ECOG performance status, more often ≥3 organ involvement and more symptomatic brain metastases. Lactate dehydrogenase levels were similar between both groups. The unadjusted median overall survival (mOS) in postapproval clinical trial patients was 8.7 (95% CI, 8.1-10.4) months compared to 7.2 (95% CI, 6.5-7.7) months (P < 0.01) in real-world patients. With the Cox hazard regression model, survival was adjusted for prognostic factors, which led to a statistically insignificant difference in mOS for trial and real-world patients of 8.7 (95% CI, 7.9-10.4) months compared to 7.3 (95% CI, 6.3-7.9) months, respectively. The PSM model resulted in 310 matched patients with similar survival (P = 0.9). Clinical outcomes of both data sources were similar. Registries could be a complementary data source to postapproval clinical trials to establish information on clinical outcomes in specific subpopulations.


Asunto(s)
Neoplasias Encefálicas/secundario , Melanoma/complicaciones , Neoplasias Cutáneas/complicaciones , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Metástasis de la Neoplasia , Sistema de Registros , Neoplasias Cutáneas/patología , Adulto Joven
6.
Clin Pharmacol Ther ; 108(3): 553-556, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32559312

RESUMEN

Timely and successful drug development for rare cancer populations, such as pediatric oncology, requires consolidated efforts in the spirit of shared responsibility. In order to advance tailored development efforts, the concept of multistakeholder Strategy Forum involving industry, academia, patient organizations, and regulators has been developed. In this study, we review the first five pediatric oncology Strategy Forums co-organized by the European Medicines Agency between 2017 and 2020, reflecting on the outcomes and the evolution of the concept over time and providing an outline of how a "safe space" for multistakeholder engagement facilitated by regulators could be of potential value beyond pediatric oncology drug development.


Asunto(s)
Antineoplásicos/uso terapéutico , Toma de Decisiones , Aprobación de Drogas , Prioridades en Salud , Participación de los Interesados , Factores de Edad , Antineoplásicos/efectos adversos , Europa (Continente) , Agencias Gubernamentales , Necesidades y Demandas de Servicios de Salud , Humanos , Evaluación de Necesidades , Seguridad del Paciente , Formulación de Políticas , Medición de Riesgo
7.
Int J Cancer ; 147(12): 3461-3470, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-32559817

RESUMEN

The aim was to provide evidence on systemically treated patients with advanced melanoma not represented in phase III trials to support clinical decision-making. Analysis were performed on advanced melanoma patients diagnosed between 2014 and 2017 in the Netherlands, treated with immune- or targeted therapy, who met ≥1 trial exclusion criteria. These criteria were derived from the KEYNOTE-006 and CHECKMATE-067/-066 phase III trials. Prognostic importance of factors associated with overall survival (OS) was assessed with the Kaplan-Meier method, Cox models, predicted OS probabilities of prognostic subgroups and a conditional inference survival tree (CIST). A nationwide population-based registry was used as data source. Of 2536 systemically treated patients with advanced melanoma, 1004 (40%) patients were ineligible for phase IIII trials. Ineligible patients had a poorer median OS (mOS) compared to eligible patients (8.8 vs 23 months). Eligibility criteria strongly associated with OS in systemically treated ineligible patients were Eastern Cooperative Oncology Group Performance Score (ECOG PS) ≥2, brain metastases (BM) and lactate dehydrogenase (LDH) of >500 U/L. Patients with ECOG PS of ≥2 with or without symptomatic BM had a predicted mOS of 6.5 and 11.3 months and a 3-year survival probability of 9.3% and 23.6%, respectively. The CIST showed the strongest prognostic covariate for survival was LDH, followed by ECOG PS. The prognosis of patients with LDH of >500 U/L is poor, but long-term survival is possible. The prognosis of ineligible patients with advanced melanoma in real-world was very heterogeneous and highly dependent on LDH value, ECOG PS and symptomatic BM.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/patología , Terapia Molecular Dirigida/métodos , Adulto , Anciano , Anciano de 80 o más Años , Toma de Decisiones Clínicas , Ensayos Clínicos Fase III como Asunto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Países Bajos , Selección de Paciente , Pronóstico , Modelos de Riesgos Proporcionales , Estudios Prospectivos , Análisis de Supervivencia , Resultado del Tratamiento , Adulto Joven
8.
Eur J Cancer ; 48(2): 237-42, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22030452

RESUMEN

On 13 July 2011 the European Commission issued a marketing authorisation valid throughout the European Union (EU) for ipilimumab for the treatment of advanced (unresectable or metastatic) melanoma in adults who have received prior therapy. Ipilimumab is a monoclonal antibody that specifically blocks the inhibitory signal of cytotoxic T lymphocyte antigen 4 (CTLA-4), resulting in T cell activation, proliferation and lymphocyte infiltration into tumours, leading to tumour cell death. The recommended induction regimen of ipilimumab is 3mg/kg administered intravenously over a 90 min period every 3 weeks for a total of four doses. In a phase 3 trial in patients with advanced melanoma, median overall survival for ipilimumab was 10 months versus 6 months for gp100, an experimental melanoma vaccine (Hazard ratio (HR) 0.66; 95% confidence interval (CI): 0.51, 0.87; p = 0.0026). Ipilimumab was most commonly associated with adverse reactions resulting from increased or excessive immune activity. Most of these, including severe reactions, resolved following initiation of appropriate medical therapy or withdrawal of ipilimumab. The most common side-effects (affecting more than 10% of patients) were diarrhoea, rash, pruritus, fatigue, nausea, vomiting, decreased appetite and abdominal pain. The objective of this paper is to summarise the scientific review of the application leading to approval in the EU. The detailed scientific assessment report and product information, including the summary of product characteristics (SmPC), are available on the European Medicines Agency (EMA) website (www.ema.europa.eu).


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Factores Inmunológicos/uso terapéutico , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Unión Europea , Humanos , Ipilimumab , Melanoma/inmunología , Melanoma/secundario , Neoplasias Cutáneas/inmunología
9.
Cancer Genet Cytogenet ; 153(1): 77-80, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15325100

RESUMEN

We summarize and briefly discuss recent findings with respect to the amplification and overexpression of candidate oncogenes in 17p11.2 ~p12 in high-grade osteosarcomas. Amplification of this region occurs in about 25% of cases. The amplification profiles are often complex and suggest the involvement of more than one oncogene. The 17p11.2 ~ p12 region harbors many low-copy repeats (LCRs). We propose LCR-mediated repeated duplication by mitotic nonallelic homologous recombination as mechanism for the generation of the amplifications in this region. Genes PMP22 and COPS3 and three expressed sequence tags from within 17p11.2 ~ p12 have been found to be frequently overexpressed and consistently overexpressed after amplification, which identifies them as candidate oncogenes in this region. Overexpression of COPS3 has been linked to TP53 protein degradation and, being equivalent to TP53 mutation, the induction of genomic instability, which frequently occurs in high-grade osteosarcoma. These findings may serve as a framework for future work aimed to identify the causative oncogenes in 17p11.2 ~p12, to clarify the mechanism of their amplification, and to determine their importance in osteosarcoma tumorigenesis.


Asunto(s)
Neoplasias Óseas/genética , Cromosomas Humanos Par 17/genética , Regulación Neoplásica de la Expresión Génica , Osteosarcoma/genética , Complejo del Señalosoma COP9 , Transformación Celular Neoplásica/genética , ADN de Neoplasias/genética , Etiquetas de Secuencia Expresada , Amplificación de Genes , Perfilación de la Expresión Génica , Humanos , Modelos Genéticos , Proteínas de la Mielina/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Hibridación de Ácido Nucleico , Oncogenes , Proteínas Proto-Oncogénicas/genética , Proteína p53 Supresora de Tumor/metabolismo
10.
Cancer Genet Cytogenet ; 152(2): 113-8, 2004 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-15262428

RESUMEN

The peripheral myelin protein (PMP22) gene is highly expressed in peripheral Schwann cells and encodes an important constituent of the myelin sheath. It is also expressed at lower levels in other normal tissues in which the protein is supposed to be involved in cell growth regulation. We recently reported frequent amplification and overexpression of PMP22 in high-grade osteosarcoma. Here, we analyzed PMP22 expression in five osteosarcoma tumors and three osteosarcoma cell lines. In normal Schwann cells, transcription of PMP22 starts at three promoters, P1A, P1B, and P2, which results in the synthesis of three alternatively spliced transcripts that all code for the same protein. We found a comparable expression pattern in normal osteoblasts. However, promoter P1A-driven transcripts were absent in all investigated tumors and cell lines and, compared to normal osteoblasts, the P1B/P2 transcript ratio was found to be increased in two of three cases with PMP22 overexpression and decreased in all five cases without overexpression. In normal Schwann cells and in NIH3T3 cells, PMP22 expression increases upon serum starvation-induced growth arrest. In contrast to this, serum withdrawal caused a considerable decrease of PMP22 expression in the osteosarcoma cell lines. We conclude that the different PMP22 expression in osteosarcoma may result in alternative availability of the PMP22 protein during the cell cycle and aberrant regulation of cell growth control in osteosarcoma tumorigenesis.


Asunto(s)
Neoplasias Óseas/genética , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de la Mielina/genética , Osteosarcoma/genética , Empalme Alternativo , Animales , Apoptosis , Ciclo Celular , Humanos , Ratones , Células 3T3 NIH/citología , Células 3T3 NIH/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Regiones Promotoras Genéticas , ARN Mensajero/genética , Células de Schwann/citología , Células de Schwann/metabolismo , Suero/metabolismo , Inanición , Transcripción Genética , Células Tumorales Cultivadas
11.
Cancer Genet Cytogenet ; 152(1): 8-14, 2004 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-15193436

RESUMEN

Osteosarcomas are malignant tumors of the bone that are characterized by complex genetic changes, including loss and amplification of chromosome regions. Region 17p11.2 approximately p12 is frequently found to be amplified in this tumor, suggesting the presence of an oncogene (or oncogenes) important in osteosarcoma tumorigenesis. We had previously determined amplification profiles for this region. Reasoning that amplification of a causative oncogene in a tumor should result in increased expression of that gene, we have now determined the expression status of genes and expressed sequence tags (ESTs) in 17p11.2 approximately p12. We constructed a 17p11.2 approximately p12-specific macroarray containing 40 genes and 21 ESTs from this region, which was used for expression profiling of 11 osteosarcoma samples (9 tumors and 2 cell lines) and of normal human osteoblasts. Compared to normal osteoblasts, genes with at least threefold increased expression were considered to be overexpressed in the tumor. Genes PMP22 and COPS3, EST AA126939 (encoding part of the hypothetical protein FLJ20343), and two anonymous ESTs (AA918483 and R02360) were found to be most consistently overexpressed after amplification. By real-time reverse transcriptase polymerase chain reaction, we could confirm the overexpression status of PMP22 and COPS3 but not of FLJ20343. We conclude that PMP22 and COPS3, and possibly also the three ESTs, are candidate amplification targets in 17p11.2 approximately p12 in osteosarcoma.


Asunto(s)
Neoplasias Óseas/genética , Cromosomas Humanos Par 17/genética , Amplificación de Genes , Osteoblastos/metabolismo , Osteosarcoma/genética , Adulto , Neoplasias Óseas/patología , Complejo del Señalosoma COP9 , Mapeo Cromosómico , Humanos , Proteínas de la Membrana/genética , Microcuerpos , Osteosarcoma/patología , Proteínas Proto-Oncogénicas/genética
12.
Cancer Genet Cytogenet ; 140(2): 162-6, 2003 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-12645656

RESUMEN

We reported previously the amplification of DNA markers in 17p12 in 3 of 60 high-grade gliomas. To detect additional cases, we screened in total 104 gliomas of various types and grades by Southern blot analysis using marker 745R, which is within the commonly amplified region. However, no other caseswith significant amplification (amplification level > 4) were found. To investigate in detail the extent of the amplifications in the three tumors, which were all glioblastomas, we determined 17p11.2 approximately p12 amplification profiles by semiquantitative polymerase chain reaction using 15 microsatellite markers and seven candidate genes. Distinct and high-level amplifications, with maximum levels ranging from 15 to 38, were found in these tumors. The 0.8 Mb-region between D17S1525 and MAP2K4 in 17p12 proved to be commonly amplified in these tumors. In one tumor, a heterogeneous distribution of the amplification in 17p12 was found, suggesting that it is a late event during glioma tumorigenesis. Another tumor showed additional high-level amplification of PMP22 and D17S1843 in 17p11.2. From the high-level amplifications we conclude that at least one, but possibly more, putative oncogenes are present in 17p11.2 approximately p12 whose amplifications and/or overexpressions contribute to glioma tumorigenesis.


Asunto(s)
Cromosomas Humanos Par 17/genética , Amplificación de Genes , Glioma/genética , Southern Blotting , Neoplasias Óseas/genética , Transformación Celular Neoplásica/genética , Aberraciones Cromosómicas , Cromosomas Humanos Par 17/ultraestructura , ADN de Neoplasias/genética , Humanos , Repeticiones de Microsatélite , Osteosarcoma/genética , Reacción en Cadena de la Polimerasa
13.
Cancer Genet Cytogenet ; 139(2): 91-6, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12550767

RESUMEN

Amplification of region 17p11.2 approximately p12 has been found in 13%-29% of high-grade osteosarcomas, suggesting the presence of an oncogene or oncogenes that may contribute to their development. To determine the location of these putative oncogenes, we established 17p11.2 approximately p12 amplification profiles by semiquantitative PCR, using 15 microsatellite markers and seven candidate genes in 19 high-grade osteosarcomas. Most of the tumors displayed complex amplification profiles, with frequent involvement of marker D17S2041 in 17p12 and TOP3A in 17p11.2 and, in some cases, very high-level amplification of PMP22 and MAPK7 in 17p11.2. Our findings suggest that multiple amplification targets, including PMP22, TOP3A, and MAPK7 or genes close to these candidate oncogenes, may be present in 17p11.2 approximately p12 and thus contribute to osteosarcoma tumorigenesis.


Asunto(s)
Neoplasias Óseas/genética , Cromosomas Humanos Par 17/ultraestructura , ADN-Topoisomerasas de Tipo I/genética , Amplificación de Genes , Proteínas de la Membrana/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Oncogenes , Osteosarcoma/genética , Neoplasias Óseas/patología , Mapeo Cromosómico , Cromosomas Humanos Par 17/genética , ADN de Neoplasias/genética , Marcadores Genéticos , Humanos , Repeticiones de Microsatélite , Proteína Quinasa 7 Activada por Mitógenos , Osteosarcoma/patología , Reacción en Cadena de la Polimerasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...