Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Dysphagia ; 38(2): 711-718, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35972695

RESUMEN

The need for multidisciplinary and multiprofessional management of dysphagia is constantly increasing and creating a major challenge for healthcare professionals and society, especially in terms of professional expertise and human resources. The distribution of tasks among the dysphagia team members, which includes phoniatricians, otolaryngologists, and speech-language therapists, is flexible and overlapping. For assessing dysphagia, the (fibreoptic) flexible endoscopic evaluation of swallowing (FEES), with or without videofluoroscopy, is a pivotal diagnostic tool. This position paper aims to illustrate the phoniatrician's role in performing a FEES, which is an indispensable component of the diagnostic workup of patients suffering from oropharyngeal dysphagia. It is based on the current collaborative expert view of the Swallowing Committee of the Union of European Phoniatricians and a literature review. A FEES is one of the core competences of phoniatricians due to their endoscopic expertise and experience in the field of dysphagia and diseases of the upper aerodigestive tract. Therefore, the phoniatrician is an important member of the dysphagia team, for the medical diagnostics of the aerodigestive tract and dysphagia as well as for FEES. Phoniatric competence is especially important for head and neck cancer patients, infants, and complex cases.


Asunto(s)
Trastornos de Deglución , Lactante , Humanos , Trastornos de Deglución/diagnóstico , Deglución , Endoscopía , Tecnología de Fibra Óptica , Personal de Salud
2.
Artículo en Alemán | MEDLINE | ID: mdl-37843595

RESUMEN

BACKGROUND: Newborn hearing screening (NHS) was introduced nationwide by the Federal Joint Committee (Gemeinsamer Bundesausschuss, G­BA) in 2009. In this process, quality targets were also set in the pediatrics directive. In order to review the quality NHS in Germany, the G­BA commissioned a consortium to conduct an initial evaluation for the years 2011 and 2012 and a follow-up evaluation for 2017 and 2018. METHODS: The evaluations were based on NHS screening parameters (Sammelstatistiken) that must be documented by all obstetrics and neonatology departments as NHS providers and can also be compiled through cooperation with hearing screening centers (HSCs). Additional data were collected through questionnaires and interviews and routine data were used to evaluate the screening process. RESULTS: In 13 federal states, a total of 15 HSCs are involved in the screening process. Across Germany, an NHS screening rate of 86.1% was documented in 2018 (82.4% in 2012), but this differed significantly between the federal states. The specified quality targets could not yet be implemented everywhere. For example, only less than half of the obstetric departments achieved the specified screening rate of over 95%. A comparison of data from the follow-up evaluation and the first evaluation showed that the structural quality of NHS had improved, while the process quality remained the same or had deteriorated. The refer rate (children who were discharged without passing the screening) increased from 5.3% to 6.0%. DISCUSSION: To improve the quality of NHS, HSCs should be established nationwide and a second screening should be carried out more consistently before discharge in the case of a refer result in the initial screening.


Asunto(s)
Audición , Tamizaje Neonatal , Recién Nacido , Humanos , Niño , Alemania , Tamizaje Neonatal/métodos
3.
Pediatr Blood Cancer ; 69(9): e29755, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35723448

RESUMEN

BACKGROUND: Hearing loss is a potential side effect from childhood cancer treatment. We described the severity of hearing loss assessed by audiometry in a representative national cohort of childhood cancer survivors (CCS) and identified clinical risk factors. PROCEDURE: We included all CCS from the Swiss Childhood Cancer Registry who were diagnosed ≤18 age and treated with platinum-based chemotherapy between 1990 and 2014. We extracted audiograms, treatment-related information, and demographic data from medical records. Two reviewers independently assessed the severity of hearing loss at latest follow-up using the Münster Ototoxicity Scale. We used ordered logistic regression to identify clinical risk factors for severity of hearing loss. RESULTS: We analyzed data from 270 CCS. Median time from cancer diagnosis to last audiogram was 5 years (interquartile range 2.5-8.1 years). We found 53 (20%) CCS with mild, 78 (29%) with moderate, and 75 (28%) with severe hearing loss. Higher severity grades were associated with (a) younger age at cancer diagnosis (odds ratio [OR] 5.4, 95% confidence interval [CI]: 2.5-12.0 for <5 years); (b) treatment in earlier years (OR 4.8, 95% CI: 2.1-11.0 for 1990-1995); (c) higher cumulative cisplatin doses (OR 13.5, 95% CI: 4.7-38.8 for >450 mg/m2 ); (d) concomitant cranial radiation therapy (CRT) (OR 4.4, 95% CI: 2.5-7.8); and (e) hematopoietic stem cell transplantation (HSCT) (OR 2.7, 95% CI: 1.0-7.2). CONCLUSION: Three of four CCS treated with platinum-based chemotherapy experienced some degree of hearing loss. We recommend closely monitoring patient's hearing function if treated at a young age with high cumulative cisplatin doses, and concomitant CRT as part of long-term care.


Asunto(s)
Antineoplásicos , Supervivientes de Cáncer , Pérdida Auditiva , Neoplasias , Antineoplásicos/efectos adversos , Carboplatino , Niño , Cisplatino , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/epidemiología , Humanos , Neoplasias/terapia , Platino (Metal)/uso terapéutico
4.
Gesundheitswesen ; 84(2): 117-125, 2022 Feb.
Artículo en Alemán | MEDLINE | ID: mdl-33951738

RESUMEN

BACKGROUND: The aim of the newborn hearing screening (NHS) is to identify and treat children with bilateral hearing disorders early. The NHS is regulated in Germany by the Pediatric Directive, which recommends an evaluation after 5 years. This evaluation was performed for the first time nationwide for children born between 2011 and 2012 regarding structural, process and result quality. OBJECTIVES: Challenges in the collection of appropriate data as basis for evaluation are described and possible improvements are suggested. METHODS: All maternity and neonatology wards performing the NHS were identified and their documentations of the NHS analysed. In addition, all pediatric audiologists were identified to gather data on children with bilateral permanent congenital hearing disorder. RESULTS: The identification of relevant maternity and neonatology wards was very burdensome. More than half of them were not aware that NHS had to be documented. There was no documentation on more than 15% of the children that were to be screened. Furthermore, data concerning bilateral congenital hearing disorders was only accessible for 60% of the expected number of affected children. CONCLUSIONS: Data required for the evaluation of the NHS regarding structural, process and result quality were incomplete and missing. The database for evaluations should be defined precisely and structures needed to obtain meaningful results have to be established in advance. Nevertheless, the evaluation of the NHS provides meaningful results concerning the screening process in Germany.


Asunto(s)
Pruebas Auditivas , Tamizaje Neonatal , Niño , Femenino , Alemania/epidemiología , Audición , Trastornos de la Audición/diagnóstico , Trastornos de la Audición/epidemiología , Hospitales , Humanos , Recién Nacido , Embarazo
5.
Pharmacogenomics J ; 20(2): 294-305, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31666714

RESUMEN

Ototoxicity is a common side effect of platinum treatment and manifests as irreversible, high-frequency sensorineural hearing loss. Genetic association studies have suggested a role for SNPs in genes related to the disposition of cisplatin or deafness. In this study, 429 pediatric patients that were treated with cisplatin were genotyped for 10 candidate SNPs. Logistic regression analyses revealed that younger age at treatment (≤5 years vs >15 years: OR: 9.1; 95% CI: 3.8-21.5; P = 5.6 × 10-7) and higher cumulative dose of cisplatin (>450 vs ≤300 mg/m2: OR: 2.4; 95% CI: 1.3-4.6; P = 0.007) confer a significant risk of ototoxicity. Of the SNPs investigated, none of them were significantly associated with an increase of ototoxicity. In the meta-analysis, ACYP2 rs1872328 (OR: 3.94; 95% CI: 1.04-14.03; P = 0.04) and SLC22A2 rs316019 (OR: 1.46; 95% CI: 1.07-2.00; P = 0.02) were associated with ototoxicity. In order to increase the understanding of the association between SNPs and ototoxicity, we propose a polygenic model, which takes into account multiple interacting genes of the cisplatin pathway that together confer an increased risk of ototoxicity.


Asunto(s)
Antineoplásicos/efectos adversos , Cisplatino/efectos adversos , Estudios de Asociación Genética/métodos , Variación Genética/genética , Internacionalidad , Ototoxicidad/genética , Adolescente , Niño , Preescolar , Femenino , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/epidemiología , Pérdida Auditiva/genética , Humanos , Lactante , Recién Nacido , Masculino , Neoplasias/tratamiento farmacológico , Neoplasias/epidemiología , Neoplasias/genética , Ototoxicidad/epidemiología , Estudios Retrospectivos , Adulto Joven
6.
Lancet Oncol ; 20(1): e29-e41, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30614474

RESUMEN

Childhood, adolescent, and young adult (CAYA) cancer survivors treated with platinum-based drugs, head or brain radiotherapy, or both have an increased risk of ototoxicity (hearing loss, tinnitus, or both). To ensure optimal care and reduce consequent problems-such as speech and language, social-emotional development, and learning difficulties-for these CAYA cancer survivors, clinical practice guidelines for monitoring ototoxicity are essential. The implementation of surveillance across clinical settings is hindered by differences in definitions of hearing loss, recommendations for surveillance modalities, and remediation. To address these deficiencies, the International Guideline Harmonization Group organised an international multidisciplinary panel, including 32 experts from ten countries, to evaluate the quality of evidence for ototoxicity following platinum-based chemotherapy and head or brain radiotherapy, and formulate and harmonise ototoxicity surveillance recommendations for CAYA cancer survivors.


Asunto(s)
Antineoplásicos/efectos adversos , Supervivientes de Cáncer , Atención a la Salud/normas , Neoplasias/tratamiento farmacológico , Ototoxicidad/diagnóstico , Ototoxicidad/prevención & control , Adolescente , Antineoplásicos/uso terapéutico , Supervivientes de Cáncer/estadística & datos numéricos , Niño , Irradiación Craneana/efectos adversos , Medicina Basada en la Evidencia , Humanos , Neoplasias/radioterapia , Ototoxicidad/etiología , Ototoxicidad/terapia , Compuestos de Platino/efectos adversos , Vigilancia de la Población , Adulto Joven
7.
Strahlenther Onkol ; 193(11): 910-920, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28887665

RESUMEN

PURPOSE: To analyze the incidence and degree of sensorineural hearing loss (SNHL) resulting from different radiation techniques, fractionation dose, mean cochlear radiation dose (Dmean), and total cisplatin dose. MATERIAL AND METHODS: In all, 29 children with medulloblastoma (58 ears) with subclinical pretreatment hearing thresholds participated. Radiotherapy (RT) and cisplatin had been applied sequentially according to the HIT MED Guidance. Audiological outcomes up to the latest follow-up (median 2.6 years) were compared. RESULTS: Bilateral high-frequency SNHL was observed in 26 patients (90%). No significant differences were found in mean hearing threshold between left and right ears at any frequency. A significantly better audiological outcome (p < 0.05) was found after tomotherapy at the 6 kHz bone-conduction threshold (BCT) and left-sided 8 kHz air-conduction threshold (ACT) than after a combined radiotherapy technique (CT). Fraction dose was not found to have any impact on the incidence, degree, and time-to-onset of SNHL. Patients treated with CT had a greater risk of SNHL at high frequencies than tomotherapy patients even though Dmean was similar. Increase in severity of SNHL was seen when the total cisplatin dose reached above 210 mg/m2, with the highest abnormal level found 8-12 months after RT regardless of radiation technique or fraction dose. CONCLUSION: The cochlear radiation dose should be kept as low as possible in patients who receive simultaneous cisplatin-based chemotherapy. The risk of clinically relevant HL was shown when Dmean exceeds 45 Gy independent of radiation technique or radiation regime. Cisplatin ototoxicity was shown to have a dose-dependent effect on bilateral SNHL, which was more pronounced in higher frequencies.


Asunto(s)
Neoplasias Cerebelosas/terapia , Quimioradioterapia/efectos adversos , Quimioradioterapia/métodos , Cisplatino/administración & dosificación , Cisplatino/efectos adversos , Fraccionamiento de la Dosis de Radiación , Pérdida Auditiva Bilateral/etiología , Pérdida Auditiva Sensorineural/etiología , Meduloblastoma/terapia , Traumatismos por Radiación/etiología , Radioterapia de Intensidad Modulada/efectos adversos , Adolescente , Umbral Auditivo/efectos de los fármacos , Umbral Auditivo/efectos de la radiación , Niño , Preescolar , Terapia Combinada , Femenino , Humanos , Masculino , Estudios Retrospectivos
8.
Pediatr Hematol Oncol ; 34(2): 120-129, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28590156

RESUMEN

Cisplatin and carboplatin are effective antineoplastic agents. They are also considered to be potentially highly ototoxic. To date, no long-term follow-up data from well-documented cohorts with substantial numbers of childhood cancer survivors (CCS) with platinum-related hearing loss are available. Therefore, in this study, we studied the reversibility of ototoxicity from discontinuation of treatment onwards in a national cohort of platinum-treated survivors with hearing loss at the end of cancer treatment. Of the 168 CCS with follow-up audiograms, we longitudinally evaluated the course of hearing function in 61 CCS who showed hearing impairment at discontinuation of treatment according to the Münster criteria (>20 dB at ≥4-8 kHz). Survivors were treated with platinum (median total cumulative dose cisplatin: 480 mg/m2 and median total cumulative dose carboplatin: 2520 mg/m2). Median follow-up time was 5.5 years (range: 1.0-28.8 years). The results showed that none of these survivors revealed improvement of hearing function even till 28.8 years after discontinuation of treatment (grade <2b during long-term follow-up). An increase in hearing loss with two or three Münster degrees was observed in five of 61 survivors after 1.6-19.6 years. Overall, this indicates that ototoxicity after platinum treatment may be irreversible and that longitudinal clinical audiological monitoring and care is required in long-term survivors of childhood cancer on a large scale.


Asunto(s)
Supervivientes de Cáncer , Carboplatino/efectos adversos , Quimioradioterapia/efectos adversos , Cisplatino/efectos adversos , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/fisiopatología , Neoplasias/terapia , Adolescente , Adulto , Carboplatino/administración & dosificación , Quimioradioterapia/métodos , Niño , Preescolar , Cisplatino/administración & dosificación , Estudios Transversales , Femenino , Pérdida Auditiva/epidemiología , Humanos , Lactante , Estudios Longitudinales , Masculino , Neoplasias/epidemiología
9.
Telemed J E Health ; 23(1): 49-54, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27267769

RESUMEN

INTRODUCTION: The success of a newborn hearing screening program depends on successful tracking and follow-up to ensure that children who have had positive screening results in the first few days of life receive appropriate and timely diagnostic and intervention services. The easy availability, through a suitable infrastructure, of the data necessary for the tracking, diagnosis, and care of children concerned is a major key to enhancing the quality and efficiency of newborn hearing screening programs. MATERIALS AND METHODS: Two systems for the automated two-way transmission of newborn hearing screening and configuration data, based on mobile communication technology, for the screening devices MADSEN AccuScreen® and Natus Echo-Screen® were developed and tested in a field study. Radio modem connections were compared with conventional analogue modem transmissions from Natus Echo-Screen devices for duration, transmission rate, number of lost connections, and frequency of use. RESULTS: The average session duration was significantly lower with the MADSEN AccuScreen (12 s) and Natus Echo-Screen both with radio modem (15 s) than the Natus Echo-Screen with analogue modem (108 s). The transmission rate was significantly higher (898 and 1,758 vs. 181 bytes/s) for the devices with radio modems. Both radio modem devices had significantly lower rates of broken connections after initial connection (2.1 and 0.9 vs. 5.5%). An increase in the frequency of data transmission from the clinics with mobile radio devices was found. CONCLUSIONS: The use of mobile communication technology in newborn hearing screening devices offers improvements in the average session duration, transmission rate, and reliability of the connection over analogue solutions. We observed a behavioral change in clinical staff using the new technology: the data exchange with the tracking center is more often used. The requirements for on-site support were reduced. These savings outweigh the small increase in costs for the Internet service provider.


Asunto(s)
Pruebas Auditivas/instrumentación , Modems/instrumentación , Telemedicina/instrumentación , Pruebas Auditivas/normas , Humanos , Recién Nacido , Modems/normas , Reproducibilidad de los Resultados , Factores de Tiempo
10.
Eur Arch Otorhinolaryngol ; 273(8): 2127-31, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27071773

RESUMEN

Previous studies demonstrated that there is a significant change in speaking fundamental frequency after testosterone therapy in female-to-male gender dysphoric individuals. It is yet an open question how the satisfaction with voice alteration can be predicted because until now it is not clear whether a testosterone therapy is sufficiently effective. The aim of the current study was not only to measure satisfaction with voice, but additionally to detect factors that predict or explain satisfaction with voice after testosterone therapy. Therefore, nine female-to-male gender dysphoric individuals were examined during the first year of testosterone treatment at different points of time. The patients underwent several voice analyses within 1 year and had to fill out several questionnaires concerning their voice, depressive symptoms, quality of life and voice handicap index. Multiple regression analyses were performed to find the factors that explained satisfaction with altered voice after 1 year. The difference of voice frequency in semitones before the treatment and after 1 year is the only significant predictor for satisfaction after 1 year (B = 0.442; SE = 0.049) and more important than the absolute fundamental frequency.


Asunto(s)
Andrógenos/uso terapéutico , Satisfacción del Paciente , Acústica del Lenguaje , Testosterona/uso terapéutico , Transexualidad , Voz/efectos de los fármacos , Adulto , Femenino , Humanos , Masculino , Calidad de Vida , Análisis de Regresión , Habla/efectos de los fármacos , Habla/fisiología , Encuestas y Cuestionarios , Transexualidad/psicología , Voz/fisiología , Calidad de la Voz
11.
Eur Arch Otorhinolaryngol ; 273(4): 959-65, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26650551

RESUMEN

Female-to-male gender dysphoric individuals rarely access medical services for voice problems arising out of hormonal treatment leading to "voice reassignment". The aim of this study was a close monitoring of voice deepening in the first year following the commencement of testosterone treatment. Voice recordings from nine female-to-male (FTM) were analyzed with Praat software and values for speaking fundamental frequency (SFF) were calculated. Audio recordings were made prior to and within the first year (mean 55.2 weeks) of testosterone treatment at a mean of 35.4 different time points. The values for speaking fundamental frequency were compared with values taken from 21 biological men with healthy voices. The 10th to 90th percentile range of FTM overlapped with those of biological men after about 36 weeks. The mean SFF change was a decrease of 8.78 seminotes at week 52 and at this point in time no significant difference between SSF in FTM and biological men was found. Testosterone treatment led to significant voice deepening within the first year with the degree of change decreasing over time. Mean SFF change in the first year was almost a sixth and thus less than one octave but nonetheless reached an SFF comparable with biological men.


Asunto(s)
Procedimientos de Reasignación de Sexo/métodos , Testosterona/administración & dosificación , Transexualidad , Calidad de la Voz/efectos de los fármacos , Adulto , Andrógenos/administración & dosificación , Femenino , Alemania , Humanos , Masculino , Espectrografía del Sonido/métodos , Transexualidad/diagnóstico , Transexualidad/fisiopatología , Resultado del Tratamiento
12.
Front Genet ; 14: 1214736, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37671045

RESUMEN

Genetic heterogeneity makes it difficult to identify the causal genes for hearing loss. Studies from previous decades have mapped numerous genetic loci, providing critical supporting evidence for gene discovery studies. Despite widespread sequencing accessibility, many historically mapped loci remain without a causal gene. The DFNA33 locus was mapped in 2009 and coincidentally contains ATP11A, a gene recently associated with autosomal dominant hearing loss and auditory neuropathy type 2. In a rare opportunity, we genome-sequenced a member of the original family to determine whether the DFNA33 locus may also be assigned to ATP11A. We identified a deep intronic variant in ATP11A that showed evidence of functionally normal splicing. Furthermore, we re-assessed haplotypes from the originally published DFNA33 family and identified two double recombination events and one triple recombination event in the pedigree, a highly unlikely occurrence, especially at this scale. This brief research report also serves as a call to the community to revisit families who have previously been involved in gene mapping studies, provide closure, and resolve these historical loci.

13.
J Cancer Surviv ; 2023 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-37736773

RESUMEN

PURPOSE: Auditory complications are potential side effects from childhood cancer treatment. Yet, limited evidence exists about the impact of auditory complications-particularly tinnitus-on health-related quality of life (HRQoL) among childhood cancer survivors (CCS). We determined the prevalence of hearing loss and tinnitus in the European PanCareLIFE cohort of CCS and examined its effect on HRQoL. METHODS: We included CCS from four European countries who were diagnosed at age ≤ 18 years; survived ≥ 5 years; and aged 25-44 years at study. We assessed HRQoL (Short Form 36), hearing loss, and tinnitus using questionnaires. We used multivariable linear regression to examine associations between these two auditory complications and HRQoL adjusting for socio-demographic and clinical factors. RESULTS: Our study population consisted of 6,318 CCS (53% female; median age at cancer diagnosis 9 years interquartile range [IQR] 5-13 years) with median age at survey of 31 years (IQR 28-35 years). Prevalence was 7.5% (476/6,318; confidence interval [CI]: 6.9-8.2) for hearing loss and 7.6% (127/1,668; CI: 6.4-9.0) for tinnitus. CCS with hearing loss had impaired physical (coefficient [coef.] -4.3, CI: -7.0 to -1.6) and mental (coef. -3.2, CI: -5.5 to -0.8) HRQoL when compared with CCS with normal hearing. Tinnitus was associated with impaired physical (coef. -8.2, CI: -11.8 to -4.7) and mental (coef. -5.9, CI: -8.8 to -3.1) HRQoL. CONCLUSION: We observed reduced HRQoL among CCS with hearing loss and tinnitus. Our findings indicate timely treatment of hearing loss and tinnitus may contribute to quality of life of survivors. IMPLICATIONS FOR CANCER SURVIVORS: CCS who experience auditory complications should be counseled about possible therapeutic and supportive measures during follow-up care.

14.
BMJ Open ; 13(5): e070259, 2023 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-37202136

RESUMEN

INTRODUCTION: Individuals with intellectual disabilities (ID) often suffer from hearing loss, in most cases undiagnosed or inappropriately treated. The implementation of a programme of systematic hearing screening, diagnostics, therapy initiation or allocation and long-term monitoring within the living environments of individuals with ID (nurseries, schools, workshops, homes), therefore, seems beneficial. METHODS AND ANALYSIS: The study aims to assess the effectiveness and costs of a low-threshold screening programme for individuals with ID. Within this programme 1050 individuals with ID of all ages will undergo hearing screening and an immediate reference diagnosis in their living environment (outreach cohort). The recruitment of participants in the outreach group will take place within 158 institutions, for example, schools, kindergartens and places of living or work. If an individual fails the screening assessment, subsequent full audiometric diagnostics will follow and, if hearing loss is confirmed, initiation of therapy or referral to and monitoring of such therapy. A control cohort of 141 participants will receive an invitation from their health insurance provider via their family for the same procedure but within a clinic (clinical cohort). A second screening measurement will be performed with both cohorts 1 year later and the previous therapy outcome will be checked. It is hypothesised that this programme leads to a relevant reduction in the number of untreated or inadequately treated cases of hearing loss and strengthens the communication skills of the newly or better-treated individuals. Secondary outcomes include the age-dependent prevalence of hearing loss in individuals with ID, the costs associated with this programme, cost of illness before-and-after enrolment and modelling of the programme's cost-effectiveness compared with regular care. ETHICS AND DISSEMINATION: The study has been approved by the Institutional Ethics Review Board of the Medical Association of Westphalia-Lippe and the University of Münster (No. 2020-843 f-S). Participants or guardians will provide written informed consent. Findings will be disseminated through presentations, peer-reviewed journals and conferences. TRIAL REGISTRATION NUMBER: DRKS00024804.


Asunto(s)
Sordera , Pérdida Auditiva , Discapacidad Intelectual , Humanos , Pérdida Auditiva/diagnóstico , Audiometría , Investigación , Audición
15.
Am J Pathol ; 176(3): 1169-80, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20110413

RESUMEN

The use of the effective antineoplastic agent cisplatin is limited by its serious side effects, such as oto- and nephrotoxicity. Ototoxicity is a problem of special importance in children, because deafness hampers their language and psychosocial development. Recently, organic cation transporters (OCTs) were identified in vitro as cellular uptake mechanisms for cisplatin. In the present study, we investigated in an in vivo model the role of OCTs in the development of cisplatin oto- and nephrotoxicity. The functional effects of cisplatin treatment on kidney (24 hours excretion of glucose, water, and protein) and hearing (auditory brainstem response) were studied in wild-type and OCT1/2 double-knockout (KO) mice. No sign of ototoxicity and only mild nephrotoxicity were observed after cisplatin treatment of knockout mice. Comedication of wild-type mice with cisplatin and the organic cation cimetidine protected from ototoxicity and partly from nephrotoxicity. For the first time we showed that OCT2 is expressed in hair cells of the cochlea. Furthermore, cisplatin-sensitive cell lines from pediatric tumors showed no expression of mRNA for OCTs, indicating the feasibility of therapeutic approaches aimed to reduce cisplatin toxicities by competing OCT2-mediated cisplatin uptake in renal proximal tubular and cochlear hair cells. These findings are very important to establish chemotherapeutical protocols aimed to maximize the antineoplastic effect of cisplatin while reducing the risk of toxicities.


Asunto(s)
Cisplatino/toxicidad , Enfermedades del Oído/inducido químicamente , Enfermedades del Oído/metabolismo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Sustancias Protectoras/farmacología , Animales , Umbral Auditivo/efectos de los fármacos , Nitrógeno de la Urea Sanguínea , Peso Corporal/efectos de los fármacos , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Línea Celular Tumoral , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Cóclea/patología , Transportador de Cobre 1 , Enfermedades del Oído/patología , Enfermedades del Oído/fisiopatología , Glucosa/metabolismo , Humanos , Riñón/efectos de los fármacos , Riñón/patología , Enfermedades Renales/fisiopatología , Pruebas de Función Renal , Masculino , Ratones , Ratones Noqueados , Proteínas de Transporte de Catión Orgánico/genética , Transportador 2 de Cátion Orgánico , Platino (Metal)/metabolismo , Estría Vascular/efectos de los fármacos , Estría Vascular/metabolismo , Estría Vascular/patología
16.
Eur J Hum Genet ; 29(11): 1611-1624, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34140649

RESUMEN

Array technology to genotype single-nucleotide variants (SNVs) is widely used in genome-wide association studies (GWAS), clinical diagnostics, and linkage studies. Arrays have undergone a tremendous growth in both number and content over recent years making a comprehensive comparison all the more important. We have compared 28 genotyping arrays on their overall content, genome-wide coverage, imputation quality, presence of known GWAS loci, mtDNA variants and clinically relevant genes (i.e., American College of Medical Genetics (ACMG) actionable genes, pharmacogenetic genes, human leukocyte antigen (HLA) genes and SNV density). Our comparison shows that genome-wide coverage is highly correlated with the number of SNVs on the array but does not correlate with imputation quality, which is the main determinant of GWAS usability. Average imputation quality for all tested arrays was similar for European and African populations, indicating that this is not a good criterion for choosing a genotyping array. Rather, the additional content on the array, such as pharmacogenetics or HLA variants, should be the deciding factor. As the research question of a study will in large part determine which class of genes are of interest, there is not just one perfect array for all different research questions. This study can thus help as a guideline to determine which array best suits a study's requirements.


Asunto(s)
Pruebas Genéticas/normas , Técnicas de Genotipaje/normas , Análisis de Secuencia por Matrices de Oligonucleótidos/normas , Pruebas Genéticas/métodos , Estudio de Asociación del Genoma Completo/métodos , Estudio de Asociación del Genoma Completo/normas , Técnicas de Genotipaje/métodos , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Juego de Reactivos para Diagnóstico/normas , Sensibilidad y Especificidad
17.
JAMA Oncol ; 7(10): 1550-1558, 2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34383016

RESUMEN

IMPORTANCE: Ototoxicity is an irreversible direct and late effect of certain childhood cancer treatments. Audiologic surveillance during therapy as part of the supportive care pathway enables early detection of hearing loss, decision-making about ongoing cancer treatment, and, when applicable, the timely use of audiologic interventions. Pediatric oncologic clinical practice and treatment trials have tended to be driven by tumor type and tumor-specific working groups. Internationally accepted standardized recommendations for monitoring hearing during treatment have not previously been agreed on. OBJECTIVE: To provide standard recommendations on hearing loss monitoring during childhood cancer therapy for clinical practice. METHODS: An Ototoxicity Task Force was formed under the umbrella of the International Society of Paediatric Oncology, consisting of international audiologists, otolaryngologists, and leaders in the field of relevant pediatric oncology tumor groups. Consensus meetings conducted by experts were organized, aimed at providing standardized recommendations on age-directed testing, timing, and frequency of monitoring during cancer treatment based on literature and consensus. Consensus statements were prepared by the core group, adapted following several videoconferences, and finally agreed on by the expert panel. FINDINGS: The consensus reached was that children who receive ototoxic cancer treatment (platinum agents, cranial irradiation, and/or brain surgery) require a baseline case history, monitoring of their middle ear and inner ear function, and assessment of tinnitus at each audiologic follow-up. As a minimum, age-appropriate testing should be performed before and at the end of treatment. Ideally, audiometry with counseling before each cisplatin cycle should be considered in the context of the individual patient, specific disease, feasibility, and available resources. CONCLUSIONS AND RELEVANCE: This is an international multidisciplinary consensus report providing standardized supportive care recommendations on hearing monitoring in children undergoing potentially ototoxic cancer treatment. The recommendations are intended to improve the care of children with cancer and facilitate comparative research on the timing and development of hearing loss caused by different cancer treatment regimens.


Asunto(s)
Pérdida Auditiva , Neoplasias , Niño , Cisplatino/uso terapéutico , Irradiación Craneana , Pérdida Auditiva/inducido químicamente , Pérdida Auditiva/diagnóstico , Humanos , Oncología Médica , Neoplasias/tratamiento farmacológico
18.
PLoS One ; 15(12): e0243998, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33326475

RESUMEN

The treatment of children with posterior fossa brain tumours (PFBT) impacts their long term functional and imaging outcomes. This study aimed to evaluate academic achievement correlated with long-term sequelae after different PFBT treatment modalities. The study cohort consisted of 110 survivors (median age at diagnosis 10.1 years and median time of follow up 13.2 years) who completed hearing questionnaires, neurological assessment and MRI of the brain ≥5 years after the end of treatment. There were three treatment groups. A cisplatin group which underwent cisplatin chemotherapy, radiotherapy and surgery (medulloblastoma N = 40), a radiotherapy group which underwent radiotherapy and surgery (astrocytoma/ependymoma N = 30), and a surgery group (astrocytoma N = 40). Academic achievement was correlated to the age at diagnosis, ototoxicity, Karnofsky score (KS), and MRI findings (Fazekas Score (FS)- treatment related parenchymal changes). For a modelled age at diagnosis of five years, the cisplatin group had lower academic achievements compared to the radiotherapy (p = 0.028) and surgery (p = 0.014) groups. Academic achievements evaluated at a modelled age of 10 years at diagnosis did not significantly differ among the treatment groups. The cisplatin group exhibited a higher occurrence of ototoxicity than the radiotherapy (p<0.019) and surgery groups (p<0.001); however, there was no correlation between ototoxicity and academic achievements (p = 0.722) in older age at diagnosis. The radiotherapy group exhibited lower KS than the surgery group (p<0.001). KS significantly influenced academic achievements in all groups (p<0.000). The cisplatin group exhibited higher FS than the surgery group (p<0.001) while FS did not correlate with academic achievement (p = 0.399). Older age is a protective factor for academic achievements irrespective of a treatment modality.


Asunto(s)
Éxito Académico , Supervivientes de Cáncer/educación , Glioma/epidemiología , Neoplasias Infratentoriales/epidemiología , Adolescente , Factores de Edad , Antineoplásicos/efectos adversos , Niño , Cisplatino/efectos adversos , Cisplatino/uso terapéutico , Femenino , Glioma/cirugía , Glioma/terapia , Humanos , Neoplasias Infratentoriales/cirugía , Neoplasias Infratentoriales/terapia , Masculino , Procedimientos Neuroquirúrgicos/efectos adversos , Radioterapia/efectos adversos
19.
Eur J Cancer ; 138: 212-224, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32905960

RESUMEN

BACKGROUND: Irreversible sensorineural hearing loss is a common side effect of platinum treatment with the potential to significantly impair the neurocognitive, social and educational development of childhood cancer survivors. Genetic association studies suggest a genetic predisposition for cisplatin-induced ototoxicity. Among other candidate genes, thiopurine methyltransferase (TPMT) is considered a critical gene for susceptibility to cisplatin-induced hearing loss in a pharmacogenetic guideline. The aim of this cross-sectional cohort study was to confirm the genetic associations in a large pan-European population and to evaluate the diagnostic accuracy of the genetic markers. METHODS: Eligibility criteria required patients to be aged less than 19 years at the start of chemotherapy, which had to include cisplatin and/or carboplatin. Patients were assigned to three phenotype categories: no, minor and clinically relevant hearing loss. Fourteen variants in eleven candidate genes (ABCC3, OTOS, TPMT, SLC22A2, NFE2L2, SLC16A5, LRP2, GSTP1, SOD2, WFS1 and ACYP2) were investigated. Multinomial logistic regression was performed to model the relationship between genetic predictors and platinum ototoxicity, adjusting for clinical risk factors. Additionally, measures of the diagnostic accuracy of the genetic markers were determined. RESULTS: 900 patients were included in this study. In the multinomial logistic regression, significant unique contributions were found from SLC22A2 rs316019, the age at the start of platinum treatment, cranial radiation and the interaction term [platinum compound]∗[cumulative dose of cisplatin]. The predictive performance of the genetic markers was poor compared with the clinical risk factors. CONCLUSIONS: PanCareLIFE is the largest study of cisplatin-induced ototoxicity to date and confirmed a role for the polyspecific organic cation transporter SLC22A2. However, the predictive value of the current genetic candidate markers for clinical use is negligible, which puts the value of clinical factors for risk assessment of cisplatin-induced ototoxicity back into the foreground.


Asunto(s)
Antineoplásicos/efectos adversos , Supervivientes de Cáncer , Carboplatino/efectos adversos , Cisplatino/efectos adversos , Pérdida Auditiva Sensorineural/genética , Audición/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Transportador 2 de Cátion Orgánico/genética , Variantes Farmacogenómicas , Polimorfismo de Nucleótido Simple , Adolescente , Edad de Inicio , Niño , Preescolar , Estudios Transversales , Europa (Continente) , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Pérdida Auditiva Sensorineural/inducido químicamente , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Lactante , Recién Nacido , Masculino , Ototoxicidad , Pruebas de Farmacogenómica , Estudios Prospectivos , Estudios Retrospectivos , Medición de Riesgo , Factores de Riesgo
20.
Data Brief ; 32: 106227, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32939381

RESUMEN

Genetic association studies suggest a genetic predisposition for cisplatin-induced ototoxicity. Among other candidate genes, thiopurine methyltransferase (TPMT) is considered a critical gene for susceptibility to cisplatin-induced hearing loss in a pharmacogenetic guideline. The PanCareLIFE cross-sectional cohort study evaluated the genetic associations in a large pan-European population and assessed the diagnostic accuracy of the genetic markers. 1,112 pediatric cancer survivors who had provided biomaterial for genotyping were screened for participation in the pharmacogenetic association study. 900 participants qualified for inclusion. Based on the assessment of original audiograms, patients were assigned to three phenotype categories: no, minor, and clinically relevant hearing loss. Fourteen variants in eleven candidate genes (ABCC3, OTOS, TPMT, SLC22A2, NFE2L2, SLC16A5, LRP2, GSTP1, SOD2, WFS1, and ACYP2) were genotyped. The genotype and phenotype data represent a resource for conducting meta-analyses to derive a more precise pooled estimate of the effects of genes on the risk of hearing loss due to platinum treatment.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA