Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 192
Filtrar
1.
Br J Haematol ; 196(1): 19-30, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34124782

RESUMEN

With the focus of leukaemia management shifting to the implications of low-level disease burden, increasing attention is being paid on the development of highly sensitive methodologies required for detection. There are various techniques capable of identification of measurable residual disease (MRD) either evidencing as relevant mutation detection [e.g. nucleophosmin 1 (NPM1) mutation] or trace levels of leukaemic clonal populations. The vast majority of these methods only permit detection of a single clone or mutation. However, mass spectrometry and next-generation sequencing enable the interrogation of multiple genes simultaneously, facilitating a more complete genomic profile. In the present review, we explore the methodologies of both techniques in conjunction with the important advantages and limitations associated with each assay. We also highlight the evidence and the various instances where either technique has been used and propose future strategies for MRD detection.


Asunto(s)
Biomarcadores de Tumor , Análisis Mutacional de ADN/métodos , Leucemia/diagnóstico , Leucemia/etiología , Mutación , Neoplasia Residual/diagnóstico , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/normas , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/normas , Humanos , Espectrometría de Masas/métodos , Espectrometría de Masas/normas , Tasa de Mutación , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
3.
J Mol Diagn ; 23(9): 1185-1194, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34186176

RESUMEN

The 1p/19q codeletion is the result of a translocation between chromosome 1 (Chr1p) and chromosome 19 (Chr19q) with the loss of derivative (1;19)(p10;q10) chromosome. The 1p/19q codeletion has predictive and prognostic significance, and it is essential for the classification of gliomas. In routine practice, the fluorescence in situ hybridization (FISH) diagnosis of 1p/19q codeletion is sometimes unexpected. This study aimed to develop a next-generation sequencing panel for the concurrent definition of the 1p/19q codeletion and IDH1/IDH2 mutation status to resolve these equivocal cases. A total of 65 glioma samples were investigated using a 1p/19q-single-nucleotide polymorphism (SNP)-IDH panel. The panel consists of 192 amplicons, including SNPs mapping to Chr1 and Chr19 and amplicons for IDH1/IDH2 analysis. The 1p/19q SNP-IDH panel consistently identified IDH1/IDH2 mutations. In 49 of 60 cases (81.7%), it provided the same 1p/19q results obtained by FISH. In the remaining 11 cases, the 1p/19q SNP-IDH panel uncovered partial chromosome imbalances as a result of interstitial amplification or deletion of the regions where the FISH probes map, leading to a mistaken overdiagnosis of 1p/19q codeletion by FISH. The 1p/19q SNP-IDH next-generation sequencing panel allows reliable analysis of the 1p/19q codeletion and IDH1/IDH2 mutation at the same time. The panel not only allows resolution of difficult cases but also represents a cost-effective alternative to standard molecular diagnostics procedures.


Asunto(s)
Neoplasias Encefálicas/genética , Cromosomas Humanos Par 19/genética , Cromosomas Humanos Par 1/genética , Eliminación de Gen , Glioma/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Hibridación Fluorescente in Situ/métodos , Isocitrato Deshidrogenasa/genética , Sobrediagnóstico , Translocación Genética/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/patología , Estudios de Cohortes , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/métodos , Femenino , Glioma/patología , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Hibridación Fluorescente in Situ/economía , Masculino , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular/economía , Técnicas de Diagnóstico Molecular/métodos , Polimorfismo de Nucleótido Simple , Reproducibilidad de los Resultados , Adulto Joven
4.
Am J Hum Genet ; 108(4): 656-668, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33770507

RESUMEN

Genetic studies in underrepresented populations identify disproportionate numbers of novel associations. However, most genetic studies use genotyping arrays and sequenced reference panels that best capture variation most common in European ancestry populations. To compare data generation strategies best suited for underrepresented populations, we sequenced the whole genomes of 91 individuals to high coverage as part of the Neuropsychiatric Genetics of African Population-Psychosis (NeuroGAP-Psychosis) study with participants from Ethiopia, Kenya, South Africa, and Uganda. We used a downsampling approach to evaluate the quality of two cost-effective data generation strategies, GWAS arrays versus low-coverage sequencing, by calculating the concordance of imputed variants from these technologies with those from deep whole-genome sequencing data. We show that low-coverage sequencing at a depth of ≥4× captures variants of all frequencies more accurately than all commonly used GWAS arrays investigated and at a comparable cost. Lower depths of sequencing (0.5-1×) performed comparably to commonly used low-density GWAS arrays. Low-coverage sequencing is also sensitive to novel variation; 4× sequencing detects 45% of singletons and 95% of common variants identified in high-coverage African whole genomes. Low-coverage sequencing approaches surmount the problems induced by the ascertainment of common genotyping arrays, effectively identify novel variation particularly in underrepresented populations, and present opportunities to enhance variant discovery at a cost similar to traditional approaches.


Asunto(s)
Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/normas , Variación Genética/genética , Genética de Población/economía , África , Análisis Mutacional de ADN/métodos , Genética de Población/métodos , Genoma Humano/genética , Estudio de Asociación del Genoma Completo , Equidad en Salud , Humanos , Microbiota , Secuenciación Completa del Genoma/economía , Secuenciación Completa del Genoma/normas
5.
Future Oncol ; 17(13): 1601-1609, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33415992

RESUMEN

Aim: FLABRA evaluated the prevalence of BRCA mutations, genetic counseling and management approaches in patients with ovarian cancer in Latin America. Patients & methods: Patients with ovarian cancer from six Latin-American countries were enrolled. Tumor samples were tested for BRCA mutations (BRCAmut). In cases with BRCAmut, blood samples were analyzed to determine germline versus somatic mutations. Medical records were reviewed for counseling approach and treatment plan. Results: From 472 patients enrolled, 406 samples yielded conclusive results: 282 were BRCA wild-type (BRCAwt), 115 were BRCAmut and nine were variants of uncertain significance. In total, 110/115 were tested for germline mutations (77 germline and 33 somatic). Conclusion: Tumor testing to identify mutations in BRCA1/2 in ovarian cancer can help optimize treatment choices, meaning fewer patients require germline testing and genetic counseling, a scant resource in Latin America. Clinical trial registration: NCT02984423 (ClinicalTrials.gov).


Asunto(s)
Carcinoma Epitelial de Ovario/diagnóstico , Pruebas Genéticas/estadística & datos numéricos , Neoplasias Ováricas/diagnóstico , Adulto , Anciano , Proteína BRCA1/genética , Proteína BRCA2/genética , Carcinoma Epitelial de Ovario/epidemiología , Carcinoma Epitelial de Ovario/genética , Carcinoma Epitelial de Ovario/terapia , Estudios Transversales , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/estadística & datos numéricos , Femenino , Asesoramiento Genético/economía , Asesoramiento Genético/estadística & datos numéricos , Pruebas Genéticas/economía , Humanos , América Latina/epidemiología , Persona de Mediana Edad , Neoplasias Ováricas/epidemiología , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Medicina de Precisión/métodos , Prevalencia , Estudios Prospectivos , Adulto Joven
6.
J Mol Diagn ; 22(12): 1430-1437, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32961317

RESUMEN

Detection of KRAS, NRAS, and BRAF mutations in tumor tissue is currently used to predict resistance to treatment with anti-epidermal growth factor receptor (EGFR) antibodies in patients with metastatic colorectal cancer (mCRC). Liquid biopsies are minimally invasive, and cell-free circulating tumor DNA (ctDNA) mutation analyses may better represent tumor heterogeneity. This study examined the incorporation of liquid biopsy RAS/BRAF ctDNA analyses into diagnostic strategies to determine mCRC patient eligibility for anti-EGFR therapy. Tumor tissue and liquid biopsies were collected from 100 mCRC patients with liver-only metastases in a multicenter prospective clinical trial. Three diagnostic strategies incorporating droplet digital PCR ctDNA analyses were compared with routine tumor tissue RAS/BRAF mutation profiling using decision tree analyses. Tissue DNA mutations in KRAS, NRAS, and BRAF were present in 54%, 0%, and 3% of mCRC patients, respectively. A 93% concordance was observed between tissue DNA and liquid biopsy ctDNA mutations. The proportion of patients with RAS/BRAF alterations increased from 57% to 60% for diagnostic strategies that combined tissue and liquid biopsy mutation analyses. Consecutive RAS/BRAF ctDNA analysis followed by tissue DNA analysis in case of a liquid biopsy-negative result appeared to be the most optimal diagnostic strategy to comprehensively determine eligibility for anti-EGFR therapy in a cost-saving manner. These results highlight the potential clinical utility of liquid biopsies for detecting primary resistance to anti-EGFR-targeted therapies.


Asunto(s)
ADN Tumoral Circulante/genética , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/secundario , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Biomarcadores de Tumor/genética , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/métodos , Exactitud de los Datos , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Femenino , GTP Fosfohidrolasas/genética , Humanos , Biopsia Líquida/economía , Masculino , Proteínas de la Membrana/genética , Reacción en Cadena de la Polimerasa/economía , Reacción en Cadena de la Polimerasa/métodos , Estudios Prospectivos , Sensibilidad y Especificidad
7.
PLoS One ; 15(8): e0237790, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32810185

RESUMEN

This study determined the frequency and factors associated with EGFR testing rates and erlotinib treatment as well as associated survival outcomes in patients with non small cell lung cancer in Kentucky. Data from the Kentucky Cancer Registry (KCR) linked with health claims from Medicaid, Medicare and private insurance groups were evaluated. EGFR testing and erlotinib prescribing were identified using ICD-9 procedure codes and national drug codes in claims, respectively. Logistic regression analysis was performed to determine factors associated with EGFR testing and erlotinib prescribing. Cox-regression analysis was performed to determine factors associated with survival. EGFR mutation testing rates rose from 0.1% to 10.6% over the evaluated period while erlotinib use ranged from 3.4% to 5.4%. Factors associated with no EGFR testing were older age, male gender, enrollment in Medicaid or Medicare, smoking, and geographic region. Factors associated with not receiving erlotinib included older age, male gender, enrollment in Medicare or Medicaid, and living in moderate to high poverty. Survival analysis demonstrated EGFR testing or erlotinib use was associated with a higher likelihood of survival. EGFR testing and erlotinib prescribing were slow to be implemented in our predominantly rural state. While population-level factors likely contributed, patient factors, including geographic location (areas with high poverty rates and rural regions) and insurance type, were associated with lack of use, highlighting rural disparities in the implementation of cancer precision medicine.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Clorhidrato de Erlotinib/uso terapéutico , Pruebas Genéticas/estadística & datos numéricos , Neoplasias Pulmonares/tratamiento farmacológico , Reclamos Administrativos en el Cuidado de la Salud/estadística & datos numéricos , Adulto , Factores de Edad , Anciano , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/estadística & datos numéricos , Prescripciones de Medicamentos/economía , Prescripciones de Medicamentos/estadística & datos numéricos , Utilización de Medicamentos/economía , Utilización de Medicamentos/estadística & datos numéricos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Femenino , Pruebas Genéticas/economía , Disparidades en Atención de Salud/economía , Humanos , Kentucky/epidemiología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Masculino , Medicaid/economía , Medicaid/estadística & datos numéricos , Medicare/economía , Medicare/estadística & datos numéricos , Persona de Mediana Edad , Mutación , Pobreza/estadística & datos numéricos , Medicina de Precisión/economía , Medicina de Precisión/estadística & datos numéricos , Sistema de Registros/estadística & datos numéricos , Estudios Retrospectivos , Factores Sexuales , Análisis de Supervivencia , Estados Unidos , Adulto Joven
8.
J Mol Diagn ; 22(11): 1344-1355, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32818599

RESUMEN

Antimicrobial resistance in Neisseria gonorrhoeae persists as a major public health concern globally. We developed and evaluated a multiplex assay that relied on high-resolution melting (HRM) technology as a rapid, simple, and cost-effective method for simultaneously detecting and identifying different molecular markers associated with extended-spectrum cephalosporins (ESCs) and azithromycin (AZM) resistance in N. gonorrhoeae. Forty-eight well-characterized N. gonorrhoeae clinical isolates were selected for initial assay establishment. The multiplex HRM assays were able to accurately identify different nucleotide variations of the antimicrobial resistance determinants related to ESCs and AZM resistance. Specificity and cross-reactivity were assessed by testing 15 nongonococcal strains. Then, the assay was validated on 218 archived DNA specimens that had been sequenced using whole-genome sequencing technology. Compared with whole-genome sequencing, these assays had a sensitivity of 98.6%, with a specificity of 99.2%. For further validation of the assay's performance, a total of 338 samples (156 clinical isolates and 182 clinical specimens) were screened using the multiplex HRM assay. The results showed good concordance with the results of PCR sequencing. Given its rapidity (within 90 minutes), ease of performing, and low cost (<$1.00 per sample), this method may be applied to large-scale epidemiologic programs for increasing surveillance of ESCs and AZM resistance in N. gonorrhoeae.


Asunto(s)
Antibacterianos/farmacología , Azitromicina/farmacología , Cefalosporinas/farmacología , Análisis Mutacional de ADN/métodos , Farmacorresistencia Bacteriana/efectos de los fármacos , Farmacorresistencia Bacteriana/genética , Neisseria gonorrhoeae/efectos de los fármacos , Neisseria gonorrhoeae/genética , Temperatura de Transición , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Exactitud de los Datos , Límite de Detección , Pruebas de Sensibilidad Microbiana , Neisseria gonorrhoeae/aislamiento & purificación , Secuenciación Completa del Genoma/economía , Secuenciación Completa del Genoma/métodos
9.
JAMA Dermatol ; 156(11): 1177-1184, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32697281

RESUMEN

Importance: The effectiveness of immune checkpoint inhibitors (ICIs) and BRAF and MEK inhibitors has improved advanced melanoma recovery. However, it is unknown whether these novel therapies are cost-effective for newly diagnosed advanced melanoma with unknown BRAF status. Objective: To compare the cost-utility of these novel agents and their combinations with or without BRAF gene testing guidance for treating newly diagnosed advanced melanoma with unknown BRAF status. Design and Setting: A decision-analytic model was adopted to project the outcomes of 8 strategies containing different ICIs and BRAF and MEK inhibitors for newly diagnosed advanced melanoma with unknown BRAF pathogenic variant status. The key clinical data were derived from the CheckMate 067, KEYNOTE-006, COMBI-d, and COMBI-v trials, and the cost and health preference data were derived from the literature. Costs were estimated from the US payer perspective. Main Outcomes and Measures: Costs, quality-adjusted life-years (QALYs), incremental cost-utility ratio (ICUR), and incremental net health benefits were calculated. Subgroup, 1-way, and probabilistic sensitivity analyses were performed. Results: Of the 8 competing strategies, nivolumab plus ipilimumab without patient selection based on BRAF pathogenic variant testing yielded the most significant health outcome, and the nivolumab strategy was the cheapest option. The nivolumab, pembrolizumab, and nivolumab plus ipilimumab strategies formed the cost-effective frontier, which showed the ordered ICURs were $8593 (SD, $592 995)/QALY for pembrolizumab vs nivolumab and $125 593 (SD, $5 751 223)/QALY for nivolumab plus ipilimumab vs pembrolizumab. Other strategies, including the BRAF testing-guided strategies (BRAF pathogenic variant testing followed by corresponding regimens for BRAF wild and pathogenic variant tumors), were dominated or extended dominated. The most influential parameters were the treatment efficacy of these new regimens. Conclusions and Relevance: For newly diagnosed advanced melanoma with unknown BRAF pathogenic variant status, nivolumab plus ipilimumab and pembrolizumab strategies are likely to be the most cost-effective options. BRAF and MEK inhibitors might be productively placed in a second-line setting after BRAF pathogenic variant is confirmed.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Análisis Costo-Beneficio , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Melanoma/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales Humanizados/economía , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/economía , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Toma de Decisiones Clínicas/métodos , Análisis Mutacional de ADN/economía , Técnicas de Apoyo para la Decisión , Costos de los Medicamentos , Femenino , Pruebas Genéticas , Humanos , Inhibidores de Puntos de Control Inmunológico/economía , Inhibidores de Puntos de Control Inmunológico/farmacología , Ipilimumab/economía , Ipilimumab/farmacología , Ipilimumab/uso terapéutico , Estimación de Kaplan-Meier , Masculino , Melanoma/diagnóstico , Melanoma/mortalidad , Melanoma/patología , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Modelos Económicos , Estadificación de Neoplasias , Nivolumab/economía , Nivolumab/farmacología , Nivolumab/uso terapéutico , Supervivencia sin Progresión , Inhibidores de Proteínas Quinasas/economía , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/genética , Años de Vida Ajustados por Calidad de Vida , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Adulto Joven
10.
Cancer Med ; 9(18): 6507-6514, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32700475

RESUMEN

PURPOSE: Referrals for Lynch syndrome (LS) assessment have traditionally been based on personal and family medical history. The introduction of universal screening practices has allowed for referrals based on immunohistochemistry tests for mismatch repair (MMR) protein expression. This study aims to characterize the effect of universal screening in a publicly funded healthcare system with comparison to patients referred by traditional criteria, from January 2012 to March 2017. METHODS: Patient files from the time of initiation of universal screening from 2012 to 2017 were reviewed. Patients were sorted into two groups: (a) universally screened and (b) referred by traditional methods. Mutation detection rates, analysis of traditional testing criteria met, and cascade carrier testing were evaluated. RESULTS: The mutation detection rate of the universal screening group was higher than the traditionally referred group (45/228 (19.7%) vs 50/390 (12.5%), P = .05), though each were able to identify unique patients. An analysis of testing criteria met by each patient showed that half of referred patients from the universal screening group could not meet any traditional testing criteria. CONCLUSION: The implementation of universal screening in a publicly funded system will increase efficiency in detecting patients with LS. The resources available for genetic testing and counseling may be more limited in public systems, thus inclusion of secondary screening with BRAF and MLH1 promoter hypermethylation testing is key to further optimizing efficiency.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/diagnóstico , Reparación de la Incompatibilidad de ADN , Análisis Mutacional de ADN , Enzimas Reparadoras del ADN/genética , Detección Precoz del Cáncer , Pruebas Genéticas , Mutación , Programas Nacionales de Salud , Colombia Británica/epidemiología , Neoplasias Colorrectales Hereditarias sin Poliposis/economía , Neoplasias Colorrectales Hereditarias sin Poliposis/epidemiología , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , Detección Precoz del Cáncer/economía , Femenino , Financiación Gubernamental , Predisposición Genética a la Enfermedad , Pruebas Genéticas/economía , Humanos , Masculino , Programas Nacionales de Salud/economía , Valor Predictivo de las Pruebas , Sector Público , Reproducibilidad de los Resultados
11.
Biomedica ; 40(2): 283-295, 2020 06 15.
Artículo en Inglés, Español | MEDLINE | ID: mdl-32673457

RESUMEN

Introduction: Cystic fibrosis is an autosomal recessive genetic disease classified as a highcost orphan disease. Objective: To determine the cost-effectiveness ratio of the diagnostic test for the CFTR gene-sequencing in asymptomatic family carriers in the first, second, and third degree of consanguinity. Materials and methods: We conducted a systematic search evaluating operative characteristics of the diagnostic test and decision-tree models in cost-effectiveness studies. A decision-tree model was elaborated taking prevention of future conceptions as a unit of analysis. We obtained the costs of the disease from the high-cost report of the Ministerio de Salud y Protección Social. The costs of the test were referenced by national laboratories. We carried out a deterministic and probabilistic sensitivity analysis with a third-payer perspective and a one-year horizon. Results: An ICER of USD$ 5051.10 was obtained as the incremental cost for obtaining 10.89% more probability of avoiding the birth of a child with cystic fibrosis per screened couple. For family members in second and third degrees, the ICER was USD$ 19,380.94 and USD$ 55,913.53, respectively, evidenced when applying the GDP per capita. This technology was cost-effective in 39%, 61.18%, and 74.36% for 1, 2, and 3 GDP per capita in first degree of consanguinity relatives. Conclusions: The genetic test for the detection of CFTR gene carriers was cost-effective depending on the threshold of availability to pay and the assumptions and limitations established in the model.


Introducción. La fibrosis quística es una enfermedad genética de carácter autosómico recesivo clasificada como enfermedad huérfana de alto costo. Objetivo. Determinar la razón de costo-efectividad de la prueba diagnóstica de secuenciación del gen CFTR para los portadores asintomáticos familiares en primer, segundo y tercer grados de consanguinidad. Materiales y métodos. Se hizo una búsqueda sistemática sobre la evaluación de las características operativas de la prueba diagnóstica y los modelos de árbol de decisiones en estudios de costo-efectividad. Se elaboró un modelo de árbol de decisiones tomando como unidad de análisis la prevención de futuras concepciones. Los costos de la enfermedad se obtuvieron del reporte de alto costo del Ministerio de Salud de Colombia. Los costos de la prueba se obtuvieron de laboratorios nacionales. Se hizo un análisis de sensibilidad, determinístico y probabilístico, con la perspectiva del tercer pagador y horizonte a un año. Resultados. Se obtuvo una razón incremental de costo-efectividad (RICE) de USD$5.051,10 por obtener 10,89 % más de probabilidades de evitar el nacimiento de un niño enfermo con fibrosis quística por pareja. Para los familiares de segundo y tercer grados, se encontró una RICE de USD$ 19.380,94 y USD$ 55.913,53, respectivamente, al aplicar el PIB per cápita. Esta tecnología fue costo-efectiva en 39 %, 61,18 % y 74,36 % para 1, 2 y 3 PIB per cápita en familiares de primer grado de consanguinidad. Conclusiones. La prueba genética de detección de portadores del gen CFTR resultó costo-efectiva dependiendo del umbral de la disponibilidad de pagar, y de los supuestos y limitaciones establecidas en el modelo.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Análisis Mutacional de ADN/economía , Tamización de Portadores Genéticos/economía , Enfermedades Asintomáticas , Sesgo , Colombia/epidemiología , Análisis Costo-Beneficio , Fibrosis Quística/economía , Fibrosis Quística/epidemiología , Fibrosis Quística/genética , Fibrosis Quística/prevención & control , Árboles de Decisión , Tamización de Portadores Genéticos/métodos , Asesoramiento Genético , Humanos , Reembolso de Seguro de Salud , Aceptación de la Atención de Salud , Probabilidad , Sensibilidad y Especificidad , Análisis de Secuencia de ADN/economía
12.
J Appl Lab Med ; 5(4): 643-655, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32407536

RESUMEN

BACKGROUND: The JAK2 V617F variant is diagnostic for myeloproliferative neoplasms, a group of clonal disorders of hematopoietic stem and progenitor cells. Although several approaches have been developed to detect the variant, a gold standard diagnostic method has not yet been defined. We describe a simple, fast, and cost-effective PCR-based approach that enhances test specificity and sensitivity by blocking the amplification of the large excess of wild-type DNA. METHODS: The method involves using an oligo peptide nucleic acid (PNA) perfectly matching its corresponding DNA sequence. The PCR protocol was optimized by collecting a detailed thermodynamic data set on PNA-DNA wild-type duplexes by circular dichroism melting experiments. The specificity and sensitivity of PNA clamping PCR were assessed by genotyping 50 patients with myeloproliferative neoplasm who carried the JAK2 V617F variant and 50 healthy donors. RESULTS: The optimized protocol enabled selective amplification of the variant alleles, achieving maximum sensitivity (100%) and specificity (100%). Analytical sensitivity was 0.05% of variant alleles as assessed by serial dilutions of DNA from the HEL cell line (which carries the JAK2 V617F variant) mixed to wild-type DNA from healthy donors. The JAK2 V617F variant test performed according to this method has better diagnostic performance than its 2 main PCR-based competitors, at much lower cost. CONCLUSIONS: High sensitivity and specificity and cost-effectiveness make PNA clamping PCR a useful testing platform for the detection of minor allele variants in small-scale diagnostic laboratories. It promises to improve patient care while enabling significant healthcare savings.


Asunto(s)
Análisis Mutacional de ADN/métodos , Janus Quinasa 2/genética , Trastornos Mieloproliferativos/diagnóstico , Ácidos Nucleicos de Péptidos/economía , Reacción en Cadena de la Polimerasa/métodos , Línea Celular Tumoral , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , Femenino , Humanos , Masculino , Mutación , Trastornos Mieloproliferativos/genética , Reacción en Cadena de la Polimerasa/economía , Sensibilidad y Especificidad
13.
Cancer Prev Res (Phila) ; 13(8): 643-648, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32409595

RESUMEN

Global interest in genetic testing for cancer susceptibility genes (CSG) has surged with falling costs, increasing awareness, and celebrity endorsement. Current access to genetic testing is based on clinical criteria/risk model assessment which uses family history as a surrogate. However, this approach is fraught with inequality, massive underutilization, and misses 50% CSG carriers. This reflects huge missed opportunities for precision prevention. Early CSG identification enables uptake of risk-reducing strategies in unaffected individuals to reduce cancer risk. Population-based genetic testing (PGT) can overcome limitations of clinical criteria/family history-based testing. Jewish population studies show population-based BRCA testing is feasible, acceptable, has high satisfaction, does not harm psychologic well-being/quality of life, and is extremely cost-effective, arguing for changing paradigm to PGT in the Jewish population. Innovative approaches for delivering pretest information/education are needed to facilitate informed decision-making for PGT. Different health systems will need context-specific implementation strategies and management pathways, while maintaining principles of population screening. Data on general population PGT are beginning to emerge, prompting evaluation of wider implementation. Sophisticated risk prediction models incorporating genetic and nongenetic data are being used to stratify populations for ovarian cancer and breast cancer risk and risk-adapted screening/prevention. PGT is potentially cost-effective for panel testing of breast and ovarian CSGs and for risk-adapted breast cancer screening. Further research/implementation studies evaluating the impact, clinical efficacy, psychologic and socio-ethical consequences, and cost-effectiveness of PGT are needed.


Asunto(s)
Neoplasias de la Mama/prevención & control , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Neoplasias Ováricas/prevención & control , Medicina de Precisión/métodos , Biomarcadores de Tumor/genética , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/genética , Análisis Costo-Beneficio , Consejo/economía , Consejo/métodos , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/métodos , Detección Precoz del Cáncer/economía , Detección Precoz del Cáncer/métodos , Femenino , Pruebas Genéticas/economía , Heterocigoto , Humanos , Judíos/genética , Mutación , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/epidemiología , Neoplasias Ováricas/genética , Educación del Paciente como Asunto/economía , Educación del Paciente como Asunto/métodos , Medicina de Precisión/economía , Prevalencia , Medición de Riesgo/economía , Medición de Riesgo/métodos
14.
Expert Rev Mol Diagn ; 20(6): 601-610, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32064968

RESUMEN

INTRODUCTION: The 'one biomarker/one drug' scenario is unsustainable because cancer is a complex disorder that involves a number of molecular defects. In the past decade, major technological advances have lowered the overall cost and increased the efficiency of next-generation sequencing (NGS). AREAS COVERED: We review recent regulations on NGS and complementary diagnostics in Japan, mainly focusing on high-quality studies that utilized these new diagnostic modalities and were published within the last 5 years. We highlight significant changes in regulation, and explain the direction of efforts to translate the results of NGS and complementary diagnostics into clinical practice. EXPERT OPINION: NGS holds a number of advantages over conventional companion and complementary diagnostics that enable simultaneous analyzes of multiple cancer genes to detect actionable mutations. Parallel technological developments and regulatory changes have led to the rapid adoption of NGS into clinical practice. NGS-based genomic data have been leveraged to better understand the characteristics of a disease that affects its patient's response to a given therapy. As NGS-based tests become more widespread, however, Japanese authorities will face significant challenges particularly with respect to the complexity of genomic data, which will have to be managed if NGS is to benefit patients.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento/tendencias , Legislación de Dispositivos Médicos , Técnicas de Diagnóstico Molecular/tendencias , Neoplasias/tratamiento farmacológico , Medicina de Precisión/métodos , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/métodos , Análisis Mutacional de ADN/tendencias , Bases de Datos de Ácidos Nucleicos , Aprobación de Recursos/legislación & jurisprudencia , Pruebas Dirigidas al Consumidor/economía , Pruebas Dirigidas al Consumidor/legislación & jurisprudencia , Farmacorresistencia Microbiana/genética , Equipos y Suministros/clasificación , Enfermedades Genéticas Congénitas/tratamiento farmacológico , Enfermedades Genéticas Congénitas/genética , Agencias Gubernamentales/organización & administración , Necesidades y Demandas de Servicios de Salud , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Secuenciación de Nucleótidos de Alto Rendimiento/instrumentación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Japón , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Técnicas de Diagnóstico Molecular/economía , Técnicas de Diagnóstico Molecular/métodos , Terapia Molecular Dirigida , Mutación , Programas Nacionales de Salud , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Neoplasias/genética
15.
Expert Rev Mol Diagn ; 20(6): 575-582, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32011193

RESUMEN

INTRODUCTION: Among non-small cell lung cancer (NSCLC) patients, there is one molecularly defined subgroup harboring activating mutations in the epidermal growth factor receptor gene (EGFR), which results in constitutive activation of its intrinsic kinase activity. Consistent data have demonstrated that these patients have a better outcome when treated with specific tyrosine-kinase inhibitors (EGFR-TKIs). Therefore, analysis of EGFR mutational status for treatment guidance is mandatory in this context. AREAS COVERED: Herein we review the clinical development and technical features of cobas® EGFR Mutation Test v2 as a companion diagnostic test (CDx) for therapy with EGFR-TKIs, such as gefitinib, in advanced NSCLC. We also discuss the pros and cons of the current version of the CDx and its performance in both tissue and plasma samples. EXPERT OPINION: The RT-PCR based cobas® EGFR Mutation Test v2 is a reliable and rapid solution for EGFR mutational status assessment at the time of diagnosis in advanced NSCLC that allows eligibility of patients for EGFR-TKI treatment. This test determines EGFR mutations with acceptable sensitivity in tissue or plasma samples. Pre-analytical considerations like tumor cell content, tumor burden or location of metastasis should be considered to better interpret results in the clinical contexture.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Análisis Mutacional de ADN/métodos , Mutación con Ganancia de Función , Genes erbB-2 , Neoplasias Pulmonares/diagnóstico , Terapia Molecular Dirigida , Proteínas de Neoplasias/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Exones/genética , Gefitinib/uso terapéutico , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Reacción en Cadena en Tiempo Real de la Polimerasa/economía , Sensibilidad y Especificidad , Manejo de Especímenes
16.
Value Health ; 23(1): 114-126, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31952666

RESUMEN

BACKGROUND: Monoclonal antibodies against epidermal growth factor receptor (EGFR) have proved beneficial for the treatment of metastatic colorectal cancer (mCRC), particularly when combined with predictive biomarkers of response. International guidelines recommend anti-EGFR therapy only for RAS (NRAS,KRAS) wild-type tumors because tumors with RAS mutations are unlikely to benefit. OBJECTIVES: We aimed to review the cost-effectiveness of RAS testing in mCRC patients before anti-EGFR therapy and to assess how well economic evaluations adhere to guidelines. METHODS: A systematic review of full economic evaluations comparing RAS testing with no testing was performed for articles published in English between 2000 and 2018. Study quality was assessed using the Quality of Health Economic Studies scale, and the British Medical Journal and the Philips checklists. RESULTS: Six economic evaluations (2 cost-effectiveness analyses, 2 cost-utility analyses, and 2 combined cost-effectiveness and cost-utility analyses) were included. All studies were of good quality and adopted the perspective of the healthcare system/payer; accordingly, only direct medical costs were considered. Four studies presented testing strategies with a favorable incremental cost-effectiveness ratio under the National Institute for Clinical Excellence (£20 000-£30 000/QALY) and the US ($50 000-$100 000/QALY) thresholds. CONCLUSIONS: Testing mCRC patients for RAS status and administering EGFR inhibitors only to patients with RAS wild-type tumors is a more cost-effective strategy than treating all patients without testing. The treatment of mCRC is becoming more personalized, which is essential to avoid inappropriate therapy and unnecessarily high healthcare costs. Future economic assessments should take into account other parameters that reflect the real world (eg, NRAS mutation analysis, toxicity of biological agents, genetic test sensitivity and specificity).


Asunto(s)
Neoplasias Colorrectales/economía , Neoplasias Colorrectales/genética , Análisis Mutacional de ADN/economía , Genes ras , Costos de la Atención en Salud , Mutación , Pruebas de Farmacogenómica/economía , Variantes Farmacogenómicas , Medicina de Precisión/economía , Antineoplásicos Inmunológicos/economía , Antineoplásicos Inmunológicos/uso terapéutico , Toma de Decisiones Clínicas , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Análisis Costo-Beneficio , Costos de los Medicamentos , Receptores ErbB/antagonistas & inhibidores , Predisposición Genética a la Enfermedad , Humanos , Metástasis de la Neoplasia , Selección de Paciente , Fenotipo , Valor Predictivo de las Pruebas , Años de Vida Ajustados por Calidad de Vida
17.
Trends Cancer ; 6(1): 31-39, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31952779

RESUMEN

Molecular tests assist at various stages of cancer patient management, including providing diagnosis, predicting prognosis, identifying therapeutic targets, and determining hereditary cancer risk. The current testing paradigm involves germline testing in a subset of patients determined to be at high risk for having a hereditary cancer syndrome, and tumor-only sequencing for treatment decisions in advanced cancer patients. A major limitation of tumor-only sequencing is its inability to distinguish germline versus somatic mutations. Tumor-normal sequencing has emerged as a comprehensive analysis for both hereditary cancer predisposition and somatic profiling. Here, we review recent studies involving tumor-normal sequencing, discuss its benefits in clinical care, challenges for its implementation, and novel insights it has provided regarding tumor biology and germline contribution to cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Análisis Mutacional de ADN/tendencias , Pruebas Genéticas/tendencias , Síndromes Neoplásicos Hereditarios/diagnóstico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/antagonistas & inhibidores , ADN Tumoral Circulante/sangre , ADN Tumoral Circulante/genética , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/métodos , Predisposición Genética a la Enfermedad , Pruebas Genéticas/economía , Pruebas Genéticas/métodos , Mutación de Línea Germinal , Humanos , Biopsia Líquida/economía , Biopsia Líquida/métodos , Terapia Molecular Dirigida/métodos , Terapia Molecular Dirigida/tendencias , Síndromes Neoplásicos Hereditarios/sangre , Síndromes Neoplásicos Hereditarios/genética , Síndromes Neoplásicos Hereditarios/terapia , Medicina de Precisión/métodos , Medicina de Precisión/tendencias , Pronóstico
18.
Methods Mol Biol ; 2072: 39-50, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31541437

RESUMEN

Transposable elements can be highly mutagenic because when they transpose they can insert into genes and disrupt their function, a propensity which has been exploited in many organisms to generate tagged mutant alleles. The Mutator (Mu) family transposon is a family of DNA-type transposons in maize with a particularly high duplication frequency, which results in large numbers of new mutations in lineages that carry active Mu elements. Here we describe a rapid and cost-effective Miseq-based Mu transposon profiling pipeline. This method can also be used for identifying flanking sequences of other types of long insertions such as T-DNAs.


Asunto(s)
Análisis Mutacional de ADN , Elementos Transponibles de ADN , Secuenciación de Nucleótidos de Alto Rendimiento , Mutagénesis Insercional , Zea mays/genética , Biología Computacional/economía , Biología Computacional/métodos , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , Análisis Mutacional de ADN/métodos , Bases de Datos Genéticas , Duplicación de Gen , Sitios Genéticos , Genoma de Planta , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Programas Informáticos
19.
Acta Clin Belg ; 75(3): 212-220, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31007159

RESUMEN

Objectives: The most cost-effective newborn screening strategy for cystic fibrosis (CF) for Flanders, Belgium, is unknown. The aim of this study was to assess the cost-effectiveness of four existing newborn screening strategies for CF: IRT-DNA (immunoreactive trypsinogen, cystic fibrosis transmembrane conductance regulator (CFTR) gene mutation analysis), IRT-PAP (pancreatitis-associated protein), IRT-PAP-DNA, and IRT-PAP-DNA-EGA (extended CFTR gene analysis).Methods: Using data from published literature, the cost-effectiveness of the screening strategies was calculated for a hypothetical cohort of 65,606 newborns in Flanders, Belgium. A healthcare payer perspective was used, and the direct medical costs associated with screening were taken into account. The robustness of the model outcomes was assessed in sensitivity analyses.Results: The IRT-PAP strategy was the most cost-effective strategy in terms of costs per CF case detected (€9314 per CF case detected). The IRT-DNA strategy was more costly (€13,966 per CF case detected), but with an expected sensitivity of 93.4% also the most effective strategy, and was expected to detect 2.2 more cases of CF than the IRT-PAP strategy. The incremental cost-effectiveness ratio of IRT-DNA vs. IRT-PAP was €54,180/extra CF case detected. The IRT-PAP-DNA strategy and the IRT-PAP-DNA-EGA strategy were both strongly dominated by the IRT-PAP strategy.Conclusion: The IRT-PAP strategy was the most cost-effective strategy in terms of costs per CF case detected. However, the strategy did not fulfil the European Cystic Fibrosis Society guidelines for sensitivity and positive predictive value. Therefore, the more costly and more effective IRT-DNA strategy may be the most appropriate newborn screening strategy for Flanders.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/diagnóstico , Tamizaje Neonatal/economía , Proteínas Asociadas a Pancreatitis/metabolismo , Tripsinógeno/metabolismo , Bélgica , Análisis Químico de la Sangre/economía , Análisis Costo-Beneficio , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Análisis Mutacional de ADN/economía , Técnicas de Apoyo para la Decisión , Humanos , Recién Nacido , Tamizaje Neonatal/métodos , Sensibilidad y Especificidad
20.
J Clin Pathol ; 72(6): 406-411, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30872385

RESUMEN

AIMS: BRAF V600E detection assists in the diagnosis of hairy cell leukaemia (HCL); however, testing practices vary. We evaluated the clinical utility of 5 BRAF mutation testing strategies for use on bone marrow trephines (BMT). METHODS: 11 HCL, 5 HCL 'mimic', 2 treated HCL and 10 normal BMT specimens were tested for mutant BRAF, comparing Sanger sequencing, pyrosequencing, amplicon-based next generation sequencing (NGS), automated (Idylla) PCR and immunohistochemistry (IHC). RESULTS: PCR and IHC were cheaper and identified V600E in 100 % of HCL cases. Pyrosequencing detected the mutation in 91%, NGS in 55% of cases and Sanger sequencing in 27%. All assays gave wild-type BRAF results in HCL mimics and normal BMT samples. CONCLUSIONS: PCR and IHC were most sensitive and cost-effective, but these have limited scope for multiplexing and are likely to be replaced by NGS gene panels or whole genome sequencing in the medium to long term.


Asunto(s)
Biomarcadores de Tumor/genética , Médula Ósea/enzimología , Análisis Mutacional de ADN/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunohistoquímica , Leucemia de Células Pilosas/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Automatización de Laboratorios , Biopsia , Médula Ósea/patología , Examen de la Médula Ósea , Análisis Costo-Beneficio , Análisis Mutacional de ADN/economía , Costos de la Atención en Salud , Secuenciación de Nucleótidos de Alto Rendimiento/economía , Humanos , Inmunohistoquímica/economía , Leucemia de Células Pilosas/economía , Leucemia de Células Pilosas/enzimología , Leucemia de Células Pilosas/patología , Valor Predictivo de las Pruebas , Reacción en Cadena en Tiempo Real de la Polimerasa/economía , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA