Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 237
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Pharmacoeconomics ; 42(5): 527-568, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38489077

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) is the most common type of lung cancer, with up to 32% of patients with NSCLC harboring an epidermal growth factor receptor (EGFR) mutation. NSCLC harboring an EGFR mutation has a dedicated treatment pathway, with EGFR tyrosine kinase inhibitors and platinum-based chemotherapy often being the therapy of choice. OBJECTIVE: The aim of this study was to systemically review and summarize economic models of first-line treatments used for locally advanced or metastatic NSCLC harboring EGFR mutations, as well as to identify areas for improvement for future models. METHODS: Literature searches were conducted via Ovid in PubMed, MEDLINE, MEDLINE In-Process, Embase, Evidence-Based Medicine Reviews: Health Technology Assessment, Evidence-Based Medicine Reviews: National Health Service Economic Evaluation Database, and EconLit. An initial search was conducted on 19 December 2022 and updated on 11 April 2023. Studies were selected according to predefined criteria using the Population, Intervention, Comparator, Outcome and Study design (PICOS) framework. RESULTS: Sixty-seven articles were included in the review, representing 59 unique studies. The majority of included models were cost-utility analyses (n = 52), with the remaining studies being cost-effectiveness analyses (n = 4) and a cost-minimization analysis (n = 1). Two studies incorporated both a cost-utility and cost-minimization analysis. Although the model structure across studies was consistently reported, justification for this choice was often lacking. CONCLUSIONS: Although the reporting of economic models in NSCLC harboring EGFR mutations is generally good, many of these studies lacked sufficient reporting of justification for structural choices, performing extensive sensitivity analyses and validation in economic evaluations. In resolving such gaps, the validity of future models can be increased to guide healthcare decision making in rare indications.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Análise Custo-Benefício , Receptores ErbB , Neoplasias Pulmonares , Modelos Econômicos , Mutação , Inibidores de Proteínas Quinases , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/economia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/economia , Neoplasias Pulmonares/patologia , Receptores ErbB/genética , Inibidores de Proteínas Quinases/economia , Inibidores de Proteínas Quinases/uso terapêutico , Antineoplásicos/economia , Antineoplásicos/uso terapêutico
2.
Brasília; CONITEC; fev. 2024.
Não convencional em Português | BRISA | ID: biblio-1551262

RESUMO

INTRODUÇÃO: O câncer de pulmão foi o segundo tipo de câncer mais diagnosticado (11,4%) no mundo e a principal causa de morte por câncer (18%) durante o ano de 2020, totalizando 2,2 milhões de novos casos e 1,8 milhões de mortes no ano. Considera-se para fins terapêuticos e prognósticos a divisão em dois principais tipos histológicos: câncer de pulmão células não pequenas células (CPCNP), que representa cerca de 85% de todos os casos de câncer de pulmão, e câncer de pulmão pequenas células (CPPC), associado a aproximadamente 15% dos casos. O comportamento biológico dos tumores malignos pode estar relacionado à expressão molecular, principalmente de proteínas envolvidas no crescimento e sobrevivência celulares, de modo que a segurança e a eficácia do tratamento guardam relação não só com o subtipo histopatológico como também com as características moleculares do tumor. Assim, para melhor direcionamento da terapia (por exemplo, erlotinibe e gefinitibe), torna-se importante diferenciar os subtipos histológicos, e identificar a presença de mutações específicas, como, por exemplo formas alteradas no gene do EGFR, como se recomenda nas Diretrizes Diagnósticas e Terapêuticas do Câncer de Pulmão do Ministério da Saúde. Entretanto, o Ministério da Saúde não recomenda nenhuma técnica específica para identificação de mutação no gene do EGFR, sendo que o rt-PCR é considerado o teste de referência em muitos contextos clínicos e estudos, seguido pelo sequenciamento de nova geração (NGS). PERGUNTA: Em pacientes diagnosticados com CPCNP, qual a acurácia diagnóstica, custoefetividade e impacto orçamentário do diagnóstico molecular por meio da técnica de rtPCR para identificação de mutação do EGFR em comparação ao NGS ou a não realização de rt-PCR? EVIDÊNCIAS CIENTÍFICAS: A revisão sistemática identificou 19 estudos observacionais de acurácia diagnóstica (teste de referência: NGS). A sensibilidade e especificidade do rt-PCR em comparação ao NGS foi de 92,0% e 97,0%, respectivamente. Apenas um estudo reportou dados de sobrevida global para a comparação de rt-PCR positivo e negativo para mutação de EGFR, apresentando uma diferença não significativa estatisticamente, de 19,2 versus 11,6 meses (Log rank p=0,143). Todos os estudos apresentaram ao menos um domínio com alto risco de viés e nenhuma preocupação quanto à aplicabilidade. AVALIAÇÃO ECONÔMICA: Na análise de custo-efetividade/utilidade comparou-se a realização de rt-PCR com a não realização do teste para um horizonte temporal por toda vida. A análise sugere que a realização do rt-PCR de mutação do EGFR para auxiliar na padronização do melhor tratamento de pacientes com CPCNP e sem tratamento prévio na fase metastática, em comparação a não realização do teste, sugere um modesto benefício clínico de 0,043 ano de vida ajustado pela qualidade (QALY) e 0,040 ano de vida (AV) ganho e uma economia de R$ 3.138,25. Portanto, rt-PCR dominou a não realização do teste (razão de custo-efetividade incremental, RCEI, não representada em casos de dominância). Ademais, as análises de sensibilidade determinística revelaram que para QALY e AV, as proporções de pacientes que usam o erlotinibe e gefitinibe são as variáveis que mais impactam na RCEI, seguida pela proporção de resultados positivos obtidos pela rt-PCR de mutação do EGFR. AVALIAÇÃO DE IMPACTO ORÇAMENTÁRIO: Considerando um horizonte temporal de cinco anos, utilizando demanda aferida (dados de APAC de 2015 a 2021) foi estimado o quantitativo de pacientes com carcinoma pulmonar de células não pequenas avançado. Ademais, foi estimada uma taxa de difusão conservadora, tendo um aumento de 10% ao ano para a tecnologia em análise. Desta forma, observou-se que a incorporação do rt-PCR para identificação de mutação do EGFR em pacientes com CPCNP, sem tratamento prévio na fase metastática, pode gerar um incremento de R$ 1.553.250,00 no primeiro ano, chegando a um incremento de R$ 8.193.750,00 no quinto ano de análise (total acumulado de cerca de R$ 24 milhões em cinco anos) em análise que não considera as potenciais economias com a incorporação do teste. Em análise de sensibilidade considerando preço 20% menor, coerente com preço informado por um dos fabricantes contatados, o impacto orçamentário acumulado em cinco anos seria de cerca de R$ 20 milhões. MONITORAMENTO DO HORIZONTE TECNOLÓGICO: No Monitoramento do Horizonte Tecnológico foi encontrada 1 tecnologia, a Allele-Discriminating Priming System (kit - ADPS), da empresa Genecast, sem identificação de licenças sanitárias Anvisa e FDA. CONSIDERAÇÕES FINAIS: A evidência disponível sobre o uso de rt-PCR para identificação de mutação EGFR em comparação ao NGS sugere elevada acurácia do rt-PCR, com sensibilidade e especificidade de 92,0% e 97,0%, respectivamente. A análise de custoefetividade sugere que a realização da rt-PCR de mutação do EGFR em pacientes com CPCNP, em comparação a não realização do teste, é dominante para QALY e AV. A análise de impacto orçamentário da incorporação da rt-PCR de mutação do EGFR em pacientes com CPCNP, no SUS, sugere um incremento de cerca de R$ 21 a 40 milhões em cinco anos de análise, a depender do preço do kit e market share considerado, em análise que não considera a potencial economia com a incorporação do teste. RECOMENDAÇÃO PRELIMINAR DA CONITEC: Os membros do Comitê de Procedimentos e Produtos, presentes na 123ª Reunião Ordinária da Conitec realizada no dia 05 de outubro de 2023, deliberaram por unanimidade que a matéria fosse disponibilizada em consulta pública com recomendação preliminar favorável à incorporação do diagnóstico molecular por meio da técnica de rt-PCR para identificação de mutação do EGFR em pacientes com câncer de pulmão de células não pequenas. CONSULTA PÚBLICA: A consulta pública nº 51 foi realizada entre os dias 12 de dezembro de 2023 e 02 de janeiro de 2024. Foram recebidas 50 contribuições, sendo 6 classificadas como técnico-científicas e 44 como de experiência ou opinião. Todas as contribuições foram favoráveis à incorporação de rt-PCR para identificação de mutação do EGFR em pacientes com CPCNP. Referente às contribuições técnico-científicas, os estudos sugeridos para inclusão no PTC não contemplavam os critérios de elegibilidade da pergunta PIROS e todas as demais contribuições corroboram os resultados apresentados no relatório de recomendação. Quanto às contribuições de experiência e opinião, das 44 recebidas, uma foi desconsiderada por tratar de outra consulta pública. Também foram recebidos dois anexos. As contribuições recebidas e anexos foram submetidos à análise de conteúdo temática. Das 43 contribuições consideradas, todas expressaram concordância em relação à recomendação preliminar da Conitec, portanto, mostraramse favoráveis à incorporação do procedimento em avaliação. Os participantes que tiveram experiência com o rt-PCR relataram como efeitos positivos a importância do uso e acesso ao teste para os pacientes usuários do SUS terem um melhor direcionamento no tratamento do câncer de pulmão de células não pequenas. Enquanto efeitos negativos e dificuldades no uso do rt-PCR, foram apontados a dificuldade no acesso e o custo. RECOMENDAÇÃO FINAL DA CONITEC: Os membros do Comitê de Produtos e Procedimentos presentes na 126ª Reunião Ordinária da Conitec deliberaram, por unanimidade, recomendar a incorporação de rt-PCR para identificação da mutação do receptor do fator de crescimento epidérmico (EGFR) em pacientes com câncer de pulmão de células não pequenas. Foi assinado o Registro de Deliberação nº 876/2024. DECISÃO: e incorporar, no âmbito do Sistema Único de Saúde - SUS, o RT-PCR para identificação de mutação do receptor do fator de crescimento epidérmico (EGFR) em pacientes com câncer de pulmão de células não pequenas, publicada no Diário Oficial da União nº 46, seção 1, página 54, em 07 de março de 2024.


Assuntos
Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , Genes erbB-1 , Reação em Cadeia da Polimerase em Tempo Real/instrumentação , Receptores ErbB/genética , Sistema Único de Saúde , Brasil , Análise Custo-Benefício/economia
3.
J Med Econ ; 27(1): 219-229, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38269536

RESUMO

AIMS: This study described treatment patterns, healthcare resource utilization (HRU) and costs among advanced or metastatic non-small cell lung cancer (a/mNSCLC) patients with different epidermal growth factor receptor (EGFR) mutation types. MATERIALS AND METHODS: This retrospective study leveraged NeoGenomics NeoNucleus linked with IQVIA PharMetrics Plus between 01 January 2016 to 30 April 2021 (study period). Patients with evidence of a/mNSCLC between 01 July 2016 to 31 March 2021 (selection window) with EGFR test results indicating exon 19 deletion (exon19del), exon 21 L858R (L858R), or exon 20 insertion (exon20i) mutations were included; date of first observed evidence of a/mNSCLC was the index date. Treatment patterns, all-cause HRU and costs during ≥1 month follow-up were reported for each cohort (exon19del, L858R, and exon20i). RESULTS: A total of 106 exon19del, 75 L858R, and 13 exon20i patients met the study criteria. The prevalence of hospitalization was highest in the exon20i cohort (76.9%), followed by L858R (62.7%) and exon19del (55.7%) cohorts. A higher proportion of patients had evidence of hospice/end-of-life care in the exon20i (30.8%) and L858R (29.3%) cohorts relative to the exon19del cohort (22.6%). The exon20i cohort had higher median total healthcare costs per patient per month ($27,069) relative to exon19del ($17,482) and L858R ($17,763). EGFR tyrosine kinase inhibitors (TKI) were the most frequently observed treatment type for exon19del and L858R cohorts, while chemotherapy was the most observed treatment in exon20i cohort. LIMITATIONS: The sample size for the study cohorts was small, thus no statistical comparisons were conducted. CONCLUSIONS: This is one of the first real-world studies to describe HRU and costs among a/mNSCLC patients by specific EGFR mutation type. HRU and costs varied between EGFR mutation types and were highest among exon20i cohort, potentially reflecting higher disease burden and unmet need among patients with this mutation.


Patients with non-small cell lung cancer (NSCLC) in an advanced or metastatic stage (a/mNSCLC) where cancer has spread to other parts of the body have high chance of dying within five years. Treatment and management of a/mNSCLC also incurs significant healthcare resource utilization (HRU) and costs. Patients with a/mNSCLC may have their epidermal growth factor receptor (EGFR) gene mutated with different variations. Our study described what a/mNSCLC patients were treated with, their HRU and healthcare costs separately for the following three types of EGFR mutations: exon 19 deletion (exon19del), exon 21 L858R (L858R), or exon 20 insertion (exon20i). Our study found that patients with exon19del or L858R mutation were commonly treated with EGFR tyrosine kinase inhibitors (TKIs), while exon20i patients were mostly treated with chemotherapy due to lack of targeted treatment for exon20i during the time when the study was conducted. HRU and healthcare costs were highest for patients with exon20i, which shows that patients with exon20i face high burden and have a need for new treatment options.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Estudos Retrospectivos , Inibidores de Proteínas Quinases/uso terapêutico , Mutação , Receptores ErbB/genética , Custos de Cuidados de Saúde
4.
Support Care Cancer ; 32(1): 67, 2023 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-38150163

RESUMO

PURPOSE: The ADAURA trial demonstrated the superiority of osimertinib over a placebo with regard to disease-free survival, showing it to be indicated as an adjuvant therapy for treatment of non-small cell lung cancer with mutated epidermal growth factor receptor (EGFR). The aim of the present study was to conduct a cost-utility analysis and an analysis of the budgetary impact of adjuvant therapy with osimertinib in patients with non-small cell lung cancer with mutated EGFR who had undergone resection surgery with curative intent. METHODS: Analyses were based on the outcomes of the ADAURA clinical trial and were conducted through a Spanish National Health Service perspective. The outcome measures used were quality-adjusted life years (QALY). RESULTS: The average overall cost of adjuvant treatment with osimertinib over a period of 100 months in the overall sample of trial patients (stages IB-IIIA) was 220,961 €, compared with 197,849 € in the placebo group. Effectiveness, estimated according to QALY, was 6.26 years in the osimertinib group and 5.96 years in the placebo group, with the incremental cost-utility ratio being 77,040 €/QALY. With regard to the budgetary impact, it was estimated that, in 2021, approximately 1130 patients would be subsidiaries to receive osimertinib. This pertains to a difference of 17,375,330 € over 100 months to fund this treatment relative to no treatment. CONCLUSION: Taking into account a Spanish threshold of 24,000 €/QALY, the reduction in the acquisition cost of osimertinib will have to be greater than 10%, to obtain a cost-effective alternative.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma de Pequenas Células do Pulmão , Humanos , Análise Custo-Benefício , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Medicina Estatal , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Receptores ErbB/genética
5.
Thorac Cancer ; 14(25): 2548-2557, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37525557

RESUMO

BACKGROUND: This study aimed to comprehensively evaluate the efficacy and toxicity of afatinib in patients with sarcopenia, an important prognostic factor for treatment efficacy and toxicity in patients with cancer. METHODS: The clinical features of patients with advanced NSCLC treated with frontline afatinib between 2014 and 2018 at a medical center in Taiwan were retrospectively reviewed. Sarcopenia was evaluated based on the total cross-sectional area of skeletal muscles assessed by computed tomography (CT) imaging at the L3 level. Baseline characteristics, response rates, survival rates, and adverse events (AEs) were compared between sarcopenic and nonsarcopenic patients. RESULTS: A total of 176 patients evaluated for sarcopenia by CT and treated with afatinib were enrolled in the current study. Sarcopenia was significantly associated with good performance status, low body mass index (BMI), low body surface area (BSA), and low total mass area (TMA). Sarcopenia did not influence the response rate (69.2% vs. 72.0%, p = 0.299), progression-free survival (median 15.9 vs. 14.9 months, p = 0.791), or overall survival (median 26.5 vs. 27.2 months, p = 0.441). However, BSA ≤ 1.7 and the 40 mg afatinib dose were associated with dose reduction. TMA was the only independent factor for afatinib discontinuation due to AEs. CONCLUSION: Sarcopenia was not associated with treatment efficacy or toxicity among patients with NSCLC harboring common mutations treated with afatinib, indicating sarcopenic patients should not be excluded from afatinib treatment. Other factors, such as BSA and TMA, were associated with dose reduction and afatinib discontinuation, respectively, which may require additional evaluations in future studies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Sarcopenia , Humanos , Afatinib/uso terapêutico , Neoplasias Pulmonares/tratamento farmacológico , Estudos Retrospectivos , Sarcopenia/induzido quimicamente , Inibidores de Proteínas Quinases/uso terapêutico , Receptores ErbB/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resultado do Tratamento , Mutação
6.
Int J Mol Sci ; 24(14)2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37511192

RESUMO

Assessment of the quality and current performance of computed tomography (CT) radiomics-based models in predicting epidermal growth factor receptor (EGFR) mutation status in patients with non-small-cell lung carcinoma (NSCLC). Two medical literature databases were systematically searched, and articles presenting original studies on CT radiomics-based models for predicting EGFR mutation status were retrieved. Forest plots and related statistical tests were performed to summarize the model performance and inter-study heterogeneity. The methodological quality of the selected studies was assessed via the Radiomics Quality Score (RQS). The performance of the models was evaluated using the area under the curve (ROC AUC). The range of the Risk RQS across the selected articles varied from 11 to 24, indicating a notable heterogeneity in the quality and methodology of the included studies. The average score was 15.25, which accounted for 42.34% of the maximum possible score. The pooled Area Under the Curve (AUC) value was 0.801, indicating the accuracy of CT radiomics-based models in predicting the EGFR mutation status. CT radiomics-based models show promising results as non-invasive alternatives for predicting EGFR mutation status in NSCLC patients. However, the quality of the studies using CT radiomics-based models varies widely, and further harmonization and prospective validation are needed before the generalization of these models.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Tomografia Computadorizada por Raios X , Humanos , Carcinoma Pulmonar de Células não Pequenas/diagnóstico por imagem , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Receptores ErbB/genética , Interpretação de Imagem Assistida por Computador , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Mutação , Tomografia Computadorizada por Raios X/métodos
7.
Ann Lab Med ; 43(6): 605-613, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37387493

RESUMO

Background: In non-small cell lung cancer (NSCLC), epidermal growth factor receptor (EGFR) mutation testing of tumor tissue should be conducted at diagnosis. Alternatively, circulating tumor DNA can be used to detect EGFR mutation. We compared the cost and clinical effect of three strategies according to the application of the EGFR test. Methods: Decision models were developed to compare the cost-effectiveness of tissue-only, tissue-first, and plasma-first diagnostic strategies as first- and second-line treatments for NSCLC from the perspective of the Korean national healthcare payer. Progression-free survival (PFS), overall survival (OS), and direct medical costs were assessed. A one-way sensitivity analysis was performed. Results: The plasma-first strategy correctly identified numerous patients in the first- and second-line treatments. This strategy also decreased the cost of biopsy procedures and complications. Compared with that when using the other two strategies, the plasma-first strategy increased PFS by 0.5 months. The plasma-first strategy increased OS by 0.9 and 1 month compared with that when using the tissue-only and tissue-first strategies, respectively. The plasma-first strategy was the least expensive first-line treatment but the most expensive second-line treatment. First-generation tyrosine kinase inhibitor and the detection rate of the T790M mutation in tissues were the most cost-influential factors. Conclusions: The plasma-first strategy improved PFS and OS, allowing for a more accurate identification of candidates for targeted therapy for NSCLC and decreased biopsy- and complication-related costs.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Análise de Custo-Efetividade , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Mutação , Inibidores de Proteínas Quinases/uso terapêutico
8.
Cancer Med ; 12(11): 12683-12704, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37017510

RESUMO

BACKGROUND: Advanced lung cancer patients exposed to breakthrough therapies like EGFR tyrosine kinase inhibitors (EGFR-TKI) may experience social inequalities in survival, partly from differences in care. This study examined survival by neighborhood-level socioeconomic and sociodemographic status, and geographical location of advanced lung cancer patients who received gefitinib, an EGFR-TKI, as first-line palliative treatment. Differences in the use and delay of EGFR-TKI treatment were also examined. METHODS: Lung cancer patients receiving gefitinib from 2001 to 2019 were identified from Quebec's health administrative databases. Accounting for age and sex, estimates were obtained for the median survival time from treatment to death, the probability of receiving osimertinib as a second EGFR-TKI, and the median time from biopsy to receiving first-line gefitinib. RESULTS: Among 457 patients who received first-line treatment with gefitinib, those living in the most materially deprived areas had the shortest median survival time (ratio, high vs. low deprivation: 0.69; 95% CI: 0.47-1.04). The probability of receiving osimertinib as a second EGFR-TKI was highest for patients from immigrant-dense areas (ratio, high vs. lowdensity: 1.95; 95% CI: 1.26-3.36) or from Montreal (ratio, other urban areas vs. Montreal: 0.39; 95% CI: 0.16-0.71). The median wait time for gefitinib was 1.27 times longer in regions with health centers peripheral to large centers in Quebec or Montreal in comparison to regions with university-affiliated centers (95% CI: 1.09-1.54; n = 353). CONCLUSION: This study shows that real-world variations in survival and treatment exist among advanced lung cancer patients in the era of breakthrough therapies and that future research on inequalities should also focus on this population.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Gefitinibe/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Cloridrato de Erlotinib/efeitos adversos , Quebeque/epidemiologia , Determinantes Sociais da Saúde , Inibidores de Proteínas Quinases/efeitos adversos , Receptores ErbB/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/epidemiologia , Canadá/epidemiologia , Mutação
10.
JCO Precis Oncol ; 7: e2200294, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36634300

RESUMO

PURPOSE: Many patients with actionable driver oncogenes (ADOs) are never identified and thus never receive targeted treatment. This study evaluated the economic impact and the potential life-years gained (LYG) that can be attributed to the extent of next-generation sequencing (NGS) testing in the United States compared with single-gene testing (SGT) in patients with metastatic nonsquamous non-small-cell lung cancer in the United States. METHODS: A model was developed to evaluate incremental rates of SGT or NSG testing on the basis of LYG and cost per LYG. ADOs included for NGS included EGFR, ALK, ROS1, BRAF, RET, MET, and NTRK. SGT included EGFR and ALK. Assumptions were made for expected incidence of ADOs. Survival distributions were fit to published trial averages of median and 5-year overall survival. Treatment costs were estimated from drug cost averages. Reimbursement costs were based on data from the Center for Medicare and Medicaid Services. RESULTS: Each incremental 10% increase in NGS testing produces an average of 2,627.4 additional LYG, with an average cost savings per LYG of $75 US dollars (USD). Replacing SGT at the current rate of 80% with NGS testing would result in an average additional 21,09.6 LYG and reduce cost per LYG by an average of $599 USD. If 100% of eligible patients were tested with NGS and each identified patient had matched treatment, the total average cost per LYG would be $16,641.57 USD. CONCLUSION: On the basis of current evidence, population-level simulations demonstrate that clinically relevant gains in survival with non-negligible reduction in costs are obtainable from widespread adoption of NGS testing and appropriate treatment matching for patients with advanced nonsquamous non-small-cell lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Idoso , Estados Unidos/epidemiologia , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/epidemiologia , Carcinoma Pulmonar de Células não Pequenas/genética , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Tirosina Quinases/uso terapêutico , Proteínas Proto-Oncogênicas/uso terapêutico , Medicare , Receptores Proteína Tirosina Quinases , Receptores ErbB/genética , Sequenciamento de Nucleotídeos em Larga Escala
11.
J Manag Care Spec Pharm ; 29(2): 172-186, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36373869

RESUMO

BACKGROUND: Lung cancer is a leading cause of cancer morbidity and death in the United States. Non-small cell lung cancer (NSCLC) accounts for 85% of lung cancer cases, and oncogenic mutations in the gene encoding the epidermal growth factor receptor (EGFR) are among its most common genetic causes. Although NSCLC tumors harboring more common oncogenic EGFR mutations can be effectively treated with EGFR tyrosine kinase inhibitors (TKIs), those harboring EGFR exon 20 insertion mutations respond poorly to treatment with therapies approved for advanced NSCLC, including TKIs. Mobocertinib, a first-in-class potent, oral, irreversible TKI, is effective in this population. OBJECTIVE: To estimate the budget impact, for a US health plan with 10 million members, of introducing mobocertinib for the treatment of patients with locally advanced or metastatic NSCLC harboring EGFR exon 20 insertion mutations who have been previously treated with platinum-based chemotherapy. METHODS: A budget impact model was developed to compare 2 scenarios: a reference scenario in which 50% of patients received amivantamab and 50% received physician's choice/usual care therapy and an alternative scenario in which mobocertinib replaced the physician's choice/usual care option. The model had a 5-year time horizon in the base case. The model included epidemiologic inputs to estimate the size of the treatment-eligible population; clinical inputs to estimate treatment duration and efficacy, as well as adverse event frequency; and cost inputs for treatment acquisition and administration, management of adverse events, monitoring, and terminal care. The duration and cost of subsequent therapies were also considered. Budget impact was reported as a total cost, as per-member per-year costs, and as per-member per-month (PMPM) costs. To assess the robustness of model estimates and identify cost drivers, one-way sensitivity analyses and a range of scenario analyses were conducted. RESULTS: The model estimated an eligible treatment population of 55 patients (11 per year) over a 5-year time horizon. In the base case, the estimated budget impact of introducing mobocertinib was $5,615,808, or $0.01 PMPM. Model findings were robust to one-way sensitivity analyses and a range of sensitivity analyses; none of these analyses led to a PMPM budget impact of more than $0.06. Cost drivers included the percentage of eligible patients, the median duration of physician's choice/usual care therapy, patient weight, and the percentage of patients who undergo molecular testing. CONCLUSIONS: The estimated budget impact of mobocertinib is low, primarily because NSCLC harboring EGFR exon 20 insertion mutations is rare. DISCLOSURES: Dr Hernandez is an employee of Takeda Pharmaceuticals America, Inc. Dr Young was an employee of Takeda Pharmaceuticals America, Inc., at the time this study was conducted. This study and the editorial assistance were funded by Takeda Pharmaceuticals America, Inc.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Humanos , Estados Unidos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Mutagênese Insercional , Custos de Medicamentos , Receptores ErbB/genética , Preparações Farmacêuticas , Éxons/genética , Mutação , Inibidores de Proteínas Quinases/uso terapêutico
12.
BMC Cancer ; 22(1): 889, 2022 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-35964032

RESUMO

BACKGROUND: This study aimed to develop and externally validate contrast-enhanced (CE) T1-weighted MRI-based radiomics for the identification of epidermal growth factor receptor (EGFR) mutation, exon-19 deletion and exon-21 L858R mutation from MR imaging of spinal bone metastasis from primary lung adenocarcinoma. METHODS: A total of 159 patients from our hospital between January 2017 and September 2021 formed a primary set, and 24 patients from another center between January 2017 and October 2021 formed an independent validation set. Radiomics features were extracted from the CET1 MRI using the Pyradiomics method. The least absolute shrinkage and selection operator (LASSO) regression was applied for selecting the most predictive features. Radiomics signatures (RSs) were developed based on the primary training set to predict EGFR mutations and differentiate between exon-19 deletion and exon-21 L858R. The RSs were validated on the internal and external validation sets using the Receiver Operating Characteristic (ROC) curve analysis. RESULTS: Eight, three, and five most predictive features were selected to build RS-EGFR, RS-19, and RS-21 for predicting EGFR mutation, exon-19 deletion and exon-21 L858R, respectively. The RSs generated favorable prediction efficacies for the primary (AUCs, RS-EGFR vs. RS-19 vs. RS-21, 0.851 vs. 0.816 vs. 0.814) and external validation (AUCs, RS-EGFR vs. RS-19 vs. RS-21, 0.807 vs. 0.742 vs. 0.792) sets. CONCLUSIONS: Radiomics features from the CE MRI could be used to detect the EGFR mutation, increasing the certainty of identifying exon-19 deletion and exon-21 L858R mutations based on spinal metastasis MR imaging.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Ósseas , Neoplasias Pulmonares , Biomarcadores , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/genética , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/genética , Imageamento por Ressonância Magnética/métodos , Mutação
13.
BMC Cancer ; 22(1): 759, 2022 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-35820813

RESUMO

BACKGROUND: Circulating cell free DNA (cfDNA) testing of plasma for EGFR somatic variants in lung cancer patients is being widely implemented and with any new service, external quality assessment (EQA) is required to ensure patient safety. An international consortium, International Quality Network for Pathology (IQNPath), has delivered a second round of assessment to measure the accuracy of cfDNA testing for lung cancer and the interpretation of the results. METHODS: A collaboration of five EQA provider organisations, all members of IQNPath, have delivered the assessment during 2018-19 to a total of 264 laboratories from 45 countries. Bespoke plasma reference material containing a range of EGFR mutations at varying allelic frequencies were supplied to laboratories for testing and reporting according to routine procedures. The genotyping accuracy and clinical reporting was reviewed against standardised criteria and feedback was provided to participants. RESULTS: The overall genotyping error rate in the EQA was found to be 11.1%. Low allelic frequency samples were the most challenging and were not detected by some testing methods, resulting in critical genotyping errors. This was reflected in higher false negative rates for samples with variant allele frequencies (VAF) rates less than 1.5% compared to higher frequencies. A sample with two different EGFR mutations gave inconsistent detection of both mutations. However, for one sample, where two variants were present at a VAF of less than 1% then both mutations were correctly detected in 145/263 laboratories. Reports often did not address the risk that tumour DNA may have not been tested and limitations of the methodologies provided by participants were insufficient. This was reflected in the average interpretation score for the EQA being 1.49 out of a maximum of 2. CONCLUSIONS: The variability in the standard of genotyping and reporting highlighted the need for EQA and educational guidance in this field to ensure the delivery of high-quality clinical services where testing of cfDNA is the only option for clinical management.


Assuntos
Ácidos Nucleicos Livres , Neoplasias Pulmonares , Receptores ErbB/genética , Frequência do Gene , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/genética , Mutação
15.
Clin Trials ; 19(3): 267-273, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35575012

RESUMO

OBJECTIVE: Although patient-reported symptoms and side effects are increasingly measured in cancer clinical trials, an appropriate assessment frequency has not yet been established. To determine whether differences in assessment frequency affect the apparent incidence and severity of patient-reported symptoms using two well-established patient-reported outcome measures used within the same clinical trial. METHODS: We examined patient-reported outcome results from AURA3 (NCT02151981), a randomized open-label study comparing Tagrisso (osimertinib) with platinum-based chemotherapy in patients with previously treated estimated glomerular filtration rate/T790M mutation-positive metastatic non-small cell lung cancer. The outcome of interest was the proportion of patients in each arm that reported worsening of nausea, vomiting, fatigue, diarrhea, constipation, and appetite loss from baseline measured using the patient-reported outcome-common terminology criteria for adverse event (weekly) or European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (every 6 weeks). RESULTS: Similar trends were observed for all six symptoms investigated. Using nausea in the chemotherapy arm as an example, 76% of patients reported any worsening from baseline based on weekly patient-reported outcome-common terminology criteria for adverse event assessments. When using an every 6-week assessment of Quality of Life Questionnaire Core 30 nausea and restricting analysis to an every 6-week assessment for patient-reported outcome-common terminology criteria for adverse event nausea, the proportion of chemotherapy arm patients reporting any worsening of nausea was 40% for both measures. Across the six patient-reported symptomatic adverse events, we observed differential proportions when comparing frequent versus sparse assessment. CONCLUSION: This analysis demonstrates that more frequent assessment of patient-reported symptomatic adverse events will lead to improved detection, and therefore a more complete understanding of the tolerability of experimental anti-cancer therapies.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/genética , Receptores ErbB/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Mutação , Náusea/induzido quimicamente , Medidas de Resultados Relatados pelo Paciente , Inibidores de Proteínas Quinases/uso terapêutico , Qualidade de Vida
16.
Asian Pac J Cancer Prev ; 23(4): 1155-1158, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35485670

RESUMO

BACKGROUND: Thyrosin kinase inhibitors (TKIs) is approved for the first line treatment of non-small cell lung cancer (NSCLC) patients with  epidermal growth factor receptor (EGFR) mutation. This study performed to assess clinical effectiveness and safety of Erlova (generic form of Erlotinib). METHODS: Somatic mutations of EGFR gene were studied in tumor tissue by polymerase chain reaction (PCR) and bi-directional sequencing in 513 chemonaive and histologically verified lung adenocarcinoma Iranian patients. Patients  with EGFR mutation received Erlova at 150 mg/day  as first line treatment. Primary endpoint was progression free survival (PFS). RESULTS: About 21% (n=109) cases had EGFR mutation. Most EGFR mutations were  occurred at exon 19. Among them, sixty nine patients treated with Erlova. Median PFS was 11.4 months and objective response rate (ORR) was about  88%. Most frequent treatment related adverse events was  skin rash. CONCLUSION: Our findings showed Erlova had remarkable effectiveness. In  mutation-positive patients with EGFR, Erlova can be used  safely instead of  other tyrosine-kinase inhibitors.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Receptores ErbB/genética , Genes erbB-1 , Humanos , Irã (Geográfico) , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Inibidores de Proteínas Quinases/farmacologia , Resultado do Tratamento
17.
BMC Health Serv Res ; 22(1): 470, 2022 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-35397521

RESUMO

BACKGROUND: Guidelines in 2013 and 2014 recommended Epidermal Growth Factor Receptor (EGFR) testing for metastatic lung adenocarcinoma patients as the efficacy of targeted therapies depends on the mutations. However, adherence to these guidelines and the corresponding costs have not been well-studied. METHODS: We identified 2362 patients at least 65 years old newly diagnosed with metastatic lung adenocarcinoma from January 2013 to December 2015 using the SEER-Medicare database. We examined the utilization patterns of EGFR testing and targeted therapies including erlotinib and afatinib. We further examined the costs of both EGFR testing and targeted therapy in terms of Medicare costs and patient out-of-pocket (OOP) costs. RESULTS: The EGFR testing rate increased from 38% in 2013 to 51% and 49% in 2014 and 2015 respectively. The testing rate was 54% among the 394 patients who received erlotinib, and 52% among the 42 patients who received afatinib. The median Medicare and OOP costs for testing were $1483 and $293. In contrast, the costs for targeted therapy were substantially higher with median 30-day costs at $6114 and $240 for erlotinib and $6239 and $471 for afatinib. CONCLUSION: This population-based study suggests that testing guidelines improved the use of EGFR testing, although there was still a large proportion of patients receiving targeted therapy without testing. The costs of targeted therapy were substantially higher than the testing costs, highlighting the need to improve adherence to testing guidelines in order to improve clinical outcomes while reducing the economic burden for both Medicare and patients.


Assuntos
Adenocarcinoma de Pulmão , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Adenocarcinoma de Pulmão/induzido quimicamente , Adenocarcinoma de Pulmão/tratamento farmacológico , Adenocarcinoma de Pulmão/genética , Afatinib/uso terapêutico , Idoso , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/genética , Receptores ErbB/uso terapêutico , Cloridrato de Erlotinib/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Medicare , Mutação , Inibidores de Proteínas Quinases/efeitos adversos , Estados Unidos
18.
BMC Health Serv Res ; 22(1): 431, 2022 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-35365136

RESUMO

BACKGROUND: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been widely used in the treatment of EGFR mutation non-small-cell lung cancer. The Chinese government has made great efforts to improve the availability and affordability of these drugs. The aim of this study was to investigate the trends in the consumption and cost of EGFR TKIs in Nanjing, a developed city in China, and evaluate the influence of health insurance coverage and national price negotiation on drug consumption. METHODS: Data about EGFR TKIs applications in 2010-2019 were extracted from Jiangsu Medicine Information Institute. Five types of EGFR TKIs were included. Consumption was expressed in defined daily doses (DDDs) and expenditure. The correlation between defined daily cost (DDC) and DDDs was analyzed by Pearson's correlation test. RESULTS: The DDC, number of DDDs and expenditure of EGFR TKIs changed little from 2010 to 2015. National price negotiation was initiated as a policy and low-price generic gefitinib came into the market in 2016. Three types of EGFR TKIs moved into the coverage of the national health insurance since 2017. Hence, the DDC decreased, and the number of DDDs increased significantly year by year since 2016. The first generation TKIs always made up of comprised the majority of the total consumption. The predominantly prescribed TKIs were gefitinib and icotinib. DDC was negatively correlated with the number of DDDs. The number of DDDs increased significantly after health insurance enrollment, price negotiation and generic drug replacement. CONCLUSION: The consumption of EGFT TKIs has increased and the DDC of EGFR TKIs has decreased since 2016. These trends may be attributed to drug reimbursement, price negotiation and generic drug replacement. Further efforts are needed to translate the high consumption of EGFR TKIs into clinical benefits.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/genética , Receptores ErbB/uso terapêutico , Humanos , Negociação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico
19.
J Natl Compr Canc Netw ; 20(7): 774-782.e4, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35385830

RESUMO

BACKGROUND: This study sought to determine whether exclusionary EGFR mutation testing followed by next-generation sequencing (NGS) is a cost-efficient and timely strategy in areas with high prevalence rates of EGFR mutation. METHODS: We developed a decision tree model to compare exclusionary EGFR testing followed by NGS and up-front NGS. Patients entered the model upon diagnosis of metastatic lung adenocarcinoma. Gene alterations with FDA-approved targeted therapies included EGFR, ALK, ROS1, BRAF, RET, MET, NTRK, and KRAS. Model outcomes were testing-related costs; time-to-test results; monetary loss, taking both costs and time into consideration; and percentage of patients who could be treated by FDA-approved therapies. Stacked 1-way and 3-way sensitivity analyses were performed. RESULTS: Exclusionary EGFR testing incurred testing-related costs of US $1,387 per patient, a savings of US $1,091 compared with the costs of up-front NGS. The time-to-test results for exclusionary EGFR testing and up-front NGS were 13.0 and 13.6 days, respectively. Exclusionary EGFR testing resulted in a savings of US $1,116 in terms of net monetary loss, without a reduction of patients identified with FDA-approved therapies. The EGFR mutation rate and NGS cost had the greatest impact on minimizing monetary loss. Given that the tissue-based NGS turnaround time was shortened to 7 days, up-front NGS testing would become the best strategy if its price could be reduced to US $568 in Taiwan. CONCLUSIONS: In areas with high prevalence rates of EGFR mutation, exclusionary EGFR testing followed by NGS, rather than up-front NGS, is currently a cost-efficient strategy for metastatic lung adenocarcinoma.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Adenocarcinoma de Pulmão/diagnóstico , Adenocarcinoma de Pulmão/genética , Receptores ErbB/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/epidemiologia , Neoplasias Pulmonares/genética , Mutação , Prevalência , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas/genética
20.
Oncologist ; 27(5): 407-413, 2022 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-35285487

RESUMO

INTRODUCTION: The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor osimertinib was recently approved for resected EGFR-mutant stages IB-IIIA non-small cell lung cancer due to improved disease-free survival (DFS) in this population compared with placebo. This study aimed to evaluate the cost-effectiveness (CE) of this strategy. MATERIALS AND METHODS: We constructed a Markov model using post-resection health state transitions with digitized DFS data from the ADAURA trial to compare cost and quality-adjusted life years (QALYs) of 3 years of adjuvant osimertinib versus placebo over a 10-year time horizon. An overall survival (OS) benefit of 5% was assumed. Costs and utility values were derived from Medicare reimbursement data and literature. A CE threshold of 3 times the gross domestic product per capita was used. Sensitivity analyses were performed. RESULTS: The incremental cost-effectiveness ratio for adjuvant osimertinib was $317 119 per QALY-gained versus placebo. Initial costs of osimertinib are higher in years 1-3. Costs due to progressive disease (PD) are higher in the placebo group through the first 6.5 years. Average pre-PD, post-PD, and total costs were $2388, $379 047, and $502 937, respectively, in the placebo group, and $505 775, $255 638, and $800 697, respectively, in the osimertinib group. Sensitivity analysis of OS gains reaches CE with an hazard ratio (HR) of 0.70-0.75 benefit of osimertinib over placebo. A 50% discount to osimertinib drug cost yielded an ICER of $115 419. CONCLUSIONS: Three-years of adjuvant osimertinib is CE if one is willing to pay $317 119 more per QALY-gained. Considerable OS benefit over placebo or other economic interventions will be needed to reach CE.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Acrilamidas , Idoso , Compostos de Anilina , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Análise Custo-Benefício , Receptores ErbB/genética , Receptores ErbB/uso terapêutico , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Medicare , Mutação , Inibidores de Proteínas Quinases/uso terapêutico , Anos de Vida Ajustados por Qualidade de Vida , Estados Unidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA