Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
ESMO Open ; 8(6): 102051, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37951129

RESUMEN

BACKGROUND: In the setting of localized colon cancer (CC), circulating tumor DNA (ctDNA) monitoring in plasma has shown potential for detecting minimal residual disease (MRD) and predicting a higher risk of recurrence. With the tumor-only sequencing approach, however, germline variants may be misidentified as somatic variations, precluding the possibility of tracking in up to 11% of patients due to a lack of known somatic mutations. In this study, we assess the potential value of adding white blood cells (WBCs) to tumor tissue sequencing to enhance the accuracy of sequencing results. PATIENTS AND METHODS: A total of 148 patients diagnosed with localized CC were prospectively recruited at the Hospital Clínico Universitario in Valencia (Spain). Employing a custom 29-gene panel, sequencing was conducted on tumor tissue, plasma and corresponding WBCs. Droplet digital PCR and amplicon-based NGS were performed on plasma samples post-surgery to track MRD. Oncogenic somatic variants were identified by annotating with COSMIC, OncoKB and an internal repository of pathogenic mutations database. A variant prioritization analysis, mainly characterized by the match of oncogenic mutations with the evidence levels defined in OncoKB, was carried out to select specific targeted therapies. RESULTS: Utilizing paired tumor and WBCs sequencing, we identified somatic mutations in all patients (100%) within our cohort, compared to 89% using only tumor tissue. Consequently, the top 10 most frequently mutated genes for plasma monitoring were altered. The sequencing of WBCs identified 9% of patients with pathogenic mutations in the germline, with APC and TP53 being the most frequently mutated genes. Additionally, mutations in genes related to clonal hematopoiesis of indeterminate potential were detected in 27% of the cohort, with TP53, KRAS, and KMT2C being the most frequently altered genes. There were no observed differences in the sensitivity of monitoring MRD using ddPCR or amplicon-based NGS (p = 1). Ultimately, 41% of the patients harbored potentially targetable alterations at diagnosis. CONCLUSION: The germline testing method not only enhanced sequencing results and raised the proportion of patients eligible for plasma monitoring, but also uncovered the existence of pathogenic germline variations, thereby aiding in the identification of patients at a higher risk of hereditary cancer syndromes.


Asunto(s)
ADN Tumoral Circulante , Neoplasias del Colon , Humanos , ADN Tumoral Circulante/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , ADN de Neoplasias/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Células Germinativas/patología
2.
ESMO Open ; 8(6): 102032, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37852035
3.
Ann Oncol ; 34(10): 885-898, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37597578

RESUMEN

BACKGROUND: HER2 mutations are targetable alterations in patients with hormone receptor-positive (HR+) metastatic breast cancer (MBC). In the SUMMIT basket study, patients with HER2-mutant MBC received neratinib monotherapy, neratinib + fulvestrant, or neratinib + fulvestrant + trastuzumab (N + F + T). We report results from 71 patients with HR+, HER2-mutant MBC, including 21 (seven in each arm) from a randomized substudy of fulvestrant versus fulvestrant + trastuzumab (F + T) versus N + F + T. PATIENTS AND METHODS: Patients with HR+ HER2-negative MBC with activating HER2 mutation(s) and prior cyclin-dependent kinase 4/6 inhibitor (CDK4/6i) therapy received N + F + T (oral neratinib 240 mg/day with loperamide prophylaxis, intramuscular fulvestrant 500 mg on days 1, 15, and 29 of cycle 1 then q4w, intravenous trastuzumab 8 mg/kg then 6 mg/kg q3w) or F + T or fulvestrant alone. Those whose disease progressed on F + T or fulvestrant could cross-over to N + F + T. Efficacy endpoints included investigator-assessed objective response rate (ORR), clinical benefit rate (RECIST v1.1), duration of response, and progression-free survival (PFS). Plasma and/or formalin-fixed paraffin-embedded tissue samples were collected at baseline; plasma was collected during and at end of treatment. Extracted DNA was analyzed by next-generation sequencing. RESULTS: ORR for 57 N + F + T-treated patients was 39% [95% confidence interval (CI) 26% to 52%); median PFS was 8.3 months (95% CI 6.0-15.1 months). No responses occurred in fulvestrant- or F + T-treated patients; responses in patients crossing over to N + F + T supported the requirement for neratinib in the triplet. Responses were observed in patients with ductal and lobular histology, 1 or ≥1 HER2 mutations, and co-occurring HER3 mutations. Longitudinal circulating tumor DNA sequencing revealed acquisition of additional HER2 alterations, and mutations in genes including PIK3CA, enabling further precision targeting and possible re-response. CONCLUSIONS: The benefit of N + F + T for HR+ HER2-mutant MBC after progression on CDK4/6is is clinically meaningful and, based on this study, N + F + T has been included in the National Comprehensive Cancer Network treatment guidelines. SUMMIT has improved our understanding of the translational implications of targeting HER2 mutations with neratinib-based therapy.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Fulvestrant , Receptor ErbB-2 , Trastuzumab
4.
ESMO Open ; 8(4): 101612, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37467659
5.
ESMO Open ; 8(3): 101562, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37196398
6.
ESMO Open ; 8(2): 101203, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37018871
7.
ESMO Open ; 8(1): 100763, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36603523
8.
ESMO Open ; 7(6): 100643, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36403559
9.
ESMO Open ; 7(4): 100556, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35961192
10.
ESMO Open ; 7(3): 100479, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35472677
11.
ESMO Open ; 7(2): 100411, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35219243
12.
ESMO Open ; 7(1): 100352, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34920292
13.
ESMO Open ; 6(6): 100285, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34752996
14.
ESMO Open ; 6(5): 100247, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34411970
15.
ESMO Open ; 6(3): 100186, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34098228
17.
ESMO Open ; 6(3): 100116, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33887688
18.
ESMO Open ; 6(2): 100061, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33639602
19.
Ann Oncol ; 32(5): 590-599, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33609722

RESUMEN

Gastroesophageal adenocarcinoma (GEA) and squamous esophageal cancer (ESCC) are responsible for >1 million deaths annually globally. Until now, patients with metastatic GEA and ESCC could anticipate survival of <1 year. Anti- programmed cell death protein 1 (anti-PD-1) monotherapy has demonstrated modest efficacy in previously treated GEA and ESCC. In 2020, four pivotal trials have established anti-PD-1 therapy as a new standard of care for selected GEA and ESCC patients as first-line advanced and adjuvant therapy. In this review, we discuss the recent results of the CheckMate 649, ATTRACTION-4, KEYNOTE-590 and CheckMate 577 trials. We consider these results in the context of current standards of care and historical trials of immune checkpoint blockade in GEA and ESCC. We explore biomarker selection for anti-PD-1 therapy and appraise the future of combination therapies. In CheckMate 649, treatment with oxaliplatin-fluoropyrimidine chemotherapy plus nivolumab in patients with combined positive score ≥5 GEA tumors provided a clinically meaningful and statistically significant improvement in overall survival. The ATTRACTION-4 trial did not see a similar overall survival benefit, despite a clear improvement in progression-free survival. We review potential explanations for this result. KEYNOTE-590 showed profoundly improved survival when pembrolizumab was added to cisplatin-fluoropyrimidine chemotherapy in ESCC patients with combined positive score ≥10 tumors; this benefit was less convincing in unselected ESCC. Finally, CheckMate 577 provides proof-of-concept for the improvement in disease-free survival with adjuvant nivolumab in high-risk resected GEA and ESCC following trimodality therapy. Immune checkpoint blockade has come of age in GEA and ESCC, and will now be integrated into first-line and earlier lines of therapy, providing benefit for a larger proportion of patients. Biomarker standardization will be critical to select the patients most likely to benefit from treatment. For patients with immune evasive tumors, novel combinations under development show promise; however, global trials are needed.


Asunto(s)
Neoplasias Esofágicas , Neoplasias Gástricas , Anticuerpos Monoclonales Humanizados , Terapia Combinada , Neoplasias Esofágicas/tratamiento farmacológico , Humanos , Nivolumab/uso terapéutico , Neoplasias Gástricas/tratamiento farmacológico
20.
ESMO Open ; 6(1): 100009, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33399075
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...