13 research outputs found

    KRAS-mutant non-small cell lung cancer (NSCLC) therapy based on tepotinib and omeprazole combination

    Full text link
    Background KRAS-mutant non-small cell lung cancer (NSCLC) shows a relatively low response rate to chemotherapy, immunotherapy and KRAS-G12C selective inhibitors, leading to short median progression-free survival, and overall survival. The MET receptor tyrosine kinase (c-MET), the cognate receptor of hepatocyte growth factor (HGF), was reported to be overexpressed in KRAS-mutant lung cancer cells leading to tumor-growth in anchorage-independent conditions. Methods Cell viability assay and synergy analysis were carried out in native, sotorasib and trametinib-resistant KRAS-mutant NSCLC cell lines. Colony formation assays and Western blot analysis were also performed. RNA isolation from tumors of KRAS-mutant NSCLC patients was performed and KRAS and MET mRNA expression was determined by real-time RT-qPCR. In vivo studies were conducted in NSCLC (NCI-H358) cell-derived tumor xenograft model. Results Our research has shown promising activity of omeprazole, a V-ATPase-driven proton pump inhibitor with potential anti-cancer properties, in combination with the MET inhibitor tepotinib in KRAS-mutant G12C and non-G12C NSCLC cell lines, as well as in G12C inhibitor (AMG510, sotorasib) and MEK inhibitor (trametinib)-resistant cell lines. Moreover, in a xenograft mouse model, combination of omeprazole plus tepotinib caused tumor growth regression. We observed that the combination of these two drugs downregulates phosphorylation of the glycolytic enzyme enolase 1 (ENO1) and the low-density lipoprotein receptor-related protein (LRP) 5/6 in the H358 KRAS G12C cell line, but not in the H358 sotorasib resistant, indicating that the effect of the combination could be independent of ENO1. In addition, we examined the probability of recurrence-free survival and overall survival in 40 early lung adenocarcinoma patients with KRAS G12C mutation stratified by KRAS and MET mRNA levels. Significant differences were observed in recurrence-free survival according to high levels of KRAS mRNA expression. Hazard ratio (HR) of recurrence-free survival was 7.291 (p = 0.014) for high levels of KRAS mRNA expression and 3.742 (p = 0.052) for high MET mRNA expression. Conclusions We posit that the combination of the V-ATPase inhibitor omeprazole plus tepotinib warrants further assessment in KRAS-mutant G12C and non G12C cell lines, including those resistant to the covalent KRAS G12C inhibitors

    Comprehensive cross-platform comparison of methods for non-invasive EGFR mutation testing : results of the RING observational trial.

    Get PDF
    Abstract Several platforms for noninvasive EGFR testing are currently used in the clinical setting with sensitivities ranging from 30% to 100%. Prospective studies evaluating agreement and sources for discordant results remain lacking. Herein, seven methodologies including two next-generation sequencing (NGS)-based methods, three high-sensitivity PCR-based platforms, and two FDA-approved methods were compared using 72 plasma samples, from EGFR-mutant non-small-cell lung cancer (NSCLC) patients progressing on a first-line tyrosine kinase inhibitor (TKI). NGS platforms as well as high-sensitivity PCR-based methodologies showed excellent agreement for EGFR-sensitizing mutations (K = 0.80-0.89) and substantial agreement for T790M testing (K = 0.77 and 0.68, respectively). Mutant allele frequencies (MAFs) obtained by different quantitative methods showed an excellent reproducibility (intraclass correlation coefficients 0.86-0.98). Among other technical factors, discordant calls mostly occurred at mutant allele frequencies (MAFs) ≤ 0.5%. Agreement significantly improved when discarding samples with MAF ≤ 0.5%. EGFR mutations were detected at significantly lower MAFs in patients with brain metastases, suggesting that these patients risk for a false-positive result. Our results support the use of liquid biopsies for noninvasive EGFR testing and highlight the need to systematically report MAFs. Keywords: NGS; circulating free DNA; epidermal growth factor receptor; non-small-cell lung cancer; osimertinib; tyrosine kinase inhibitor

    Epidemiological trends of HIV/HCV coinfection in Spain, 2015-2019

    Get PDF
    Altres ajuts: Spanish AIDS Research Network; European Funding for Regional Development (FEDER).Objectives: We assessed the prevalence of anti-hepatitis C virus (HCV) antibodies and active HCV infection (HCV-RNA-positive) in people living with HIV (PLWH) in Spain in 2019 and compared the results with those of four similar studies performed during 2015-2018. Methods: The study was performed in 41 centres. Sample size was estimated for an accuracy of 1%. Patients were selected by random sampling with proportional allocation. Results: The reference population comprised 41 973 PLWH, and the sample size was 1325. HCV serostatus was known in 1316 PLWH (99.3%), of whom 376 (28.6%) were HCV antibody (Ab)-positive (78.7% were prior injection drug users); 29 were HCV-RNA-positive (2.2%). Of the 29 HCV-RNA-positive PLWH, infection was chronic in 24, it was acute/recent in one, and it was of unknown duration in four. Cirrhosis was present in 71 (5.4%) PLWH overall, three (10.3%) HCV-RNA-positive patients and 68 (23.4%) of those who cleared HCV after anti-HCV therapy (p = 0.04). The prevalence of anti-HCV antibodies decreased steadily from 37.7% in 2015 to 28.6% in 2019 (p < 0.001); the prevalence of active HCV infection decreased from 22.1% in 2015 to 2.2% in 2019 (p < 0.001). Uptake of anti-HCV treatment increased from 53.9% in 2015 to 95.0% in 2019 (p < 0.001). Conclusions: In Spain, the prevalence of active HCV infection among PLWH at the end of 2019 was 2.2%, i.e. 90.0% lower than in 2015. Increased exposure to DAAs was probably the main reason for this sharp reduction. Despite the high coverage of treatment with direct-acting antiviral agents, HCV-related cirrhosis remains significant in this population

    Significado clínico-patológico de las mutaciones de los genes c-Kit y PDGFRa en tumores del estroma del tracto gastrointestinal

    No full text
    Los tumores del estroma gastrointestinal (GIST) se caracterizan por la hiperexpresión del receptor de membrana c-KIT (CD117) que se detecta en un 95% de los GIST. Recientemente, se ha descrito que entre el 5-7% de GIST expresan PDGFRα. En nuestro estudio se quiere observar la relación directa entre las mutaciones y tipo de mutaciones obtenidas en los dos genes a estudiar (c-KIT y PDGFRα) con la evolución clínico-histopatológica de los pacientes. Para ello se han recogido un total de 173 casos incluidos en parafina diagnosticados en el periodo comprendido entre los años 1985 y 2007. Del total de casos, 83 pertenecen al Hospital Clínico de Valencia, 24 casos de la Fundación Instituto Valenciano de Oncología y 65 casos de consulta. El estudio clínico está basado en el análisis de las historias clínicas de los pacientes; valorando los parámetros: sexo, edad, localización del tumor, tamaño tumoral, progresión tumoral y tratamiento. El análisis anatomopatológico se realizó sobre muestras de tejido tumoral incluidas en parafina; se realizaron técnicas convencionales de H&E para el recuento mitósico, histología, presencia de necrosis y pleomorfismo. Además son clasificados según el riesgo histológico siguiendo los criterios de Fletcher (mitosis y tamaño) o Miettinen que incluye además la localización. En el estudio inmunohistoquímico cabe destacar que los casos anteriores a 1998 se han estudiado en dos micromatrices titulares y el resto de casos en las laminillas convencionales. La inmunoreactividad se realiza en los marcadores c-KIT, Ki-67, MDM2, cromogranina, CD99, CD34, sinaptofisina, vimentina, P53, AML, desmina, S-100, PDGFRa. Para nuestro estudio de biología molecular necesitamos extraer ADN; posteriormente se lleva a cabo la PCR de los exones 9, 11, 13 y 17 del gen c-KIT, y 12 y 18 del PDGFRα, se secuencia y se estudian para caracterizar el tipo de alteración génica. A nivel de resultados, de los 173 casos, finalmente 143 son GIST. La mediana de seguimiento ha sido de 48 meses, con un mínimo de 0 días (debido al fallecimiento del paciente en el momento de la intervención) y un máximo de 245 meses. Los parámetros clínicos e histopatológicos han sido: 84 varones y 59 hembras; una mediana de edad de 63 años (24-86); con una localización mayoritaria en el estómago, seguido del intestino delgado y una mediana tumoral de 7,2cm (0,1-40cm); la histología celular mayoritaria es la fusocelular; la mediana de recuento mitósico ha sido de 4 (0-80mitosis en 50HPF). El marcador inmunohistoquímico por excelencia de los GIST es KIT (CD 117); destacando que un 91% de los tumores son positivos. Obteniendo en el resto de marcadores: 61,9% de positividad para PDGFRα, 52,9% para Ki-67 y 42,3% para P53. Después del estudio de los exones tanto de c-KIT como de PDGFRα hemos hallado un total de 98 mutaciones de los 143 pacientes (68%). 88 de las mutaciones (90%) se producen en el gen c-KIT y únicamente 10 mutaciones en el gen PDGFRα (10%). Las mutaciones en c-KIT ocurren en el exón 11 con un total de 75 mutaciones (80% del total de mutaciones), ocho en el exón 9 de c-KIT (8% del total de mutaciones), una en el exón 13 (1% del total de mutaciones) y dos en el 17(2%). Respecto al gen PDGFRα se han hallado 9 mutaciones en el exón 18 (9% del total de mutaciones). Se han obtenido como parámetros independientes de progresión la clasificación de Fletcher (p<0,0001), la histología del tumor (p=0,006), el marcador inmunohistoquímico Ki-67 (p=0,019), el recuento mitósico superior a 5 mitosis en 50 HPF (p=0,007) y la presencia de mutación en c-KIT (p=0,003). Y para la supervivencia el recuento mitósico (p=0,001) y la resección incompleta del tumor (p=0,003).Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the gastrointestinal tract. The tumors characteristically harbor c-KIT or PDGFRa mutations, and mutant tumors respond to imatinib mesylate. We have investigated the prognostic relevance of the type and position of the mutations, in addition to other clinicopathologic factors, in a large series of 145 patients with GIST. The median age for the group, 84 males and 59 females, was 63 years (range, 24 to 86 years The median tumor size was 7,2 cm (range, 2 to 26 cm), and there were 71 gastric tumors, 36 small intestinal, 18 extragastrointestinal and 18 with another primary location. In most tumors the mitotic rate was low: Spindle cell type was present in 97 of cases, 15 epithelioid and 24 mixed cell types. Regarding immunohistochemistry expression of c-KIT (CD117) was observed in 91% of the tumors. Mutations were detected in 98 tumors (68%): 88 cases involving c-KIT and 10 cases involving PDFGRa. The median follow-up was 52 months with an overrall survival of 45%. Fletcher classification, cellularity, Ki-67, mitotic rate and mutation in c-KIT was constituted independent prognostic factor for progression and mitotic rate and incomplete surgery to overall survival

    Cancer epigenetic biomarkers in liquid biopsy for high incidence malignancies

    Get PDF
    Simple Summary Apart from genetic changes, cancer is characterized by epigenetic alterations, which indicate modifications in the DNA (such as DNA methylation) and histones (such as methylation and acetylation), as well as gene expression regulation by non-coding (nc)RNAs. These changes can be used in biological fluids (liquid biopsies) for diagnosis, prognosis and prediction of cancer drug response. Although these alterations are not widely used as biomarkers in the clinical practice yet, increasing number of commercial kits and clinical trials are expected to prove that epigenetic changes are able to offer valuable information for cancer patients. Early alterations in cancer include the deregulation of epigenetic events such as changes in DNA methylation and abnormal levels of non-coding (nc)RNAs. Although these changes can be identified in tumors, alternative sources of samples may offer advantages over tissue biopsies. Because tumors shed DNA, RNA, and proteins, biological fluids containing these molecules can accurately reflect alterations found in cancer cells, not only coming from the primary tumor, but also from metastasis and from the tumor microenvironment (TME). Depending on the type of cancer, biological fluids encompass blood, urine, cerebrospinal fluid, and saliva, among others. Such samples are named with the general term "liquid biopsy" (LB)

    Cancer epigenetic biomarkers in liquid biopsy for high incidence malignancies

    No full text
    Simple Summary Apart from genetic changes, cancer is characterized by epigenetic alterations, which indicate modifications in the DNA (such as DNA methylation) and histones (such as methylation and acetylation), as well as gene expression regulation by non-coding (nc)RNAs. These changes can be used in biological fluids (liquid biopsies) for diagnosis, prognosis and prediction of cancer drug response. Although these alterations are not widely used as biomarkers in the clinical practice yet, increasing number of commercial kits and clinical trials are expected to prove that epigenetic changes are able to offer valuable information for cancer patients. Early alterations in cancer include the deregulation of epigenetic events such as changes in DNA methylation and abnormal levels of non-coding (nc)RNAs. Although these changes can be identified in tumors, alternative sources of samples may offer advantages over tissue biopsies. Because tumors shed DNA, RNA, and proteins, biological fluids containing these molecules can accurately reflect alterations found in cancer cells, not only coming from the primary tumor, but also from metastasis and from the tumor microenvironment (TME). Depending on the type of cancer, biological fluids encompass blood, urine, cerebrospinal fluid, and saliva, among others. Such samples are named with the general term "liquid biopsy" (LB)

    Prognostic Impact of let-7e MicroRNA and Its Target Genes in Localized High-Risk Intestinal GIST: A Spanish Group for Research on Sarcoma (GEIS) Study

    No full text
    MicroRNAs (miRNAs) are small non-coding RNAs that negatively regulate gene expression at the post-transcriptional level, and they have been described as being associated with tumor prognosis. Here, miRNA profiling was planned to explore new molecular prognostic biomarkers in localized intestinal high-risk GIST. Paraffin tumor blocks of 14 and 86 patients were used in the discovery and expansion sets, respectively. GeneChip miRNA v3.0 was employed to identify the miRNAs differentially expressed between relapsed and non-relapsed patient samples, which were validated in the expansion set, by qRT-PCR. RT2 Profiler PCR Array was used for the screening of let-7e targets. Expression levels were correlated with relapse-free survival and overall survival. In the discovery set, 39 miRNAs were significantly deregulated, let-7e and miR-550 being the most underexpressed and overexpressed miRNAs in the relapsed group, respectively. In the expansion set, the underexpression of let-7e or the overexpression of 4 of its target genes (ACVR1B, CASP3, COL3A1, and COL5A2) were statistically associated with worse relapse-free survival. The expression of let-7e and 4 of its target genes are potential prognostic biomarkers in high-risk localized intestinal GIST. The expression of these genes is a potential molecular tool useful for a more accurate prognosis in this subset of GIST patients.The project was funded by the Instituto de Salud Carlos III (ISCIII (Carlos III Health Institute))-Fondo Europeo de Desarrollo Regional; FEDER (European Regional Development Fund (ERDF)), through a public competitive call (project reference PI15/01254; principal investigator Javier Martín Broto) and by the Consejería de la Salud-Junta de Andalucia–(Health Council–Regional Government of Andalusia)-Proyectos de investigación coordinados 2016, through a competitive public call (project reference PC-0537-2016-0537; principal investigator Nadia Hindi).Ye

    Signature-driven repurposing of Midostaurin for combination with MEK1/2 and KRASG12C inhibitors in lung cancer

    No full text
    Drug combinations are key to circumvent resistance mechanisms compromising response to single anti-cancer targeted therapies. The implementation of combinatorial approaches involving MEK1/2 or KRASG12C inhibitors in the context of KRAS-mutated lung cancers focuses fundamentally on targeting KRAS proximal activators or effectors. However, the antitumor effect is highly determined by compensatory mechanisms arising in defined cell types or tumor subgroups. A potential strategy to find drug combinations targeting a larger fraction of KRAS-mutated lung cancers may capitalize on the common, distal gene expression output elicited by oncogenic KRAS. By integrating a signature-driven drug repurposing approach with a pairwise pharmacological screen, here we show synergistic drug combinations consisting of multi-tyrosine kinase PKC inhibitors together with MEK1/2 or KRASG12C inhibitors. Such combinations elicit a cytotoxic response in both in vitro and in vivo models, which in part involves inhibition of the PKC inhibitor target AURKB. Proteome profiling links dysregulation of MYC expression to the effect of both PKC inhibitor-based drug combinations. Furthermore, MYC overexpression appears as a resistance mechanism to MEK1/2 and KRASG12C inhibitors. Our study provides a rational framework for selecting drugs entering combinatorial strategies and unveils MEK1/2- and KRASG12C-based therapies for lung cancer
    corecore