Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 333
Filtrar
1.
Oncogene ; 40(43): 6143-6152, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34508175

RESUMEN

MYCN drives aggressive behavior and refractoriness to chemotherapy, in several tumors. Since MYCN inactivation in clinical settings is not achievable, alternative vulnerabilities of MYCN-driven tumors need to be explored to identify more effective and less toxic therapies. We previously demonstrated that PARP inhibitors enhance MYCN-induced replication stress and promote mitotic catastrophe, counteracted by CHK1. Here, we showed that PARP and CHK1 inhibitors synergized to induce death in neuroblastoma cells and in primary cultures of SHH-dependent medulloblastoma, their combination being more effective in MYCN amplified and MYCN overexpressing cells compared to MYCN non-amplified cells. Although the MYCN amplified IMR-32 cell line carrying the p.Val2716Ala ATM mutation showed the highest sensitivity to the drug combination, this was not related to ATM status, as indicated by CRISPR/Cas9-based correction of the mutation. Suboptimal doses of the CHK1 inhibitor MK-8776 plus the PARP inhibitor olaparib led to a MYCN-dependent accumulation of DNA damage and cell death in vitro and significantly reduced the growth of four in vivo models of MYCN-driven tumors, without major toxicities. Our data highlight the combination of PARP and CHK1 inhibitors as a new potential chemo-free strategy to treat MYCN-driven tumors, which might be promptly translated into clinical trials.


Asunto(s)
Neoplasias Cerebelosas/tratamiento farmacológico , Meduloblastoma/tratamiento farmacológico , Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/tratamiento farmacológico , Ftalazinas/administración & dosificación , Piperazinas/administración & dosificación , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Cerebelosas/genética , Neoplasias Cerebelosas/patología , Sinergismo Farmacológico , Femenino , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Meduloblastoma/genética , Meduloblastoma/patología , Ratones , Mutación , Neuroblastoma/genética , Neuroblastoma/patología , Ftalazinas/farmacología , Piperazinas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Cell Death Dis ; 12(9): 821, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34462431

RESUMEN

Metabolic reprogramming is an integral part of the growth-promoting program driven by the MYC family of oncogenes. However, this reprogramming also imposes metabolic dependencies that could be exploited therapeutically. Here we report that the pyrimidine biosynthetic enzyme dihydroorotate dehydrogenase (DHODH) is an attractive therapeutic target for MYCN-amplified neuroblastoma, a childhood cancer with poor prognosis. Gene expression profiling and metabolomic analysis reveal that MYCN promotes pyrimidine nucleotide production by transcriptional upregulation of DHODH and other enzymes of the pyrimidine-synthesis pathway. Genetic and pharmacological inhibition of DHODH suppresses the proliferation and tumorigenicity of MYCN-amplified neuroblastoma cell lines. Furthermore, we obtain evidence suggesting that serum uridine is a key factor in determining the efficacy of therapeutic agents that target DHODH. In the presence of physiological concentrations of uridine, neuroblastoma cell lines are highly resistant to DHODH inhibition. This uridine-dependent resistance to DHODH inhibitors can be abrogated by dipyridamole, an FDA-approved drug that blocks nucleoside transport. Importantly, dipyridamole synergizes with DHODH inhibition to suppress neuroblastoma growth in animal models. These findings suggest that a combination of targeting DHODH and nucleoside transport is a promising strategy to overcome intrinsic resistance to DHODH-based cancer therapeutics.


Asunto(s)
Dihidroorotato Deshidrogenasa/metabolismo , Amplificación de Genes , Terapia Molecular Dirigida , Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/genética , Nucleósidos/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Compuestos de Bifenilo/farmacología , Carbazoles/farmacología , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Amplificación de Genes/efectos de los fármacos , Humanos , Masculino , Ratones Endogámicos NOD , Ratones SCID , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/sangre , Neuroblastoma/patología , Transcripción Genética/efectos de los fármacos , Uridina/sangre
3.
Adv Drug Deliv Rev ; 176: 113900, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34324884

RESUMEN

The recent approval of messenger RNA (mRNA)-based vaccines to combat the SARS-CoV-2 pandemic highlights the potential of both conventional mRNA and self-amplifying mRNA (saRNA) as a flexible immunotherapy platform to treat infectious diseases. Besides the antigen it encodes, mRNA itself has an immune-stimulating activity that can contribute to vaccine efficacy. This self-adjuvant effect, however, will interfere with mRNA translation and may influence the desired therapeutic outcome. To further exploit its potential as a versatile therapeutic platform, it will be crucial to control mRNA's innate immune-stimulating properties. In this regard, we describe the mechanisms behind the innate immune recognition of mRNA and provide an extensive overview of strategies to control its innate immune-stimulating activity. These strategies range from modifications to the mRNA backbone itself, optimization of production and purification processes to the combination with innate immune inhibitors. Furthermore, we discuss the delicate balance of the self-adjuvant effect in mRNA vaccination strategies, which can be both beneficial and detrimental to the therapeutic outcome.


Asunto(s)
Amplificación de Genes/inmunología , Inmunidad Innata/inmunología , Inmunoterapia/métodos , ARN Mensajero/inmunología , Vacunas Sintéticas/inmunología , Animales , COVID-19/genética , COVID-19/inmunología , COVID-19/prevención & control , Amplificación de Genes/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Inmunoterapia/tendencias , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas de ARNm
4.
Cancer Invest ; 39(3): 235-239, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33538211

RESUMEN

Comprehensive molecular testing of individual tumors has led to the identification of novel molecularly defined cancer therapies and treatment indications. Given low frequencies of many molecular alterations, efficacy of therapies used to target them are often undefined, especially in the context of rare malignancies. Here we describe the first reported case of MET amplification in sinonasal undifferentiated carcinoma (SNUC), a rare cancer with a poor prognosis. The patient was treated with crizotinib, a tyrosine kinase inhibitor that targets c-MET, and experienced a complete response. Our report demonstrates the potential of employing precision oncology approaches in SNUC and other rare cancers.


Asunto(s)
Carcinoma/terapia , Crizotinib/farmacología , Neoplasias del Seno Maxilar/terapia , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biopsia , Carcinoma/diagnóstico por imagen , Carcinoma/genética , Carcinoma/patología , Femenino , Amplificación de Genes/efectos de los fármacos , Humanos , Neoplasias del Seno Maxilar/diagnóstico por imagen , Neoplasias del Seno Maxilar/genética , Neoplasias del Seno Maxilar/patología , Terapia Molecular Dirigida , Tomografía Computarizada por Tomografía de Emisión de Positrones , Proteínas Proto-Oncogénicas c-met/genética
5.
Cell ; 184(2): 384-403.e21, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33450205

RESUMEN

Many oncogenic insults deregulate RNA splicing, often leading to hypersensitivity of tumors to spliceosome-targeted therapies (STTs). However, the mechanisms by which STTs selectively kill cancers remain largely unknown. Herein, we discover that mis-spliced RNA itself is a molecular trigger for tumor killing through viral mimicry. In MYC-driven triple-negative breast cancer, STTs cause widespread cytoplasmic accumulation of mis-spliced mRNAs, many of which form double-stranded structures. Double-stranded RNA (dsRNA)-binding proteins recognize these endogenous dsRNAs, triggering antiviral signaling and extrinsic apoptosis. In immune-competent models of breast cancer, STTs cause tumor cell-intrinsic antiviral signaling, downstream adaptive immune signaling, and tumor cell death. Furthermore, RNA mis-splicing in human breast cancers correlates with innate and adaptive immune signatures, especially in MYC-amplified tumors that are typically immune cold. These findings indicate that dsRNA-sensing pathways respond to global aberrations of RNA splicing in cancer and provoke the hypothesis that STTs may provide unexplored strategies to activate anti-tumor immune pathways.


Asunto(s)
Antivirales/farmacología , Inmunidad/efectos de los fármacos , Empalmosomas/metabolismo , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/patología , Inmunidad Adaptativa/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Femenino , Amplificación de Genes/efectos de los fármacos , Humanos , Intrones/genética , Ratones , Terapia Molecular Dirigida , Proteínas Proto-Oncogénicas c-myc/metabolismo , Empalme del ARN/efectos de los fármacos , Empalme del ARN/genética , ARN Bicatenario/metabolismo , Transducción de Señal/efectos de los fármacos , Empalmosomas/efectos de los fármacos , Neoplasias de la Mama Triple Negativas/genética
6.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33443214

RESUMEN

Tandem gene amplification is a frequent and dynamic source of antibiotic resistance in bacteria. Ongoing expansions and contractions of repeat arrays during population growth are expected to manifest as cell-to-cell differences in copy number (CN). As a result, a clonal bacterial culture could comprise subpopulations of cells with different levels of antibiotic sensitivity that result from variable gene dosage. Despite the high potential for misclassification of heterogenous cell populations as either antibiotic-susceptible or fully resistant in clinical settings, and the concomitant risk of inappropriate treatment, CN distribution among cells has defied analysis. Here, we use the MinION single-molecule nanopore sequencer to uncover CN heterogeneity in clonal populations of Escherichia coli and Acinetobacter baumannii grown from single cells isolated while selecting for resistance to an optimized arylomycin, a member of a recently discovered class of Gram-negative antibiotic. We found that gene amplification of the arylomycin target, bacterial type I signal peptidase LepB, is a mechanism of unstable arylomycin resistance and demonstrate in E. coli that amplification instability is independent of RecA. This instability drives the emergence of a nonuniform distribution of lepB CN among cells with a range of 1 to at least 50 copies of lepB identified in a single clonal population. In sum, this remarkable heterogeneity, and the evolutionary plasticity it fuels, illustrates how gene amplification can enable bacterial populations to respond rapidly to novel antibiotics. This study establishes a rationale for further nanopore-sequencing studies of heterogeneous cell populations to uncover CN variability at single-molecule resolution.


Asunto(s)
Acinetobacter baumannii/genética , Antibacterianos/farmacología , Farmacorresistencia Microbiana/genética , Escherichia coli/genética , Amplificación de Genes/efectos de los fármacos , Proteínas de la Membrana/genética , Secuenciación de Nanoporos/métodos , Péptidos Cíclicos/genética , Serina Endopeptidasas/genética , Variaciones en el Número de Copia de ADN , Proteínas de Unión al ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Heterogeneidad Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Pruebas de Sensibilidad Microbiana , Mutación , Secuenciación de Nanoporos/instrumentación , Rec A Recombinasas/metabolismo
7.
Int J Mol Sci ; 22(2)2021 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-33435440

RESUMEN

Lung cancer is the leading cause of death for malignancy worldwide. Its molecular profiling has enriched our understanding of cancer initiation and progression and has become fundamental to provide guidance on treatment with targeted therapies. Testing the presence of driver mutations in specific genes in lung tumors has thus radically changed the clinical management and outcomes of the disease. Numerous studies performed with traditional sequencing methods have investigated the occurrence of such mutations in lung cancer, and new insights regarding their frequency and clinical significance are continuously provided with the use of last generation sequencing technologies. In this review, we discuss the molecular epidemiology of the main druggable genetic alterations in non-small cell lung cancer, namely EGFR, KRAS, BRAF, MET, and HER2 mutations or amplification, as well as ALK and ROS1 fusions. Furthermore, we investigated the predictive impact of these alterations on the outcomes of modern targeted therapies, their global prognostic significance, and their mutual interaction in cases of co-occurrence.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Mutación/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/epidemiología , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/epidemiología , Epidemiología Molecular , Terapia Molecular Dirigida , Proteínas de Fusión Oncogénica/genética
8.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-35008802

RESUMEN

Despite constant advances in the field of pediatric oncology, the survival rate of high-risk neuroblastoma patients remains poor. The molecular and genetic features of neuroblastoma, such as MYCN amplification and stemness status, have established themselves not only as potent prognostic and predictive factors but also as intriguing targets for personalized therapy. Novel thiosemicarbazones target both total level and activity of a number of proteins involved in some of the most important signaling pathways in neuroblastoma. In this study, we found that di-2-pyridylketone 4-cyclohexyl-4-methyl-3-thiosemicarbazone (DpC) potently decreases N-MYC in MYCN-amplified and c-MYC in MYCN-nonamplified neuroblastoma cell lines. Furthermore, DpC succeeded in downregulating total EGFR and phosphorylation of its most prominent tyrosine residues through the involvement of NDRG1, a positive prognostic marker in neuroblastoma, which was markedly upregulated after thiosemicarbazone treatment. These findings could provide useful knowledge for the treatment of MYC-driven neuroblastomas that are unresponsive to conventional therapies.


Asunto(s)
Quelantes del Hierro/farmacología , Neuroblastoma/metabolismo , Piridinas/farmacología , Transducción de Señal , Tiosemicarbazonas/farmacología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Forma de la Célula/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Receptores ErbB/metabolismo , Amplificación de Genes/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Biológicos , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/patología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estrés Fisiológico/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
9.
Int J Mol Sci ; 23(1)2021 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-35008821

RESUMEN

Tropomyosin receptor kinase (TRK) and receptor tyrosine kinase (RTK class VII) expression are important in many human diseases, especially cancers, including colorectal, lung, and gastric cancer. Using RNA sequencing analysis, we evaluated the mRNA expression and mutation profiles of gastric cancer patients with neurotropic tropomyosin receptor kinase (NTRK) 1-3 overexpression (defined as a ≥2.0-fold change). Furthermore, we screened eight TRK inhibitors in NCI-N87, SNU16, MKN28, MKN7, and AGS cells. Among these inhibitors, entrectinib showed the highest inhibitory activity; therefore, this drug was selected for analysis of its therapeutic mechanisms in gastric cancer. Entrectinib treatment induced apoptosis in NTRK1-3-expressing and VEGFR2-expressing NCI-N87 and AGS cells, but it had no effect on NTRK1-3-, VEGFR2-, TGFBR1-, and CD274-expressing MKN7 cells. SNU16 and MKN28 cells with low NTRK1-3 expression were not affected by entrectinib. Therefore, a mechanistic study was conducted in NCI-N87 (high expression of NTRK1-3 but mutation of NTRK3), AGS (high expression of NTRK1-3) and MKN28 (low expression of NTRK1-3) gastric cancer cell lines. Entrectinib treatment significantly reduced expression levels of phosphorylated NFκB, AKT, ERK, and ß-catenin in NCI-N87 and AGS cells, whereas it upregulated the expression levels of ECAD in NCI-N87 cells. Together, these results suggest that entrectinib has anti-cancer activity not only in GC cells overexpressing pan NTRK but also in VEGFR2 GC cells via the inhibition of the pan NTRK and VEGFR signaling pathways.


Asunto(s)
Apoptosis , Benzamidas/farmacología , Transición Epitelial-Mesenquimal , Indazoles/farmacología , Receptor trkA/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Adulto , Anciano , Anciano de 80 o más Años , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptor trkA/antagonistas & inhibidores , Neoplasias Gástricas/genética
10.
Cell Death Dis ; 11(12): 1029, 2020 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-33268769

RESUMEN

Medulloblastoma (MB) is the most common malignant pediatric brain tumor that can be categorized into four major molecular subgroups. Group 3 MB with MYC amplification (MYCamp-G3-MB) has been shown to be highly aggressive and exhibited worst prognosis, indicating the need for novel effective therapy most urgently. A few epigenetic targeted therapeutic strategies have recently been proven to effectively treat preclinical models of MYCamp-G3-MB, including BET inhibition, HDAC inhibition and SETD8 inhibition, unveiling a promising direction for further investigation. In this study, we carried out systemic bioinformatic analyses of public-available MB datasets as well as functional genomic screening datasets of primary MYCamp-G3-MB lines to search for other potential therapeutic targets within epigenetic modulators. We identified SSRP1, a subunit of histone-chaperone FACT complex, to be the top drug target candidate as it is highly cancer-dependent in whole-genome CRISPR-Cas9 screening across multiple MYCamp-G3-MB lines; significantly upregulated in MYCamp-G3-MB compared to normal cerebellum and most of the rest MB subtypes; its higher expression is correlated with worse prognosis; and it has a blood-brain-barrier penetrable targeted drug that has entered early phase human clinical trials already. Then we utilized RNA-interference approach to verify the cancer-dependency of SSRP1 in multiple MYCamp-G3-MB lines and further confirmed the therapeutic efficacy of FACT-targeted curaxin drug CBL0137 on treating preclinical models of MYCamp-G3-MB in vitro and in vivo, including an orthotopic intracranial xenograft model. Mechanistically, transcriptome analyses showed CBL0137 preferentially suppressed cell-cycle and DNA-repair related biological processes. Moreover, it selectively disrupted transcription of MYC and NEUROD1, two critical oncogenic transcription factors of MYCamp-G3-MB, via depleting FACT complex from their promoter regions. In summary, our study demonstrates FACT-targeted CBL0137 works effectively on treating MYCamp-G3-MB, presenting another promising epigenetic-targeted therapeutic strategy against the most devastating form of MB.


Asunto(s)
Carbazoles/uso terapéutico , Proteínas de Unión al ADN/metabolismo , Amplificación de Genes , Proteínas del Grupo de Alta Movilidad/metabolismo , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/genética , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Elongación Transcripcional/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carbazoles/farmacología , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/antagonistas & inhibidores , Humanos , Meduloblastoma/patología , Ratones Desnudos , Factores de Transcripción Otx/metabolismo , Pronóstico , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética/efectos de los fármacos , Factores de Elongación Transcripcional/antagonistas & inhibidores , Transcriptoma/genética
11.
Sci Rep ; 10(1): 18864, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139774

RESUMEN

Kochia, a major weed species, has evolved resistance to four herbicide modes of action. Herbicide resistance appears to spread quickly, which could result in diminished standing genetic variation, reducing the ability of populations to adapt further. Here we used double digest restriction enzyme associated sequencing to determine the level of gene flow among kochia populations and whether selection for glyphosate resistance reduces genetic variation. Canadian Prairie populations show little to no genetic differentiation (FST = 0.01) and no correlation between genetic and geographic distance (r2 = - 0.02 p = 0.56), indicating high gene flow and no population structure. There is some evidence that kochia populations are genetically depauperate compared to other weed species, but genetic diversity did not differ between glyphosate susceptible and resistant populations or individuals. The inbreeding coefficients suggest there are 23% fewer heterozygotes in these populations than expected with random mating, and no variation was found within the chloroplast. These results indicate that any alleles for herbicide resistance can be expected to spread quickly through kochia populations, but there is little evidence this spread will reduce the species' genetic variation or limit the species' ability to respond to further selection pressure.


Asunto(s)
Chenopodiaceae/genética , Variación Genética/genética , Resistencia a los Herbicidas/genética , 3-Fosfoshikimato 1-Carboxiviniltransferasa/genética , Canadá , Chenopodiaceae/efectos de los fármacos , Variaciones en el Número de Copia de ADN/genética , Amplificación de Genes/efectos de los fármacos , Dosificación de Gen/genética , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Flujo Génico , Glicina/análogos & derivados , Glicina/farmacología , Herbicidas/farmacología , Glifosato
12.
Medicine (Baltimore) ; 99(46): e23053, 2020 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-33181670

RESUMEN

The human epidermal growth factor receptor 2 (HER2) is amplified in approximately 20% of breast cancers, and HER2 receptor targeting therapy is associated with a significant improvement in disease-free and overall survival. In several clinical trials, the pathologic complete response (pCR) rate was significantly increased with combined pertuzumab and trastuzumab treatment in HER2-amplified breast cancer. Although the efficacy and safety of anti-HER2 dual blockade therapy has been reported, the markers that predict the response are still unclear. This study aimed to investigate the relationship between the level of HER2 amplification and the pCR in trastuzumab and pertuzumab neoadjuvant therapy.Twenty-two HER2-amplified early breast cancer patients who had received neoadjuvant docetaxel, carboplatin, trastuzumab, and pertuzumab (TCHP) therapy were included in this study. HER2/CEP17 ratio and average HER2 copy number were measured by fluorescence in situ hybridization analysis. The relationship between level of HER2 amplification and tumor pCR status was investigated.The median age was 47.5 years (range, 36-62). 31.8% of the patients were hormone receptor (HR) positive and 68.2%% of the patients were HR negative. The pCR (ypN0/is ypN0) rate in the breast and axilla was 68.2%. The patients who experienced a pCR had a median HER2/CEP17 ratio of 7.08 (range, 3.16-10.40) and average HER2 copy number of 17.00 (range, 5.85-37.50). The patients who did not experience a pCR had a median ratio of 4.70 (range, 1.06-9.00) and median HER2 copy number of 12.00 (range, 5.85-20.95) (P = .030, P = .174), respectively.pCR was highly correlated with HER2/CEP17 ratio in neoadjuvant anti-HER2 dual blockade. This suggests that the HER2/CEP17 ratio can be used as a predictive marker for pCR in neoadjuvant trastuzumab and pertuzumab therapy.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Receptor ErbB-2/efectos de los fármacos , Trastuzumab/uso terapéutico , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Carboplatino/uso terapéutico , Cromosomas Humanos Par 17 , Docetaxel/uso terapéutico , Femenino , Amplificación de Genes/efectos de los fármacos , Dosificación de Gen , Humanos , Hibridación Fluorescente in Situ , Persona de Mediana Edad , Terapia Neoadyuvante , Resultado del Tratamiento
13.
Cancer Sci ; 111(10): 3813-3823, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32735723

RESUMEN

The third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) osimertinib is approved for untreated, or previously EGFR-TKI-treated T790M-positive EGFR-mutated non-small cell lung carcinoma (NSCLC). We investigated the heterogeneity of responses to osimertinib and its underlying mechanisms. A patient with EGFR-L858R-mutated NSCLC was treated with erlotinib. Following treatment, he developed brain and multiple bone metastases and was eventually diagnosed with NSCLC with EGFR-T790M mutation. The responses of various tumor specimens to osimertinib were heterogeneous. We investigated EGFR-T790M and MET amplification using PCR and FISH in autopsy specimens of the cervical spine, lumbar spine, and brain. We established the KNZ osimertinib-resistant (KNZ_OR) tumor cell line with MET amplification using a cervical spine lesion that was intrinsically resistant to osimertinib. We evaluated the effects of MET knockdown and MET inhibitor on KNZ_OR cell sensitivity to osimertinib in vitro and in vivo. Osimertinib-resistant lesions (cervical spine and brain) showed EGFR-L858R and MET amplification, but not EGFR-T790M, whereas osimertinib-sensitive lesions (lumbar spine) showed EGFR-L858R and -T790, but not MET amplification. Osimertinib decreased the association of amplified MET with L858R-mutated EGFR but increased that with human epidermal growth factor receptor 3 in KNZ_OR cells. MET knockdown or MET inhibitor sensitized KNZ_OR cells to osimertinib in vitro, indicating that MET amplification induced osimertinib resistance. Combination with osimertinib plus crizotinib induced tumor shrinkage in the KNZ_OR xenograft model. Hence, MET amplification might induce heterogeneous responses to osimertinib in EGFR-mutated NSCLC. Further investigations on mutated EGFR and amplified MET might lead to the development of effective therapies.


Asunto(s)
Acrilamidas/administración & dosificación , Compuestos de Anilina/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-met/genética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Receptores ErbB/genética , Femenino , Amplificación de Genes/efectos de los fármacos , Xenoinjertos , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Persona de Mediana Edad , Mutación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/efectos adversos
14.
Cancer ; 126(17): 4002-4012, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32584426

RESUMEN

BACKGROUND: A high percentage of small cell lung cancer (SCLC) cases harbor cell cycle-related gene mutations and RICTOR amplification. Based on underlying somatic mutations, the authors have conducted a phase 2 biomarker-driven, multiarm umbrella study. METHODS: The SCLC Umbrella Korea StudiES (SUKSES) is an adaptive platform trial that undergoes continual modification according to the observed outcomes. This study included 286 patients with SCLC who failed platinum therapy and who had known genomic profiles based on a predesigned screening trial. Patients with MYC amplification or CDKN2A and TP53 co-alterations were allocated to adavosertib (SUKSES protocol C [SUKSES-C]; 7 patients) and those with RICTOR amplification were allocated to vistusertib (SUKSES-D; 4 patients). Alternatively, patients who were without any predefined biomarkers were assigned to a non-biomarker-selected arm: adavosertib (SUKSES-N1; 21 patients) or AZD2811NP (SUKSES-N3; 15 patients). RESULTS: Patients in the SUKSES-C and SUKSES-N1 arms demonstrated no objective response. Three patients presented with stable disease (SD) in SUKSES-C and 6 patients in SUKSES-N1. The median progression-free survival (PFS) was 1.3 months (95% confidence interval, 0.9 months to not available) for SUKSES-C and 1.2 months (95% CI, 1.1-1.4 months) for SUKSES-N1. Patients in the SUKSES-D arm demonstrated no objective response and no SD, with a PFS of 1.2 months (95% CI, 1.0 months to not available). The SUKSES-N3 arm had 5 patients with SD and a PFS of 1.6 months (95% CI, 0.9-1.7 months), without an objective response. Grade≥3 adverse events (graded according to National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.03]) were observed as follows: 3.2% in the SUKSES-C and SUKSES-N1 arms and 50.0% in the SUKSES-D arm. Target-related neutropenia (grade≥3) was observed in approximately 60.0% of patients in the AZD2811NP arm using the current dosing schedule. CONCLUSIONS: To the best of the authors' knowledge, the current study is the first biomarker-driven umbrella study conducted in patients with recurrent SCLC. Although the current study demonstrated the limited clinical efficacy of monotherapy, novel biomarker approaches using other cell cycle inhibitor(s) or combinations warrant further investigation.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-myc/genética , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Proteína p53 Supresora de Tumor/genética , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Benzamidas/administración & dosificación , Biomarcadores de Tumor/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Femenino , Amplificación de Genes/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Morfolinas/administración & dosificación , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Platino (Metal)/efectos adversos , Supervivencia sin Progresión , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Pirimidinonas/administración & dosificación , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología
15.
Biochem Biophys Res Commun ; 526(3): 586-591, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32247608

RESUMEN

Taxanes are applied as potent chemotherapeutic agents in the treatment of patients with esophageal cancer, but their usefulness is limited, partly because of acquisition of chemoresistance. In our previous study, we established three taxane resistant esophageal cancer cell lines; significant ABCB1 upregulations were found in all three. However, the responsible mechanism(s) still remains an open question. In this study, we explored possible mechanisms that might contribute to upregulation of ABCB1 in taxane resistant cells. ABCB1 gene amplification was found in taxane resistant cell line RTE-1P, but expressional upregulation cannot be explained only by gene amplification, because gene amplification is one order of magnitude or less whereas gene expression is more than two orders of magnitude. In the parental TE-1, ABCB1 expression was upregulated after treatment with 5-azadeoxycytidine and/or trichostatin A; epigenetic mechanisms may be deeply involved. ABCB1 has two promoters; a downstream promoter was found to play the dominant role in taxane resistant esophageal cancer cell lines. Analyses of CpG islands demonstrated that taxane resistant cells showed unmethylated CGI whereas parental cells were dominantly methylated. In conclusion, we propose that both the ABCB1 gene amplification and aberrations in epigenetic mechanisms are responsible for acquisition of taxane resistance in esophageal cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Hidrocarburos Aromáticos con Puentes/farmacología , Resistencia a Antineoplásicos , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Taxoides/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Línea Celular Tumoral , Epigénesis Genética/efectos de los fármacos , Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos
16.
Cancer Discov ; 10(2): 306-325, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31776131

RESUMEN

Acquired chromosomal DNA copy gains are a feature of many tumors; however, the mechanisms that underpin oncogene amplification are poorly understood. Recent studies have begun to uncover the importance of epigenetic states and histone lysine methyltransferases (KMT) and demethylases (KDM) in regulating transient site-specific DNA copy-number gains (TSSG). In this study, we reveal a critical interplay between a myriad of lysine methyltransferases and demethylases in modulating H3K4/9/27 methylation balance to control extrachromosomal amplification of the EGFR oncogene. This study further establishes that cellular signals (hypoxia and EGF) are able to directly promote EGFR amplification through modulation of the enzymes controlling EGFR copy gains. Moreover, we demonstrate that chemical inhibitors targeting specific KMTs and KDMs are able to promote or block extrachromosomal EGFR amplification, which identifies potential therapeutic strategies for controlling EGFR copy-number heterogeneity in cancer, and, in turn, drug response. SIGNIFICANCE: This study identifies a network of epigenetic factors and cellular signals that directly control EGFR DNA amplification. We demonstrate that chemical inhibitors targeting enzymes controlling this amplification can be used to rheostat EGFR copy number, which uncovers therapeutic opportunities for controlling EGFR DNA amplification heterogeneity and the associated drug response.This article is highlighted in the In This Issue feature, p. 161.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Metilación de ADN/genética , Histonas/metabolismo , Neoplasias/genética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Histona Demetilasas con Dominio de Jumonji/metabolismo , Lisina/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
17.
Nat Commun ; 10(1): 2030, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-31048689

RESUMEN

Acquired resistance to MEK1/2 inhibitors (MEKi) arises through amplification of BRAFV600E or KRASG13D to reinstate ERK1/2 signalling. Here we show that BRAFV600E amplification and MEKi resistance are reversible following drug withdrawal. Cells with BRAFV600E amplification are addicted to MEKi to maintain a precise level of ERK1/2 signalling that is optimal for cell proliferation and survival, and tumour growth in vivo. Robust ERK1/2 activation following MEKi withdrawal drives a p57KIP2-dependent G1 cell cycle arrest and senescence or expression of NOXA and cell death, selecting against those cells with amplified BRAFV600E. p57KIP2 expression is required for loss of BRAFV600E amplification and reversal of MEKi resistance. Thus, BRAFV600E amplification confers a selective disadvantage during drug withdrawal, validating intermittent dosing to forestall resistance. In contrast, resistance driven by KRASG13D amplification is not reversible; rather ERK1/2 hyperactivation drives ZEB1-dependent epithelial-to-mesenchymal transition and chemoresistance, arguing strongly against the use of drug holidays in cases of KRASG13D amplification.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/genética , Masculino , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Privación de Tratamiento , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
18.
Eur J Cancer ; 112: 66-79, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30947024

RESUMEN

BACKGROUND: Induction chemotherapy plays an important role in the management of patients with high-risk neuroblastoma. Predictors of response to induction therapy are largely lacking. We sought to describe clinical and biological features associated with induction response. METHODS: Patients from four consecutive COG high-risk trials were included. Response was evaluated by the 1993 International Neuroblastoma Response Criteria. The primary end-point was end-induction partial response (PR) or better. Univariate analyses were performed to compare response as a function of clinical or biologic predictors. A multivariate logistic regression model using significant predictors from univariate analyses was constructed to model PR or better. RESULTS: The analytic cohort included 1242 patients. End-induction response ≥PR was significantly associated with higher event-free and overall survival. Baseline factors associated with ≥PR included age <18 months (87.4% with ≥PR vs. 78.7% if older; p = 0.0103), International Neuroblastoma Staging System non-stage 4 (89.0% vs. 78.4% if stage 4; p = 0.0016), MYCN amplification (85.5% vs. 77.1% if non-amplified; p = 0.0006), 1p loss of heterozygosity (LOH; 85.6% vs. 76.0% if no LOH; p = 0.0085), no 11q LOH (84.8% vs. 70.9% if 11q LOH; p = 0.0004) and high mitosis-karyorrhexis index (MKI; 84.5% vs. 77.5% if low-intermediate MKI; p = 0.0098). On multivariable analysis (n = 407), the absence of 11q LOH was the only factor that remained significantly associated with ≥PR (odds ratio: 1.962 vs. 11q LOH; 95% confidence interval 1.104-3.487; p = 0.0216). CONCLUSIONS: Improved end-induction response in high-risk neuroblastoma is associated with longer survival. Patients with 11q LOH are less likely to respond to induction therapies and should be prioritised for novel approaches in future trials.


Asunto(s)
Neuroblastoma/tratamiento farmacológico , Preescolar , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Amplificación de Genes/efectos de los fármacos , Amplificación de Genes/genética , Humanos , Lactante , Modelos Logísticos , Pérdida de Heterocigocidad/efectos de los fármacos , Pérdida de Heterocigocidad/genética , Masculino , Proteína Proto-Oncogénica N-Myc/genética , Terapia Neoadyuvante/métodos , Estadificación de Neoplasias/métodos , Neuroblastoma/genética , Neuroblastoma/patología , Proteínas Nucleares/genética , Pronóstico , Factores de Riesgo
19.
PLoS One ; 14(4): e0215504, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31009485

RESUMEN

Inorganic arsenic is an environmental human carcinogen of several organs including the urinary tract. RWPE-1 cells are immortalized, non-tumorigenic, human prostate epithelia that become malignantly transformed into the CAsE-PE line after continuous in vitro exposure to 5µM arsenite over a period of months. For insight into in vitro arsenite transformation, we performed RNA-seq for differential gene expression and targeted sequencing of KRAS. We report >7,000 differentially expressed transcripts in CAsE-PE cells compared to RWPE-1 cells at >2-fold change, q<0.05 by RNA-seq. Notably, KRAS expression was highly elevated in CAsE-PE cells, with pathway analysis supporting increased cell proliferation, cell motility, survival and cancer pathways. Targeted DNA sequencing of KRAS revealed a mutant specific allelic imbalance, 'MASI', frequently found in primary clinical tumors. We found high expression of a mutated KRAS transcript carrying oncogenic mutations at codons 12 and 59 and many silent mutations, accompanied by lower expression of a wild-type allele. Parallel cultures of RWPE-1 cells retained a wild-type KRAS genotype. Copy number analysis and sequencing showed amplification of the mutant KRAS allele. KRAS is expressed as two splice variants, KRAS4a and KRAS4b, where variant 4b is more prevalent in normal cells compared to greater levels of variant 4a seen in tumor cells. 454 Roche sequencing measured KRAS variants in each cell type. We found KRAS4a as the predominant transcript variant in CAsE-PE cells compared to KRAS4b, the variant expressed primarily in RWPE-1 cells and in normal prostate, early passage, primary epithelial cells. Overall, gene expression data were consistent with KRAS-driven proliferation pathways found in spontaneous tumors and malignantly transformed cell lines. Arsenite is recognized as an important environmental carcinogen, but it is not a direct mutagen. Further investigations into this in vitro transformation model will focus on genomic events that cause arsenite-mediated mutation and overexpression of KRAS in CAsE-PE cells.


Asunto(s)
Arsenitos/envenenamiento , Transformación Celular Neoplásica/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Amplificación de Genes/efectos de los fármacos , Mutación , Próstata/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Carcinógenos Ambientales/envenenamiento , Línea Celular , Transformación Celular Neoplásica/genética , Células Epiteliales/metabolismo , Exones/genética , Amplificación de Genes/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Masculino , Próstata/patología
20.
Cell Oncol (Dordr) ; 42(4): 449-458, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30838525

RESUMEN

PURPOSE: Despite the development of molecular targeted therapies, few advances have been made in the treatment of lung squamous cell carcinoma (SCC). SOX2 amplification is one of the most common genetic alterations in SCC. Here, we investigated the effects of THZ1, a potent cyclin-dependent kinase 7 (CDK7) inhibitor that plays a key role in gene transcription, in SCC. METHODS: Lung SCC-derived cell viabilities were assessed using a CCK-8 assay. SOX2 expression and RNAPII-CTD phosphorylation levels after THZ1 treatment were determined by Western blotting. The effect of SOX2 suppression using shRNA was assessed by flow cytometry. Gene expression patterns after THZ1 treatment of lung SCC-derived cells were identified using microarray-based mRNA profiling. RESULTS: We found that THZ1 treatment led to suppression of cell growth and apoptotic cell death in SOX2-amplified SCC-derived cells only, whereas the modest growth-inhibitory effect of cisplatin did not differ according to SOX2 amplification status. We also found that THZ1 decreased the phosphorylation of the carboxyl-terminal domain of RNA polymerase II and the expression of several genes. Specifically, we found that the expression of transcription-associated genes, including SOX2, was down-regulated by THZ1 in SOX2-amplified SCC cells. This inhibition of SOX2 expression resulted in suppression of the growth of these cells. CONCLUSIONS: From our data, we conclude that THZ1 may effectively control the proliferation and survival of SOX2-amplified SCC cells through a decrease in global transcriptional activity, suggesting that CDK7 inhibition leading to transcription suppression may be a promising therapeutic option for lung SCC with a SOX2 amplification.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Amplificación de Genes , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Factores de Transcripción SOXB1/genética , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Amplificación de Genes/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , ARN Polimerasa II/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transcripción Genética/efectos de los fármacos , Quinasa Activadora de Quinasas Ciclina-Dependientes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...