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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Genomics ; 17(1): 64, 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37454130

RESUMEN

BACKGROUND: Female breast cancer remains the second leading cause of cancer-related death in the USA. The heterogeneity in the tumor morphology across the cohort and within patients can lead to unpredictable therapy resistance, metastasis, and clinical outcome. Hence, supplementing classic pathological markers with intrinsic tumor molecular markers can help identify novel molecular subtypes and the discovery of actionable biomarkers. METHODS: We conducted a large multi-institutional genomic analysis of paired normal and tumor samples from breast cancer patients to profile the complex genomic architecture of breast tumors. Long-term patient follow-up, therapeutic regimens, and treatment response for this cohort are documented using the Breast Cancer Collaborative Registry. The majority of the patients in this study were at tumor stage 1 (51.4%) and stage 2 (36.3%) at the time of diagnosis. Whole-exome sequencing data from 554 patients were used for mutational profiling and identifying cancer drivers. RESULTS: We identified 54 tumors having at least 1000 mutations and 185 tumors with less than 100 mutations. Tumor mutational burden varied across the classified subtypes, and the top ten mutated genes include MUC4, MUC16, PIK3CA, TTN, TP53, NBPF10, NBPF1, CDC27, AHNAK2, and MUC2. Patients were classified based on seven biological and tumor-specific parameters, including grade, stage, hormone receptor status, histological subtype, Ki67 expression, lymph node status, race, and mutational profiles compared across different subtypes. Mutual exclusion of mutations in PIK3CA and TP53 was pronounced across different tumor grades. Cancer drivers specific to each subtype include TP53, PIK3CA, CDC27, CDH1, STK39, CBFB, MAP3K1, and GATA3, and mutations associated with patient survival were identified in our cohort. CONCLUSIONS: This extensive study has revealed tumor burden, driver genes, co-occurrence, mutual exclusivity, and survival effects of mutations on a US Midwestern breast cancer cohort, paving the way for developing personalized therapeutic strategies.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Pronóstico , Mutación , Biomarcadores de Tumor/genética , Fosfatidilinositol 3-Quinasa Clase I/genética
2.
J Pathol ; 260(3): 289-303, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37186300

RESUMEN

Breast cancer invasion and metastasis result from a complex interplay between tumor cells and the tumor microenvironment (TME). Key oncogenic changes in the TME include aberrant synthesis, processing, and signaling of hyaluronan (HA). Hyaluronan-mediated motility receptor (RHAMM, CD168; HMMR) is an HA receptor enabling tumor cells to sense and respond to this aberrant TME during breast cancer progression. Previous studies have associated RHAMM expression with breast tumor progression; however, cause and effect mechanisms are incompletely established. Focused gene expression analysis of an internal breast cancer patient cohort confirmed that increased RHAMM expression correlates with aggressive clinicopathological features. To probe mechanisms, we developed a novel 27-gene RHAMM-related signature (RRS) by intersecting differentially expressed genes in lymph node (LN)-positive patient cases with the transcriptome of a RHAMM-dependent model of cell transformation, which we validated in an independent cohort. We demonstrate that the RRS predicts for poor survival and is enriched for cell cycle and TME-interaction pathways. Further analyses using CRISPR/Cas9-generated RHAMM-/- breast cancer cells provided direct evidence that RHAMM promotes invasion in vitro and in vivo. Immunohistochemistry studies highlighted heterogeneous RHAMM protein expression, and spatial transcriptomics associated the RRS with RHAMM-high microanatomic foci. We conclude that RHAMM upregulation leads to the formation of 'invasive niches', which are enriched in RRS-related pathways that drive invasion and could be targeted to limit invasive progression and improve patient outcomes. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias de la Mama , Humanos , Femenino , Neoplasias de la Mama/patología , Ácido Hialurónico/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Receptores de Hialuranos/metabolismo , Microambiente Tumoral
3.
Support Care Cancer ; 27(4): 1365-1373, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30112722

RESUMEN

SIGNIFICANCE: Poor sleep quality is a common and persistent problem reported by women with breast cancer (BC). Empirical evidence identifies many risk factors for self-reported sleep deficiency, but inconsistencies limit translation to practice. PURPOSE: To increase understanding of risk factors predicting self-reported poor sleep quality in women with BC who completed the Breast Cancer Collaborative Registry (BCCR) questionnaire. METHODS: This cross-sectional study recruited women with a first diagnosis of BC (n = 1302) at five sites in Nebraska and South Dakota. Women completed the BCCR that includes numerous variables as well as the Pittsburgh Sleep Quality Index (PSQI) and SF36v2 (n = 1260). Descriptive statistics and non-parametric correlations were used to determine associations and create predictive models of sleep quality with BCCR variables and SF36v2 subscales. RESULTS: Most women were white (93.7%) and married (71.5%); mean age was 60.1 (21-90) years. Poor sleep was self-reported by 53% of women. Seven variables were highly associated with sleep quality (p ≤ 0.001). The first model found younger age, lower physical activity, and higher fatigue were the strongest combined and independent variables predicting poor sleep quality (F = 23.0 (p < .001), R2 = 0.103). Participants self-reported lower health status on most SF36v2 subscales [Z = 44.9 (11.6) to 49.1 (10.1)]. A second model found that all subscales were predictors of poor sleep; vitality, mental health, bodily pain, and general health were the strongest predictors (F = 101.3 (p < .001), R2 = 0.26). CONCLUSIONS: Results confirm previously identified risk factors and reveal inconsistencies in other variables. Clinicians need to routinely screen for the identified risk factors of self-reported poor sleep quality.


Asunto(s)
Neoplasias de la Mama/fisiopatología , Dolor en Cáncer/complicaciones , Dolor en Cáncer/fisiopatología , Sistema de Registros , Trastornos del Sueño-Vigilia/etiología , Trastornos del Sueño-Vigilia/fisiopatología , Sueño/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/psicología , Dolor en Cáncer/psicología , Estudios Transversales , Fatiga/etiología , Fatiga/psicología , Femenino , Humanos , Persona de Mediana Edad , Calidad de Vida , Autoinforme , Trastornos del Sueño-Vigilia/psicología , Encuestas y Cuestionarios , Adulto Joven
4.
Breast J ; 25(5): 948-952, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31187577

RESUMEN

There are less data available on the effect of the ACA on breast cancer care beyond the screening level. A retrospective review at participating iCaRe2/BCCR institutions was completed before and after ACA. Post-ACA, patients were older, more urban, and more likely to be insured through Medicaid. Increased imaging use was noted post-ACA. These patients were less likely to be diagnosed with late-stage cancers, received fewer mastectomies, and were more likely to have radiation.


Asunto(s)
Neoplasias de la Mama/diagnóstico por imagen , Neoplasias de la Mama/terapia , Patient Protection and Affordable Care Act/estadística & datos numéricos , Neoplasias de la Mama/epidemiología , Femenino , Accesibilidad a los Servicios de Salud/estadística & datos numéricos , Humanos , Mamografía/estadística & datos numéricos , Mastectomía/estadística & datos numéricos , Medicaid/estadística & datos numéricos , Persona de Mediana Edad , Estudios Retrospectivos , Ultrasonografía Mamaria/estadística & datos numéricos , Estados Unidos/epidemiología , Población Urbana
5.
J Circadian Rhythms ; 17: 6, 2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31303884

RESUMEN

INTRODUCTION: Women diagnosed with breast cancer (BC) are at increased risk of sleep deficiency. Approximately 30-60% of these women report poor sleep during and following surgery, chemotherapy, radiation therapy, and anti-estrogen therapy. The purpose of this study was to examine the relationship between genetic variation in circadian rhythm genes and self-reported sleep quality in women with BC. METHODS: This cross-sectional study recruited women with a first diagnosis of breast cancer at five sites in Nebraska and South Dakota. Sixty women were included in the study. Twenty-six circadian genes were selected for exome sequencing using the Nextera Rapid Capture Expanded Exome kit. 414 variants had a minor allele frequency of ≥5% and were included in the exploratory analysis. The association between Pittsburgh Sleep Quality Index (PSQI) score and genetic variants was determined by two-sample t-test or ANOVA. RESULTS: Twenty-five variants were associated with the PSQI score at p < 0.10, of which 19 were significant at p<0.05, although the associations did not reach statistical significance after adjustment for multiple comparisons. Variants associated with PSQI were from genes CSNK1D & E, SKP1, BHLHE40 & 41, NPAS2, ARNTL, MYRIP, KLHL30, TIMELESS, FBXL3, CUL1, PER1&2, RORB. Two genetic variants were synonymous or missense variants in the BHLHE40 and TIMELESS genes, respectively. CONCLUSIONS: These exploratory results demonstrate an association of genetic variants in circadian rhythm pathways with self-reported sleep in women with BC. Testing this association is warranted in a larger replication population.

7.
Breast Cancer Res Treat ; 151(1): 219-24, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25833210

RESUMEN

Partner and localizer of BRCA2 (PALB2), plays an important functional role in DNA damage repair. Recent studies indicate that germline mutations in PALB2 predispose individuals to a high risk of developing familial breast cancer. Therefore, comprehensive identification of PALB2 germline mutations is potentially important for understanding their roles in tumorigenesis and for testing their potential utility as clinical targets. Most of the previous studies of PALB2 have focused on familial breast cancer cases with normal/wild-type BRCA1 and BRCA2 (BRCAx). We hypothesize that PALB2 genetic mutations also exist in individuals with BRCA mutations (BRCA+). To test this hypothesis, PALB2 germline mutations were screened in 107 exome data sets collected from familial breast cancer families who were either BRCA1+ or BRCAx. Two novel heterozygous mutations predicted to alter the function of PALB2 were identified (c.2014G>C, p.E672Q and c.2993G>A, p.G998E). Notably, both of these mutations co-existed in BRCA1+ and BRCA1x families. These studies show that mutations in PALB2 can occur independent of the status of BRCA1 mutations, and they highlight the importance to include BRCA1+ families in PALB2 mutation screens.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Masculino , Linaje
8.
J Natl Compr Canc Netw ; 13(7): 880-915, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26150582

RESUMEN

Breast cancer is the most frequently diagnosed malignancy in women in the United States and is second only to lung cancer as a cause of cancer death. To assist women who are at increased risk of developing breast cancer and their physicians in the application of individualized strategies to reduce breast cancer risk, NCCN has developed these guidelines for breast cancer risk reduction.


Asunto(s)
Neoplasias de la Mama/prevención & control , Conducta de Reducción del Riesgo , Femenino , Humanos , Factores de Riesgo
10.
BMC Cancer ; 14: 470, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24969172

RESUMEN

BACKGROUND: Genetic predisposition is the primary risk factor for familial breast cancer. For the majority of familial breast cancer, however, the genetic predispositions remain unknown. All newly identified predispositions occur rarely in disease population, and the unknown genetic predispositions are estimated to reach up to total thousands. Family unit is the basic structure of genetics. Because it is an autosomal dominant disease, individuals with a history of familial breast cancer must carry the same genetic predisposition across generations. Therefore, focusing on the cases in lineages of familial breast cancer, rather than pooled cases in disease population, is expected to provide high probability to identify the genetic predisposition for each family. METHODS: In this study, we tested genetic predispositions by analyzing the family-specific variants in familial breast cancer. Using exome sequencing, we analyzed three families and 22 probands with BRCAx (BRCA-negative) familial breast cancer. RESULTS: We observed the presence of family-specific, novel, deleterious germline variants in each family. Of the germline variants identified, many were shared between the disease-affected family members of the same family but not found in different families, which have their own specific variants. Certain variants are putative deleterious genetic predispositions damaging functionally important genes involved in DNA replication and damaging repair, tumor suppression, signal transduction, and phosphorylation. CONCLUSIONS: Our study demonstrates that the predispositions for many BRCAx familial breast cancer families can lie in each disease family. The application of a family-focused approach has the potential to detect many new predispositions.


Asunto(s)
Genes BRCA1 , Genes BRCA2 , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/genética , Exoma , Femenino , Humanos , Modelos Biológicos , Linaje
11.
BMC Cancer ; 14: 342, 2014 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-24884718

RESUMEN

BACKGROUND: BRCA1 plays an essential role in maintaining genome stability. Inherited BRCA1 germline mutation (BRCA1+) is a determined genetic predisposition leading to high risk of breast cancer. While BRCA1+ induces breast cancer by causing genome instability, most of the knowledge is known about somatic genome instability in breast cancer cells but not germline genome instability. METHODS: Using the exome-sequencing method, we analyzed the genomes of blood cells in a typical BRCA1+ breast cancer family with an exon 13-duplicated founder mutation, including six breast cancer-affected and two breast cancer unaffected members. RESULTS: We identified 23 deleterious mutations in the breast cancer-affected family members, which are absent in the unaffected members. Multiple mutations damaged functionally important and breast cancer-related genes, including transcriptional factor BPTF and FOXP1, ubiquitin ligase CUL4B, phosphorylase kinase PHKG2, and nuclear receptor activator SRA1. Analysis of the mutations between the mothers and daughters shows that most mutations were germline mutation inherited from the ancestor(s) while only a few were somatic mutation generated de novo. CONCLUSION: Our study indicates that BRCA1+ can cause genome instability with both germline and somatic mutations in non-breast cells.


Asunto(s)
Proteína BRCA1/genética , Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Inestabilidad Genómica , Adulto , Proteína BRCA1/sangre , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/sangre , Análisis Mutacional de ADN , Exones , Femenino , Efecto Fundador , Predisposición Genética a la Enfermedad , Herencia , Humanos , Masculino , Persona de Mediana Edad , Madres , Mutación , Núcleo Familiar , Linaje , Fenotipo
12.
Cancers (Basel) ; 16(8)2024 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-38672660

RESUMEN

Breast cancer (BC) is a complex disease affecting one in eight women in the USA. Advances in population genomics have led to the development of polygenic risk scores (PRSs) with the potential to augment current risk models, but replication is often limited. We evaluated 2 robust PRSs with 313 and 3820 SNPs and the effects of multiple genotype imputation replications in BC cases and control populations. Biological samples from BC cases and cancer-free controls were drawn from three European ancestry cohorts. Genotyping on the Illumina Global Screening Array was followed by stringent quality control measures and 20 genotype imputation replications. A total of 468 unrelated cases and 4337 controls were scored, revealing significant differences in mean PRS percentiles between cases and controls (p < 0.001) for both SNP sets (313-SNP PRS: 52.81 and 48.07; 3820-SNP PRS: 55.45 and 49.81), with receiver operating characteristic curve analysis showing area under the curve values of 0.596 and 0.603 for the 313-SNP and 3820-SNP PRS, respectively. PRS fluctuations (from ~2-3% up to 9%) emerged across imputation iterations. Our study robustly reaffirms the predictive capacity of PRSs for BC by replicating their performance in an independent BC population and showcases the need to average imputed scores for reliable outcomes.

13.
Breast J ; 19(5): 520-8, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23800003

RESUMEN

Genetic predisposition plays a key role in the development of familial breast cancer. In spite of strong familial clustering of the disease and extensive efforts made during the past decade; however, progress has been slow in identifying genetic predisposition for the majority of familial breast cancer families. The question arises therefore as to whether current approaches are adequate in identifying the unknown genetic predisposition. We analyzed eight members of a BRCA1-, BRCA2-, p53-, and PTEN-negative breast cancer family, of which five had breast cancer, one is an obligate gene carrier, and two were unaffected. We sequenced the entire coding region of the genome for each member using exome sequencing to identify nonsynonymous variants. We identified 55 nonsynonymous germline variants affecting 49 genes in multiple members of the family, of which 22 are predicted to have damaging effects. We validated 20 of the 22 selected variants in the family by Sanger sequencing. Two variants in KAT6B, an acetal transferase gene, were identified in six family members of which five were affected with breast cancer and one is the unaffected obligate carrier. We further examined the presence of the identified variants in a cohort of 40 additional breast cancer cases from 22 familial breast cancer families, but none of the 22 variants was detected in these cases. Sequencing the entire coding exons in KAT6B detects no variants in these cases. Our results show that genetic predisposition for familial breast cancer can be rich in an affected family, but the predisposition can be family-specific. As such, it will be difficult to detect them by applying population-based approach. Our study supports the concept that focusing on each affected family will be required to determine the genetic predisposition for many familial breast cancer families whose genetic dispositions remain unknown.


Asunto(s)
Neoplasias de la Mama/congénito , Predisposición Genética a la Enfermedad , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/genética , Femenino , Genes BRCA1 , Genes BRCA2 , Histona Acetiltransferasas/genética , Humanos , Masculino , Persona de Mediana Edad , Mutación
14.
Nat Genet ; 30(3): 285-9, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11836499

RESUMEN

The breast cancer tumor-suppressor gene, BRCA1, encodes a protein with a BRCT domain-a motif that is found in many proteins that are implicated in DNA damage response and in genome stability. Phosphorylation of BRCA1 by the DNA damage-response proteins ATM, ATR and hCds1/Chk2 changes in response to DNA damage and at replication-block checkpoints. Although cells that lack BRCA1 have an abnormal response to DNA damage, the exact role of BRCA1 in this process has remained unclear. Here we show that BRCA1 is essential for activating the Chk1 kinase that regulates DNA damage-induced G2/M arrest. Thus, BRCA1 controls the expression, phosphorylation and cellular localization of Cdc25C and Cdc2/cyclin B kinase-proteins that are crucial for the G2/M transition. We show that BRCA1 regulates the expression of both Wee1 kinase, an inhibitor of Cdc2/cyclin B kinase, and the 14-3-3 family of proteins that sequesters phosphorylated Cdc25C and Cdc2/cyclin B kinase in the cytoplasm. We conclude that BRCA1 regulates key effectors that control the G2/M checkpoint and is therefore involved in regulating the onset of mitosis.


Asunto(s)
Daño del ADN , Fase G2/fisiología , Genes BRCA1/fisiología , Mitosis/fisiología , Proteínas Quinasas/metabolismo , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Activación Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Pruebas de Precipitina , Transducción de Señal , Células Tumorales Cultivadas
15.
PLoS One ; 18(9): e0292423, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37769019

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0050281.].

16.
Breast Cancer Res ; 14(2): R60, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22494620

RESUMEN

INTRODUCTION: In response to gamma-irradiation (IR)-induced double-strand DNA breaks, cells undergo cell-cycle arrest, allowing time for DNA repair before reentering the cell cycle. G2/M checkpoint activation involves activation of ataxia telangiectasia mutated (ATM)/ATM- and rad3-related (ATR) kinases and inhibition of Cdc25 phosphatases, resulting in inhibition of Cdc2 kinase and subsequent G2/M cell-cycle arrest. Previous studies from our laboratory showed that the G2/M checkpoint activation after IR exposure of MCF-7 breast cancer cells is dependent on the activation of extracellular signal-regulated protein kinase 1 and 2 (ERK1/2) signaling. In the present studies, we investigated the role of Ras-related C3 botulinum toxin substrate 1 (Rac1) guanosine triphosphatase (GTPase) in IR-induced G2/M checkpoint response and ERK1/2 activation, as well as in cell survival after IR. METHODS: With Rac1-specific inhibitor, dominant negative mutant Rac1 (N17Rac1) and specific small interfering RNA, the effect of Rac1 on IR-induced G2/M checkpoint response and ERK1/2 activation was examined in human breast cancer cells. In addition, the effect of Rac1 on cell survival after irradiation was assessed by using Rac1-specific inhibitor. RESULTS: IR exposure of MCF-7 breast cancer cells was associated with a marked activation of Rac1 GTPase. Furthermore, inhibition of Rac1 by using specific inhibitor, dominant-negative Rac1 mutant, or specific siRNA resulted in attenuation of IR-induced G2/M arrest and concomitant diminution of IR-induced activation of ATM, ATR, Chk1, and Chk2 kinases, as well as phosphorylation of Cdc2-Tyr15. Moreover, Rac1 inhibition or decreased Rac1 expression also abrogated IR-induced phosphorylation of mitogen-activated protein kinase kinase 1 and 2 (MEK1/2) and ERK1/2. Ultimately, inhibition of Rac1 markedly increased cellular sensitivity to IR exposure, which involves induction of apoptosis. CONCLUSION: Studies in this report suggest that Rac1 GTPase plays an essential role in the activation of IR-induced ERK1/2 signaling and subsequent G2/M checkpoint response. Furthermore, results also support a role for Rac1 in promoting cell survival after irradiation treatment.


Asunto(s)
Neoplasias de la Mama/radioterapia , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Puntos de Control de la Fase M del Ciclo Celular/efectos de la radiación , Proteína de Unión al GTP rac1/metabolismo , Aminoquinolinas/farmacología , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteína Quinasa CDC2 , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Ciclina B/metabolismo , Quinasas Ciclina-Dependientes , Proteínas de Unión al ADN/metabolismo , Activación Enzimática/efectos de la radiación , Inhibidores Enzimáticos/farmacología , Femenino , Rayos gamma , Humanos , MAP Quinasa Quinasa 1/metabolismo , MAP Quinasa Quinasa 2/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosforilación , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/farmacología , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Tirosina/metabolismo , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Proteína de Unión al GTP rac1/genética
18.
JCO Precis Oncol ; 6: e2100463, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35476550

RESUMEN

PURPOSE: The 80-gene molecular subtyping signature (80-GS) reclassifies a proportion of immunohistochemistry (IHC)-defined luminal breast cancers (estrogen receptor-positive [ER+], human epidermal growth factor receptor 2-negative [HER2-]) as Basal-Type. We report the association of 80-GS reclassification with neoadjuvant treatment response and 5-year outcome in patients with breast cancer. METHODS: Neoadjuvant Breast Registry Symphony Trial (NBRST; NCT01479101) is an observational, prospective study that included 1,069 patients with early-stage breast cancer age 18-90 years who received neoadjuvant therapy. Pathologic complete response (pCR) and 5-year distant metastasis-free survival (DMFS) and overall survival (OS) were assessed in 477 patients with IHC-defined ER+, HER2- tumors and in a reference group of 229 patients with IHC-defined triple-negative breast cancer (TNBC). RESULTS: 80-GS reclassified 15% of ER+, HER2- tumors (n = 73) as Basal-Type (ER+/Basal), which had similar pCR compared with TNBC/Basal tumors (34% v 38%; P = .52), and significantly higher pCR than ER+/Luminal A (2%; P < .001) and ER+/Luminal B (6%; P < .001) tumors. The 5-year DMFS (%, [95% CI]) was significantly lower for patients with ER+/Basal tumors (66% [52.6 to 77.3]), compared with those with ER+/Luminal A tumors (92.3% [85.2 to 96.1]) and ER+/Luminal B tumors (73.5% [44.5 to 79.3]). Importantly, patients with ER+/Basal or TNBC/Basal tumors that had a pCR exhibited significantly improved DMFS and OS compared with those with residual disease. By contrast, patients with ER+/Luminal B tumors had comparable 5-year DMFS and OS whether or not they achieved pCR. CONCLUSION: Significant differences in chemosensitivity and 5-year outcome suggest patients with ER+/Basal molecular subtype may benefit from neoadjuvant regimens optimized for patients with TNBC/Basal tumors compared with patients with ER+/Luminal subtype. These data highlight the importance of identifying this subset of patients to improve treatment planning and long-term survival.


Asunto(s)
Terapia Neoadyuvante , Neoplasias de la Mama Triple Negativas , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Humanos , Persona de Mediana Edad , Estudios Prospectivos , Receptor ErbB-2 , Receptores de Estrógenos/genética , Receptores de Progesterona/análisis , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adulto Joven
19.
Epigenetics ; 15(12): 1325-1335, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32508251

RESUMEN

Superoxide dismutase 3 (SOD3) is a secreted antioxidant enzyme that regulates reactive oxygen species in the microenvironment. It is also a potential tumour suppressor gene that is significantly downregulated in breast cancer. We have previously shown that its mRNA expression is inversely correlated with relapse free survival in breast cancer patients. This study aimed to investigate the correlation of SOD3 promoter DNA methylation with its expression in different molecular subtypes of breast carcinoma. We found that SOD3 expression was significantly reduced in breast carcinoma samples compared to normal tissues with the lowest levels observed in Luminal B subtype. Pyrosequencing analysis showed significant increase in methylation levels in the SOD3 promoter region (-108 and -19 from the TSS) in tumours vs normal tissues (53.6% vs 25.2%). The highest degree of correlation between methylation and SOD3 expression levels was observed in Luminal B subtype (Spearman's R = -0.540, P < 0.00093). In this subtype, the -78 CpG position is the most significantly methylated site. The Spearman's coefficient analysis also indicated the most significant correlation of DNA methylation at this site with SOD3 gene expression levels in tumours vs. normal tissues (R = -0.5816, P < 6.9E-12). Moreover, copy number variation analysis of TCGA database revealed that the more aggressive Triple Negative and Her2+ subtypes had higher levels of SOD3 gene deletion. The predominantly down-regulated expression pattern of SOD3 and the various genetic and epigenetic deregulations of its expression suggest that loss of this antioxidant promotes an advantageous tumour-promoting microenvironment in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma/genética , Metilación de ADN , Superóxido Dismutasa/genética , Neoplasias de la Mama/metabolismo , Regulación hacia Abajo , Epigénesis Genética , Femenino , Regulación Neoplásica de la Expresión Génica , Historia Medieval , Humanos , Regiones Promotoras Genéticas , Superóxido Dismutasa/metabolismo
20.
Oncotarget ; 11(3): 304, 2020 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-32076492

RESUMEN

[This corrects the article DOI: 10.18632/oncotarget.2500.].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA