Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Breast Cancer Res Treat ; 183(3): 525-547, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32685993

RESUMO

PURPOSE: Previous studies indicate that breast cancer molecular subtypes differ with respect to their dependency on autophagy, but our knowledge of the differential expression and prognostic significance of autophagy-related biomarkers in breast cancer is limited. METHODS: Immunohistochemistry (IHC) was performed on tissue microarrays from a large population of 3992 breast cancer patients divided into training and validation cohorts. Consensus staining scores were used to evaluate the expression levels of autophagy proteins LC3B, ATG4B, and GABARAP and determine the associations with clinicopathological variables and molecular biomarkers. Survival analyses were performed using the Kaplan-Meier function and Cox proportional hazards regression models. RESULTS: We found subtype-specific expression differences for ATG4B, with its expression lowest in basal-like breast cancer and highest in Luminal A, but there were no significant associations with patient prognosis. LC3B and GABARAP levels were highest in basal-like breast cancers, and high levels were associated with worse outcomes across all subtypes (DSS; GABARAP: HR 1.43, LC3B puncta: HR 1.43). High ATG4B levels were associated with ER, PR, and BCL2 positivity, while high LC3B and GABARAP levels were associated with ER, PR, and BCL2 negativity, as well as EGFR, HER2, HER3, CA-IX, PD-L1 positivity, and high Ki67 index (p < 0.05 for all associations). Exploratory multi-marker analysis indicated that the combination of ATG4B and GABARAP with LC3B could be useful for further stratifying patient outcomes. CONCLUSIONS: ATG4B levels varied across breast cancer subtypes but did not show prognostic significance. High LC3B expression and high GABARAP expression were both associated with poor prognosis and with clinicopathological characteristics of aggressive disease phenotypes in all breast cancer subtypes.


Assuntos
Neoplasias da Mama , Proteínas Reguladoras de Apoptose , Autofagia , Proteínas Relacionadas à Autofagia/genética , Biomarcadores Tumorais , Neoplasias da Mama/genética , Cisteína Endopeptidases , Feminino , Humanos , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Proteínas Associadas aos Microtúbulos/genética , Prognóstico
2.
Int J Gynecol Pathol ; 38(6): 552-561, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30059451

RESUMO

Tubo-ovarian transitional cell carcinoma (TCC) is grouped with high-grade serous carcinoma (HGSC) in the current World Health Organization classification. TCC is associated with BRCA mutations and a better prognosis compared with HGSC. Previous papers examining the immunohistochemical features of TCC have studied limited numbers of samples. No marker reflecting the biological difference between TCC and HGSC is known. We collected a large cohort of TCC to determine whether TCC and HGSC could be distinguished by immunohistochemistry. A tissue microarray was built from 89 TCC and a control cohort of 232 conventional HGSC. Immunohistochemistry was performed, scored, and statistically analyzed for routine markers of HGSC and urothelial tumors: PAX8, WT1, p53, p16, ER, p63, and GATA3. Using scoring cutoffs commonly employed in clinical practice, the immunohistochemical profile of TCC was indistinguishable from HGSC for all markers. However, more detailed scoring criteria revealed statistically significant differences between the 2 groups of tumors with respect to ER, PAX8, and WT1. HGSC showed more diffuse and intense staining for PAX8 (P=0.004 and 0.001, respectively) and WT1 (P=0.002 and 0.002, respectively); conversely, TCC showed more intense staining for ER (P=0.007). TCC and HGSC therefore show subtle differences in their immunohistochemical profiles which might reflect underlying (epi)genetic differences. Further studies using proteomic analysis will focus on the identification of differentially expressed proteins that might serve as markers of TCC-like differentiation, which could help explain biologic differences between TCC and HGSC and might identify other cases of HGSC with a better prognosis.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células de Transição/patologia , Proteínas de Neoplasias/metabolismo , Neoplasias Ovarianas/patologia , Proteômica , Carcinoma de Células de Transição/metabolismo , Estudos de Coortes , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Ovarianas/metabolismo , Análise Serial de Tecidos
3.
Histopathology ; 73(2): 306-313, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29660837

RESUMO

AIMS: Gynandroblastoma is a rare ovarian sex cord-stromal tumour characterised by the presence of both male (Sertoli and/or Leydig cells) and female (granulosa cells) components. We investigated the mutational status of DICER1, FOXL2 and AKT1 genes at hot-spot regions that are known to be the key driving events in the development of Sertoli-Leydig cell tumour (SLCT), adult granulosa cell tumour (aGCT) and juvenile granulosa cell tumour (jGCT), respectively, to gain insights into the molecular pathogenesis of gynandroblastoma. METHODS AND RESULTS: Sixteen cases of gynandroblastoma were studied. All contained SLCT or Sertoli cell tumour components. aGCT and jGCT components were identified in seven and 10 cases, respectively, with one presenting both components. Heterozygous hot-spot mutations in the RNase IIIb domain of DICER1 were discovered in three cases, including one case with heterologous mucinous elements, all of which were composed of moderately or poorly differentiated SLCT and jGCT components, and harboured the mutations in both histological components. None of the 16 cases displayed mutations at the p.C134W (c.402C→G) of FOXL2 or within the pleckstrin-homology domain of AKT1. All cases showed FOXL2 immunostaining in both male and female components. CONCLUSION: DICER1 hot-spot mutation is the key-driving event in a subset of gynandroblastomas containing components of SLCT and jGCT. Gynandroblastomas composed of SLCT and jGCT may represent morphological variants of SLCT. The molecular basis of gynandroblastoma containing a component of aGCT is different from pure aGCT.


Assuntos
RNA Helicases DEAD-box/genética , Ribonuclease III/genética , Tumores do Estroma Gonadal e dos Cordões Sexuais/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Pessoa de Meia-Idade , Mutação , Adulto Jovem
4.
Clin Exp Metastasis ; 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-38217840

RESUMO

Melanoma is a highly immunogenic malignancy with an elevated mutational burden, diffuse lymphocytic infiltration, and one of the highest response rates to immune checkpoint inhibitors (ICIs). However, over half of all late-stage patients treated with ICIs will either not respond or develop progressive disease. Spatial imaging technologies are being increasingly used to study the melanoma tumor microenvironment (TME). The goal of such studies is to understand the complex interplay between the stroma, melanoma cells, and immune cell-types as well as their association with treatment response. Investigators seeking a better understanding of the role of cell location within the TME and the importance of spatial expression of biomarkers are increasingly turning to highly multiplexed imaging approaches to more accurately measure immune infiltration as well as to quantify receptor-ligand interactions (such as PD-1 and PD-L1) and cell-cell contacts. CyTOF-IMC (Cytometry by Time of Flight - Imaging Mass Cytometry) has enabled high-dimensional profiling of melanomas, allowing researchers to identify complex cellular subpopulations and immune cell interactions with unprecedented resolution. Other spatial imaging technologies, such as multiplexed immunofluorescence and spatial transcriptomics, have revealed distinct patterns of immune cell infiltration, highlighting the importance of spatial relationships, and their impact in modulating immunotherapy responses. Overall, spatial imaging technologies are just beginning to transform our understanding of melanoma biology, providing new avenues for biomarker discovery and therapeutic development. These technologies hold great promise for advancing personalized medicine to improve patient outcomes in melanoma and other solid malignancies.

5.
Artigo em Inglês | MEDLINE | ID: mdl-37173094

RESUMO

Family physicians provide comprehensive care for the community and are an integral part of the healthcare system. Canada is experiencing a shortage of family physicians, driven in part by overbearing expectations of family physicians, limited support and resources, antiquated physician compensation, and high clinic operating costs. An additional factor contributing to this scarcity is the shortage of medical school and family medicine residency spots, which have not kept pace with population demand. We analysed and compared data on provincial populations and numbers of physicians, residency spots and medical school seats across Canada. Family physician shortages are the highest in the territories (>55%), Quebec (21.5%) and British Columbia (17.7%). Among the provinces, Ontario, Manitoba, Saskatchewan and British Columbia have the fewest family physicians per 100 000 persons in the population. Among the provinces that offer medical education, British Columbia and Ontario have the fewest medical school seats per population, while Quebec has the most. British Columbia has the smallest medical class size and the least number of family medicine residency spots as a function of population, and one of the highest percentages of provincial residents without family doctors. Paradoxically, Quebec has a relatively large medical class size and a high number of family medicine residency spots as a function of population, but also one of the highest percentages of provincial residents without family doctors. Possible strategies to improve the current shortage include encouraging Canadian medical students and international medical graduates to consider family medicine, and reducing administrative burdens for current physicians. Other steps include creating a national data framework, understanding physician needs to guide effective policy changes, increasing seats in medical schools and family residency programmes, providing financial incentives and facilitating entry into family medicine for international medical graduates.


Assuntos
Educação Médica , Médicos de Família , Humanos , Medicina de Família e Comunidade/educação , Colúmbia Britânica , Biópsia
6.
Nat Med ; 29(8): 2121-2132, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37414899

RESUMO

Fecal microbiota transplantation (FMT) represents a potential strategy to overcome resistance to immune checkpoint inhibitors in patients with refractory melanoma; however, the role of FMT in first-line treatment settings has not been evaluated. We conducted a multicenter phase I trial combining healthy donor FMT with the PD-1 inhibitors nivolumab or pembrolizumab in 20 previously untreated patients with advanced melanoma. The primary end point was safety. No grade 3 adverse events were reported from FMT alone. Five patients (25%) experienced grade 3 immune-related adverse events from combination therapy. Key secondary end points were objective response rate, changes in gut microbiome composition and systemic immune and metabolomics analyses. The objective response rate was 65% (13 of 20), including four (20%) complete responses. Longitudinal microbiome profiling revealed that all patients engrafted strains from their respective donors; however, the acquired similarity between donor and patient microbiomes only increased over time in responders. Responders experienced an enrichment of immunogenic and a loss of deleterious bacteria following FMT. Avatar mouse models confirmed the role of healthy donor feces in increasing anti-PD-1 efficacy. Our results show that FMT from healthy donors is safe in the first-line setting and warrants further investigation in combination with immune checkpoint inhibitors. ClinicalTrials.gov identifier NCT03772899 .


Assuntos
Transplante de Microbiota Fecal , Melanoma , Animais , Camundongos , Transplante de Microbiota Fecal/métodos , Inibidores de Checkpoint Imunológico , Fezes/microbiologia , Melanoma/terapia , Imunoterapia , Resultado do Tratamento
7.
Materials (Basel) ; 14(24)2021 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-34947276

RESUMO

Medical devices with antimicrobial properties are a potential long-term solution to the high rate of multi-drug-resistant healthcare-associated infections. Silver nanoparticles (AgNPs) are an established agent for effectively eliminating a wide range of microbial strains. AgNPs have been commonly incorporated into traditional plastic materials; however, recently, there has been increased interest in using AgNPs combined with 3D-printing technology for medical devices due to the accessibility and customizability of 3D-printed products. This study reports a novel method of utilizing acetone to partially dissolve 3D-printed polymer acrylonitrile butadiene styrene (ABS) plastic to attach a layer of AgNPs. The antimicrobial properties of this AgNP-coated surface were tested against several microbial strains prevalent in healthcare-associated infections. AgNP-coated ABS (AgNP-ABS) plastic demonstrated significant elimination of viable bacteria within 4 h for all tested bacterial species (Acinetobacter baumannii, non-pathogenic and pathogenic Escherichia coli, Pseudomonas aeruginosa, and methicillin-resistant Staphylococcus aureus) and within 19 h for the tested fungus Candida albicans. The longevity of adhesion of AgNPs to the ABS plastic was assessed by checking antibacterial activity against A. baumannii after repeat use cycles. AgNP-ABS plastic showed decreased antibacterial efficacy with repeated use but maintained the ability to eliminate microbes within 3 h for up to eight use cycles. The AgNP-coated ABS plastic showed efficacy as an antimicrobial surface, and future studies will consider its applicability in the production of medical devices.

8.
Hum Pathol ; 101: 40-52, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32360491

RESUMO

The current World Health Organization classification does not distinguish transitional cell carcinoma of the ovary (TCC) from conventional tubo-ovarian high-grade serous carcinoma (HGSC), despite evidence suggesting improved prognosis for patients with TCC; instead, it is considered a morphologic variant of HGSC. The immunohistochemical (IHC) markers applied to date do not distinguish between TCC and HGSC. Therefore, we sought to compare the proteomic profiles of TCC and conventional HGSC to identify proteins enriched in TCC. Prognostic biomarkers in HGSC have proven to be elusive, and our aim was to identify biomarkers of TCC as a way of reliably and reproducibly identifying patients with a favorable prognosis and better response to chemotherapy compared with those with conventional HGSC. Quantitative global proteome analysis was performed on archival material of 12 cases of TCC and 16 cases of HGSC using SP3 (single-pot, solid phase-enhanced, sample preparation)-Clinical Tissue Proteomics, a recently described protocol for full-proteome analysis from formalin-fixed paraffin-embedded tissues. We identified 430 proteins that were significantly enriched in TCC over HGSC. Unsupervised co-clustering perfectly distinguished TCC from HGSC based on protein expression. Pathway analysis showed that proteins associated with cell death, necrosis, and apoptosis were highly expressed in TCCs, whereas proteins associated with DNA homologous recombination, cell mitosis, proliferation and survival, and cell cycle progression pathways had reduced expression. From the proteomic analysis, three potential biomarkers for TCC were identified, claudin-4 (CLDN4), ubiquitin carboxyl-terminal esterase L1 (UCHL1), and minichromosome maintenance protein 7 (MCM7), and tested by IHC analysis on tissue microarrays. In agreement with the proteomic analysis, IHC expression of those proteins was stronger in TCC than in HGSC (p < 0.0001). Using global proteomic analysis, we are able to distinguish TCC from conventional HGSC. Follow-up studies will be necessary to confirm that these molecular and morphologic differences are clinically significant.


Assuntos
Biomarcadores Tumorais/análise , Carcinoma de Células de Transição/diagnóstico , Cistadenocarcinoma Seroso/diagnóstico , Neoplasias das Tubas Uterinas/diagnóstico , Neoplasias Ovarianas/diagnóstico , Feminino , Humanos , Proteômica/métodos
9.
Am J Surg Pathol ; 43(5): 628-638, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30986800

RESUMO

Sertoli-Leydig cell tumors (SLCTs) are rare ovarian sex cord-stromal neoplasms. The only known recurrent genetic abnormality is DICER1 mutation, with rare mutations reported in FOXL2. We set out to establish a molecular classifier using DICER1 and FOXL2 somatic mutation status and clinicopathologic features in 42 SLCTs. Five tumors (12%) were well differentiated, 31 (74%) moderately differentiated, and 6 (14%) poorly differentiated. Eight (19%) had heterologous elements, and 2 (5%) showed retiform differentiation; all 10 were moderately differentiated. DICER1 RNase IIIb domain mutations were identified in 18/41 (44%; 17 moderately, 1 poorly differentiated), including all cases with retiform or heterologous elements. FOXL2 c.402C>G (p.C134W) mutation was identified in 8/42 (19%) tumors (5 moderately, 3 poorly differentiated). DICER1 and FOXL2 mutations were mutually exclusive. Median age for the cohort was 47 years (range, 15 to 90 y). Patients with DICER1 mutations were younger (median, 24.5 y; range, 15 to 62 y) than patients with FOXL2 mutation (median, 79.5 y; range, 51 to 90 y) (P<0.0001). Nine of 10 tumors with retiform or heterologous elements occurred in premenopausal patients (median, 26.5 y; range, 15 to 57 y). Patients with tumors that were wild type for DICER1 and FOXL2 (15/42, 37%) had an intermediate age (median, 51 y; range, 17 to 74 y). All tumors were FOXL2 positive by immunohistochemistry. Patients with FOXL2 mutation trended toward presenting more often with abnormal bleeding (P=0.13); DICER1-mutant patients trended toward having more androgenic symptoms (P=0.22). Our data suggest at least 3 molecular subtypes of SLCT with distinct clinicopathologic features: DICER1 mutant (younger, more androgenic symptoms, moderately/poorly differentiated, retiform or heterologous elements), FOXL2 mutant (postmenopausal, abnormal bleeding, moderately/poorly differentiated, no retiform or heterologous elements), and DICER1/FOXL2 wild type (intermediate age, no retiform or heterologous elements, including all well-differentiated tumors).


Assuntos
Biomarcadores Tumorais/genética , RNA Helicases DEAD-box/genética , Proteína Forkhead Box L2/genética , Mutação , Neoplasias Ovarianas/genética , Ribonuclease III/genética , Tumor de Células de Sertoli-Leydig/genética , Adolescente , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Diferenciação Celular , Feminino , Predisposição Genética para Doença , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/classificação , Neoplasias Ovarianas/patologia , Fenótipo , Tumor de Células de Sertoli-Leydig/classificação , Tumor de Células de Sertoli-Leydig/patologia , Adulto Jovem
10.
Hum Pathol ; 92: 39-47, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31220479

RESUMO

LINE-1 (L1) retrotransposons are mobile genetic elements capable of "copy-and-pasting" their own sequences into random genomic loci, and one of the proteins it uses to achieve mobility is LINE-1 open reading frame 1 protein (L1ORF1p). L1ORF1p expression is found across many epithelial cancers, including small cohorts of ovarian and endometrial cancers, and is highly expressed in cancers with mutant p53 expressions. Here we aimed to gain insights into L1ORF1p expression levels within specific histotypes of ovarian cancers: high-grade serous (n = 585), low-grade serous (n = 26), clear cell (n = 132), endometrioid (n = 148), and mucinous (n = 32) ovarian cancers, as well as endometrial cancers (n = 607) using tissue microarray (TMA's). We demonstrated that L1ORF1p expression is associated with advanced stage and serous histotype in gynecological cancers. Like previous studies, we found a higher proportion of L1ORF1p expression in cases with aberrant p53 expression. We evaluated the expression of L1ORF1p in serous tubal intraepithelial carcinomas (STICs) (n = 6) and p53 signature lesions (n = 2) in fallopian tubes. Three STIC cases displayed aberrant p53 overexpression with corresponding L1ORF1p expression in the same tissues, but such correlation was not seen in the two p53 signature lesions, suggesting that L1 protein may be expressed after dysplastic transformation. The remaining three STIC cases have TP53 nonsense mutations with absent p53 expression but a strong and clear L1ORF1p expression within the STIC lesions. While L1ORF1p may not be prognostic in gynecological cancers, it may be useful clinically as a diagnostic IHC marker for p53 null STIC lesions and this warrants further investigation.


Assuntos
Adenocarcinoma de Células Claras/metabolismo , Carcinoma Endometrioide/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Desoxirribonuclease I/metabolismo , Neoplasias do Endométrio/metabolismo , Neoplasias Ovarianas/metabolismo , Adenocarcinoma de Células Claras/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma Endometrioide/patologia , Cistadenocarcinoma Seroso/patologia , Neoplasias do Endométrio/patologia , Feminino , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/patologia , Proteína Supressora de Tumor p53/metabolismo , Adulto Jovem
12.
J Pathol Clin Res ; 3(4): 279-293, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29085668

RESUMO

Molecular subclassification of endometrial carcinoma (EC) with Proactive Molecular Risk Classifier for Endometrial Cancer (ProMisE) identifies four subtypes [DNA polymerase epsilon (POLE) mutant, mismatch repair-deficient, p53 wild-type (wt), and p53 abnormal]. The aim of this study was to evaluate additional EC biomarkers in the context of these subtypes. Tissue microarrays encompassing 460 previously characterized ECs were assessed for L1-cell adhesion molecule (L1CAM), progesterone receptor (PR), estrogen receptor (ER) alpha, stathmin, and phosphatase and tensin homolog (PTEN), by immunohistochemistry (IHC). Associations with clinicopathological parameters, molecular subtype, and outcomes were determined. About 413 ECs (75% endometrioid, >15% serous) had complete data. L1CAM overexpression was found in 16%, associated with older age, lower body mass index (BMI), advanced stage, grade 3 (97%), non-endometrioid histology (84%), deep myometrial invasion, lymphovascular space invasion (LVSI), and ER-negative, PR-negative status. Tumours overexpressing L1CAM were associated with poor outcomes {hazard ratio (HR) [95% confidence interval (CI)] 3.35 [2.10-5.23] for disease-specific survival [DSS], p < 0.0001}. PR positivity was associated with younger women, higher BMI, early stage (77% stage I), low grade (61%), endometrioid histology (90%) without LVSI or nodal disease, ER positivity (90%), p53wt tumours (55%), and favourable outcomes [HR (CI) 0.39 (0.25-0.62) for DSS, p < 0.0001]. ER positive tumours were early stage (73%), low grade, endometrioid histology, with improved DSS. Stathmin and PTEN IHC were not associated with outcomes. There was minimal agreement between IHC and mutation status for PTEN. L1CAM overexpression was significantly associated with the p53 abnormal molecular subtype, which accounted for more than 70% of the tumours overexpressing L1CAM. PR expression also correlated with molecular subtype, with most PR negative tumours being p53 abnormal. Multivariable analysis demonstrated that only ProMisE subtype [overall survival (OS), DSS, and progression-free survival] and age (OS only) maintained an association with outcomes. The prognostic significance of the single biomarkers tested could be explained based on their being covariable with the ProMisE molecular subtype.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA