Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Ann Surg Oncol ; 29(10): 6350-6358, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35802213

RESUMO

BACKGROUND: Atypical lobular hyperplasia (ALH) and classic lobular carcinoma in situ encompass a spectrum of proliferative lesions known as lobular neoplasia (LN). When imaging-concordant and found in isolation on core needle biopsy (CNB), LN infrequently upgrades to carcinoma on surgical excision, and routine excision is not indicated. Upgrade rates in the setting of synchronous carcinoma are not well studied. PATIENTS AND METHODS: Patients with radiology-pathology concordant synchronous LN and separately biopsied ipsilateral (n = 35) or contralateral (n = 15) carcinoma who underwent excision between 2010 and 2021 were retrospectively identified. Frequency of upgrade, to either invasive or in situ carcinoma, was quantified, and factors associated with upgrade were assessed using Fisher's exact test. RESULTS: The median age was 55 (range 33-74) years. The upgrade rate of LN was 6% and not significantly different between ipsilateral (2.9%) and contralateral (13.3%) carcinoma (p = 0.15). All upgraded LN lesions were ALH on CNB and detected as non-mass enhancement on magnetic resonance imaging (MRI). No additional disease was demonstrated after excision at the site of the original LN CNB in 22.9% (8 out of 35) of ipsilateral and 13.3% (2 out of 15) of contralateral patients. Upgrade was not associated with family history, menopausal status, imaging modality used to detect LN, or extent of LN on CNB (p > 0.05). CONCLUSIONS: Our results demonstrate a low upgrade rate (6%) in our study cohort of LN with synchronous ipsilateral or contralateral carcinoma, which suggests that not all LN mandates excision with synchronous carcinoma. Larger, multi-institution studies are needed to validate these findings.


Assuntos
Carcinoma de Mama in situ , Neoplasias da Mama , Carcinoma in Situ , Carcinoma Lobular , Lesões Pré-Cancerosas , Adulto , Idoso , Biópsia com Agulha de Grande Calibre , Carcinoma de Mama in situ/patologia , Carcinoma de Mama in situ/cirurgia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/cirurgia , Carcinoma in Situ/patologia , Carcinoma Lobular/patologia , Feminino , Humanos , Hiperplasia/cirurgia , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/patologia , Estudos Retrospectivos
2.
Mod Pathol ; 32(5): 666-674, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30455417

RESUMO

Interval colorectal cancers may arise from missed or incompletely excised precursors or from a unique rapid progression pathway. We compared the clinicopathologic and molecular profiles of interval and matched non-interval colorectal cancer to determine whether interval colorectal cancers harbor any unique genetic characteristics. Fifty one of 982 colorectal cancer (5.2%) were categorized as interval colorectal cancer, defined as colorectal cancer detected in a diagnostic examination prior to the next recommended colonoscopy and at least 1 year after the last colonoscopy. Clinicopathologic characteristics of interval colorectal cancer were compared to non-interval colorectal cancer matched 1:1 on age, gender, and tumor location. Molecular profile of a subset of interval colorectal cancer (n = 20) and matched (1:2) non-interval colorectal cancer (n = 40) were evaluated using next generation sequencing. Interval colorectal cancer were more likely to occur in the right colon (55% vs. 35%; p = 0.02) and in patients > 70 years of age (55% vs. 34%; p = 0.002). Clinicopathologic features and aberrant DNA mismatch repair protein expression were not significantly different between interval and matched non-interval colorectal cancer. The frequency and spectrum of genetic alterations was also similar in interval and matched non-interval colorectal cancer. Similar findings were seen when analysis was restricted to interval colorectal cancer diagnosed <5 years after last colonoscopy (n = 42). Interval and non-interval colorectal cancers share similar clinicopathologic and genetic profiles when matched for tumor location. Interval colorectal cancers and are more likely to develop from missed or incompletely excised precursors rather than a unique rapid progression pathway.


Assuntos
Adenocarcinoma/genética , Adenocarcinoma/patologia , Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Colectomia , Colonoscopia , Neoplasias Colorretais/cirurgia , Progressão da Doença , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Margens de Excisão , Pessoa de Meia-Idade , Lesões Pré-Cancerosas/cirurgia , Valor Preditivo dos Testes , Fatores de Risco , Fatores de Tempo , Adulto Jovem
3.
Gynecol Oncol ; 152(2): 426-433, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30503267

RESUMO

Most ovarian carcinomas are high-grade serous carcinomas (HGSC) that contain TP53 mutations, present at advanced stage, and eventually become resistant to chemotherapy. The rapid evolution of this disease has been attributed to an origin in the distal fallopian tube, in the form of serous tubal intraepithelial carcinomas (STICs). This has led to a disease model where malignancy develops first in the tube and spreads to the peritoneum or regional lymph nodes. However, although most early or incidentally discovered HGSCs manifest in the tube with STICs, many advanced HGSCs are not accompanied by a malignancy in the fimbria. To resolve this paradox, the focus has shifted to earlier, premalignant serous proliferations (ESPs) in the tubes, which lack the cytomorphologic features of malignancy but contain TP53 mutations. These have been termed p53 signatures or serous tubal intraepithelial lesions (STILs). Although they have not been presumed to have cancer-causing potential by themselves, some ESPs have recently been shown to share identical TP53 mutations with concurrent HGSCs, indicating a shared lineage between these early mucosal changes and metastatic malignancy. This discovery supports a paradigm by which HGSCs can emerge not only from STICs but also from exfoliated precursor cells (precursor escape) that eventually undergo malignant transformation within the peritoneal cavity. This paradigm unifies both localized and widespread HGSCs to a visible pre-existing cellular alteration in the tubal epithelium, and highlights a consistent and necessary biologic event (TP53 mutation) rarely encountered in the ovary or secondary Mullerian system. This dual pathway to HGSCs underscores the subtle nature of many serous cancer origins in the tube, explains contrasting clinico-pathologic presentations, and explains why, until recently, the fallopian tube was unappreciated as the principal origin of HGSCs. Moreover, it highlights additional challenges faced in preventing or intercepting HGSCs at a curable stage.


Assuntos
Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/patologia , Neoplasias das Tubas Uterinas/genética , Neoplasias das Tubas Uterinas/patologia , Carcinoma in Situ/genética , Carcinoma in Situ/patologia , Feminino , Genes p53 , Humanos , Mutação , Gradação de Tumores , Proteína Supressora de Tumor p53/genética
4.
J Pathol ; 246(3): 344-351, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30043522

RESUMO

The distal Fallopian tube is a site of origin for many 'ovarian' high-grade serous carcinomas (HGSCs) with intraepithelial carcinomas (STICs) that share identical TP53 mutations with metastatic tumors. TP53 mutation-positive early serous proliferations (ESPs) comprise a spectrum including p53 signatures and serous tubal intraepithelial lesions (STILs) and are not considered malignant; however, ESPs are often the only abnormality found in Fallopian tubes of women with metastatic HGSC. The purpose of this study was to determine if a relationship exists between isolated ESPs and concurrent metastatic HGSCs in the absence of STIC. Fallopian tubes from 32 HGSCs without a co-existing STIC/HGSC in the endosalpinx were exhaustively sectioned. The presence of either STIC/HGSC or ESP in the endosalpinx was documented and DNA from tissues containing ESPs, concurrent HGSC, and control epithelia were interrogated for TP53 mutations by targeted amplicon-based sequencing with average coverage reads >4000 across DNA replicate samples. Serial sectioning revealed a previously unrecognized STIC/HGSC in 3 of 32 (9.3%) and ESPs in 13 (40.6%). Twelve contained TP53 mutations. Nine (75%) shared identical TP53 mutations with concurrent HGSCs, four at high (≥ 5%) and five at low (< 5%) allele frequency. All control epithelia were TP53 mutation-negative. This study, for the first time, indicates lineage identity between ESPs in the distal tube and some metastatic HGSCs via a shared site-specific TP53 mutation. It supports a novel serous carcinogenic sequence in which an ESP could eventually culminate in a metastatic serous cancer via 'precursor escape' and would explain the apparent sudden onset of cancers without co-existing STICs. This paradigm for serous cancer development underscores the likelihood that multiple precursor types in the Fallopian tube contribute to serous cancer development with implications for the evolution, pathologic classification, and prevention of this lethal malignancy. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Carcinoma in Situ/patologia , Linhagem da Célula , Proliferação de Células , Células Epiteliais/patologia , Neoplasias das Tubas Uterinas/patologia , Tubas Uterinas/patologia , Neoplasias Císticas, Mucinosas e Serosas/secundário , Neoplasias Ovarianas/patologia , Lesões Pré-Cancerosas/patologia , Idoso , Carcinoma in Situ/genética , Neoplasias das Tubas Uterinas/genética , Feminino , Predisposição Genética para Doença , Humanos , Pessoa de Meia-Idade , Mutação , Gradação de Tumores , Neoplasias Císticas, Mucinosas e Serosas/genética , Neoplasias Ovarianas/genética , Fenótipo , Lesões Pré-Cancerosas/genética , Proteína Supressora de Tumor p53/genética
6.
Eur J Epidemiol ; 32(5): 393-407, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28510098

RESUMO

Previous studies suggest that abnormal energy balance status may dysregulate intestinal epithelial homeostasis and promote colorectal carcinogenesis, yet little is known about how host energy balance and obesity influence enterocyte differentiation during carcinogenesis. We hypothesized that the association between high body mass index (BMI) and colorectal carcinoma incidence might differ according to tumor histopathologic differentiation status. Using databases of the Nurses' Health Study and Health Professionals Follow-up Study, and duplication-method Cox proportional hazards models, we prospectively examined an association between BMI and the incidence of colorectal carcinoma subtypes classified by differentiation features. 120,813 participants were followed for 26 or 32 years and 1528 rectal and colon cancer cases with available tumor pathological data were documented. The association between BMI and colorectal cancer risk significantly differed depending on the presence or absence of poorly-differentiated foci (Pheterogeneity = 0.006). Higher BMI was associated with a higher risk of colorectal carcinoma without poorly-differentiated foci (≥30.0 vs. 18.5-22.4 kg/m2: multivariable-adjusted hazard ratio, 1.87; 95% confidence interval, 1.49-2.34; Ptrend < 0.001), but not with risk of carcinoma with poorly-differentiated foci (Ptrend = 0.56). This differential association appeared to be consistent in strata of tumor microsatellite instability or FASN expression status, although the statistical power was limited. The association between BMI and colorectal carcinoma risk did not significantly differ by overall tumor differentiation, mucinous differentiation, or signet ring cell component (Pheterogeneity > 0.03, with the adjusted α of 0.01). High BMI was associated with risk of colorectal cancer subtype containing no poorly-differentiated focus. Our findings suggest that carcinogenic influence of excess energy balance might be stronger for tumors that retain better intestinal differentiation throughout the tumor areas.


Assuntos
Índice de Massa Corporal , Diferenciação Celular/genética , Neoplasias Colorretais/patologia , Obesidade , Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais/genética , Epigenômica , Seguimentos , Humanos , Incidência , Instabilidade de Microssatélites , Epidemiologia Molecular , Obesidade/complicações , Patologia Molecular , Fatores de Risco , Estados Unidos/epidemiologia
7.
Appl Immunohistochem Mol Morphol ; 32(1): 17-23, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37937544

RESUMO

The clinical utility of the proliferation marker Ki67 in breast cancer treatment and prognosis is an active area of research. Studies have suggested that differences in pre-analytic and analytic factors contribute to low analytical validity of the assay, with scoring methods accounting for a large proportion of this variability. Use of standard scoring methods is limited, in part due to the time intensive nature of such reporting protocols. Therefore, use of digital image analysis tools may help to both standardize reporting and improve workflow. In this study, digital image analysis was utilized to quantify Ki67 indices in 280 breast biopsy and resection specimens during routine clinical practice. The supervised Ki67 indices were then assessed for agreement with a manual count of 500 tumor cells. Agreement was excellent, with an intraclass correlation coefficient of 0.96 for the pathologist-supervised analysis. This study illustrates an example of a rapid, accurate workflow for implementation of digital image analysis in Ki67 scoring in breast cancer.


Assuntos
Neoplasias da Mama , Humanos , Feminino , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/patologia , Antígeno Ki-67 , Processamento de Imagem Assistida por Computador/métodos , Diagnóstico por Imagem , Projetos de Pesquisa , Biomarcadores Tumorais/análise
8.
Cancers (Basel) ; 14(9)2022 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-35565427

RESUMO

Controversy persists regarding metformin's role in cancer therapy. Our recent work suggested metformin acts by impacting the tumor microenvironment (TME), normalizing the epigenetic profile of cancer-associated mesenchymal stem cells (CA-MSC). As CA-MSC can negatively impact tumor immune infiltrates, we evaluated metformin's impact on the human TME, focusing on the interplay of stroma and immune infiltrates. Tumor samples from (i) 38 patients treated with metformin and chemotherapy and (ii) 44 non-metformin matched controls were included in a tissue microarray (TMA). The TMA was used to compare the presence of CA-MSC, desmoplasia and immune infiltrates in the TME. In vitro and in vivo models examined metformin's role in alteration of the CA-MSC phenotype. The average percentage of CA-MSC was significantly lower in metformin-treated than in chemotherapy alone-treated tumors (p = 0.006). There were fewer regulatory T-cells in metformin-treated tumors (p = 0.043). Consistent with CA-MSC's role in excluding T-cells from tumor islets, the T-cells were primarily present within the tumor stroma. Evaluation of metformin's impact in vitro suggested that metformin cannot reverse a CA-MSC phenotype; however, the in vivo model where metformin was introduced prior to the establishment of the CA-MSC phenotype supported that metformin can partially prevent the reprogramming of normal MSC into CA-MSC. Metformin treatment led to a decrease in both the presence of protumorigenic CA-MSC and in immune exclusion of T cells, leading to a more immune-permissive environment. This suggests clinical utility in prevention and in treatment for early-stage disease and putatively in immune therapy.

9.
Cancer Res ; 82(24): 4680-4693, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36219681

RESUMO

Ovarian clear cell carcinoma (OCCC) is a deadly and treatment-resistant cancer, which arises within the unique microenvironment of endometriosis. In this study, we identified a subset of endometriosis-derived mesenchymal stem cells (enMSC) characterized by loss of CD10 expression that specifically support OCCC growth. RNA sequencing identified alterations in iron export in CD10-negative enMSCs and reciprocal changes in metal transport in cocultured OCCC cells. CD10-negative enMSCs exhibited elevated expression of iron export proteins hephaestin and ferroportin and donate iron to associated OCCCs, functionally increasing the levels of labile intracellular iron. Iron is necessary for OCCC growth, and CD10-negative enMSCs prevented the growth inhibitory effects of iron chelation. In addition, enMSC-mediated increases in OCCC iron resulted in a unique sensitivity to ferroptosis. In vitro and in vivo, treatment with the ferroptosis inducer erastin resulted in significant death of cancer cells grown with CD10-negative enMSCs. Collectively, this work describes a novel mechanism of stromal-mediated tumor support via iron donation. This work also defines an important role of endometriosis-associated MSCs in supporting OCCC growth and identifies a critical therapeutic vulnerability of OCCC to ferroptosis based on stromal phenotype. SIGNIFICANCE: Endometriosis-derived mesenchymal stem cells support ovarian clear cell carcinoma via iron donation necessary for cancer growth, which also confers sensitivity to ferroptosis-inducing therapy.


Assuntos
Adenocarcinoma de Células Claras , Endometriose , Células-Tronco Mesenquimais , Neoplasias Ovarianas , Humanos , Feminino , Endometriose/metabolismo , Endometriose/patologia , Neoplasias Ovarianas/patologia , Ferro , Adenocarcinoma de Células Claras/metabolismo , Células-Tronco Mesenquimais/metabolismo , Microambiente Tumoral
10.
Hematol Oncol Clin North Am ; 32(6): 915-928, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30390765

RESUMO

This article summarizes the pathogenesis of ovarian carcinoma, focusing on the paradox of high-grade serous carcinogenesis. The fallopian tube is the prime site of origin in early serous cancers. Because a subset of serous cancers is associated with early serous proliferations absent intramucosal carcinomas, "precursor escape" is emerging, whereby some advanced cancers trace their roots to early serous proliferations. This has parallels in the endometriosis model and opens up a novel mechanism by which advanced malignancy could emerge without an obvious tubal carcinoma. The impact of this concept on classification of serous cancer and expectations from preventive strategies are discussed.


Assuntos
Cistadenocarcinoma Seroso , Neoplasias das Tubas Uterinas , Células-Tronco Neoplásicas , Neoplasias Ovarianas , Evasão Tumoral/imunologia , Cistadenocarcinoma Seroso/imunologia , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patologia , Neoplasias das Tubas Uterinas/imunologia , Neoplasias das Tubas Uterinas/metabolismo , Neoplasias das Tubas Uterinas/patologia , Feminino , Humanos , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia
11.
Oncoimmunology ; 7(7): e1442999, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29900052

RESUMO

Inhibitors targeting the PDCD1 (programmed cell death 1, PD-1) immune checkpoint pathway have revolutionized cancer treatment strategies. The TIME (Tumor Immunity in the MicroEnvironment) classification based on tumor CD274 (PDCD1 ligand 1, PD-L1) expression and tumor-infiltrating lymphocytes (TIL) has been proposed to predict response to immunotherapy. It remains to be determined clinical, pathological, and molecular features of TIME subtypes of colorectal cancer. Using 812 colon and rectal carcinoma cases from the Nurses' Health Study and Health Professionals Follow-up Study, we examined the association of tumor characteristics and survival outcomes with four TIME subtypes (TIME 1, CD274low/TILabsent; TIME 2, CD274high/TILpresent; TIME 3, CD274low/TILpresent; and TIME 4, CD274high/TILabsent). In survival analyses, Cox proportional hazards models were adjusted for potential confounders, including microsatellite instability (MSI) status, CpG island methylator phenotype (CIMP) status, LINE-1 methylation level, and KRAS, BRAF, and PIK3CA mutation status. TIME subtypes 1, 2, 3 and 4 had 218 (27%), 117 (14%), 103 (13%), and 374 (46%) colorectal cancer cases, respectively. Compared with TIL-absent subtypes (TIME 1 and 4), TIL-present subtypes (TIME 2 and 3) were associated with high-level MSI, high-degree CIMP, BRAF mutation, and higher amounts of neoantigens (p < 0.001). TIME subtypes were not significantly associated with colorectal cancer-specific or overall survival. In conclusion, TIL-present TIME subtypes of colorectal cancer are associated with high levels of MSI and neoantigen load, supporting better responsiveness to cancer immunotherapy. Further studies examining tumor molecular alterations and additional factors in the tumor microenvironment may inform development of immunoprevention and immunotherapy strategies.

12.
Artigo em Inglês | MEDLINE | ID: mdl-29552640

RESUMO

Precision medicine has a goal of customizing disease prevention and treatment strategies. Under the precision medicine paradigm, each patient has unique pathologic processes resulting from cellular genomic, epigenomic, proteomic, and metabolomic alterations, which are influenced by pharmacological, environmental, microbial, dietary, and lifestyle factors. Hence, to realize the promise of precision medicine, multi-level research methods that can comprehensively analyze many of these variables are needed. In order to address this gap, the integrative field of molecular pathology and population data science (i.e., molecular pathological epidemiology) has been developed to enable such multi-level analyses, especially in gastrointestinal cancer research. Further integration of pharmacology can improve our understanding of drug effects, and inform decision-making of drug use at both the individual and population levels. Such integrative research demonstrated potential benefits of aspirin in colorectal carcinoma with PIK3CA mutations, providing the basis for new clinical trials. Evidence also suggests that HPGD (15-PDGH) expression levels in normal colon and the germline rs6983267 polymorphism that relates to tumor CTNNB1 (ß-catenin)/WNT signaling status may predict the efficacy of aspirin for cancer chemoprevention. As immune checkpoint blockade targeting the CD274 (PD-L1)/PDCD1 (PD-1) pathway for microsatellite instability-high (or mismatch repair-deficient) metastatic gastrointestinal or other tumors has become standard of care, potential modifying effects of dietary, lifestyle, microbial, and environmental factors on immunotherapy need to be studied to further optimize treatment strategies. With its broad applicability, our integrative approach can provide insights into the interactive role of medications, exposures, and molecular pathology, and guide the development of precision medicine.

13.
Transplantation ; 89(5): 500-8, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20118845

RESUMO

BACKGROUND: Severed donor heart lymphatics are not anastomosed to recipient lymphatics in cardiac transplantation. We evaluated the effects of cellular infiltrates of T cells and macrophages on the morphology of lymphatics in heart grafts. METHODS: Dark agouti hearts were transplanted to Lewis or control dark agouti rats on subtherapeutic doses of cyclosporin. Transplants were examined by immunohistology and quantitative immunofluorescence microscopy using lymphatic endothelial hyaluronan receptor-1 as a lymphatic marker and CD8 and CD68 as markers for cellular infiltration at selected intervals from 1 to 8 weeks posttransplantation. RESULTS: Allograft inner myocardial lymphatic density decreased by more than 30-fold at 1 week and recovered to only 15% of the native level at 8 weeks posttransplantation. In contrast, allograft lymphatics in and near the epicardium showed no significant density decline but increased in size by more than 5-fold at 2 weeks, and sustained approximately 3-fold increase at 8 weeks posttransplantation. Lymphatic changes correlated temporally with the extent of T cell and macrophage infiltration in allografts, which peaked at 2 to 3 weeks posttransplantation. When grafts were retransplanted from allogeneic to isogeneic recipients at 3 weeks posttransplantation, inner lymphatic density returned close to native level within 2 weeks after retransplantation. CONCLUSIONS: This is the first characterization of regional and morphologic effects of immunologic responses on heart lymphatics after transplantation. Elimination of alloimmune responses produces rapid restoration of inner lymphatic vessels, suggesting that lymphatics injured during rejection can recover when rejection is reversed during the posttransplantation course.


Assuntos
Transplante de Coração/patologia , Transplante de Coração/fisiologia , Sistema Linfático/patologia , Regeneração/fisiologia , Anastomose Cirúrgica , Animais , Aorta/cirurgia , Transplante de Coração/imunologia , Isoantígenos/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Artéria Pulmonar/cirurgia , Ratos , Ratos Endogâmicos Lew , Ratos Endogâmicos , Receptores de Superfície Celular/análise , Transplante Heterotópico/métodos , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA