Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Oncologist ; 28(8): 737-e693, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-36952231

RESUMEN

BACKGROUND: Combination immunotherapy is now considered the standard first-line therapy for patients with metastatic clear cell renal cell carcinoma (mccRCC) after multiple clinical trials demonstrated improved overall survival compared with single-agent tyrosine kinase inhibitors. Cabozantinib modulates critical components of the immune system, such as decreasing regulatory T cells and increasing T-effector cell populations, and is approved for the treatment of mRCC. Avelumab is a human IgG1 monoclonal antibody that binds to programmed death-ligand 1 protein and inhibits the interaction with PD-1. This phase I trial assessed the safety and clinical activity of avelumab and cabozantinib combination therapy in mccRCC. METHODS: This study was a phase I, 3+3 dose escalation clinical trial. The primary endpoint was the safety and identification of the recommended phase II dose (RP2D). Secondary endpoints included objective response rate (ORR) and radiographic progression-free survival (rPFS). There were 3 dose cohorts: cabozantinib 20, 40, and 60 mg/day, each combined with avelumab (10 mg/kg intravenously every 2 weeks). An additional 3 patients were included in the final dose cohort as a confirmation of the RP2D. No dose modifications were allowed for avelumab, but dose delays were permitted. Both dose reductions and holds were allowed for cabozantinib. Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1, was used to determine ORR, and treatment beyond progression was allowed. RESULTS: Twelve patients with newly diagnosed mccRCC were enrolled from July 2018 until March 2020. Three patients were enrolled in the 20 and 40 mg cohorts each, and 6 were enrolled in the 60 mg cohort. The International Metastatic RCC Database Consortium (IMDC) risk categories for these patients were: 4 patients (favorable risk), 6 patients (intermediate risk), and 2 patients (poor risk). No dose-limiting toxicities (DLTs) were observed in any cohort. Six patients developed serious adverse events related to study treatment after the DLT window period. Immune-related adverse events (iRAEs) were reported in 11 patients; fatigue and diarrhea were the most common (each with n = 4, 33.3%), followed by maculopapular rash and hand-foot syndrome (each with n = 3, 25%). Dose reductions were required in 5 of 6 patients in the cabozantinib 60 mg cohort after the DLT period. One patient discontinued avelumab due to irAE (nephritis), while none discontinued cabozantinib due to toxicity. The ORR was 50%, with one complete response (CR) and 5 partial responses (PR). The disease control rate (CR + PR + stable disease) was noted in 92% of the patients. Radiological PFS survival rate at 6 and 12 months was reported in 67.7% and 33.5% of patients, respectively. CONCLUSION: Combination therapy with avelumab and cabozantinib is safe and showed preliminary clinical activity in mccRCC. Even though the DLT was not met in any of the 3 cohorts, the recommended RP2D dose for the combination is cabozantinib 40 mg/day due to a high incidence of grade 2 toxicity for cabozantinib 60 mg/day after the DLT period. (ClinicalTrials.gov Identifier: NCT03200587).


Asunto(s)
Carcinoma de Células Renales , Humanos , Carcinoma de Células Renales/patología , Anticuerpos Monoclonales Humanizados/uso terapéutico
2.
Mod Pathol ; 36(5): 100125, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36870308

RESUMEN

Triple-negative apocrine carcinomas (TNACs) are rare breast tumors with limited studies evaluating their molecular characteristics and clinical behavior. We performed a histologic, immunohistochemical, genetic, and clinicopathologic assessment of 42 invasive TNACs (1 with a focal spindle cell component) from 41 patients, 2 pure apocrine ductal carcinomas in situ (A-DCIS), and 1 A-DCIS associated with spindle cell metaplastic carcinoma (SCMBC). All TNACs had characteristic apocrine morphology and expressed androgen receptor (42/42), gross cystic disease fluid protein 15 (24/24), and CK5/6 (16/16). GATA3 was positive in most cases (16/18, 89%), and SOX10 was negative (0/22). TRPS1 was weakly expressed in a minority of tumors (3/14, 21%). Most TNACs had low Ki67 proliferation (≤10% in 67%, 26/39), with a median index of 10%. Levels of tumor infiltrating lymphocytes were low (≤10% in 93%, 39/42, and 15% in 7%, 3/42). Eighteen percent of TNACs presented with axillary nodal metastasis (7/38). No patients treated with neoadjuvant chemotherapy achieved pathologic complete response (0%, 0/10). Nearly all patients with TNAC (97%, n = 32) were without evidence of disease at the time of study (mean follow-up of 62 months). Seventeen invasive TNACs and 10 A-DCIS (7 with paired invasive TNAC) were profiled by targeted capture-based next-generation DNA sequencing. Pathogenic mutations in phosphatidylinositol 3-kinase pathway genes PIK3CA (53%) and/or PIK3R1 (53%) were identified in all TNACs (100%), including 4 (24%) with comutated PTEN. Ras-MAPK pathway genes, including NF1 (24%), and TP53 were mutated in 6 tumors each (35%). All A-DCIS shared mutations, such as phosphatidylinositol 3-kinase aberrations and copy number alterations with paired invasive TNACs or SCMBC, and a subset of invasive carcinomas showed additional mutations in tumor suppressors (NF1, TP53, ARID2, and CDKN2A). Divergent genetic profiles between A-DCIS and invasive carcinoma were identified in 1 case. In summary, our findings support TNAC as a morphologically, immunohistochemically, and genetically homogeneous subgroup of triple-negative breast carcinomas and suggest overall favorable clinical behavior.


Asunto(s)
Neoplasias de la Mama , Carcinoma in Situ , Carcinoma Intraductal no Infiltrante , Neoplasias de la Mama Triple Negativas , Humanos , Femenino , Carcinoma Intraductal no Infiltrante/patología , Neoplasias de la Mama/patología , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Factores de Transcripción , Fosfatidilinositol 3-Quinasas , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proteínas Represoras
3.
Curr Treat Options Oncol ; 24(9): 1199-1212, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37407886

RESUMEN

OPINION STATEMENT: PRCC is a unique histologic entity compared to other forms of renal cell carcinoma, harboring distinct molecular drivers. The WHO 2022 classification is further emphasizing the molecular biology by making molecular classifications of PRCC subclassifications and discontinuing the morphologic type 1 and type 2 classification system. We agree with this functional classification system and encourage all future clinical trials to only include patients with similar diagnosis instead of conducting basket trials (including all nccRCC together) which limits the scientific value of those conclusions. Based on recent disease-specific clinical trial (S1500, PAPMET), the current standard of care for patients with treatment naïve PRCC is cabozantinib. Prospective clinical trials clearly establish that immune checkpoint inhibitor therapy has meaningful activity in PRCC. The data to date include only single-arm clinical trials of combination immune therapy. Despite the positive and encouraging results, we need validation through randomized studies because of the overestimation of effect size seen in single-arm trials. These randomized trials are currently underway and enrolling. We strongly encourage all physicians to support these studies and enroll patients with PRCC to these trials in order to continue improving the standard of care.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/etiología , Carcinoma de Células Renales/terapia , Neoplasias Renales/diagnóstico , Neoplasias Renales/etiología , Neoplasias Renales/terapia , Ensayos Clínicos como Asunto
4.
Mod Pathol ; 35(4): 549-553, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34663915

RESUMEN

The College of American Pathologists/American Society of Clinical Oncology recommends HER2 testing prior to initiation of targeted therapy for patients with advanced Gastroesophageal adenocarcinoma (GEA), using immunohistochemistry (IHC) followed by fluorescence in situ hybridization (FISH) in cases with an equivocal (score 2 + ) result on IHC. The FISH results are considered indeterminate if the HER2/CEP17 ratio is <2.0 with an average CEP17 copy number of ≥3.0 and a HER2 copy number ≥4.0 and ≤6.0 after counting additional tumor cells. Indeterminate results may be resolved by using an alternative chromosome 17 probe such as RAI1. The purpose of this study is to review our experience with RAI1 alternate probe in HER2 FISH testing of GEA in a large reference laboratory setting. Esophageal, gastroesophageal, and gastric adenocarcinomas received for HER2 FISH testing in our lab between 9/2018 and 1/2020 were included. HER2/CEP17 and HER2/ RAI1 ratios, and the average HER2, CEP17, RAI1 signals per cell were recorded. 328 GEA had HER2 testing performed in our lab during the study period. 101 (30.8%) were amplified, 169 (51.5%) were non-amplified and 58 (17.7%) were indeterminate. Following RAI1 testing, 42 (72.4%) of 58 indeterminate cases were reclassified as non-amplified and 16 (27.6%) were reclassified as amplified, increasing the total amplified cases to 117 (35.7%). The correlation between the average CEP17 and RAI1 copy number for all cases was weak (R2 = 0.095). In summary, using the alternate probe RAI1 reclassifies 27.6% of original HER2 FISH indeterminate gastroesophageal carcinomas as amplified, which makes them eligible for targeted therapies.


Asunto(s)
Adenocarcinoma , Neoplasias de la Mama , Adenocarcinoma/genética , Biomarcadores de Tumor/genética , Cromosomas Humanos Par 17/genética , Femenino , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ/métodos , Receptor ErbB-2/genética
5.
Oncologist ; 26(12): 1006-e2129, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34423501

RESUMEN

LESSONS LEARNED: Long-term safety of radium-223 with enzalutamide was confirmed in this clinical trial. PSA-PFS2 was prolonged with the combination compared with enzalutamide alone. BACKGROUND: Previously, we showed the combination of radium-223 and enzalutamide to be safe and associated with improved efficacy based on a concomitant decline in serum bone metabolism markers compared with enzalutamide alone in a phase II trial of men with metastatic castration-resistant prostate cancer (mCRPC) [1]. METHODS: Secondary endpoints were not included in our initial report, and we include them herein, after a median follow-up of 22 months. These objectives included long-term safety, prostate-specific antigen (PSA)-progression-free survival (PFS), and radiographic progression-free survival; PSA-PFS2 (time from start of protocol therapy to PSA progression on subsequent therapy); time to next therapy (TTNT); and overall survival (OS). Survival analysis and log-rank tests were performed using the R statistical package v.4.0.2 (https://www.r-project.org). Statistical significance was defined as p < .05. RESULTS: Of 47 patients (median age, 68 years), 35 received the combination and 12 enzalutamide alone. After a median follow-up of 22 months, final safety results did not show any increase in fractures or other adverse events in the combination arm. PSA-PFS2 was significantly improved, and other efficacy parameters were numerically improved in the combination over the enzalutamide arm. CONCLUSION: The combination of enzalutamide and radium-223 was found to be safe and associated with promising efficacy in men with mCRPC. These hypothesis-generating results portend well for the ongoing phase III PEACE III trial in this setting.


Asunto(s)
Feniltiohidantoína , Neoplasias de la Próstata , Anciano , Benzamidas , Castración , Humanos , Masculino , Nitrilos , Feniltiohidantoína/uso terapéutico , Radio (Elemento)
6.
Oncologist ; 26(9): 751-760, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34157173

RESUMEN

PURPOSE: Progression from metastatic castration-sensitive prostate cancer (mCSPC) to a castration-resistant (mCRPC) state heralds the lethal phenotype of prostate cancer. Identifying genomic alterations associated with mCRPC may help find new targets for drug development. In the majority of patients, obtaining a tumor biopsy is challenging because of the predominance of bone-only metastasis. In this study, we hypothesize that machine learning (ML) algorithms can identify clinically relevant patterns of genomic alterations (GAs) that distinguish mCRPC from mCSPC, as assessed by next-generation sequencing (NGS) of circulating cell-free DNA (cfDNA). EXPERIMENTAL DESIGN: Retrospective clinical data from men with metastatic prostate cancer were collected. Men with NGS of cfDNA performed at a Clinical Laboratory Improvement Amendments (CLIA)-certified laboratory at time of diagnosis of mCSPC or mCRPC were included. A combination of supervised and unsupervised ML algorithms was used to obtain biologically interpretable, potentially actionable insights into genomic signatures that distinguish mCRPC from mCSPC. RESULTS: GAs that distinguish patients with mCRPC (n = 187) from patients with mCSPC (n = 154) (positive predictive value = 94%, specificity = 91%) were identified using supervised ML algorithms. These GAs, primarily amplifications, corresponded to androgen receptor, Mitogen-activated protein kinase (MAPK) signaling, Phosphoinositide 3-kinase (PI3K) signaling, G1/S cell cycle, and receptor tyrosine kinases. We also identified recurrent patterns of gene- and pathway-level alterations associated with mCRPC by using Bayesian networks, an unsupervised machine learning algorithm. CONCLUSION: These results provide clinical evidence that progression from mCSPC to mCRPC is associated with stereotyped concomitant gain-of-function aberrations in these pathways. Furthermore, detection of these aberrations in cfDNA may overcome the challenges associated with obtaining tumor bone biopsies and allow contemporary investigation of combinatorial therapies that target these aberrations. IMPLICATIONS FOR PRACTICE: The progression from castration-sensitive to castration-resistant prostate cancer is characterized by worse prognosis and there is a pressing need for targeted drugs to prevent or delay this transition. This study used machine learning algorithms to examine the cell-free DNA of patients to identify alterations to specific pathways and genes associated with progression. Detection of these alterations in cell-free DNA may overcome the challenges associated with obtaining tumor bone biopsies and allow contemporary investigation of combinatorial therapies that target these aberrations.


Asunto(s)
Ácidos Nucleicos Libres de Células , Neoplasias de la Próstata Resistentes a la Castración , Teorema de Bayes , Biomarcadores de Tumor , Progresión de la Enfermedad , Humanos , Aprendizaje Automático , Masculino , Metástasis de la Neoplasia , Fosfatidilinositol 3-Quinasas , Neoplasias de la Próstata Resistentes a la Castración/genética , Estudios Retrospectivos
7.
J Urol ; 205(3): 709-717, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33080152

RESUMEN

PURPOSE: Five programmed cell death protein 1 or its ligand (L1) inhibitors are approved for treatment of platinum refractory, locally advanced/unresectable or metastatic urothelial carcinoma. However, their comparative effectiveness is unknown. We compared time to initiation of third therapy or death, and overall survival with different programmed cell death protein 1/L1 inhibitors in patients with platinum refractory metastatic urothelial carcinoma. MATERIALS AND METHODS: Patient-level data were extracted from a real-world de-identified database. Comparative effectiveness was inferred via Cox proportional hazards model, weighted by matching weights. Each patient's propensity for each treatment was modeled via random forest, based on potential drivers of treatment selection. A propensity score for each therapy was used to calculate a matching weight, targeting the same estimand as 1:1 matching of treatment groups with balance among potential confounders. Eligibility criteria included diagnosis of metastatic urothelial carcinoma, receipt of first line treatment with a platinum based chemotherapy, followed by initiation of single agent programmed cell death protein 1/L1 inhibitor after disease progression from August 1, 2016 through May 1, 2019. RESULTS: Overall, 609 patients were eligible for analysis. Median time to initiation of third therapy or death with atezolizumab, nivolumab and pembrolizumab was 4.2, 5.3 and 4.5 months, respectively, and median overall survival was 6.4, 8.0 and 8.3 months, respectively. Matching weighted analyses did not show strong evidence of differences among programmed cell death protein 1/L1 inhibitors in terms of time to initiation of third therapy or death and overall survival. CONCLUSIONS: In this large real-world cohort, effectiveness in terms of time to initiation of third therapy or death and overall survival with programmed cell death protein 1/L1 inhibitors in patients with platinum refractory locally advanced/unresectable or metastatic urothelial carcinoma was similar.


Asunto(s)
Carcinoma de Células Transicionales/tratamiento farmacológico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Anciano , Carboplatino/administración & dosificación , Carcinoma de Células Transicionales/mortalidad , Carcinoma de Células Transicionales/patología , Cisplatino/administración & dosificación , Investigación sobre la Eficacia Comparativa , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Puntaje de Propensión , Tasa de Supervivencia , Neoplasias de la Vejiga Urinaria/mortalidad , Neoplasias de la Vejiga Urinaria/patología
8.
Adv Anat Pathol ; 28(6): 396-407, 2021 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-34561376

RESUMEN

Fumarate hydratase-deficient renal cell carcinoma (FH-deficient RCC) is typically considered a high-grade, aggressive subtype of RCC that frequently arises in the setting of hereditary leiomyomatosis-renal cell carcinoma (HLRCC) syndrome. Increasing experience with HLRCC-associated RCC and FH-deficient RCC has resulted in recognition of tumors with lower grade morphologic features, overlapping with those of succinate dehydrogenase-deficient RCC and other low-grade oncocytic tumors. In this review article, we report a previously unpublished case that was recently encountered in our practice and review cases in the current literature with an aim of getting a better understanding of these oncocytic tumors and their morphologic spectrum. The 13 cases reviewed were approximately equally distributed across males and females, occurred at a younger age, and were more frequently seen in the right kidney, with both unifocal and multifocal presentations. While most presented an exclusive, low-grade oncocytic morphology, in 4 cases they were associated with either separate high-grade tumors, or as a secondary pattern in an otherwise conventional high-grade FH-deficient RCC. Loss of FH and 2 succinyl cysteine (2SC) positivity by immunohistochemistry supported their diagnosis, and are recommended to be performed alongside CD117, CK7, and CK20 in to aid classification in challenging oncocytic tumors. When occurring in isolation, these tumors are distinctive from their high-grade counterparts, with no reported adverse outcomes in cases reported thus far. As such, accurate diagnosis of this low-grade pattern among FH-deficient RCCs is worthwhile not only due to its association with HLRCC and need of genetic counseling and surveillance, but also due to more favorable prognosis. Finally, increasing experience with the low-grade end of the morphologic spectrum of FH deficient RCC reiterates that not all tumors of this subtype of RCC have a uniformly aggressive outcome.


Asunto(s)
Productos Biológicos , Carcinoma de Células Renales , Neoplasias Renales , Leiomiomatosis , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/genética , Diagnóstico Diferencial , Femenino , Fumarato Hidratasa/genética , Humanos , Neoplasias Renales/diagnóstico , Neoplasias Renales/genética , Leiomiomatosis/diagnóstico , Leiomiomatosis/genética , Masculino
9.
Semin Diagn Pathol ; 38(3): 212-221, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33840529

RESUMEN

Since the discovery of association of SMARCB1 mutations with malignant rhabdoid tumors and renal medullary carcinoma, mutations in genes of the SWI/SNF chromatin remodeling complex have been increasingly identified across a diverse spectrum of neoplasms. As a group, SWI/SNF complex subunit mutations are now recognized to be the second most frequent type of mutations across tumors. SMARCB1 mutations were originally reported in malignant rhabdoid tumors of the kidney and thought to be pathognomonic for this tumor. However, more broadly, recognition of typical rhabdoid cytomorphology and SMARCB1 mutations beyond rhabdoid tumors has changed our understanding of the pathobiology of these tumors. While mutations of SWI/SNF complex are diagnostic of rhabdoid tumors and renal medullary carcinoma, their clinical relevance extends to potential prognostic and predictive utility in other tumors as well. Beyond SMARCB1, the PBRM1 and ARID1A genes are the most frequently altered members of the SWI/SNF complex in genitourinary neoplasms, especially in clear cell renal cell carcinoma and urothelial carcinoma. In this review, we provide an overview of alterations in the SWI/SNF complex encountered in genitourinary neoplasms and discuss their increasing clinical importance.


Asunto(s)
Carcinoma de Células Transicionales , Neoplasias Renales , Tumor Rabdoide , Neoplasias de la Vejiga Urinaria , Proteínas Cromosómicas no Histona/genética , ADN Helicasas , Humanos , Inmunohistoquímica , Neoplasias Renales/genética , Proteínas Nucleares , Tumor Rabdoide/genética
10.
Mod Pathol ; 31(9): 1429-1441, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29765141

RESUMEN

In recent years, there has been increased interest in carcinomas of the urologic tract, that demonstrate association with the polyoma virus BK arising in immunosuppressed individuals, though the nature of this association is uncertain. To begin to understand this phenomenon, we reviewed the clinical, morphological, and immunohistochemical features of 11 carcinomas of the urologic tract, mainly urothelial (N = 9) and collecting duct carcinomas (N = 2), occurring during immunosuppression, and expressing polyoma virus T-antigen by immunohistochemistry. These were compared to a control group of carcinomas (N = 8), also arising during immunosuppression, but without T-antigen expression. A subset of both groups were also studied by hybrid capture-based DNA sequencing, probing not only for 479 cancer-related human genes, but also for polyoma and other viral sequences. Polyoma T-antigen-expressing tumors arose in 7 males and 4 females, at a median age of 66, and were aggressive, high-grade tumors with more than 1 variant morphologic pattern identified in 81% of cases, and a majority (73%) presenting at high stage category (>pT3). Diffuse polyoma T-antigen staining was seen in 91% of cases, with co-localization of aberrant p53 staining in 89%. Sequencing detected a lower number of deleterious mutations among T-antigen-expressing cases (average 1.62; 1/8 with TP53 mutation) compared to control cases (average 3.5, 2/4 with TP53 mutation). Only BK virus was detected with clonal integration and breakpoints randomly distributed across the human and viral genomes in 5/5 of the polyoma T-antigen-expressing carcinomas, and in none of the controls (0/4). In summary, these findings identify aggressive clinicopathologic features of polyoma T-antigen-expressing carcinomas, document BK as the strain involved, and associate BK viral integration with T-antigen expression and p53 aberrancy. While the apparent randomness of viral insertion sites is functionally unclear, the differing rates of mutations between T-antigen-expressing and control cases is intriguing.


Asunto(s)
Carcinoma/virología , Neoplasias Renales/virología , Poliomavirus/aislamiento & purificación , Neoplasias de la Vejiga Urinaria/virología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma/metabolismo , Carcinoma/patología , Femenino , Humanos , Inmunohistoquímica , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
12.
Histopathology ; 71(1): 42-52, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28165631

RESUMEN

AIMS: Fumarate hydratase (FH)-deficient renal cell carcinoma (RCC) is a high-grade, aggressive tubulopapillary carcinoma, arising predominantly in the setting of the hereditary leiomyomatosis-RCC syndrome of familial uterocutaneous leiomyomatosis and deficiency of FH. In contrast, succinate dehydrogenase (SDH)-deficient RCC is a lower-grade oncocytic carcinoma with cytoplasmic flocculence/vacuolation and inclusions, arising mostly in individuals harbouring germline mutations of subunit B of the SDH complex (SDHB). Herein we aim to report the clinicopathologic features of a novel form of FH-deficient RCC showing a low grade oncocytic morphology, reminiscent of SDH-deficient RCC. METHODS AND RESULTS: These distinctive, low-grade oncocytic neoplasms, with solid, nested and focally tubular architecture (2-90 mm), arose in four males (aged 11-41 years). Uniform cytology of polygonal cells, with flocculent, vacuolated eosinophilic cytoplasm with scattered inclusions, fine chromatin, and inconspicuous nucleoli, was apparent. Despite these features suggestive of SDH-deficient RCC, each tumour was confirmed as an FH-deficient carcinoma with retained SDHB expression. One case showed a synchronous, anatomically separate, typical high-grade FH-deficient RCC; one other showed such a tumour at nephrectomy 4 years later. No progression has been noted at 3 and 7 years in the cases with only the SDH-like lesions; the two cases with separate, typical FH-deficient RCCs progressed. CONCLUSIONS: In summary, we characterize a novel oncocytic type of FH-deficient RCC with a striking resemblance to SDH-deficient RCC, posing a diagnostic challenge and raising concerns about sampling and multifocality for syndrome-associated cases under surveillance protocols.


Asunto(s)
Carcinoma de Células Renales/patología , Fumarato Hidratasa/deficiencia , Neoplasias Renales/patología , Adulto , Carcinoma de Células Renales/enzimología , Niño , Humanos , Neoplasias Renales/enzimología , Masculino , Succinato Deshidrogenasa
14.
Adv Anat Pathol ; 24(3): 151-160, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28398953

RESUMEN

S100P, or placental S100, is a member of a large family of S100 proteins and considered to be a promising immunohistochemical marker to support urothelial differentiation. This review synthesizes published data regarding the expression of S100P in urothelial carcinoma across histological grade and variant patterns, and in other malignancies, in an effort to summarize the state of understanding of this marker and evaluate its potential. We provide also a broad comparison of S100P with other contemporary and traditional urothelial markers and outline the potential utility of S100P in various diagnostically challenging scenarios. Taken in context, we recommend that to provide immunohistochemical support for consideration of urothelial differentiation, S100P may be included in a panel of markers (due to its high sensitivity), with better established (GATA3) and more specific (uroplakin 2) markers, for comparison with corresponding markers of other primary sites under consideration, depending on the clinical context. We emphasize that the overall most appropriate panel for any given case depends on the differential diagnosis engendered by the morphology encountered, and the constellation of clinical findings. As always with immunohistochemical panels, expected positive and negative markers for each diagnostic consideration should be included. Finally, since as of date there are no optimally sensitive or specific markers of urothelial differentiation, all final diagnoses relying on immunohistochemical support should be made in the appropriate clinical and histological context.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Transicionales/patología , Neoplasias de la Vejiga Urinaria/patología , Neoplasias Urológicas/patología , Urotelio/patología , Carcinoma de Células Transicionales/metabolismo , Humanos , Inmunohistoquímica/métodos , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias Urológicas/diagnóstico , Neoplasias Urológicas/metabolismo
15.
Adv Anat Pathol ; 23(2): 71-83, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26849813

RESUMEN

Since the publication of the World Health Organization "blue book" in 2004, several recent studies have provided new insights on the pathologic aspects of urachal neoplasms. The proposed updates include modified criteria for the diagnosis of urachal carcinoma. A uniform nomenclature for cystic tumors was lacking, and it is recommended that urachal mucinous cystic tumors should be separated and classified in a manner similar to ovarian mucinous neoplasms. The spectrum includes mucinous cystadenoma, mucinous cystic tumor of low malignant potential, mucinous cystic tumor of low malignant potential with intraepithelial carcinoma, and microscopically or frankly invasive mucinous cystadenocarcinoma, with 65% of cystic tumors classified as mucinous cystic tumor of low malignant potential. Most importantly, it has been shown that progression-free survival of noninvasive mucinous cystic tumors is significantly better than noncystic invasive adenocarcinoma. This development, along with prior descriptions of urachal villous adenoma, has also reaffirmed the occurrence of benign tumors of urachal epithelial origin. For noncystic (usual) invasive adenocarcinomas, the traditionally described histologic subtypes of enteric, mucinous, signet ring cell, not otherwise specified, and mixed remain appropriate, with 50% of tumors classified as mucinous subtype. Although this subtyping is helpful in diagnosis and differential diagnosis, the clinical significance of subtyping adenocarcinoma is still uncertain. Rare nonglandular morphologies such as urothelial, squamous, and neuroendocrine carcinoma in urachal carcinomas have been described in detail with proposals for their own set of diagnostic criteria. These criteria are based on unique features of urachal nonglandular carcinomas. Among the immunomarkers studied, only ß-catenin and CK7 may be of help in the distinction of urachal from colorectal adenocarcinoma. Awareness of the expression profile of immunomarkers such as CDX2, P504S (racemase), PSMA, claudin-18, and REG IV in urachal tumors and in tumors in the differential diagnosis is important to avoid overreliance of these markers in the diagnosis. Limited studies have identified KRAS mutations interestingly only in mucinous adenocarcinoma and exclusive of MSI loss, and mutations in BRAF are not present. Several alternative tumor staging approaches (eg, Mayo, Ontario, TNM systems) different from the traditional staging proposed by Sheldon are used that provide better tumor distribution across stages; however, the prognostic utility of the stage substratification has yet to be validated in large prospective studies. Evidence though suggests that staging urachal cancer is most pertinent when dichotomized to tumors that have spread outside versus within the perivesical tissue. Only high tumor stage and residual tumor after surgery have been shown to be independent predictors of outcome. This review updates the contemporary classification of urachal epithelial tumors, which has informed the upcoming 2016 classification of World Health Organization tumors. We provide modified criteria for diagnosing urachal adenocarcinomas, which remains a clinico-pathologic exercise. The role of ancillary diagnostic methodology and issues pertaining to staging and prognostication are presented.


Asunto(s)
Adenocarcinoma/diagnóstico , Neoplasias de la Vejiga Urinaria/diagnóstico , Adenocarcinoma/clasificación , Humanos , Neoplasias de la Vejiga Urinaria/clasificación
16.
J Cutan Pathol ; 42(2): 102-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25407361

RESUMEN

We report three cases of desmoplastic trichoepithelioma (DTE) with overlying pseudocarcinomatous hyperplasia. All three cases developed on the face of patients in their second decade. In spite of their young age, a diagnosis of squamous cell carcinoma was considered for each case, given the unusual juxtaposition of these two well-known simulators of malignancy. Awareness of this potential association may avert a misdiagnosis of malignancy.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico , Neoplasias Faciales/diagnóstico , Neoplasias de Cabeza y Cuello/diagnóstico , Neoplasias Cutáneas/diagnóstico , Adolescente , Biopsia , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Niño , Diagnóstico Diferencial , Neoplasias Faciales/metabolismo , Neoplasias Faciales/patología , Femenino , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Hiperplasia/patología , Inmunohistoquímica , Queratina-20/análisis , Masculino , Neoplasias Cutáneas/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello , Adulto Joven
17.
Int J Surg Pathol ; 32(3): 551-555, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-37394760

RESUMEN

TFEB-amplified renal cell carcinoma (RCC), which belongs to the MITF family of RCC, is characterized by genomic amplification at the 6p21.1 locus where the TFEB gene is located. The vascular endothelial growth factor A and cyclin D3 genes are also located at this same locus. When tumors lack classic morphologic features, they may be classified as "RCC not otherwise specified (NOS)." However, it is increasingly important to accurately diagnose the RCC subtype to define the patient's individual prognosis and select the subsequent therapeutic modalities, which now include targeted agents. Therefore, knowledge of the diagnostic features of TFEB-altered RCCs, such as t(6;11) RCCs and TFEB-amplified RCCs, is critical for identifying these tumors. Herein, we present an interesting case of TFEB-amplified RCC that was initially diagnosed as RCC NOS on biopsy of a renal tumor in a community practice setting with available molecular findings demonstrating CCND3 amplification. The genetic abnormality was "accidentally" detected due to the amplification of the colocated CCND3 gene at the 6p21 locus of the TFEB gene on a limited genetic sequencing panel. This case highlights the importance of molecular tests in accurately diagnosing RCC and carefully interpreting molecular findings in the context of histomorphologic features.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/diagnóstico , Carcinoma de Células Renales/genética , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Amplificación de Genes , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Neoplasias Renales/diagnóstico , Neoplasias Renales/genética , Translocación Genética , Biomarcadores de Tumor/genética , Ciclina D3/genética , Ciclina D3/metabolismo
18.
Int J Surg Pathol ; : 10668969241260236, 2024 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-39034146

RESUMEN

Morphological features are critical in evaluation of renal tumors and directing molecular workup. The objective of this study was to review histomorphology of renal tumors with molecular alterations of known subtypes. Renal tumors in The Cancer Genome Atlas were reviewed to identify tumors with defining molecular alterations. Single representative digital slides and pathology reports were reviewed and morphologic features recorded. Sixty tumors were identified with molecular alterations in genes characteristic of defined renal cell carcinoma (RCC) subtypes. Findings included the presence of both low- and high-grade histology in TFE3 rearranged RCCs, TFEB amplified RCCs, succinate dehydrogenase (SDH) mutated RCCs and RCCs with mutations in mismatch repair genes. Three ELOC mutated RCCs were identified, one of which demonstrated infiltrative features. Pseudostratification of nuclei in fumarate hydratase mutated RCCs and nuclear grooves in SDH mutated RCCs were intriguing findings not previously reported. Mucinous features were noted in NF2, KRAS, and SDH mutated and ALK rearranged tumors. Significant morphologic overlap was noted across most categories with limited clues for subclassification. Whereas the number of diagnostic entities for kidney tumors continues to increase, many of these have overlapping features, highlighting the significant role molecular characterization currently plays and will continue to play in the future.

19.
J Neuropathol Exp Neurol ; 83(5): 338-344, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38605523

RESUMEN

EGFR amplification in gliomas is commonly defined by an EGFR/CEP7 ratio of ≥2. In testing performed at a major reference laboratory, a small subset of patients had ≥5 copies of both EGFR and CEP7 yet were not amplified by the EGFR/CEP7 ratio and were designated high polysomy cases. To determine whether these tumors are more closely related to traditionally defined EGFR-amplified or nonamplified gliomas, a retrospective search identified 22 out of 1143 (1.9%) gliomas with an average of ≥5 copies/cell of EGFR and CEP7 with an EGFR/CEP7 ratio of <2 displaying high polysomy. Of these cases, 4 had insufficient clinicopathologic data to include in additional analysis, 15 were glioblastomas, 2 were IDH-mutant astrocytomas, and 1 was a high-grade glial neoplasm, NOS. Next-generation sequencing available on 3 cases demonstrated one with a TERT promoter mutation, TP53 mutations in all cases, and no EGFR mutations or amplifications, which most closely matched the nonamplified cases. The median overall survival times were 42.86, 66.07, and 41.14 weeks for amplified, highly polysomic, and nonamplified, respectively, and were not significantly different (p = 0.3410). High chromosome 7 polysomic gliomas are rare but our data suggest that they may be biologically similar to nonamplified gliomas.


Asunto(s)
Neoplasias Encefálicas , Amplificación de Genes , Glioblastoma , Glioma , Humanos , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Aberraciones Cromosómicas , Receptores ErbB/genética , Glioblastoma/genética , Glioblastoma/patología , Glioma/genética , Glioma/patología , Hibridación Fluorescente in Situ , Isocitrato Deshidrogenasa/genética , Mutación/genética , Estudios Retrospectivos , Cromosomas Humanos Par 7/genética
20.
Am J Clin Pathol ; 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38940388

RESUMEN

OBJECTIVES: Artificial intelligence (AI)-based chatbots have demonstrated accuracy in a variety of fields, including medicine, but research has yet to substantiate their accuracy and clinical relevance. We evaluated an AI chatbot's answers to questions posed during a treatment planning conference. METHODS: Pathology residents, pathology faculty, and an AI chatbot (OpenAI ChatGPT [January 30, 2023, release]) answered a questionnaire curated from a genitourinary subspecialty treatment planning conference. Results were evaluated by 2 blinded adjudicators: a clinician expert and a pathology expert. Scores were based on accuracy and clinical relevance. RESULTS: Overall, faculty scored highest (4.75), followed by the AI chatbot (4.10), research-prepared residents (3.50), and unprepared residents (2.87). The AI chatbot scored statistically significantly better than unprepared residents (P = .03) but not statistically significantly different from research-prepared residents (P = .33) or faculty (P = .30). Residents did not statistically significantly improve after research (P = .39), and faculty performed statistically significantly better than both resident categories (unprepared, P < .01; research prepared, P = .01). CONCLUSIONS: The AI chatbot gave answers to medical questions that were comparable in accuracy and clinical relevance to pathology faculty, suggesting promise for further development. Serious concerns remain, however, that without the ability to provide support with references, AI will face legitimate scrutiny as to how it can be integrated into medical decision-making.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA