Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mod Pathol ; 37(5): 100467, 2024 May.
Article in English | MEDLINE | ID: mdl-38460672

ABSTRACT

Renal low-grade oncocytic tumor (LOT) is a recently recognized renal cell neoplasm designated within the "other oncocytic tumors" category in the 2022 World Health Organization classification system. Although the clinicopathologic, immunohistochemical, and molecular features reported for LOT have been largely consistent, the data are relatively limited. The morphologic overlap between LOT and other low-grade oncocytic neoplasms, particularly eosinophilic chromophobe renal cell carcinoma (E-chRCC), remains a controversial area in renal tumor classification. To address this uncertainty, we characterized and compared large cohorts of LOT (n = 67) and E-chRCC (n = 69) and revealed notable differences between the 2 entities. Clinically, LOT predominantly affected women, whereas E-chRCC showed a male predilection. Histologically, although almost all LOTs were dominated by a small-nested pattern, E-chRCC mainly showed solid and tubular architectures. Molecular analysis revealed that 87% of LOT cases harbored mutations in the tuberous sclerosis complex (TSC)-mTOR complex 1 (mTORC1) pathway, most frequently in MTOR and RHEB genes; a subset of LOT cases had chromosomal 7 and 19q gains. In contrast, E-chRCC lacked mTORC1 mutations, and 60% of cases displayed chromosomal losses characteristic of chRCC. We also explored the cell of origin for LOT and identified L1 cell adhesion molecule (L1CAM), a collecting duct and connecting tubule principal cell marker, as a highly sensitive and specific ancillary test for differentiating LOT from E-chRCC. This distinctive L1CAM immunohistochemical labeling suggests the principal cells as the cell of origin for LOT, unlike the intercalated cell origin of E-chRCC and oncocytoma. The ultrastructural analysis of LOT showed normal-appearing mitochondria and intracytoplasmic lumina with microvilli, different from what has been described for chRCC. Our study further supports LOT as a unique entity with a benign clinical course. Based on the likely cell of origin and its clinicopathologic characteristics, we propose that changing the nomenclature of LOT to "Oncocytic Principal Cell Adenoma of the Kidney" may be a better way to define and describe this entity.


Subject(s)
Adenoma, Oxyphilic , Biomarkers, Tumor , Carcinoma, Renal Cell , Kidney Neoplasms , Neural Cell Adhesion Molecule L1 , Humans , Kidney Neoplasms/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/chemistry , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/chemistry , Female , Male , Middle Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/analysis , Neural Cell Adhesion Molecule L1/metabolism , Aged , Adult , Adenoma, Oxyphilic/pathology , Adenoma, Oxyphilic/genetics , Diagnosis, Differential , Aged, 80 and over , Immunohistochemistry , Neoplasm Grading , Mutation
2.
Mod Pathol ; 37(10): 100557, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38964503

ABSTRACT

Small cell carcinomas (SMC) of the lung are now molecularly classified based on the expression of transcriptional regulators (NEUROD1, ASCL1, POU2F3, and YAP1) and DLL3, which has emerged as an investigational therapeutic target. PLCG2 has been shown to identify a distinct subpopulation of lung SMC with stem cell-like and prometastasis features and poor prognosis. We analyzed the expression of these novel neuroendocrine markers and their association with traditional neuroendocrine markers and patient outcomes in a cohort of bladder neuroendocrine carcinoma (NEC) consisting of 103 SMC and 19 large cell NEC (LCNEC) assembled in tissue microarrays. Coexpression patterns were assessed and integrated with detailed clinical annotation including overall (OS) and recurrence-free survival (RFS) and response to neoadjuvant/adjuvant chemotherapy. We identified 5 distinct molecular subtypes in bladder SMC based on the expression of ASCL1, NEUROD1, and POU2F3: ASCL1+/NEUROD1- (n = 33; 34%), ASCL1- /NEUROD1+ (n = 21; 21%), ASCL1+/NEUROD1+ (n = 17; 17%), POU2F3+ (n = 22, 22%), and ASCL1- /NEUROD1- /POU2F3- (n = 5, 5%). POU2F3+ tumors were mutually exclusive with those expressing ASCL1 and NEUROD1 and exhibited lower expression of traditional neuroendocrine markers. PLCG2 expression was noted in 33 tumors (32%) and was highly correlated with POU2F3 expression (P < .001). DLL3 expression was high in both SMC (n = 72, 82%) and LCNEC (n = 11, 85%). YAP1 expression was enriched in nonneuroendocrine components and negatively correlated with all neuroendocrine markers. In patients without metastatic disease who underwent radical cystectomy, PLCG2+ or POU2F3+ tumors had shorter RFS and OS (P < .05), but their expression was not associated with metastasis status or response to neoadjuvant/adjuvant chemotherapy. In conclusion, the NEC of the bladder can be divided into distinct molecular subtypes based on the expression of ASCL1, NEUROD1, and POU2F3. POU2F3-expressing tumors represent an ASCL1/NEUROD1-negative subset of bladder NEC characterized by lower expression of traditional neuroendocrine markers. Marker expression patterns were similar in SMC and LCNEC. Expression of PLCG2 and POU2F3 was associated with shorter RFS and OS. DLL3 was expressed at high levels in both SMC and LCNEC of the bladder, nominating it as a potential therapeutic target.

3.
Pediatr Blood Cancer ; 70(8): e30434, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37243318

ABSTRACT

BACKGROUND: Ganglioneuroblastoma intermixed (GNBI) is classified as "favorable" histology by International Neuroblastoma Pathology Classification system. However, the International Neuroblastoma Risk Group (INRG) stratifies patients using wider clinicopathological and cytogenetic/molecular parameters. While the diagnosis of GNBI is typically made on resected tumor, it may sometimes be rendered on initial biopsy. We studied GNBI noted at diagnosis to evaluate its correlation with INRG staging and other clinicopathological and molecular features. METHODS: In this retrospective study, clinical, radiological, pathological, cytogenetic, and molecular information from patients with GNBI at diagnosis seen between 1995 and 2021 was analyzed. INRG staging was performed. RESULTS: Of the 15,827 neuroblastoma specimens, GNBI was noted in 237 patients. Of these, 53 had the initial pathological diagnosis of GNBI; median follow-up 3.5 (range: 0.2-14) years. Disease was locoregional in 41 (77%, 16 stage L1 and 25 L2); none relapsed. Twelve (23%) had metastatic disease at presentation; six (50%) relapsed, and two died of disease. MYCN was amplified in two metastatic tumors. Six of 31 (19%) tumors tested had recurrent cytogenetic abnormalities and nonrecurrent somatic gene mutations in 10/23 (43%). The presence of any adverse molecular/cytogenetic findings was associated with metastatic disease (p < .05). For patients with localized GNBI undergoing both biopsy and resection, GNBI was diagnosed in both in 17/19 (90%). CONCLUSIONS: Localized GNBI at diagnosis has excellent long-term clinical outcome even without cytotoxic therapy. For localized GNBI, a biopsy sample is adequate to make the diagnosis. When associated with metastasis at diagnosis, prognosis is poorer, possibly due to associated adverse biological features.


Subject(s)
Ganglioneuroblastoma , Neuroblastoma , Humans , Infant , Ganglioneuroblastoma/diagnosis , Ganglioneuroblastoma/genetics , Ganglioneuroblastoma/pathology , Retrospective Studies , Neuroblastoma/pathology , Prognosis , Genomics , Neoplasm Staging
4.
Genes Chromosomes Cancer ; 61(9): 542-550, 2022 09.
Article in English | MEDLINE | ID: mdl-35430756

ABSTRACT

Tumors with NUTM1 fusions occur predominantly in the thoracic cavity and head and neck region. However, recent literature expanded the location of NUTM1-translocated malignancy to soft tissue, brain, and visceral organs. In this study, we describe the first series of six NUTM1-translocated carcinomas and sarcomas occurring in the genitourinary tract. The sites of origin were kidney (n = 2), bladder (n = 3), and penis (n = 1). All tumors occurred in adulthood (range: 30-78 years). The histologic features were heterogeneous, showing epithelial, spindle cell, or primitive small blue round cell morphology. Glandular architecture, keratinization, rhabdoid cells, or myxoid-to-edematous stromal component were also noted. In three cases, features were in keeping with a carcinoma (two from kidney and one from bladder), whereas the remaining three were classified as malignant undifferentiated neoplasm (MUN)/sarcoma. Fusion partners detected in four cases tested by either FISH and/or RNA sequencing were BRD4 in two kidney tumors, MXD1 in a bladder sarcoma, and MXD4 in a penile sarcoma. NUT immunostain showed diffuse spiculated positivity in five cases. Immunopositivity for various cytokeratins was noted in two tumors. The outcome of NUTM1-rearranged genitourinary malignancy was dismal: four of five cases with follow-up developed distant metastasis, and three suffered disease-specific death. In conclusion, NUTM1-rearranged carcinoma and sarcoma can affect the genitourinary tract, including kidney, bladder, and penis. Histologic features and keratin immunoexpression are highly variable. A NUTM1-fusion positive malignancy may be included in the differential diagnosis of a MUN of the genitourinary tract given the dismal outcome and the existing BET-targeted therapy for tumors with BRD3/4::NUTM1 fusion.


Subject(s)
Carcinoma , Sarcoma , Soft Tissue Neoplasms , Adult , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Biomarkers, Tumor/genetics , Carcinoma/genetics , Cell Cycle Proteins , Humans , Male , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Repressor Proteins/genetics , Sarcoma/genetics , Soft Tissue Neoplasms/genetics , Transcription Factors/genetics
5.
Mod Pathol ; 35(6): 825-835, 2022 06.
Article in English | MEDLINE | ID: mdl-34949764

ABSTRACT

The morphologic spectrum of type 1 papillary renal cell carcinoma (PRCC) is not well-defined, since a significant proportion of cases have mixed type 1 and 2 histology. We analyzed 199 cases of PRCC with any (even if focal) type 1 features, with a median follow-up of 12 years, to identify clinicopathological features associated with outcome. Ninety-five tumors (48%) of the cohort contained some type 2 component (median amount: 25%; IQR: 10%, 70%). As a group they showed high rates of progression-free (PFS) and cancer-specific survival (CSS). Tumor size, mitotic rate, lymphovascular invasion, sarcomatoid differentiation, sheet-like architecture, and lack of tumor circumscription were significantly associated with CSS (p ≤ 0.015) on univariate analysis. While predominant WHO/ISUP nucleolar grade was associated with PFS (p = 0.013) and CSS (p = 0.030), the presence of non-predominant (<50%) nucleolar grade did not show association with outcome (p = 0.7). PFS and CSS showed no significant association with the presence or the amount of type 2 morphology. We compared the molecular alterations in paired type 1 and type 2 areas in a subset of 22 cases with mixed type 1 and 2 features and identified 12 recurrently mutated genes including TERT, ARID1A, KDM6A, KMT2D, NFE2L2, MET, APC, and TP53. Among 78 detected somatic mutations, 61 (78%) were shared between the paired type 1 and type 2 areas. Copy number alterations, including chromosome 7 and 17 gains, were similar between type 1 and 2 areas. These findings support that type 2 features in a PRCC with mixed histology represent either morphologic variance or clonal evolution. Our study underscores the notion that PRCC with any classic type 1 regions is best considered as type 1 PRCC and assigned the appropriate WHO/ISUP nucleolar grade. It provides additional evidence that type 2 PRCC as a separate category should be re-assessed and likely needs to be abandoned.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , DNA Copy Number Variations , Humans , Kidney Neoplasms/pathology , Treatment Outcome
6.
Histopathology ; 81(2): 246-254, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35758203

ABSTRACT

AIM: Clinicopathologic characterisation of a contemporary series of neuroendocrine (NE) differentiation in the setting of prostatic carcinoma (PCa) was examined. METHODS AND RESULTS: We reviewed institutional databases for in-house cases with a history of PCa and histopathologic evidence of NE differentiation during the disease course. In all, 79 cases were identified: 32 primary and 47 metastases. Metastatic lesions were in liver (n = 15), lymph node (n = 9), bone (n = 6), lung (n = 3), brain (n = 1), and other sites (n = 13). In all, 63 of 76 (82%) cases with NE differentiation and available history were posttherapy: six postradiation therapy (RT), 24 post- androgen-deprivation therapy (ADT), and 33 post-RT + ADT. Morphologic assessment (n = 79): (i) 23 pure small-cell/high-grade NE carcinoma (HGNEC): 20/23 metastatic; (ii) 10 combined high-grade PCa and small-cell/HGNEC: 9/10 primary; (iii) 15 PCa with diffuse NE immunohistochemistry (IHC) marker positivity/differentiation, associated with nested to sheet-like growth of cells with abundant cytoplasm and prominent nucleoli, yet diffuse positivity for at least one prostatic and one NE IHC marker: all metastatic; (iv) 11 PCa with patchy NE differentiation, displaying more than single-cell positivity for NE IHC: five primary / six metastatic; (v) nine PCa with focal NE marker positive cells: four primary / five metastatic; (vi) 11 PCa with 'Paneth cell-like' change: all primary. CONCLUSIONS: In this contemporary series, the majority of NE differentiation in the setting of PCa was seen posttherapy. We highlight the tendencies of small-cell/HGNEC and PCa with diffuse NE differentiation by IHC to occur in metastatic settings, while morphologically combined high-grade PCa + small-cell/HGNEC and 'Paneth cell-like' change occur in primary disease.


Subject(s)
Carcinoma, Neuroendocrine , Carcinoma, Small Cell , Prostatic Neoplasms , Androgen Antagonists , Carcinoma, Neuroendocrine/pathology , Carcinoma, Small Cell/pathology , Humans , Immunohistochemistry , Male , Prostate/pathology , Prostatic Neoplasms/pathology
7.
Mod Pathol ; 34(2): 445-456, 2021 02.
Article in English | MEDLINE | ID: mdl-32879414

ABSTRACT

Mucinous tubular and spindle cell carcinoma (MTSCC) is a rare subtype of renal cell carcinoma with characteristic histologic features and chromosomal alterations. Although typically indolent, a small subset of cases has been reported to exhibit aggressive clinical behavior. We retrospectively identified 33 patients with MTSCC, consisting of 10 cases of locally advanced/metastatic MTSCC (pT3 or N1 or M1) and 23 kidney-confined MTSCC (pT1/T2) without disease recurrence or progression. Utilizing a single-nucleotide polymorphism array and a targeted next-generation sequencing platform, we examined genome-wide molecular alterations in 24 cases, including 11 available samples from 8 patients with locally advanced/metastatic MTSCC. Ten patients with locally advanced/metastatic MTSCC were 8 females (80%) and 2 males (20%). At nephrectomy, 7 of these 10 cases (70%) were pT3 or pN1 while the remaining 3 (30%) were pT1/T2. Eight patients (80%) developed metastases and common sites included lymph node (4, 40%), bone (4, 40%), and retroperitoneum (3, 30%). Four patients died of disease (40%) during follow-up. Locally advanced/metastatic MTSCCs shared typical MTSCC genomic profiles with loss of chromosomes 1, 4, 6, 8, 9, 13, 14, 15, and 22, while some exhibited additional complex genomic alterations, most frequently a relative gain of 1q (7/8). Homozygous loss of CDKN2A/B was observed in 3 (38%) locally advanced/metastatic MTSCCs. Tumor necrosis, solid nested/sheet pattern, irregular trabecular/single-file infiltration in a desmoplastic stroma, lymphovascular space invasion, and increased mitotic activity were associated with locally advanced/metastatic MTSCCs (all p < 0.05). Our findings reveal that MTSCCs with aggressive clinical behavior have progressed through clonal evolution; CDKN2A/B deletion and additional complex genomic abnormalities may contribute to this process. Recognizing the morphologic presentation of high-grade MTSCC and evaluating adverse histologic features seen in these tumors can help establish a definitive diagnosis and stratify patients for treatment and prognostication.


Subject(s)
Adenocarcinoma, Mucinous/genetics , Adenocarcinoma, Mucinous/pathology , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Aged , Female , Humans , Male , Middle Aged , Retrospective Studies
8.
J Pathol ; 248(3): 260-265, 2019 07.
Article in English | MEDLINE | ID: mdl-30838648

ABSTRACT

Inverted urothelial papilloma (IUP) and urothelial papilloma (UP) are rare urothelial neoplasms that typically follow a benign clinical course. Oncogenic mutations in FGFR3, HRAS, and the TERT promoter have been reported in these entities but no comprehensive molecular analysis has been performed. We sought to characterize the genomic landscape of IUP and UP using whole-exome and targeted next-generation sequencing. In IUP, 10 of 11 tumors harbored oncogenic hotspot mutations in HRAS and the remaining tumor had an oncogenic KRAS mutation. None of the IUP tumors harbored TERT promoter or FGFR3 mutations. In UP, 8 of 11 tumors had oncogenic KRAS mutations and two had oncogenic HRAS mutations. One UP tumor had oncogenic mutations in FGFR3, PIK3CA, and the TERT promoter, and arose in a patient with recurrent non-invasive papillary urothelial carcinomas. In contrast to urothelial carcinoma, the APOBEC mutational signature was not present in any IUP and UP tumors, and oncogenic alterations in chromatin remodeling genes were uncommon in both IUP and UP. The current study suggests that IUP and UP are driven primarily by RAS pathway activation and lack the more common genomic features of urothelial cancers. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Papilloma, Inverted/pathology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Urinary Bladder/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/pathology , Databases, Genetic , Female , Genomics , Humans , Male , Middle Aged , Mutation/genetics , Papilloma, Inverted/genetics , Promoter Regions, Genetic/genetics
9.
Mod Pathol ; 32(5): 701-709, 2019 05.
Article in English | MEDLINE | ID: mdl-30622286

ABSTRACT

Tubulocystic renal cell carcinoma, a unique tumor, was recently included as a new entity in the World Health Organization classification of renal tumors. It has variably been reported to be related to other renal cell carcinomas, including papillary renal cell carcinoma, fumarate hydratase-deficient carcinoma, and others, likely because many such carcinomas may show variable amounts of tubulocystic architecture. The published data characterizing the molecular features of these tumors are inconsistent. We studied nine "pure" tubulocystic renal cell carcinomas, as defined by International Society of Urologic Pathologists (ISUP) and World Health Organization (WHO), by targeted next-generation sequencing, and fluorescence in situ hybridization for X and Y chromosomes, to investigate if these show any unique characteristics or any overlap with known mutational/molecular profiles or copy number alterations in other subtypes of renal cell carcinoma. All nine tubulocystic carcinomas demonstrated combined losses at chromosome 9 and gains at chromosome 17, as well as, loss of chromosome Y (in 5/5). None of the tumors showed mutational profiles characteristic of other renal neoplasms, including those seen in fumarate hydratase-deficient renal cell carcinoma. Recurrent mutations in chromatin-modifying genes, KMT2C and KDM5C, were detected in two of nine tumors. Thus, tubulocystic renal cell carcinoma, if defined strictly, at the clinical and pathologic level, demonstrates genomic features distinct from other subtypes of renal cell carcinoma. These findings support the contention that tubulocystic renal cell carcinoma should be diagnosed only using strict morphological criteria and only when presenting in a "pure" form; presence of variable papillary, poorly differentiated, or other architectural patterns most likely do not belong to the category of tubulocystic renal cell carcinoma.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Neoplasms, Cystic, Mucinous, and Serous/genetics , Transcriptome , Adult , Aged , Carcinoma, Renal Cell/pathology , Chromosome Aberrations , Chromosomes, Human, X , Chromosomes, Human, Y , DNA-Binding Proteins/genetics , Female , Gene Amplification , Genetic Predisposition to Disease , Histone Demethylases/genetics , Humans , Kidney Neoplasms/pathology , Male , Middle Aged , Mutation , Neoplasms, Cystic, Mucinous, and Serous/pathology , Phenotype , United States
11.
Am J Ther ; 23(1): e252-5, 2016.
Article in English | MEDLINE | ID: mdl-25486519

ABSTRACT

St. Jude mechanical prosthesis is the most commonly used prosthetic device with least valvular complications with excellent hemodynamics. However, prosthetic valve thrombosis is one of the serious complications, with rates between 0.03% and 0.13% per patient-year depending on the type of anticoagulation used and compliance to the therapy. Transthoracic echocardiography (TTE) is the initial screening tool (class I) that would provide clues for the assessment of valvular hemodynamics. Fluoroscopy is an alternate imaging modality for the assessment of mechanical leaflet motion, especially in patients when prosthetic valves are difficult to image on TTE or transesophageal echocardiography. A complete fluoroscopic evaluation of a prosthetic valve includes assessment of valvular motion and structural integrity. Opening and closing angles can be measured fluoroscopically to determine whether a specific valve is functioning properly. We discuss a case of a 91-year-old man with thrombosis of bileaflet mechanical mitral prosthesis that was demonstrated on real-time fluoroscopy (not evident on TTE). An algorithmic approach to diagnosis and management of prosthetic heart valve thrombosis is outlined.


Subject(s)
Heart Valve Prosthesis/adverse effects , Mitral Valve/surgery , Thrombosis/diagnosis , Thrombosis/therapy , Aged , Aged, 80 and over , Echocardiography , Echocardiography, Transesophageal , Fluoroscopy , Humans , Male
12.
Skeletal Radiol ; 45(7): 929-35, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27020452

ABSTRACT

OBJECTIVE: Giant cell tumor of bone (GCTB) is a locally aggressive primary bone tumor. Its malignant counterpart is quite rare. Rarely, a conventional GCTB shows marked nuclear atypia, referred to as symplastic/pseudoanaplastic change, which can mimic sarcomatous transformation. Recently, somatic driver mutations of histone H3.3 exclusively in H3F3A have been described in GCTB. We report a series of 9 cases of GCTB with symplastic/pseudoanaplastic change, along with analysis of H3F3A variants. MATERIALS AND METHODS: Nine cases of GCTB with symplastic change were identified. Clinico-radiological features, morphological features, and immunohistochemical stain for Ki-67 stain were reviewed. H3F3A variants were also analyzed using Sanger sequencing. RESULTS: Histologically, conventional giant cell tumor areas with scattered foci of markedly atypical cells were seen in all of the cases and all showed rare if any Ki-67 labeling. One patient had received denosumab treatment and another radiation therapy. Radiological features were characteristic of conventional GCTB. Mutation in H3F3A (p.Gly34Trp [G34W]) was found in 6 of the 7 cases. Clinical follow-up ranged from 6 to 208 months. Local recurrences were seen in 4 cases (44 %). CONCLUSIONS: GCTB with symplastic/pseudoanaplastic change is an uncommon variant of conventional GCTB, which can mimic primary sarcoma or sarcomatous transformation. These tumors possess the same missense mutation in histone H3.3 as conventional GCTB.


Subject(s)
Bone Neoplasms/diagnostic imaging , Bone Neoplasms/genetics , Giant Cell Tumor of Bone/diagnostic imaging , Giant Cell Tumor of Bone/genetics , Histones/genetics , Adult , Denosumab , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Retrospective Studies
13.
Mod Pathol ; 28(6): 845-853, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25676555

ABSTRACT

Integrated sequencing analysis identified a group of tumors among clear cell renal cell carcinomas characterized by hotspot mutations in TCEB1 (a gene that contributes to the VHL complex to ubiquitinate hypoxia-inducible factor). We analyzed 11 tumors from two distinct cohorts with TCEB1 mutations along with an expanded cohort to assess whether these should be considered an entity distinct from clear cell renal cell carcinoma and clear cell papillary renal cell carcinoma. All tumors were characterized by hotspot mutations in TCEB1 Y79C/S/F/N or A100P. Morphological and immunohistochemical characteristics of the tumors were assessed by two experienced genitourinary pathologists. Clinical and pathological variables, copy number alterations, mutations, and expression signatures were compared with a cohort of TCEB1 wild-type tumors. All TCEB1-mutated tumors were VHL and PBRM1 wild type and contained distinct copy number profiles including loss of heterozygosity of chromosome 8, the location of TCEB1 (8q21.11). All tumors lacked the clear cell renal cell carcinoma signature 3p loss and contained distinct gene expression signatures. None of the clear cell papillary tumors harbored TCEB1 mutations. Pathologically, all TCEB1-mutated tumors shared characteristic features including thick fibromuscular bands transecting the tumor, pure clear cell cytology frequently with cells showing voluminous cytoplasm, and clear cell renal cell carcinoma-like acinar areas associated with infolding tubular and focally papillary architecture. The presence of voluminous cytoplasm, absence of luminal polarization of tumor nuclei, and lack of extensive cup-like distribution of carbonic anhydrase-IX expression distinguish it from clear cell papillary carcinoma. None of the patients developed metastases at last follow-up (median 48 months). In sum, TCEB1-mutated renal cell carcinoma is a distinct entity with recurrent hotspot mutations, specific copy number alterations, pathway activation, and characteristic morphological features. Further clinical follow-up is needed to determine whether these tumors are more indolent compared with the conventional clear cell renal cell carcinoma.


Subject(s)
Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mutation , Transcription Factors/genetics , Adult , Aged , Biomarkers, Tumor/analysis , DNA Mutational Analysis , Elongin , Female , Genomics , Humans , Immunohistochemistry , Male , Middle Aged
14.
Clin Imaging ; 83: 83-86, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35007888

ABSTRACT

PURPOSE: Acquired cystic kidney disease-associated renal cell carcinoma (ACKD-RCC) is a relatively recently described entity with scarce literature describing its imaging appearance (1, 2). The purpose of this study was to determine typical and potentially unique features of ACKD-RCC on CT scan that could aid lesion identification. MATERIALS AND METHODS: A retrospective review of the CT scans of 24 patients with 29 histologically proven ACKD-RCC lesions was performed. Imaging features were recorded based on consensus readings of two radiologists. RESULTS: Tumors ranged in size from 1.2 to 5 cm. Nineteen lesions were right-sided and 10 left-sided. Nineteen lesions were exophytic. One patient had bilateral lesions and three patients had multiple lesions in the same kidney. All lesions had well-defined margins with 21 round, 7 lobulated and 1 crescentic in shape. On non-enhanced exam 4 lesions were hypodense, 16 iso-dense and 9 hyperdense; 5 had gross calcifications. Twenty two patients had contrast-enhanced CT exams, with 13 lesions demonstrating homogeneous enhancement (solid pattern) and 14 having inhomogeneous enhancement (cystic or mixed solid and cystic pattern). Only 1 patient had metastatic disease. Eight patients had a history of renal transplants. CONCLUSION: ACKD-RCCs are well-defined lesions of variable size that are almost always rounded and most often exophytic. They occasionally have calcifications and are not uncommonly hyperdense on non-enhanced exam. They are most often iso-dense on non-enhanced exam and can be solid, cystic or mixed in attenuation on enhanced exam.


Subject(s)
Carcinoma, Renal Cell , Kidney Diseases, Cystic , Kidney Neoplasms , Kidney Transplantation , Carcinoma, Renal Cell/complications , Carcinoma, Renal Cell/diagnostic imaging , Carcinoma, Renal Cell/pathology , Humans , Kidney Diseases, Cystic/diagnostic imaging , Kidney Neoplasms/diagnostic imaging , Kidney Neoplasms/pathology , Retrospective Studies , Tomography, X-Ray Computed
15.
Urol Pract ; 9(5): 459-465, 2022 Sep.
Article in English | MEDLINE | ID: mdl-37145713

ABSTRACT

INTRODUCTION: Our goal was to determine whether zonal origin of anterior dominant prostate cancers is associated with clinical outcome among patients treated with radical prostatectomy. METHODS: We investigated the clinical outcomes of 197 patients with previously well-characterized anterior dominant prostatic tumors on radical prostatectomy. Univariable Cox proportional hazards models were used to test for an association between anterior peripheral zone (PZ) or transition zone (TZ) tumor location and clinical outcomes. RESULTS: Zonal origin of anterior dominant tumors: 97/197 (49%) anterior PZ, 70 (36%) TZ, 14 (7%) both zones and 16 (8%) indeterminate zone. Comparing anterior PZ and TZ tumors, there were no significant differences in Grade group, incidence of extraprostatic extension or surgical margin positivity rate. Overall, 19 (9.6%) patients experienced biochemical recurrence (BCR), including 10 with anterior PZ origin and 5 with TZ origin. Median followup time among those without BCR was 9.5 years (IQR 7.2, 12.7). BCR-free survival at 5 and 10 years was 91% and 89% for anterior PZ tumors, and 94% and 92% for TZ tumors, respectively. On univariate analysis, there was no evidence of a difference in time to BCR between anterior PZ and TZ tumor zone of origin (p=0.5). CONCLUSIONS: In this anatomically well-characterized cohort of anterior dominant prostate cancers, long-term BCR-free survival was not significantly associated with zone of origin. Future studies utilizing zone of origin as a parameter should consider separating anterior and posterior PZ localization, as outcomes may differ.

16.
Clin Genitourin Cancer ; 20(5): 431-441, 2022 10.
Article in English | MEDLINE | ID: mdl-35676169

ABSTRACT

INTRODUCTION: Small cell carcinoma of the bladder (SCCB) is a rare variant of bladder cancer with poor outcomes. We evaluated long-term outcomes of nonmetastatic (M0) and metastatic (M1) SCCB and correlated pathologic response with genomic alterations of patients treated with neoadjuvant chemotherapy (NAC). PATIENTS AND METHODS: Clinical history and pathology samples from SCCB patients diagnosed at our institution were reviewed. RESULTS: One hundred and ninety-nine SCCB patients were identified. (M0: 147 [74%]; M1: 52 [26%]). Among M0 patients, 108 underwent radical cystectomy (RC) (NAC: 71; RC only: 23; adjuvant chemotherapy: 14); 14 received chemoradiotherapy; the rest received chemotherapy alone or no cancer-directed therapy. RC-only patients had a median follow-up of 9.1 years, and median disease-free survival (DFS) and overall survival (OS) were 1.1 and 1.2 years, respectively. NAC patients had pathologic response (

Subject(s)
Carcinoma, Small Cell , Urinary Bladder Neoplasms , Carcinoma, Small Cell/pathology , Chemotherapy, Adjuvant , Cystectomy , Genomics , Humans , Neoadjuvant Therapy , Retrospective Studies , Urinary Bladder/pathology , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Xeroderma Pigmentosum Group D Protein
17.
Nat Commun ; 13(1): 6575, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36323682

ABSTRACT

Cancers arising from the bladder urothelium often exhibit lineage plasticity with regions of urothelial carcinoma adjacent to or admixed with regions of divergent histomorphology, most commonly squamous differentiation. To define the biologic basis for and clinical significance of this morphologic heterogeneity, here we perform integrated genomic analyses of mixed histology bladder cancers with separable regions of urothelial and squamous differentiation. We find that squamous differentiation is a marker of intratumoral genomic and immunologic heterogeneity in patients with bladder cancer and a biomarker of intrinsic immunotherapy resistance. Phylogenetic analysis confirms that in all cases the urothelial and squamous regions are derived from a common shared precursor. Despite the presence of marked genomic heterogeneity between co-existent urothelial and squamous differentiated regions, no recurrent genomic alteration exclusive to the urothelial or squamous morphologies is identified. Rather, lineage plasticity in bladder cancers with squamous differentiation is associated with loss of expression of FOXA1, GATA3, and PPARG, transcription factors critical for maintenance of urothelial cell identity. Of clinical significance, lineage plasticity and PD-L1 expression is coordinately dysregulated via FOXA1, with patients exhibiting morphologic heterogeneity pre-treatment significantly less likely to respond to immune checkpoint inhibitors.


Subject(s)
Carcinoma, Squamous Cell , Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Humans , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Transitional Cell/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Phylogeny , Urinary Bladder Neoplasms/pathology , Cell Lineage
18.
Can Urol Assoc J ; 15(10): 339-344, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33750519

ABSTRACT

INTRODUCTION: We compared clinicopathological characteristics and outcomes of radical nephrectomy (RN) for small renal masses (SRM) in patients with end-stage renal disease (ESRD) before or after transplant at a high-volume urologic and transplant center. METHODS: We performed a retrospective review of patients with ESRD (glomerular filtration rate [GFR] <15 mL/min) who underwent RN for suspected malignant SRM from 2000-2018. Group 1 consisted of patients who underwent RN after transplant; group 2 underwent RN prior to transplant, and group 3 underwent RN without subsequent transplant. Dominant tumor size and histopathological characteristics, recurrence, and survival outcomes were compared between groups. Chi-squared and Mann-Whitney U tests were used to compare categorical and continuous baseline and histopathologic characteristics, respectively. Univariate analysis and log rank test were used to compare RCC recurrence rates. RESULTS: We identified 34 nephrectomies in group 1, 27 nephrectomies in group 2, and 70 nephrectomies in group 3. Median time from transplant to SRM radiological diagnosis in group 1 was 87 months, and three months from diagnosis to nephrectomy for all groups. There were no statistically significant differences between pathological dominant mass size, histological subtype breakdown, grade, or stage between the groups. Rates of benign histology were similar between the groups. Univariate analysis did not reveal a statistically significant difference in recurrence-free survival between the groups (p=0.9). CONCLUSIONS: Patients undergoing nephrectomy before or after transplant for SRM have similar indolent clinicopathological characteristics and low recurrence rates. Our results suggest that chronic immunosuppression does not adversely affect SRM biology.

19.
Med Mycol Case Rep ; 24: 82-85, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31080714

ABSTRACT

Cryptococcal infections are acquired by inhalation of encapsulated yeast cells or basidiospores. While Cryptococcus has a propensity to invade the lungs and central nervous system, other sites can be affected. Laryngeal cryptococcosis is rare with less than 30 previously reported cases, which commonly occurred in apparently immunocompetent hosts on inhaled corticosteroids. We present a case of laryngeal cryptococcosis with a long-term inhaled corticosteroid use, co-infection of pulmonary Mycobacterium avium-intracellulare, and mannose-binding lectin deficiency.

20.
Elife ; 82019 04 01.
Article in English | MEDLINE | ID: mdl-30924768

ABSTRACT

While genomic sequencing routinely identifies oncogenic alterations for the majority of cancers, many tumors harbor no discernable driver lesion. Here, we describe the exceptional molecular phenotype of a genomically quiet kidney tumor, clear cell papillary renal cell carcinoma (CCPAP). In spite of a largely wild-type nuclear genome, CCPAP tumors exhibit severe depletion of mitochondrial DNA (mtDNA) and RNA and high levels of oxidative stress, reflecting a shift away from respiratory metabolism. Moreover, CCPAP tumors exhibit a distinct metabolic phenotype uniquely characterized by accumulation of the sugar alcohol sorbitol. Immunohistochemical staining of primary CCPAP tumor specimens recapitulates both the depletion of mtDNA-encoded proteins and a lipid-depleted metabolic phenotype, suggesting that the cytoplasmic clarity in CCPAP is primarily related to the presence of glycogen. These results argue for non-genetic profiling as a tool for the study of cancers of unknown driver.


Subject(s)
Carcinoma, Renal Cell/pathology , Cell Respiration , Kidney Neoplasms/pathology , Aerobiosis , Histocytochemistry , Humans , Immunohistochemistry , Metabolic Networks and Pathways , Oxidation-Reduction
SELECTION OF CITATIONS
SEARCH DETAIL