70 results on '"Gopalan, Anuradha"'
Search Results
2. Histopathologic and Molecular Characterization of IDH-Mutant Prostatic Adenocarcinoma
- Author
-
Samueli, Benzion, Al-Ahmadie, Hikmat, Chen, Ying-Bei, Gopalan, Anuradha, Sarungbam, Judy, Tickoo, Satish K., Reuter, Victor E., Fine, Samson W., and Chen, Jie-Fu
- Abstract
Gain-of-function isocitrate dehydrogenase (IDH) mutations are pathogenically significant in many tumor types and are actionable in cholangiocarcinoma, low-grade glioma, and acute myeloid leukemia. Rare IDHmutations have been described in prostatic adenocarcinoma (PCa). Recent publications have suggested that psammomatous calcifications in PCa are associated with IDH1mutations. In this retrospective study, we queried our institutional clinical sequencing database (cohort 1), and previously published PCa data sets in cBioPortal (cohort 2). Samples were stratified based on oncogenic hotspot IDHmutations at IDH1R132 and IDH2R140/R172, and other nonhotspot IDHmutations. Seventeen (0.4%) cases were identified from 4033 PCa cases in cohort 1 harboring mutually exclusive oncogenic hotspot IDH1(N = 15, 1 of which was subclonal) or IDH2(N = 2) mutations, and 20 (0.5%) cases had nonhotspot IDH1/2mutations. A histologic review of 13 cases with IDH1hotspot mutations and available material showed grade group 3 or higher disease. Immunohistochemistry was performed on cases with IDH1hotspot mutations when possible and showed AR, PSA, PSMA, and NKX3.1 positive in all the 4 cases stained. In cohort 2, 9 cases (0.3%) harboring IDH1hotspot mutations were identified from 2749 patients, and 9 cases carried nonhotspot IDH1/2mutations. The combined cohorts of 23 PCa cases with clonal IDH1hotspot mutations had no ETS fusions, SPOPhotspot mutations, and somatic or germline alterations in BRCA1/2, ATM, RB1,or AR; 19 cases with successful microsatellite instability testing were all microsatellite stable. Conversely, among 29 cases with nonhotspot IDHmutations, there were 4 with TMPRSS2::ERGfusions, 6 with SPOPhotspot mutations, and 10 with ARamplifications/hotspot mutations; 8 were microsatellite instability high. Notably, two cases with IDH1hotspot mutations had psammomatous calcifications. Our findings provide evidence that IDH1hotspot mutations serve as driver alterations in this rare yet distinct molecular subset of PCa. Further studies are warranted to correlate response to androgen deprivation and IDH inhibitors.
- Published
- 2025
- Full Text
- View/download PDF
3. Long-term Outcomes of Local and Metastatic Small Cell Carcinoma of the Urinary Bladder and Genomic Analysis of Patients Treated With Neoadjuvant Chemotherapy
- Author
-
Teo, Min Yuen, Guercio, Brendan J., Arora, Arshi, Hao, Xueli, Regazzi, Ashley M., Donahue, Timothy, Herr, Harry W., Goh, Alvin C., Cha, Eugene K., Pietzak, Eugene, Donat, Sherri M., Dalbagni, Guido, Bochner, Bernard H., Olgac, Semra, Sarungbam, Judy, Sirintrapun, S. Joseph, Chen, Ying-Bei, Gopalan, Anuradha, Fine, Samson W., Tickoo, Satish K., Reuter, Victor E., Weigelt, Britta, Schultheis, Anne M., Funt, Samuel A., Bajorin, Dean F., Solit, David B., Iyer, Gopa, Ostrovnaya, Irina, Rosenberg, Jonathan E., and Al-Ahmadie, Hikmat
- Abstract
•In small cell carcinoma of the bladder, neoadjuvant chemotherapy improves survival.•Pathologic response is associated with superior long-term survival.•Metastatic disease is associated with poor clinical outcomes.•Small cell carcinoma of the bladder harbors high tumor mutation burden.•ERCC2mutations are associated with complete response to neoadjuvant chemotherapy.
- Published
- 2022
- Full Text
- View/download PDF
4. Impact of Zone of Origin in Anterior Dominant Prostate Cancer: Long-Term Biochemical Recurrence-Free Survival in an Anatomically Well-Characterized Cohort
- Author
-
Fine, Samson W., Al-Ahmadie, Hikmat A., Vertosick, Emily, Vickers, Andrew J., Chen, Ying-Bei, Gopalan, Anuradha, Sarungbam, Judy, Sirintrapun, S. Joseph, Tickoo, Satish K., Eastham, James A., Scardino, Peter T., and Reuter, Victor E.
- Published
- 2022
- Full Text
- View/download PDF
5. Differential NEUROD1, ASCL1, and POU2F3 Expression Defines Molecular Subsets of Bladder Small Cell/Neuroendocrine Carcinoma With Prognostic Implications
- Author
-
Akbulut, Dilara, Whiting, Karissa, Teo, Min-Yuen, Tallman, Jacob E., Ozcan, Gamze Gokturk, Basar, Merve, Jia, Liwei, Rammal, Rayan, Chen, Jie-Fu, Sarungbam, Judy, Chen, Ying-Bei, Gopalan, Anuradha, Fine, Samson W., Tickoo, Satish K., Mehra, Rohit, Baine, Marina, Bochner, Bernard H., Pietzak, Eugene J., Bajorin, Dean F., Rosenberg, Jonathan E., Iyer, Gopa, Solit, David B., Reuter, Victor E., Rekhtman, Natasha, Ostrovnaya, Irina, and Al-Ahmadie, Hikmat
- 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.
- Published
- 2024
- Full Text
- View/download PDF
6. Papillary renal cell carcinoma: a single institutional study of 199 cases addressing classification, clinicopathologic and molecular features, and treatment outcome
- Author
-
Murugan, Paari, Jia, Liwei, Dinatale, Renzo G., Assel, Melissa, Benfante, Nicole, Al-Ahmadie, Hikmat A., Fine, Samson W., Gopalan, Anuradha, Sarungbam, Judy, Sirintrapun, S. Joseph, Hakimi, A. Ari, Russo, Paul, Chen, Ying-Bei, Tickoo, Satish K., and Reuter, Victor E.
- 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.
- Published
- 2022
- Full Text
- View/download PDF
7. Prognostic and therapeutic significance of COP9 signalosome subunit CSN5in prostate cancer
- Author
-
Mazzu, Ying Z., Liao, Yu-Rou, Nandakumar, Subhiksha, Jehane, Lina E., Koche, Richard P., Rajanala, Sai Harisha, Li, Ruifang, Zhao, HuiYong, Gerke, Travis A., Chakraborty, Goutam, Lee, Gwo-Shu Mary, Nanjangud, Gouri J., Gopalan, Anuradha, Chen, Yu, and Kantoff, Philip W.
- Abstract
Chromosome 8q gain is associated with poor clinical outcomes in prostate cancer, but the underlying biological mechanisms remain to be clarified. CSN5, a putative androgen receptor (AR) partner that is located on chromosome 8q, is the key subunit of the COP9 signalosome, which deactivates ubiquitin ligases. Deregulation of CSN5 could affect diverse cellular functions that contribute to tumor development, but there has been no comprehensive study of its function in prostate cancer. The clinical significance of CSN5 amplification/overexpression was evaluated in 16 prostate cancer clinical cohorts. Its oncogenic activity was assessed by genetic and pharmacologic perturbations of CSN5 activity in prostate cancer cell lines. The molecular mechanisms of CSN5 function were assessed, as was the efficacy of the CSN5 inhibitor CSN5i-3 in vitro and in vivo. Finally, the transcription cofactor activity of CSN5 in prostate cancer cells was determined. The prognostic significance of CSN5 amplification and overexpression in prostate cancer was independent of MYC amplification. Inhibition of CSN5 inhibited its oncogenic function by targeting AR signaling, DNA repair, multiple oncogenic pathways, and spliceosome regulation. Furthermore, inhibition of CSN5 repressed metabolic pathways, including oxidative phosphorylation and glycolysis in AR-negative prostate cancer cells. Targeting CSN5 with CSN5i-3 showed potent antitumor activity in vitro and in vivo. Importantly, CSN5i-3 synergizes with PARP inhibitors to inhibit prostate cancer cell growth. CSN5 functions as a transcription cofactor to cooperate with multiple transcription factors in prostate cancer. Inhibiting CSN5 strongly attenuates prostate cancer progression and could enhance PARP inhibition efficacy in the treatment of prostate cancer.
- Published
- 2022
- Full Text
- View/download PDF
8. CD274(PD-L1) Copy Number Changes (Gain) & Response to Immune Checkpoint Blockade Therapy in Carcinomas of the Urinary Tract
- Author
-
Gupta, Sounak, Vanderbilt, Chad M., Zhang, Yanming, Tickoo, Satish K., Fine, Samson W., Gopalan, Anuradha, Chen, Ying-Bei, Sirintrapun, Sahussapont J., Teo, Min Yuen, Funt, Samuel A., Iyer, Gopa, Rosenberg, Jonathan E., Bajorin, Dean F., Bochner, Bernard H., Pietzak, Eugene J., Ross, Dara S., Ladanyi, Marc, Cheville, John C., Solit, David B., Reuter, Victor E., and Al-Ahmadie, Hikmat A.
- Abstract
BACKGROUND: Immune checkpoint inhibitors are an important therapeutic option for urothelial carcinoma, but durable responses are achieved in a minority of patients. Identifying pre-treatment biomarkers that may predict response to these therapies or who exhibit intrinsic resistance, is of paramount importance.OBJECTIVE: To explore the prevalence of PD-L1copy number alteration in urothelial carcinoma and correlate with response to immune checkpoint inhibitors.METHODS: We analyzed a cohort of 1050 carcinomas of the bladder and upper urinary tract that underwent targeted next generation sequencing, prospectively. We assessed PD-L1 protein expression, copy number status (next generation sequencing/FISH), and detailed treatment response.RESULTS: We identified 9 tumors with PD-L1amplification and 9 tumors with PD-L1deletion. PD-L1 protein expression was the highest in PD-L1amplified tumors. Of the 9 patients whose tumors harbored PD-L1amplification, 6 received immunotherapy with 4 deriving clinical benefit, and 2 achieving durable response. Of the 9 patients whose tumors had PD-L1copy number losses, 4 received immunotherapy with 3 experiencing disease progression.CONCLUSIONS: PD-L1copy number alterations may serve as potential biomarkers of response to immunotherapy in urothelial carcinoma patients, if validated in larger cohorts.
- Published
- 2021
- Full Text
- View/download PDF
9. L1 Cell Adhesion Molecule (L1CAM) Expression and Molecular Alterations Distinguish Low-Grade Oncocytic Tumor From Eosinophilic Chromophobe Renal Cell Carcinoma
- Author
-
Alghamdi, Mohammed, Chen, Jie-Fu, Jungbluth, Achim, Koutzaki, Sirma, Palmer, Matthew B., Al-Ahmadie, Hikmat A., Fine, Samson W., Gopalan, Anuradha, Sarungbam, Judy, Sirintrapun, S. Joseph, Tickoo, Satish K., Reuter, Victor E., and Chen, Ying-Bei
- 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)-mTORcomplex 1 (mTORC1) pathway, most frequently in MTORand RHEBgenes; 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.
- Published
- 2024
- Full Text
- View/download PDF
10. A Pan-Cancer Study of Somatic TERTPromoter Mutations and Amplification in 30,773 Tumors Profiled by Clinical Genomic Sequencing
- Author
-
Gupta, Sounak, Vanderbilt, Chad M., Lin, Yun-Te, Benhamida, Jamal K., Jungbluth, Achim A., Rana, Satshil, Momeni-Boroujeni, Amir, Chang, Jason C., Mcfarlane, Tiffany, Salazar, Paulo, Mullaney, Kerry, Middha, Sumit, Zehir, Ahmet, Gopalan, Anuradha, Bale, Tejus A., Ganly, Ian, Arcila, Maria E., Benayed, Ryma, Berger, Michael F., Ladanyi, Marc, and Dogan, Snjezana
- Abstract
TERTgene promoter mutations are known in multiple cancer types. Other TERTalterations remain poorly characterized. Sequencing data from 30,773 tumors analyzed by a hybridization capture next-generation sequencing assay (Memorial Sloan Kettering Cancer Center Integrated Mutation Profiling of Actionable Cancer Targets) were analyzed for the presence of TERTalterations. Promoter rearrangements (500 bases upstream of the transcriptional start site), hypermethylation (n= 57), and gene expression (n= 155) were evaluated for a subset of cases. Mutually exclusive and recurrent promoter mutations were identified at three hot spots upstream of the transcriptional start site in 11.3% of cases (−124: 74%; −146: 24%; and −138: <2%). Mutually exclusive amplification events were identified in another 2.3% of cases, whereas mutually exclusive rearrangements proximal to the TERTgene were seen in 24 cases. The highest incidence of TERTpromoter mutations was seen in cutaneous melanoma (82%), whereas amplification events significantly outnumbered promoter mutations in well-differentiated/dedifferentiated liposarcoma (14.1% versus 2.4%) and adrenocortical carcinoma (13.6% versus 4.5%). Gene expression analysis suggests that the highest levels of gene expression are seen in cases with amplifications and rearrangements. Hypermethylation events upstream of the TERTcoding sequence were not mutually exclusive with known pathogenic alterations. Studies aimed at defining the prevalence and prognostic impact of TERTalterations should incorporate other pathogenic TERTalterations as these may impact telomerase function.
- Published
- 2021
- Full Text
- View/download PDF
11. Adverse histology, homozygous loss of CDKN2A/B, and complex genomic alterations in locally advanced/metastatic renal mucinous tubular and spindle cell carcinoma
- Author
-
Yang, Chen, Cimera, Robert S., Aryeequaye, Ruth, Jayakumaran, Gowtham, Sarungbam, Judy, Al-Ahmadie, Hikmat A., Gopalan, Anuradha, Sirintrapun, S. Joseph, Fine, Samson W., Tickoo, Satish K., Epstein, Jonathan I., Reuter, Victor E., Zhang, Yanming, and Chen, Ying-Bei
- 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/Bwas 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/Bdeletion 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.
- Published
- 2021
- Full Text
- View/download PDF
12. Adverse histology, homozygous loss of CDKN2A/B, and complex genomic alterations in locally advanced/metastatic renal mucinous tubular and spindle cell carcinoma
- Author
-
Yang, Chen, Cimera, Robert S., Aryeequaye, Ruth, Jayakumaran, Gowtham, Sarungbam, Judy, Al-Ahmadie, Hikmat A., Gopalan, Anuradha, Sirintrapun, S. Joseph, Fine, Samson W., Tickoo, Satish K., Epstein, Jonathan I., Reuter, Victor E., Zhang, Yanming, and Chen, Ying-Bei
- 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/Bwas 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/Bdeletion 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.
- Published
- 2021
- Full Text
- View/download PDF
13. CD274(PD-L1) Copy Number Changes (Gain) & Response to Immune Checkpoint Blockade Therapy in Carcinomas of the Urinary Tract
- Author
-
Gupta, Sounak, Vanderbilt, Chad M., Zhang, Yanming, Tickoo, Satish K., Fine, Samson W., Gopalan, Anuradha, Chen, Ying-Bei, Sirintrapun, Sahussapont J., Teo, Min Yuen, Funt, Samuel A., Iyer, Gopa, Rosenberg, Jonathan E., Bajorin, Dean F., Bochner, Bernard H, Pietzak, Eugene J, Ross, Dara S., Ladanyi, Marc, Cheville, John C., Solit, David B., Reuter, Victor E., and Al-Ahmadie, Hikmat A.
- Abstract
Immune checkpoint inhibitors are an important therapeutic option for urothelial carcinoma, but durable responses are achieved in a minority of patients. Identifying pre-treatment biomarkers that may predict response to these therapies or who exhibit intrinsic resistance, is of paramount importance. To explore the prevalence of PD-L1copy number alteration in urothelial carcinoma and correlate with response to immune checkpoint inhibitors. We analyzed a cohort of 1050 carcinomas of the bladder and upper urinary tract that underwent targeted next generation sequencing, prospectively. We assessed PD-L1 protein expression, copy number status (next generation sequencing/FISH), and detailed treatment response. We identified 9 tumors with PD-L1amplification and 9 tumors with PD-L1deletion. PD-L1 protein expression was the highest in PD-L1amplified tumors. Of the 9 patients whose tumors harbored PD-L1amplification, 6 received immunotherapy with 4 deriving clinical benefit, and two achieving durable response. Of the 9 patients whose tumors had PD-L1copy number losses, 4 received immunotherapy with 3 experiencing disease progression. PD-L1copy number alterations may serve as potential biomarkers of response to immunotherapy in urothelial carcinoma patients, if validated in larger cohorts.
- Published
- 2021
- Full Text
- View/download PDF
14. Papillary renal cell carcinoma: a single institutional study of 199 cases addressing classification, clinicopathologic and molecular features, and treatment outcome
- Author
-
Murugan, Paari, Jia, Liwei, Dinatale, Renzo G., Assel, Melissa, Benfante, Nicole, Al-Ahmadie, Hikmat A., Fine, Samson W., Gopalan, Anuradha, Sarungbam, Judy, Sirintrapun, S. Joseph, Hakimi, A. Ari, Russo, Paul, Chen, Ying-Bei, Tickoo, Satish K., and Reuter, Victor E.
- 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.
- Published
- 2021
- Full Text
- View/download PDF
15. Immunohistochemistry-based assessment of androgen receptor status and the AR-null phenotype in metastatic castrate resistant prostate cancer
- Author
-
Gupta, Sounak, Vanderbilt, Chad, Abida, Wassim, Fine, Samson W., Tickoo, Satish K., Al-Ahmadie, Hikmat A., Chen, Ying-Bei, Sirintrapun, Sahussapont J., Chadalavada, Kalyani, Nanjangud, Gouri J., Bialik, Ann, Morris, Michael J., Scher, Howard I., Ladanyi, Marc, Reuter, Victor E., and Gopalan, Anuradha
- Abstract
Background: Molecular and immunohistochemistry-based profiling of prostatic adenocarcinoma has revealed frequent Androgen Receptor(AR) gene and protein alterations in metastatic disease. This includes an AR-null non-neuroendocrine phenotype of metastatic castrate resistant prostate cancer which may be less sensitive to androgen receptor signaling inhibitors. This AR-null non-neuroendocrine phenotype is thought to be associated with TP53and RB1alterations. Herein, we have correlated molecular profiling of metastatic castrate resistant prostate cancer with AR/P53/RB immunohistochemistry and relevant clinical correlates. Design: Twenty-seven cases of metastatic castrate resistant prostate cancer were evaluated using histopathologic examination to rule out neuroendocrine differentiation. A combination of a hybridization exon-capture next-generation sequencing-based assay (n= 26), fluorescence in situ hybridization for ARcopy number status (n= 16), and immunohistochemistry for AR (n= 27), P53 (n= 24) and RB (n= 25) was used to profile these cases. Results: Of 27 metastatic castrate resistant prostate cancer cases, 17 had ARamplification and showed positive nuclear expression of AR by immunohistochemistry. Nine cases lacked ARcopy number alterations using next-generation sequencing/fluorescence in situ hybridization. A subset of these metastatic castrate resistant prostate cancer cases demonstrated the AR-null phenotype by immunohistochemistry (five cases and one additional case where next-generation sequencing failed). Common co-alterations in these cases involved the TP53, RB1, and PTENgenes and all these patients received prior therapy with androgen receptor signaling inhibitors (abiraterone and/or enzalutamide). Conclusions: Our study suggests that AR immunohistochemistry may distinguish AR-null from AR-expressing cases in the metastatic setting. AR-null status informs clinical decision-making regarding continuation of therapy with androgen receptor signaling inhibitors and consideration of other treatment options. This might be a relevant and cost-effective diagnostic strategy when there is limited access and/or limited tumor material for molecular testing.
- Published
- 2020
- Full Text
- View/download PDF
16. PD-1 Blockade in Advanced Adrenocortical Carcinoma.
- Author
-
Raj, Nitya, Zheng, Youyun, Kelly, Virginia, Katz, Seth S., Chou, Joanne, Do, Richard K.G., Capanu, Marinela, Zamarin, Dmitriy, Saltz, Leonard B., Ariyan, Charlotte E., Untch, Brian R., O'Reilly, Eileen M., Gopalan, Anuradha, Berger, Michael F., Olino, Kelly, Segal, Neil H., and Reidy-Lagunes, Diane L.
- Published
- 2020
- Full Text
- View/download PDF
17. TFEBExpression Profiling in Renal Cell Carcinomas
- Author
-
Gupta, Sounak, Argani, Pedram, Jungbluth, Achim A., Chen, Ying-Bei, Tickoo, Satish K., Fine, Samson W., Gopalan, Anuradha, Al-Ahmadie, Hikmat A., Sirintrapun, Sahussapont J., Sanchez, Alejandro, Hakimi, Abraham Ari, Mcfarlane, Tiffany, Salazar, Paulo A., Williamson, Sean R., Skala, Stephanie L., Mehra, Rohit, Hes, Ondrej, Antonescu, Cristina R., Ladanyi, Marc, Arcila, Maria E., and Reuter, Victor E.
- Abstract
TFEBis overexpressed in TFEB-rearranged renal cell carcinomas as well as in renal tumors with amplifications of TFEBat 6p21.1. As recent literature suggests that renal tumors with 6p21.1 amplification behave more aggressively than those with rearrangements of TFEB, we compared relative TFEBgene expression in these tumors. This study included 37 TFEB-altered tumors: 15 6p21.1-amplified and 22 TFEB-rearranged (including 5 cases from The Cancer Genome Atlas data set). TFEBstatus was verified using a combination of fluorescent in situ hybridization (n=27) or comprehensive molecular profiling (n=13) and digital droplet polymerase chain reaction was used to quantify TFEBmRNA expression in 6p21.1-amplified (n=9) and TFEB-rearranged renal tumors (n=19). These results were correlated with TFEB immunohistochemistry. TFEB-altered tumors had higher TFEBexpression when normalized to B2M(mean: 168.9%, n=28), compared with non–TFEB-altered controls (mean: 7%, n=18, P=0.005). Interestingly, TFEBexpression in tumors with rearrangements (mean: 224.7%, n=19) was higher compared with 6p21.1-amplified tumors (mean: 51.2%, n=9; P=0.06). Of note, classic biphasic morphology was only seen in TFEB-rearranged tumors and when present correlated with 6.8-fold higher TFEBexpression (P=0.00004). Our results suggest that 6p21.1 amplified renal tumors show increased TFEBgene expression but not as much as t(6;11) renal tumors. These findings correlate with the less consistent/diffuse expression of downstream markers of TFEBactivation (cathepsin K, melan A, HMB45) seen in the amplified neoplasms. This suggests that the aggressive biological behavior of 6p21.1 amplified renal tumors might be secondary to other genes at the 6p21.1 locus that are co-amplified, such as VEGFAand CCND3, or other genetic alterations.
- Published
- 2019
- Full Text
- View/download PDF
18. JAK2/PD-L1/PD-L2(9p24.1) amplifications in renal cell carcinomas with sarcomatoid transformation: implications for clinical management
- Author
-
Gupta, Sounak, Cheville, John, Jungbluth, Achim, Zhang, Yanming, Zhang, Lei, Chen, Ying-Bei, Tickoo, Satish, Fine, Samson, Gopalan, Anuradha, Al-Ahmadie, Hikmat, Sirintrapun, Sahussapont, Blum, Kyle, Lohse, Christine, Hakimi, A., Thompson, R., Leibovich, Bradley, Berger, Michael, Arcila, Maria, Ross, Dara, Ladanyi, Marc, Antonescu, Cristina, and Reuter, Victor
- Abstract
Amplifications of JAK2, PD-L1, and PD-L2at 9p24.1 lead to constitutive expression of PD-L1. This, coupled with JAK2-activation dependent upregulation of PD-L1and adaptive/induced expression leads to higher tumor PD-L1 expression and immune evasion. Renal tumors were therefore evaluated for 9p24.1 amplifications. A combination of next generation sequencing-based copy number analysis, fluorescence in situ hybridization for JAK2/INSL6and PD-L1/PD-L2and immunohistochemistry for phospho-STAT3 (downstream target of JAK2), PD-L1, PD-L2, and PD-1 was performed. In this study we interrogated a “Discovery” cohort of 593 renal tumors, a “Validation” cohort of 398 high-grade renal tumors, The Cancer Genome Atlas (879 cases) and other public datasets (846 cases). 9p24.1 amplifications were significantly enriched in renal tumors with sarcomatoid transformation (5.95%, 15/252) when compared to all histologic subtypes in the combined “Discovery”, “Validation” and public datasets (16/2636, 0.6%, p< 0.00001). Specifically, 9p24.1 amplifications amongst sarcomatoid tumors in public datasets, the “Discovery” and “Validation” cohorts were 7.7% (6/92), 15.1% (5/33), and 3.1% (4/127), respectively. Herein, we describe 13 cases and amplification status for these was characterized using next generation sequencing (n= 9) and/or fluorescence in situ hybridization (n= 10). Correlation with PD-L1 immunohistochemistry (n= 10) revealed constitutive expression (mean H-score: 222/300, n= 10). Analysis of outcomes based on PD-L1 expression in tumor cells performed on 282 cases (“Validation” cohort) did not reveal a significant prognostic effect and was likely reflective of advanced disease. A high incidence of constitutive PD-L1 expression in tumor cells in the “Validation” cohort (H-Score ≥250/300) was noted amongst 83 rhabdoid (6%) and 127 sarcomatoid renal tumors (7.1%). This suggests additional mechanisms of constitutive expression other than amplification events. Importantly, two patients with 9p24.1-amplified sarcomatoid renal tumors showed significant response to immunotherapy. In summary, a subset of renal tumors with sarcomatoid transformation exhibits constitutive PD-L1 overexpression and these patients should be evaluated for enhanced response to immunotherapy.
- Published
- 2019
- Full Text
- View/download PDF
19. Distinctive mechanisms underlie the loss of SMARCB1 protein expression in renal medullary carcinoma: morphologic and molecular analysis of 20 cases
- Author
-
Jia, Liwei, Carlo, Maria I., Khan, Hina, Nanjangud, Gouri J., Rana, Satshil, Cimera, Robert, Zhang, Yanming, Hakimi, A.Ari, Verma, Amit K., Al-Ahmadie, Hikmat A., Fine, Samson W., Gopalan, Anuradha, Sirintrapun, S.Joseph, Tickoo, Satish K., Reuter, Victor E., Gartrell, Benjamin A., and Chen, Ying-Bei
- Abstract
Renal medullary carcinoma is a rare but highly aggressive type of renal cancer occurring in patients with sickle cell trait or rarely with other hemoglobinopathies. Loss of SMARCB1 protein expression, a core subunit of the switch/sucrose nonfermentable (SWI/SNF) chromatin remodeling complex, has emerged as a key diagnostic feature of these tumors. However, the molecular mechanism underlying this loss remains unclear. We retrospectively identified 20 patients diagnosed with renal medullary carcinoma at two institutions from 1996 to 2017. All patients were confirmed to have sickle cell trait, and all tumors exhibited a loss of SMARCB1 protein expression by immunohistochemistry. The status of SMARCB1locus was examined by fluorescence in situ hybridization (FISH) using 3-color probes, and somatic alterations were detected by targeted next-generation sequencing platforms. FISH analysis of all 20 cases revealed 11 (55%) with concurrent hemizygous loss and translocation of SMARCB1, 6 (30%) with homozygous loss of SMARCB1, and 3 (15%) without structural or copy number alterations of SMARCB1despite protein loss. Targeted sequencing revealed a pathogenic somatic mutation of SMARCB1in one of these 3 cases that were negative by FISH. Tumors in the 3 subsets with different FISH findings largely exhibited similar clinicopathologic features, however, homozygous SMARCB1deletion was found to show a significant association with the solid growth pattern, whereas tumors dominated by reticular/cribriform growth were enriched for SMARCB1translocation. Taken together, we demonstrate that different molecular mechanisms underlie the loss of SMARCB1 expression in renal medullary carcinoma. Biallelic inactivation of SMARCB1occurs in a large majority of cases either via concurrent hemizygous loss and translocation disrupting SMARCB1or by homozygous loss.
- Published
- 2019
- Full Text
- View/download PDF
20. JAK2/PD-L1/PD-L2(9p24.1) amplifications in renal cell carcinomas with sarcomatoid transformation: implications for clinical management
- Author
-
Gupta, Sounak, Cheville, John C., Jungbluth, Achim A., Zhang, Yanming, Zhang, Lei, Chen, Ying-Bei, Tickoo, Satish K., Fine, Samson W., Gopalan, Anuradha, Al-Ahmadie, Hikmat A., Sirintrapun, Sahussapont J., Blum, Kyle A., Lohse, Christine M., Hakimi, A.Ari, Thompson, R.Houston, Leibovich, Bradley C., Berger, Michael F., Arcila, Maria E., Ross, Dara S., Ladanyi, Marc, Antonescu, Cristina R., and Reuter, Victor E.
- Abstract
Amplifications of JAK2, PD-L1, and PD-L2at 9p24.1 lead to constitutive expression of PD-L1. This, coupled with JAK2-activation dependent upregulation of PD-L1and adaptive/induced expression leads to higher tumor PD-L1 expression and immune evasion. Renal tumors were therefore evaluated for 9p24.1 amplifications. A combination of next generation sequencing-based copy number analysis, fluorescence in situ hybridization for JAK2/INSL6and PD-L1/PD-L2and immunohistochemistry for phospho-STAT3 (downstream target of JAK2), PD-L1, PD-L2, and PD-1 was performed. In this study we interrogated a “Discovery” cohort of 593 renal tumors, a “Validation” cohort of 398 high-grade renal tumors, The Cancer Genome Atlas (879 cases) and other public datasets (846 cases). 9p24.1 amplifications were significantly enriched in renal tumors with sarcomatoid transformation (5.95%, 15/252) when compared to all histologic subtypes in the combined “Discovery”, “Validation” and public datasets (16/2636, 0.6%, p< 0.00001). Specifically, 9p24.1 amplifications amongst sarcomatoid tumors in public datasets, the “Discovery” and “Validation” cohorts were 7.7% (6/92), 15.1% (5/33), and 3.1% (4/127), respectively. Herein, we describe 13 cases and amplification status for these was characterized using next generation sequencing (n= 9) and/or fluorescence in situ hybridization (n= 10). Correlation with PD-L1 immunohistochemistry (n= 10) revealed constitutive expression (mean H-score: 222/300, n= 10). Analysis of outcomes based on PD-L1 expression in tumor cells performed on 282 cases (“Validation” cohort) did not reveal a significant prognostic effect and was likely reflective of advanced disease. A high incidence of constitutive PD-L1 expression in tumor cells in the “Validation” cohort (H-Score ≥250/300) was noted amongst 83 rhabdoid (6%) and 127 sarcomatoid renal tumors (7.1%). This suggests additional mechanisms of constitutive expression other than amplification events. Importantly, two patients with 9p24.1-amplified sarcomatoid renal tumors showed significant response to immunotherapy. In summary, a subset of renal tumors with sarcomatoid transformation exhibits constitutive PD-L1 overexpression and these patients should be evaluated for enhanced response to immunotherapy.
- Published
- 2019
- Full Text
- View/download PDF
21. Distinctive mechanisms underlie the loss of SMARCB1 protein expression in renal medullary carcinoma: morphologic and molecular analysis of 20 cases
- Author
-
Jia, Liwei, Carlo, Maria, Khan, Hina, Nanjangud, Gouri, Rana, Satshil, Cimera, Robert, Zhang, Yanming, Hakimi, A., Verma, Amit, Al-Ahmadie, Hikmat, Fine, Samson, Gopalan, Anuradha, Sirintrapun, S., Tickoo, Satish, Reuter, Victor, Gartrell, Benjamin, and Chen, Ying-Bei
- Abstract
Renal medullary carcinoma is a rare but highly aggressive type of renal cancer occurring in patients with sickle cell trait or rarely with other hemoglobinopathies. Loss of SMARCB1 protein expression, a core subunit of the switch/sucrose nonfermentable (SWI/SNF) chromatin remodeling complex, has emerged as a key diagnostic feature of these tumors. However, the molecular mechanism underlying this loss remains unclear. We retrospectively identified 20 patients diagnosed with renal medullary carcinoma at two institutions from 1996 to 2017. All patients were confirmed to have sickle cell trait, and all tumors exhibited a loss of SMARCB1 protein expression by immunohistochemistry. The status of SMARCB1locus was examined by fluorescence in situ hybridization (FISH) using 3-color probes, and somatic alterations were detected by targeted next-generation sequencing platforms. FISH analysis of all 20 cases revealed 11 (55%) with concurrent hemizygous loss and translocation of SMARCB1, 6 (30%) with homozygous loss of SMARCB1, and 3 (15%) without structural or copy number alterations of SMARCB1despite protein loss. Targeted sequencing revealed a pathogenic somatic mutation of SMARCB1in one of these 3 cases that were negative by FISH. Tumors in the 3 subsets with different FISH findings largely exhibited similar clinicopathologic features, however, homozygous SMARCB1deletion was found to show a significant association with the solid growth pattern, whereas tumors dominated by reticular/cribriform growth were enriched for SMARCB1translocation. Taken together, we demonstrate that different molecular mechanisms underlie the loss of SMARCB1 expression in renal medullary carcinoma. Biallelic inactivation of SMARCB1occurs in a large majority of cases either via concurrent hemizygous loss and translocation disrupting SMARCB1or by homozygous loss.
- Published
- 2019
- Full Text
- View/download PDF
22. In Organ-confined Prostate Cancer, Tumor Quantitation Not Found to Aid in Prediction of Biochemical Recurrence
- Author
-
Ito, Yujiro, Vertosick, Emily A., Sjoberg, Daniel D., Vickers, Andrew J., Al-Ahmadie, Hikmat A., Chen, Ying-Bei, Gopalan, Anuradha, Sirintrapun, Sahussapont J., Tickoo, Satish K., Eastham, James A., Scardino, Peter T., Reuter, Victor E., and Fine, Samson W.
- Abstract
Supplemental Digital Content is available in the text.In the eighth edition AJCC staging, all organ-confined disease is assigned pathologic stage T2, without subclassification. We investigated whether total tumor volume (TTV) and/or maximum tumor diameter (MTD) of the index lesion are useful in improving prediction of biochemical recurrence (BCR) in pT2 patients. We identified 1657 patients with digital tumor maps and quantification of TTV/MTD who had pT2 disease on radical prostatectomy (RP). Multivariable Cox regression models were used to assess whether TTV and/or MTD are independent predictors of BCR when adjusting for a base model incorporating age, preoperative prostate-specific antigen, RP grade group, and surgical margin status. If either tumor quantification added significantly, we calculated and reported the c-index. Ninety-five patients experienced BCR after RP; median follow-up for patients without BCR was 5.7 years. The c-index was 0.737 for the base model. Although there was some evidence of an association between TTV and BCR (P=0.088), this did not meet conventional levels of statistical significance and only provided a limited increase in discrimination (0.743; c-index improvement: 0.006). MTD was not associated with BCR (P>0.9). In analyses excluding patients with grade group 1 on biopsy who would be less likely to undergo RP in contemporary practice (622 patients; 59 with BCR), TTV/MTD was not a statistically significant predictor (P=0.4 and 0.8, respectively). Without evidence that tumor quantitation, in the form of either TTV or MTD of the index lesion, is useful for the prediction of BCR in pT2 prostate cancer, we cannot recommend its routine reporting.
- Published
- 2019
- Full Text
- View/download PDF
23. PD-L1 Expression in Urothelial Carcinoma With Predominant or Pure Variant Histology
- Author
-
Reis, Henning, Serrette, Rene, Posada, Jennifer, Lu, Vincent, Chen, Ying-bei, Gopalan, Anuradha, Fine, Samson W., Tickoo, Satish K., Sirintrapun, Sahussapont J., Iyer, Gopa, Funt, Samuel A., Teo, Min Yuen, Rosenberg, Jonathan E., Bajorin, Dean F., Dalbagni, Guido, Bochner, Bernard H., Solit, David B., Reuter, Victor E., and Al-Ahmadie, Hikmat A.
- Abstract
The introduction of immune checkpoint blockade (ICB) therapy has transformed the management of advanced bladder cancer (BC). Despite its limitations, PD-L1 immunohistochemistry may serve as a predictive biomarker of anti-PD-L1/PD1 therapy. While urothelial carcinoma (UC) patients with predominant or pure variant histology (UCV) account for up to one-third of advanced cases, to date, most ICB BC studies have excluded patients with such histologies. To assess the potential utility of ICB in patients with UCV, we analyzed PD-L1 expression in UCV and compared 3 commonly used and commercially available PD-L1 antibodies. Full sections from 84 UCV cases were stained with clones SP263, 22C3, and SP142, all of which are considered predictive assays to identify UC patients who are more likely to respond to anti-PD-1/PD-L1 inhibitors durvalumab, pembrolizumab, and atezolizumab, respectively. Expression on tumor cells (TC) and tumor-infiltrating immune cells (IC) was assessed. Staining extent and characteristics were evaluated, and concordance among the 3 clones was determined at various cutoff points as used in previous studies in BC. We found that PD-L1 was expressed in a significant percentage of UCV cases at different cutoff points (cutoff 1% TC: 37% to 54%, cutoff 5% TC: 23% to 37%), with the highest expression in UC with squamous differentiation. These figures are equal to or higher than those for classic/pure UC (4% to 30%). The results suggest that patients with UCV may benefit from anti-PD-1/PD-L1 therapy and argue against the exclusion of UC with predominant or pure variant histology from clinical ICB studies. The highest expression in both TC and IC was observed with clone SP263, followed by 22C3 and SP142, and all clones showed strong agreement in a pairwise comparison, both in TC and IC (R-values: 0.780 to 0.901), which indicates that all 3 clones are potentially useful in the evaluation of PD-L1 expression in UCV.
- Published
- 2019
- Full Text
- View/download PDF
24. Tubulocystic renal cell carcinoma: a distinct clinicopathologic entity with a characteristic genomic profile
- Author
-
Sarungbam, Judy, Mehra, Rohit, Tomlins, Scott A., Smith, Steven C., Jayakumaran, Gowtham, Al-Ahmadie, Hikmat, Gopalan, Anuradha, Sirintrapun, Sahussapont J., Fine, Samson W., Zhang, Yanming, Amin, Mahul B., Reuter, Victor E., Chen, Ying-Bei, and Tickoo, Satish K.
- 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, KMT2Cand 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.
- Published
- 2019
- Full Text
- View/download PDF
25. Tubulocystic renal cell carcinoma: a distinct clinicopathologic entity with a characteristic genomic profile
- Author
-
Sarungbam, Judy, Mehra, Rohit, Tomlins, Scott A., Smith, Steven C., Jayakumaran, Gowtham, Al-Ahmadie, Hikmat, Gopalan, Anuradha, Sirintrapun, Sahussapont J., Fine, Samson W., Zhang, Yanming, Amin, Mahul B., Reuter, Victor E., Chen, Ying-Bei, and Tickoo, Satish K.
- 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, KMT2Cand 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.
- Published
- 2019
- Full Text
- View/download PDF
26. Analysis of the Prevalence of Microsatellite Instability in Prostate Cancer and Response to Immune Checkpoint Blockade
- Author
-
Abida, Wassim, Cheng, Michael L., Armenia, Joshua, Middha, Sumit, Autio, Karen A., Vargas, Hebert Alberto, Rathkopf, Dana, Morris, Michael J., Danila, Daniel C., Slovin, Susan F., Carbone, Emily, Barnett, Ethan S., Hullings, Melanie, Hechtman, Jaclyn F., Zehir, Ahmet, Shia, Jinru, Jonsson, Philip, Stadler, Zsofia K., Srinivasan, Preethi, Laudone, Vincent P., Reuter, Victor, Wolchok, Jedd D., Socci, Nicholas D., Taylor, Barry S., Berger, Michael F., Kantoff, Philip W., Sawyers, Charles L., Schultz, Nikolaus, Solit, David B., Gopalan, Anuradha, and Scher, Howard I.
- Abstract
IMPORTANCE: The anti–programmed cell death protein 1 (PD-1) antibody pembrolizumab is approved by the US Food and Drug Administration for the treatment of microsatellite instability–high (MSI-H) or mismatch repair–deficient (dMMR) solid tumors, but the prevalence of MSI-H/dMMR prostate cancer and the clinical utility of immune checkpoint blockade in this disease subset are unknown. OBJECTIVE: To define the prevalence of MSI-H/dMMR prostate cancer and the clinical benefit of anti–PD-1/programmed cell death 1 ligand 1 (PD-L1) therapy in this molecularly defined population. DESIGN, SETTING, AND PARTICIPANTS: In this case series, 1551 tumors from 1346 patients with prostate cancer undergoing treatment at Memorial Sloan Kettering Cancer Center were prospectively analyzed using a targeted sequencing assay from January 1, 2015, through January 31, 2018. Patients had a diagnosis of prostate cancer and consented to tumor molecular profiling when a tumor biopsy was planned or archival tissue was available. For each patient, clinical outcomes were reported, with follow-up until May 31, 2018. MAIN OUTCOMES AND MEASURES: Tumor mutation burden and MSIsensor score, a quantitative measure of MSI, were calculated. Mutational signature analysis and immunohistochemistry for MMR protein expression were performed in select cases. RESULTS: Among the 1033 patients who had adequate tumor quality for MSIsensor analysis (mean [SD] age, 65.6 [9.3] years), 32 (3.1%) had MSI-H/dMMR prostate cancer. Twenty-three of 1033 patients (2.2%) had tumors with high MSIsensor scores, and an additional 9 had indeterminate scores with evidence of dMMR. Seven of the 32 MSI-H/dMMR patients (21.9%) had a pathogenic germline mutation in a Lynch syndrome–associated gene. Six patients had more than 1 tumor analyzed, 2 of whom displayed an acquired MSI-H phenotype later in their disease course. Eleven patients with MSI-H/dMMR castration-resistant prostate cancer received anti–PD-1/PD-L1 therapy. Six of these (54.5%) had a greater than 50% decline in prostate-specific antigen levels, 4 of whom had radiographic responses. As of May 2018, 5 of the 6 responders (5 of 11 total [45.5%]) were still on therapy for as long as 89 weeks. CONCLUSIONS AND RELEVANCE: The MSI-H/dMMR molecular phenotype is uncommon yet therapeutically meaningful in prostate cancer and can be somatically acquired during disease evolution. Given the potential for durable responses to anti–PD-1/PD-L1 therapy, these findings support the use of prospective tumor sequencing to screen all patients with advanced prostate cancer for MSI-H/dMMR. Because not all patients with the MSI-H/dMMR phenotype respond, further studies should explore mechanisms of resistance.
- Published
- 2019
- Full Text
- View/download PDF
27. Morphologic and Immunohistochemical Assessment of CDH1Loss of Function Alterations in Prostatic Adenocarcinoma
- Author
-
Gupta, Sounak, Fine, Samson W., Chang, Jason, Tickoo, Satish K., Chen, Ying-Bei, Al-Ahmadie, Hikmat A., Sirintrapun, Sahussapont J., Abida, Wassim, Ladanyi, Marc, Reuter, Victor E., and Gopalan, Anuradha
- Published
- 2020
- Full Text
- View/download PDF
28. Somatic Mutations of TSC2or MTORCharacterize a Morphologically Distinct Subset of Sporadic Renal Cell Carcinoma With Eosinophilic and Vacuolated Cytoplasm
- Author
-
Chen, Ying-Bei, Mirsadraei, Leili, Jayakumaran, Gowtham, Al-Ahmadie, Hikmat A., Fine, Samson W., Gopalan, Anuradha, Sirintrapun, S. Joseph, Tickoo, Satish K., and Reuter, Victor E.
- Abstract
Supplemental Digital Content is available in the text.The differential diagnosis of renal cell neoplasms with solid or nested architecture and eosinophilic cytoplasm has become increasingly complex. Despite recent advances in classifying a number of entities exhibiting this morphology, some tumors remain in the unclassified category. Here we describe a morphologically distinct group of sporadic renal cell carcinoma (RCC) with predominantly nested architecture, eosinophilic, and remarkably vacuolated cytoplasm retrospectively identified from a cohort of previously unclassified tumors. We examined the clinicopathologic and immunohistochemical features of these tumors and investigated their mutational and copy number alterations using a targeted next-generation sequencing platform. The study included 7 patients with a mean age of 54 years (range: 40 to 68 y) and a male to female ratio of 3:4. All patients presented with a solitary renal mass and had no prior medical or family history raising concern for syndromic conditions. Tumors were well-circumscribed, unencapsulated, and comprised of nests of eosinophilic cells in a hypocellular and often edematous stroma. Tumor cells had round nuclei with prominent nucleoli and granular cytoplasm with striking vacuolization. Thick-walled vessels and calcifications were also frequently present, whereas increased mitotic activity, necrosis, foamy histiocytes or lymphocytic infiltrates were not identified. All cases were positive for PAX8, had retained expression of SDHB and FH, and exhibited a CK7−/CK20− phenotype. While cathepsin-K was positive in 5 cases, none exhibited immunoreactivity to HMB45 or Melan A, or TFE3 immunostaining. Next-generation sequencing identified somatic inactivating mutations of TSC2(3/5 tumors tested) or activating mutations of MTOR(2/5) as the primary molecular alterations, consistent with hyperactive mTOR complex 1 signaling which was further demonstrated by phospho-S6 and phospho-4E-BP1 immunostaining. Copy number analysis revealed a loss of chromosome 1 in both cases with MTORmutation. These tumors represent a novel subset of sporadic RCC characterized by alterations in TSC1-TSC2 complex or the mTOR complex 1 pathway. Recognition of their characteristic morphologic and immunophenotypic features will allow them to be readily identified and separated from the unclassified RCC category.
- Published
- 2019
- Full Text
- View/download PDF
29. Comedonecrosis Revisited
- Author
-
Fine, Samson W., Al-Ahmadie, Hikmat A., Chen, Ying-Bei, Gopalan, Anuradha, Tickoo, Satish K., and Reuter, Victor E.
- Abstract
From the advent of the Gleason grading system for prostate cancer, cancer displaying intraluminal necrotic cells and/or karyorrhexis within cribriform/solid architecture, a phenomenon termed “comedonecrosis,” has been assigned pattern 5. Intraductal carcinoma (IDC-P) shows morphologic overlap with high-grade cribriform/solid adenocarcinoma architecturally and cytologically and may also show central necrosis, yet due to the presence of basal cells at the duct periphery is not currently assigned a grade in clinical practice. On the basis of observations from routine clinical cases, we hypothesized that comedonecrosis was more significantly associated with IDC-P than invasive disease. From a large series of mapped radical prostatectomy specimens (n=933), we identified 125 high-grade (≥Gleason score 4+3=7), high-volume tumors with available slides for review. All slides were examined for the presence of unequivocal comedonecrosis. Standard immunohistochemistry for basal cell markers was performed to detect basal cell labeling in these foci. In total, 19 of 125 (15%) cases showed some ducts with comedonecrosis—9 cases with 1 focus and 10 cases with ≥2 foci; in all, a total of 73 foci of true comedonecrosis were evaluated. Immunohistochemical stains revealed labeling for basal cell markers in a basal cell distribution for at least some comedonecrosis foci in 18 of 19 (95%) cases, 12 with IDC-P exclusively and 6 with a mix of IDC-P and invasive carcinoma comedonecrosis foci. These results suggest that comedonecrosis is strongly associated with IDC-P and hence, the routine assignment of pattern 5 to carcinoma exhibiting comedonecrosis should be reconsidered.
- Published
- 2018
- Full Text
- View/download PDF
30. Distinct Genomic Copy Number Alterations Distinguish Mucinous Tubular and Spindle Cell Carcinoma of the Kidney From Papillary Renal Cell Carcinoma With Overlapping Histologic Features
- Author
-
Ren, Qinghu, Wang, Lu, Al-Ahmadie, Hikmat A., Fine, Samson W., Gopalan, Anuradha, Sirintrapun, Sahussapont J., Tickoo, Satish K., Reuter, Victor E., and Chen, Ying-Bei
- Abstract
Mucinous tubular and spindle cell carcinoma (MTSCC) of the kidney is a rare type of renal cell carcinoma that frequently exhibits histologic and immunophenotypic features overlapping with type 1 papillary renal cell carcinoma (PRCC). To clarify molecular attributes that can be used for this difficult differential diagnosis, we sought to delineate the genome-wide copy number alterations in tumors displaying classic histologic features of MTSCC in comparison to the solid variant of type 1 PRCC and indeterminate cases with overlapping histologic features. The study included 11 histologically typical MTSCC, 9 tumors with overlapping features between MTSCC and PRCC, and 6 cases of solid variant of type 1 PRCC. DNA samples extracted from macrodissected or microdissected tumor areas were analyzed for genome-wide copy number alterations using an SNP-array platform suitable for clinical archival material. All cases in the MTSCC group exhibited multiple chromosomal losses, most frequently involving chromosomes 1, 4, 6, 8, 9, 13, 14, 15, and 22, while lacking trisomy 7 or 17. In contrast, cases with overlapping morphologic features of MTSCC and PRCC predominantly showed multiple chromosomal gains, most frequently involving chromosomes 7, 16, 17, and 20, similar to the chromosomal alteration pattern that was seen in the solid variant of type 1 PRCC cases. Morphologic comparison of these molecularly characterized tumors identified histologic features that help to distinguish MTSCC from PRCC, but immunohistochemical profiles of these tumors remained overlapping, including a marker for Hippo-Yes-associated protein signaling. Characteristic patterns of genome-wide copy number alterations strongly support MTSCC and PRCC as distinct entities despite their immunohistochemical and certain morphologic overlap, and help define histologic features useful for the classification of questionable cases.
- Published
- 2018
- Full Text
- View/download PDF
31. Effect of Janus kinase (JAK) signaling inhibition on lineage plasticity and drug sensitivity in castrate resistant prostate cancer.
- Author
-
Zaidi, Samir, Chan, Joseph Minhow, Love, Jillian, Zhao, Jimmy, Setty, Manu, Lawrence, Kayla, Gopalan, Anuradha, Goodrich, David, Morris, Michael J., Chen, Yu, Karthaus, Wouter, Pe'er, Dana, and Sawyers, Charles L.
- Published
- 2023
- Full Text
- View/download PDF
32. Prostate-specific membrane antigen cleavage of vitamin B9 stimulates oncogenic signaling through metabotropic glutamate receptors
- Author
-
Kaittanis, Charalambos, Andreou, Chrysafis, Hieronymus, Haley, Mao, Ninghui, Foss, Catherine A., Eiber, Matthias, Weirich, Gregor, Panchal, Palak, Gopalan, Anuradha, Zurita, Juan, Achilefu, Samuel, Chiosis, Gabriela, Ponomarev, Vladimir, Schwaiger, Markus, Carver, Brett S., Pomper, Martin G., and Grimm, Jan
- Abstract
Prostate-specific membrane antigen (PSMA) or folate hydrolase 1 (FOLH1) is highly expressed on prostate cancer. Its expression correlates inversely with survival and increases with tumor grade. However, the biological role of PSMA has not been explored, and its role in prostate cancer remained elusive. Filling this gap, we demonstrate that in prostate cancer, PSMA initiates signaling upstream of PI3K through G protein–coupled receptors, specifically via the metabotropic glutamate receptor (mGluR). PSMA’s carboxypeptidase activity releases glutamate from vitamin B9 and other glutamated substrates, which activate mGluR I. Activated mGluR I subsequently induces activation of phosphoinositide 3-kinase (PI3K) through phosphorylation of p110β independent of PTEN loss. The p110β isoform of PI3K plays a particularly important role in the pathogenesis of prostate cancer, but the origin of its activation was so far unknown. PSMA expression correlated with PI3K–Akt signaling in cells, animal models, and patients. We interrogated the activity of the PSMA–PI3K axis through positron emission tomography and magnetic resonance imaging. Inhibition of PSMA in preclinical models inhibited PI3K signaling and promoted tumor regression. Our data present a novel oncogenic signaling role of PSMA that can be exploited for therapy and interrogated with imaging.
- Published
- 2018
- Full Text
- View/download PDF
33. Stromal hedgehog signaling maintains smooth muscle and hampers micro-invasive prostate cancer
- Author
-
Yang, Zhaohui, Peng, Yu-Ching, Gopalan, Anuradha, Gao, Dong, Chen, Yu, and Joyner, Alexandra L.
- Abstract
It is widely appreciated that reactive stroma or carcinoma-associated fibroblasts can influence epithelial tumor progression. In prostate cancer (PCa), the second most common male malignancy worldwide, the amount of reactive stroma is variable and has predictive value for tumor recurrence. By analyzing human PCa protein and RNA expression databases, we found smooth muscle cells (SMCs) are decreased in advanced tumors, whereas fibroblasts are maintained. In three mouse models of PCa, PB-MYC, ERG/PTEN and TRAMP, we found the composition of the stroma is distinct. SMCs are greatly depleted in advanced PB-MYC tumors and locally reduced in ERG/PTEN prostates, whereas in TRAMP tumors the SMC layers are increased. In addition, interductal fibroblast-like cells expand in PB-MYC and ERG/PTEN tumors, whereas in TRAMP PCa they expand little and stromal cells invade into intraductal adenomas. Fate mapping of SMCs showed that in PB-MYC tumors the cells are depleted, whereas they expand in TRAMP tumors and interestingly contribute to the stromal cells in intraductal adenomas. Hedgehog (HH) ligands secreted by epithelial cells are known to regulate prostate mesenchyme expansion differentially during development and regeneration. Any possible role of HH signaling in stromal cells during PCa progression is poorly understood. We found that HH signaling is high in SMCs and fibroblasts near tumor cells in all models, and epithelial Shh expression is decreased whereas Ihh and Dhh are increased. In human primary PCa, expression of IHH is the highest of the three HH genes, and elevated HH signaling correlates with high stromal gene expression. Moreover, increasing HH signaling in the stroma of PB-MYC PCa resulted in more intact SMC layers and decreased tumor progression (micro-invasive carcinoma). Thus, we propose HH signaling restrains tumor progression by maintaining the smooth muscle and preventing invasion by tumor cells. Our studies highlight the importance of understanding how HH signaling and stromal composition impact on PCa to optimize drug treatments.
- Published
- 2017
- Full Text
- View/download PDF
34. Prognostic Impact of Subclassification of Radical Prostatectomy Positive Margins by Linear Extent and Gleason Grade.
- Author
-
Udo, Kazuma, Cronin, Angel M., Carlino, Lauren J., Savage, Caroline J., Maschino, Alexandra C., Al-Ahmadie, Hikmat A., Gopalan, Anuradha, Tickoo, Satish K., Scardino, Peter T., Eastham, James A., Reuter, Victor E., and Fine, Samson W.
- Subjects
PROSTATECTOMY ,GLEASON grading system ,SURGICAL site ,CANCER invasiveness ,PROSTATE-specific antigen ,KAPLAN-Meier estimator ,PROBABILITY theory - Abstract
Purpose: We evaluated the relationship of progression to positive surgical margin linear length and Gleason grade at a positive surgical margin. Materials and Methods: We studied 2,150 prostatectomies done for pT2 or pT3a disease to determine grade, stage and surgical margin status. In patients with positive surgical margins we recorded the location, number, positive margin linear length and highest Gleason grade at a positive margin. The Kaplan-Meier method and log rank test were used to determine differences in progression-free probability among positive margin features. The concordance index was used to discriminate the accuracy of grouping surgical margin status as negative/positive vs positive margin linear length/highest Gleason grade. Results: A total of 207 cases (10%) showed positive surgical margins, including 93 (45%) that were pT2+ and 114 (55%) that were pT3a. Patients with pT3a and positive margins had greater prostate specific antigen and tumor volume, and Gleason score 7 or greater than those with pT2+. A total of 45 patients with positive margins progressed. We then subcategorized positive margins. Of the patients 164 (79%) had 1 positive margin. Positive margin linear length was 1 mm or less, 1.1 to 3 and greater than 3 in 104 (50%), 55 (27%) and 48 cases (23%), respectively. Two-year progression-free probability was 95%, 91%, 83% and 47% in patients with negative margins and the 3 positive margin linear length groups, respectively (p <0.001). Gleason grade at a positive margin was 3 and 4/5 in 154 (74%) and 53 patients (26%), respectively. The latter group was significantly more likely to progress (p <0.001). The overall margin status concordance index was 0.636. It was not considerably enhanced by categorizing by positive surgical margin linear length/highest Gleason grade at positive margins. Conclusions: The linear extent of and highest Gleason grade at a positive surgical margin are associated with progression. However, subcategorization does not importantly add to predictive models using margin status only. More robust markers are needed in patients with positive surgical margins to warrant routine reporting and identify those at risk for biochemical recurrence. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
35. Interobserver and intraobserver reproducibility in digital and routine microscopic assessment of prostate needle biopsies.
- Author
-
Rodriguez-Urrego, Paula A., Cronin, Angel M., Al-Ahmadie, Hikmat A., Gopalan, Anuradha, Tickoo, Satish K., Reuter, Victor E., and Fine, Samson W.
- Subjects
NEEDLE biopsy ,MEDICAL imaging systems ,PROSTATE disease diagnosis ,MICROSCOPY ,MEDICAL consultation ,PATHOLOGISTS - Abstract
Summary: Advances in whole slide digital imaging in the past decade necessitate validation of these tools in each organ system in advance of clinical adoption. We assessed reproducibility in reporting prostate needle biopsy parameters among urologic pathologists using routine and digital microscopy in a consultation/second opinion-like setting. Four urologic pathologists evaluated a single core level from 50 diagnostically challenging needle biopsy specimens by routine microscopy and whole slide digital imaging. Interobserver and intraobserver agreement were calculated for primary and secondary Gleason grades, Gleason score, tumor quantitation (percentage and size in millimeters), and perineural invasion. Interobserver agreement for routine microscopy was excellent for primary Gleason grade (κ = 0.72) and good for all other parameters (κ ranging from 0.36 to 0.55). Whole slide digital imaging assessment yielded similar agreement for all parameters. Intraobserver agreement for primary Gleason grade and Gleason score was very good to excellent for all pathologists (all κ ≥ 0.65 and ≥0.73, respectively). Size of tumor in millimeters consistently displayed higher levels of agreement than percentage of tumor across media and pathologists. Digital assessment of routinely reported cancer parameters on prostatic needle biopsy for a given scanned core level is comparable to that of routine microscopy. These findings imply that histologic interpretation using dynamic whole slide images may accurately simulate routine microscopic evaluation in the consultation setting. Implementation of whole slide digital imaging in these scenarios may significantly reduce the workload of large referral centers in the near future and impact the manner in which pathologists seek second opinion consultation on challenging cases. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
36. Prostatic Transition Zone Directed Needle Biopsies Uncommonly Sample Clinically Relevant Transition Zone Tumors.
- Author
-
Haarer, Chadwick F., Gopalan, Anuradha, Tickoo, Satish K., Scardino, Peter T., Eastham, James A., Reuter, Victor E., and Fine, Samson W.
- Subjects
PROSTATE tumors ,NEEDLE biopsy ,DIAGNOSIS ,PROSTATE cancer ,COMPARATIVE method ,PROSTATECTOMY ,BIOLOGICAL specimens ,PROSTATE-specific antigen ,UROLOGY - Abstract
Purpose: We compared prostate cancer detected in transition zone directed needle biopsies with those in corresponding radical prostatectomy specimens. Materials and Methods: We reviewed needle biopsy and radical prostatectomy slides from 61 patients in whom cancer was present on transition zone directed needle biopsy. We assessed needle biopsy cancer features as well as transition zone lesions and dominant tumor sites on radical prostatectomy. Results: Prostate cancer was detected in 25 of 61 left (41%), 23 of 61 right (38%) and 13 of 61 bilateral (21%) transition zone directed needle biopsies. On radical prostatectomy 24 of 61 cases (39.5%) had no tumor in the transition zone, 24 of 61 (39.5%) had nondominant transition zone cancer and 13 of 61 (21%) had a dominant transition zone lesion. Of cases with cancer in the left and right transition zone directed needle biopsy 18 of 38 (47%) and 17 of 36 (47%), respectively, had no transition zone tumor or showed tumor in the contralateral transition zone only at radical prostatectomy. In 8 cases the transition zone directed core was the only one with cancer on needle biopsy and 2 of 8 (25%) such cases showed dominant transition zone cancer at radical prostatectomy. Conclusions: Cancer identified in transition zone directed needle biopsy cores was not from the transition zone or did not reflect a dominant transition zone lesion in almost 80% of cases. Cancer identified in a left or right transition zone directed needle biopsy did not predict ipsilateral transition zone cancer in almost 50% of cases. These findings suggest that such biopsies do not adequately characterize transition zone tumors. Thus, care should be taken in their interpretation. [Copyright &y& Elsevier]
- Published
- 2009
- Full Text
- View/download PDF
37. The Anterior Layer of Denonvilliers’ Fascia: A Common Misconception in the Laparoscopic Prostatectomy Literature.
- Author
-
Secin, Fernando P., Karanikolas, Nicholas, Gopalan, Anuradha, Bianco, Fernando J., Shayegan, Bobby, Touijer, Karim, Olgac, Semra, Myers, Robert P., Dalbagni, Guido, and Guillonneau, Bertrand
- Subjects
PROSTATECTOMY ,SEMINAL vesicles ,DISSECTION ,LAPAROSCOPIC surgery ,BIOPSY - Abstract
Purpose: Incision of the anterior layer of Denonvilliers’ fascia is commonly cited as a key step in successful dissection of the vasa deferentia and seminal vesicles from the posterior bladder neck during laparoscopic radical prostatectomy. However, anatomical descriptions do not support the presence of Denonvilliers’ fascia anterior to the seminal vesicles. To address this inconsistency we performed a detailed anatomical study of tissue planes encountered during laparoscopic dissection of the posterior bladder neck. Materials and Methods: To grossly characterize the tissue planes encountered during laparoscopic posterior bladder neck dissection, ex vivo dissections were performed on 4 separate cystoprostatectomy specimens. Biopsies of the representative areas were obtained from 20 consecutive laparoscopic radical prostatectomy specimens by 2 dedicated uropathologists. Results: Following incision into the posterior bladder neck mucosa, longitudinally oriented fibers were readily visualized, extending from bladder neck to prostate base. Histologically this anatomical landmark represents the fusion of 2 separate tissue layers, that is an inner lamella composed of longitudinally disposed smooth muscle fibers in continuation with the longitudinal fascia of the bladder detrusor (medial fascicle of the detrusor running in between the ureters) and an outer lamella composed of fibroadipose tissue in continuation with the bladder adventitia. Conclusions: Our anatomical and histological analysis refutes the prevailing belief in the laparoscopic literature that the longitudinal muscle fibers identified during dissection of the posterior bladder neck represent the anterior layer of Denonvilliers’ fascia. They correspond to the posterior longitudinal fascia of the detrusor muscle that is externally upholstered by the bladder adventitia. [Copyright &y& Elsevier]
- Published
- 2007
- Full Text
- View/download PDF
38. Hereditary Leiomyomatosis and Renal Cell Carcinoma Syndrome–associated Renal Cancer
- Author
-
Chen, Ying-Bei, Brannon, A. Rose, Toubaji, Antoun, Dudas, Maria E., Won, Helen H., Al-Ahmadie, Hikmat A., Fine, Samson W., Gopalan, Anuradha, Frizzell, Norma, Voss, Martin H., Russo, Paul, Berger, Michael F., Tickoo, Satish K., and Reuter, Victor E.
- Abstract
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome is an autosomal dominant disorder in which germline mutations of fumarate hydratase(FH) gene confer an increased risk of cutaneous and uterine leiomyomas and renal cancer. HLRCC-associated renal cancer is highly aggressive and frequently presents as a solitary mass. We reviewed the clinicopathologic features of 9 patients with renal tumors presenting as sporadic cases but who were later proven to have FHgermline mutations. Histologically, all tumors showed mixed architectural patterns, with papillary as the dominant pattern in only 3 cases. Besides papillary, tubular, tubulopapillary, solid, and cystic elements, 6 of 9 tumors contained collecting duct carcinoma–like areas with infiltrating tubules, nests, or individual cells surrounded by desmoplastic stroma. Prominent tubulocystic carcinoma–like component and sarcomatoid differentiation were identified. Although all tumors exhibited the proposed hallmark of HLRCC (large eosinophilic nucleolus surrounded by a clear halo), this feature was often not uniformly present throughout the tumor. Prior studies have shown that a high level of fumarate accumulated in HLRCC tumor cells causes aberrant succination of cellular proteins by forming a stable chemical modification, S-(2-succino)-cysteine (2SC), which can be detected by immunohistochemistry. We thus explored the utility of detecting 2SC by immunohistochemistry in the differential diagnosis of HLRCC tumors and other high-grade renal tumors and investigated the correlation between 2SC staining and FHmolecular alterations. All confirmed HLRCC tumors demonstrated diffuse and strong nuclear and cytoplasmic 2SC staining, whereas all clear cell (184184, 100), most high-grade unclassified (9397, 96), and the large majority of “type 2” papillary (3545, 78) renal cell carcinoma cases showed no 2SC immunoreactivity. A subset of papillary (22) and rare unclassified (4) tumors showed patchy or diffuse cytoplasmic staining without nuclear labeling, unlike the pattern seen with confirmed HLRCC tumors. Sequencing revealed no germline or somatic FHalterations in 14 tumors that either exhibited only cytoplasmic 2SC staining (n=5) or were negative for 2SC (n=9), despite their HLRCC-like morphologic features. Our results emphasize the pivotal role of pathologic examination in the diagnosis of HLRCC patients and indicate immunohistochemical detection of 2SC as a useful ancillary tool in the differentiation of HLRCC renal tumors from other high-grade renal cell carcinomas.
- Published
- 2014
- Full Text
- View/download PDF
39. Epithelioid angiomyolipoma of the kidney: pathological features and clinical outcome in a series of consecutively resected tumors
- Author
-
He, Wenlei, Cheville, John C, Sadow, Peter M, Gopalan, Anuradha, Fine, Samson W, Al-Ahmadie, Hikmat A, Chen, Ying-Bei, Oliva, Esther, Russo, Paul, Reuter, Victor E, and Tickoo, Satish K
- Abstract
The 2004 World Health Organization classification of tumors defines epithelioid angiomyolipoma of kidney as a potentially malignant mesenchymal neoplasm with reported metastasis in approximately one-third of the cases. However, this conclusion was based primarily on individual case reports and small retrospective series. More recently reported larger series have shown varying results. We reviewed 437 consecutive renal angiomyolipomas with primary resection at three tertiary-care institutions with high nephrectomy volumes. Only tumors showing >80% epithelioid histology were included in this study. Tumors resected elsewhere and reviewed in consultation were not included. Twenty of these 437 (4.6%) were classified as epithelioid angiomyolipoma. The female to male ratio was 11:9, mean age 49.7 (range, 30–80) years, and mean tumor size 8.7 (range, 1–25) cm. Microscopic tumor necrosis was present in 10 (50%) tumors and mitotic activity (range, <1–5/10 high power fields) in 8 (40%); atypical mitoses were seen in only 1 (5%) tumor. Pleomorphic ganglion-like or multinucleated giant cells were seen in 18 (90%) tumors. With a mean follow-up of 82.5 (range, 1–356) months, seventeen patients were alive with no-evidence-of-disease at the time of last follow-up; two patients died of unrelated causes with no-evidence-of-disease, and one patient (5%) developed distant metastases. Our data, based on consecutively resected angiomyolipomas with long clinical follow-up, suggests that epithelioid angiomyolipomas constitute a small proportion of all angiomyolipomas, and the rate of aggressive behavior among epithelioid angiomyolipomas, even when showing morphologic features previously reported to portend aggressive clinical behavior, is very low.
- Published
- 2013
- Full Text
- View/download PDF
40. Epithelioid angiomyolipoma of the kidney: pathological features and clinical outcome in a series of consecutively resected tumors
- Author
-
He, Wenlei, Cheville, John C, Sadow, Peter M, Gopalan, Anuradha, Fine, Samson W, Al-Ahmadie, Hikmat A, Chen, Ying-Bei, Oliva, Esther, Russo, Paul, Reuter, Victor E, and Tickoo, Satish K
- Abstract
The 2004 World Health Organization classification of tumors defines epithelioid angiomyolipoma of kidney as a potentially malignant mesenchymal neoplasm with reported metastasis in approximately one-third of the cases. However, this conclusion was based primarily on individual case reports and small retrospective series. More recently reported larger series have shown varying results. We reviewed 437 consecutive renal angiomyolipomas with primary resection at three tertiary-care institutions with high nephrectomy volumes. Only tumors showing >80% epithelioid histology were included in this study. Tumors resected elsewhere and reviewed in consultation were not included. Twenty of these 437 (4.6%) were classified as epithelioid angiomyolipoma. The female to male ratio was 11:9, mean age 49.7 (range, 30–80) years, and mean tumor size 8.7 (range, 1–25) cm. Microscopic tumor necrosis was present in 10 (50%) tumors and mitotic activity (range, <1–5/10 high power fields) in 8 (40%); atypical mitoses were seen in only 1 (5%) tumor. Pleomorphic ganglion-like or multinucleated giant cells were seen in 18 (90%) tumors. With a mean follow-up of 82.5 (range, 1–356) months, seventeen patients were alive with no-evidence-of-disease at the time of last follow-up; two patients died of unrelated causes with no-evidence-of-disease, and one patient (5%) developed distant metastases. Our data, based on consecutively resected angiomyolipomas with long clinical follow-up, suggests that epithelioid angiomyolipomas constitute a small proportion of all angiomyolipomas, and the rate of aggressive behavior among epithelioid angiomyolipomas, even when showing morphologic features previously reported to portend aggressive clinical behavior, is very low.
- Published
- 2013
- Full Text
- View/download PDF
41. Clear-cell papillary renal cell carcinoma: molecular and immunohistochemical analysis with emphasis on the von Hippel–Lindau gene and hypoxia-inducible factor pathway-related proteins
- Author
-
Rohan, Stephen M, Xiao, Yonghong, Liang, Yupu, Dudas, Maria E, Al-Ahmadie, Hikmat A, Fine, Samson W, Gopalan, Anuradha, Reuter, Victor E, Rosenblum, Marc K, Russo, Paul, and Tickoo, Satish K
- Abstract
Over the past few years several investigators have independently described unique low-grade renal epithelial neoplasms with clear cytoplasm, focal to diffuse papillary architecture, and occasional leiomyomatous stromal metaplasia that are not currently recognized in the World Health Organization classification of renal tumors. These tumors have been referred to by a variety of names including clear-cell papillary renal cell carcinoma and recently “clear-cell tubulopapillary renal cell carcinoma”. On the basis of the available data, such tumors are positive for cytokeratin 7 (CK7) and carbonic anhydrase IX (CA9), while being negative for CD10, α-methylacyl-CoA racemase (AMACR), and TFE3. These tumors reportedly lack trisomies of chromosomes 7 and 17, deletions of 3p25, von Hippel–Lindau (VHL) gene mutations, and VHL promoter hypermethylation. Herein, we report on nine cases of this tumor emphasizing detailed studies of the VHL gene and hypoxia-inducible factor (HIF) pathway. Molecular studies performed included VHL mutational analysis, copy number changes assessed using single-nucleotide polymorphism arrays, and qRT-PCR for VHL mRNA expression. Immunohistochemical stains for markers of HIF pathway activation (HIF-1α, CA9, and glucose transporter-1 (GLUT-1)) as well as other relevant markers (CK7, CD10, AMACR, and TFE3) were performed. None of our tumors harbored VHL gene mutations, losses of chromosomal region 3p25, or trisomies of chromosomes 7 or 17. VHL mRNA was overexpressed in our tumors relative to normal renal tissue and clear-cell renal cell carcinoma. All cases showed strong co-expression of CK7, HIF-1α, GLUT-1, and CA9. No expression of TFE3, CD10, or AMACR was seen. The morphological, immunophenotypic, and molecular features of these unique low-grade tumors are sufficiently distinct to allow separation from other renal cell carcinoma subtypes. The co-expression of CA9, HIF-1α, and GLUT-1 in the absence of VHL gene alterations in clear-cell papillary renal cell carcinoma suggests activation of the HIF pathway by non-VHL-dependent mechanisms.
- Published
- 2011
- Full Text
- View/download PDF
42. Clear-cell papillary renal cell carcinoma: molecular and immunohistochemical analysis with emphasis on the von Hippel–Lindaugene and hypoxia-inducible factor pathway-related proteins
- Author
-
Rohan, Stephen M, Xiao, Yonghong, Liang, Yupu, Dudas, Maria E, Al-Ahmadie, Hikmat A, Fine, Samson W, Gopalan, Anuradha, Reuter, Victor E, Rosenblum, Marc K, Russo, Paul, and Tickoo, Satish K
- Abstract
Over the past few years several investigators have independently described unique low-grade renal epithelial neoplasms with clear cytoplasm, focal to diffuse papillary architecture, and occasional leiomyomatous stromal metaplasia that are not currently recognized in the World Health Organization classification of renal tumors. These tumors have been referred to by a variety of names including clear-cell papillary renal cell carcinoma and recently “clear-cell tubulopapillary renal cell carcinoma”. On the basis of the available data, such tumors are positive for cytokeratin 7 (CK7) and carbonic anhydrase IX (CA9), while being negative for CD10, α-methylacyl-CoA racemase (AMACR), and TFE3. These tumors reportedly lack trisomies of chromosomes 7 and 17, deletions of 3p25, von Hippel–Lindau(VHL) gene mutations, and VHLpromoter hypermethylation. Herein, we report on nine cases of this tumor emphasizing detailed studies of the VHLgene and hypoxia-inducible factor (HIF) pathway. Molecular studies performed included VHLmutational analysis, copy number changes assessed using single-nucleotide polymorphism arrays, and qRT-PCR for VHLmRNA expression. Immunohistochemical stains for markers of HIF pathway activation (HIF-1α, CA9, and glucose transporter-1 (GLUT-1)) as well as other relevant markers (CK7, CD10, AMACR, and TFE3) were performed. None of our tumors harbored VHLgene mutations, losses of chromosomal region 3p25, or trisomies of chromosomes 7 or 17. VHLmRNA was overexpressed in our tumors relative to normal renal tissue and clear-cell renal cell carcinoma. All cases showed strong co-expression of CK7, HIF-1α, GLUT-1, and CA9. No expression of TFE3, CD10, or AMACR was seen. The morphological, immunophenotypic, and molecular features of these unique low-grade tumors are sufficiently distinct to allow separation from other renal cell carcinoma subtypes. The co-expression of CA9, HIF-1α, and GLUT-1 in the absence of VHLgene alterations in clear-cell papillary renal cell carcinoma suggests activation of the HIF pathway by non-VHL-dependent mechanisms.
- Published
- 2011
- Full Text
- View/download PDF
43. TMPRSS2–ERG gene fusion is associated with low Gleason scores and not with high-grade morphological features
- Author
-
Fine, Samson W, Gopalan, Anuradha, Leversha, Margaret A, Al-Ahmadie, Hikmat A, Tickoo, Satish K, Zhou, Qin, Satagopan, Jaya M, Scardino, Peter T, Gerald, William L, and Reuter, Victor E
- Abstract
TMPRSS2–ERG gene rearrangement is seen in about half of clinically localized prostate cancers, yet controversy exists with regard to its prognostic implications. Similarly, the relationship of TMPRSS2–ERG fusion to Gleason score and morphology remains uncertain. We assigned Gleason scores and recorded morphological features for 521 clinically localized prostate cancers sampled in triplicate and arrayed in eight tissue microarray blocks. Fluorescence in situ hybridization was performed to delineate TMPRSS2–ERG aberrations. Using maximum Gleason score, based on three core evaluation, and overall Gleason score, based on prostatectomy sections, Fisher's exact test was performed for tumors with TMPRSS2–ERG translocation/deletion, copy number increase (≥3) of the TMPRSS2–ERG region without translocation/deletion, and copy number increase and concomitant translocation/deletion. In all, 217 (42%) translocation/deletion and 30 (5.9%) copy number increase-alone cases were detected. Among 217 translocation/deletion cases, 32 had translocation/deletion with copy number increase. In all, 237, 200, and 75 cancers had maximum core-specific Gleason score of 6, 7, and 8–10, respectively. Tumors with translocation/deletion tended toward lower Gleason scores than those without (P=0.002) with similar results for overall Gleason score (P=0.02); copy number increase cases tended toward higher Gleason scores than those without (P<0.001). Gleason score of 8–10 tumors demonstrated lower odds of translocation/deletion (odds ratio (OR) 0.38; 95% CI 0.21–0.68) and higher odds of copy number increase alone (OR 7.33; 95% CI 2.65–20.31) or copy number increase+translocation/deletion (OR 3.03; 95% CI 1.12–8.15) relative to Gleason score of <7 tumors. No significant difference in TMPRSS2–ERG incidence was observed between patients with and without cribriform glands, glomerulations, signet-ring cells, or intraductal cancer (P=0.821, 0.095, 0.132, 0.375). TMPRSS2–ERG gene fusion is associated with lower core-specific and overall Gleason scores and not with high-grade morphologies. Conversely, TMPRSS2–ERG copy number increase, with or without rearrangement, is associated with higher Gleason score. These findings indicate that translocation/deletion of TMPRSS2–ERG is not associated with histological features of aggressive prostate cancer.
- Published
- 2010
- Full Text
- View/download PDF
44. TMPRSS2–ERGgene fusion is associated with low Gleason scores and not with high-grade morphological features
- Author
-
Fine, Samson W, Gopalan, Anuradha, Leversha, Margaret A, Al-Ahmadie, Hikmat A, Tickoo, Satish K, Zhou, Qin, Satagopan, Jaya M, Scardino, Peter T, Gerald, William L, and Reuter, Victor E
- Abstract
TMPRSS2–ERGgene rearrangement is seen in about half of clinically localized prostate cancers, yet controversy exists with regard to its prognostic implications. Similarly, the relationship of TMPRSS2–ERGfusion to Gleason score and morphology remains uncertain. We assigned Gleason scores and recorded morphological features for 521 clinically localized prostate cancers sampled in triplicate and arrayed in eight tissue microarray blocks. Fluorescence in situhybridization was performed to delineate TMPRSS2–ERGaberrations. Using maximum Gleason score, based on three core evaluation, and overall Gleason score, based on prostatectomy sections, Fisher's exact test was performed for tumors with TMPRSS2–ERGtranslocation/deletion, copy number increase (≥3) of the TMPRSS2–ERGregion without translocation/deletion, and copy number increase and concomitant translocation/deletion. In all, 217 (42%) translocation/deletion and 30 (5.9%) copy number increase-alone cases were detected. Among 217 translocation/deletion cases, 32 had translocation/deletion with copy number increase. In all, 237, 200, and 75 cancers had maximum core-specific Gleason score of 6, 7, and 8–10, respectively. Tumors with translocation/deletion tended toward lower Gleason scores than those without (P=0.002) with similar results for overall Gleason score (P=0.02); copy number increase cases tended toward higher Gleason scores than those without (P<0.001). Gleason score of 8–10 tumors demonstrated lower odds of translocation/deletion (odds ratio (OR) 0.38; 95% CI 0.21–0.68) and higher odds of copy number increase alone (OR 7.33; 95% CI 2.65–20.31) or copy number increase+translocation/deletion (OR 3.03; 95% CI 1.12–8.15) relative to Gleason score of <7 tumors. No significant difference in TMPRSS2–ERGincidence was observed between patients with and without cribriform glands, glomerulations, signet-ring cells, or intraductal cancer (P=0.821, 0.095, 0.132, 0.375). TMPRSS2–ERGgene fusion is associated with lower core-specific and overall Gleason scores and not with high-grade morphologies. Conversely, TMPRSS2–ERGcopy number increase, with or without rearrangement, is associated with higher Gleason score. These findings indicate that translocation/deletion of TMPRSS2–ERGis not associated with histological features of aggressive prostate cancer.
- Published
- 2010
- Full Text
- View/download PDF
45. Frequent somatic CDH1 loss-of-function mutations in plasmacytoid variant bladder cancer
- Author
-
Al-Ahmadie, Hikmat A, Iyer, Gopa, Lee, Byron H, Scott, Sasinya N, Mehra, Rohit, Bagrodia, Aditya, Jordan, Emmet J, Gao, Sizhi Paul, Ramirez, Ricardo, Cha, Eugene K, Desai, Neil B, Zabor, Emily C, Ostrovnaya, Irina, Gopalan, Anuradha, Chen, Ying-Bei, Fine, Samson W, Tickoo, Satish K, Gandhi, Anupama, Hreiki, Joseph, Viale, Agnès, Arcila, Maria E, Dalbagni, Guido, Rosenberg, Jonathan E, Bochner, Bernard H, Bajorin, Dean F, Berger, Michael F, Reuter, Victor E, Taylor, Barry S, and Solit, David B
- Abstract
Plasmacytoid bladder cancer is an aggressive histologic variant with a high risk of disease-specific mortality. Using whole-exome and targeted sequencing, we find that truncating somatic alterations in the CDH1 gene occur in 84% of plasmacytoid carcinomas and are specific to this histologic variant. Consistent with the aggressive clinical behavior of plasmacytoid carcinomas, which frequently recur locally, CRISPR/Cas9-mediated knockout of CDH1 in bladder cancer cells enhanced cell migration.
- Published
- 2016
- Full Text
- View/download PDF
46. Testicular mixed germ cell tumors: a morphological and immunohistochemical study using stem cell markers, OCT3/4, SOX2 and GDF3, with emphasis on morphologically difficult-to-classify areas
- Author
-
Gopalan, Anuradha, Dhall, Deepti, Olgac, Semra, Fine, Samson W, Korkola, James E, Houldsworth, Jane, Chaganti, Raju S, Bosl, George J, Reuter, Victor E, and Tickoo, Satish K
- Abstract
Stem cell markers, OCT3/4, and more recently SOX2 and growth differentiation factor 3 (GDF3), have been reported to be expressed variably in germ cell tumors. We investigated the immunohistochemical expression of these markers in different testicular germ cell tumors, and their utility in the differential diagnosis of morphologically difficult-to-classify components of these tumors. A total of 50 mixed testicular germ cell tumors, 43 also containing difficult-to-classify areas, were studied. In these areas, multiple morphological parameters were noted, and high-grade nuclear details similar to typical embryonal carcinoma were considered ‘embryonal carcinoma-like high-grade’. Immunohistochemical staining for OCT3/4, c-kit, CD30, SOX2, and GDF3 was performed and graded in each component as 0, negative; 1+, 1–25%; 2+, 26–50%; and 3+, >50% positive staining cells. The different components identified in these tumors were seminoma (8), embryonal carcinoma (50), yolk sac tumor (40), teratoma (40), choriocarcinoma (3) and intra-tubular germ cell neoplasia, unclassified (35). By immunohistochemistry, the staining patterns were OCT3/4 −3+, all seminomas, embryonal carcinomas and intra-tubular germ cell neoplasia; SOX2 −3+, all embryonal carcinomas and −2 to 3+, 11/14 (79%) primitive neuroectodermal components in immature teratomas; GDF3 −2 to 3+, all yolk sac tumors, seminomas and intra-tubular germ cell neoplasia and 1 to 2+, 40/50 embryonal carcinomas. A total of 34/43 (79%) of difficult-to-classify areas stained 3+ for OCT3/4, CD30, and SOX2, similar to embryonal carcinoma. Among these areas, only ‘embryonal carcinoma-like high-grade’ nuclear details were significantly associated with such an immunophenotype. Thus, SOX2 is expressed in embryonal carcinoma and primitive neuroectoderm of teratoma, and unlike OCT3/4, not in intra-tubular germ cell neoplasia and seminoma. Therefore, it may be useful in the distinction of seminoma from embryonal carcinoma, and potentially in diagnosing early carcinomatous differentiation in seminoma. GDF3 positivity, in the absence of OCT3/4 and CD30, combined with morphological features, is helpful in the diagnosis of yolk sac tumor. ‘Embryonal carcinoma-like high-grade’ nuclear details are the most important morphological criterion for the diagnosis of embryonal carcinoma in difficult-to-classify areas.
- Published
- 2009
- Full Text
- View/download PDF
47. Testicular mixed germ cell tumors: a morphological and immunohistochemical study using stem cell markers, OCT3/4, SOX2 and GDF3, with emphasis on morphologically difficult-to-classify areas
- Author
-
Gopalan, Anuradha, Dhall, Deepti, Olgac, Semra, Fine, Samson W, Korkola, James E, Houldsworth, Jane, Chaganti, Raju S, Bosl, George J, Reuter, Victor E, and Tickoo, Satish K
- Abstract
Stem cell markers, OCT3/4, and more recently SOX2 and growth differentiation factor 3 (GDF3), have been reported to be expressed variably in germ cell tumors. We investigated the immunohistochemical expression of these markers in different testicular germ cell tumors, and their utility in the differential diagnosis of morphologically difficult-to-classify components of these tumors. A total of 50 mixed testicular germ cell tumors, 43 also containing difficult-to-classify areas, were studied. In these areas, multiple morphological parameters were noted, and high-grade nuclear details similar to typical embryonal carcinoma were considered ‘embryonal carcinoma-like high-grade’. Immunohistochemical staining for OCT3/4, c-kit, CD30, SOX2, and GDF3 was performed and graded in each component as 0, negative; 1+, 1–25%; 2+, 26–50%; and 3+, >50% positive staining cells. The different components identified in these tumors were seminoma (8), embryonal carcinoma (50), yolk sac tumor (40), teratoma (40), choriocarcinoma (3) and intra-tubular germ cell neoplasia, unclassified (35). By immunohistochemistry, the staining patterns were OCT3/4 −3+, all seminomas, embryonal carcinomas and intra-tubular germ cell neoplasia; SOX2 −3+, all embryonal carcinomas and −2 to 3+, 11/14 (79%) primitive neuroectodermal components in immature teratomas; GDF3 −2 to 3+, all yolk sac tumors, seminomas and intra-tubular germ cell neoplasia and 1 to 2+, 40/50 embryonal carcinomas. A total of 34/43 (79%) of difficult-to-classify areas stained 3+ for OCT3/4, CD30, and SOX2, similar to embryonal carcinoma. Among these areas, only ‘embryonal carcinoma-like high-grade’ nuclear details were significantly associated with such an immunophenotype. Thus, SOX2 is expressed in embryonal carcinoma and primitive neuroectoderm of teratoma, and unlike OCT3/4, not in intra-tubular germ cell neoplasia and seminoma. Therefore, it may be useful in the distinction of seminoma from embryonal carcinoma, and potentially in diagnosing early carcinomatous differentiation in seminoma. GDF3 positivity, in the absence of OCT3/4 and CD30, combined with morphological features, is helpful in the diagnosis of yolk sac tumor. ‘Embryonal carcinoma-like high-grade’ nuclear details are the most important morphological criterion for the diagnosis of embryonal carcinoma in difficult-to-classify areas.Modern Pathology (2009) 22, 1066–1074; doi:10.1038/modpathol.2009.66; published online 24 April 2009
- Published
- 2009
- Full Text
- View/download PDF
48. Estrogen and progesterone-receptor-positive stroma as a non-tumorous proliferation in kidneys: a possible metaplastic response to obstruction
- Author
-
Tickoo, Satish K, Gopalan, Anuradha, Tu, Jiangling J, Harik, Lara R, Al-Ahmadie, Hikmat A, Fine, Samson W, Olgac, Semra, and Reuter, Victor E
- Abstract
The presence of estrogen and progesterone-receptor-positive stroma is well known in renal mixed epithelial and stromal tumor, cystic nephroma, and angiomyolipoma with epithelial cysts. It has been suggested that the hormone receptor positivity in mixed epithelial and stromal tumor may be etiologically related to exogenous hormone intake—a phenomenon that has become more frequent in recent years. In the past few years, we have observed such stroma in some non-neoplastic kidneys, as well as in tumor-bearing kidneys away from the tumor. Herein we present our experience with 10 such cases. In a prospective manner, whenever we noted stroma resembling that in ovaries or müllerian organs (endometrial or cervical-like) in kidneys removed for any cause, immunohistochemical stains for estrogen and progesterone receptors were performed. There were eight males and two females among the group, with ages ranging from 11 months to 71 years. In six cases, the nephrectomies were performed for a non-functional kidney, and in three for tumors (one each of chromophobe, clear cell, and acquired cystic disease-associated renal cell carcinoma). One case was a partial nephrectomy for vesico-ureteric reflux, with upper pole hydronephrosis. Such stroma was present in nine cases as a non-mass forming proliferation around dilated, frequently inflamed pelvicalyceal system and collecting ducts. In one it was present at the periphery of an acquired cystic disease-associated renal cell carcinoma, as well as around non-tumorous cysts. The only common finding in all cases was a generalized or segmental hydronephrosis, or tumor compression-related focal obstruction. The stroma was positive for estrogen receptors in all 10 cases, and for progesterone receptors in seven. Thus, estrogen- and progesterone receptor-positive stroma can be present in the kidney, not only as a component of certain tumors, but also in association with non-neoplastic conditions. Its association with obstructive changes suggests that it may represent a metaplastic change in the renal interstitial cells surrounding these obstructed epithelial structures.
- Published
- 2008
- Full Text
- View/download PDF
49. Estrogen and progesterone-receptor-positive stroma as a non-tumorous proliferation in kidneys: a possible metaplastic response to obstruction
- Author
-
Tickoo, Satish K, Gopalan, Anuradha, Tu, Jiangling J, Harik, Lara R, Al-Ahmadie, Hikmat A, Fine, Samson W, Olgac, Semra, and Reuter, Victor E
- Abstract
The presence of estrogen and progesterone-receptor-positive stroma is well known in renal mixed epithelial and stromal tumor, cystic nephroma, and angiomyolipoma with epithelial cysts. It has been suggested that the hormone receptor positivity in mixed epithelial and stromal tumor may be etiologically related to exogenous hormone intake—a phenomenon that has become more frequent in recent years. In the past few years, we have observed such stroma in some non-neoplastic kidneys, as well as in tumor-bearing kidneys away from the tumor. Herein we present our experience with 10 such cases. In a prospective manner, whenever we noted stroma resembling that in ovaries or müllerian organs (endometrial or cervical-like) in kidneys removed for any cause, immunohistochemical stains for estrogen and progesterone receptors were performed. There were eight males and two females among the group, with ages ranging from 11 months to 71 years. In six cases, the nephrectomies were performed for a non-functional kidney, and in three for tumors (one each of chromophobe, clear cell, and acquired cystic disease-associated renal cell carcinoma). One case was a partial nephrectomy for vesico-ureteric reflux, with upper pole hydronephrosis. Such stroma was present in nine cases as a non-mass forming proliferation around dilated, frequently inflamed pelvicalyceal system and collecting ducts. In one it was present at the periphery of an acquired cystic disease-associated renal cell carcinoma, as well as around non-tumorous cysts. The only common finding in all cases was a generalized or segmental hydronephrosis, or tumor compression-related focal obstruction. The stroma was positive for estrogen receptors in all 10 cases, and for progesterone receptors in seven. Thus, estrogen- and progesterone receptor-positive stroma can be present in the kidney, not only as a component of certain tumors, but also in association with non-neoplastic conditions. Its association with obstructive changes suggests that it may represent a metaplastic change in the renal interstitial cells surrounding these obstructed epithelial structures.Modern Pathology (2008) 21, 60–65; doi:10.1038/modpathol.3800958; published online 14 September 2007
- Published
- 2008
- Full Text
- View/download PDF
50. A case of Castleman's disease that presented as a retroperitoneal mass
- Author
-
Vora, Kinjal, Dash, Atreya, Bach, Ariadne, Gopalan, Anuradha, and Russo, Paul
- Abstract
Background A 35-year -old man presented to a local emergency room with acute left-flank pain and a medical history of nephrolithiasis. There were no aggravating or relieving factors for the left-flank pain and no other presenting symptoms, and the physical examination was unremarkable.Investigations Complete blood count, urinalysis, serum tumor markers, scrotal ultrasonography, CT scan of the abdomen (with and without contrast), MRI of the abdomen.Diagnosis Unicentric Castleman's disease (hyaline-vascular type).Management Surgical exploration and excision. Pathologic and immunohistochemical work-up confirmed the diagnosis. CT scan after 7 months was normal with no evidence of recurrence.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.