193 results on '"Tickoo SK"'
Search Results
2. WHO 2022 classification of penile and scrotal cancers: updates and evolution.
- Author
-
Menon, S, Moch, H, Berney, DM, Cree, IA, Srigley, JR, Tsuzuki, T, Compérat, E, Hartmann, A, Netto, G, Rubin, MA, Gill, AJ, Turajlic, S, Tan, PH, Raspollini, MR, Tickoo, SK, and Amin, M B
- Subjects
PENILE cancer ,BASAL cell carcinoma ,HUMAN papillomavirus ,SQUAMOUS cell carcinoma ,TUMOR classification ,BIOLOGICAL nomenclature - Abstract
Squamous cell carcinoma (SCC) is the most common malignant tumour of the penis. The 2022 WHO classification reinforces the 2016 classification and subclassifies precursor lesions and tumours into human papillomavirus (HPV)‐associated and HPV‐independent types. HPV‐associated penile intraepithelial neoplasia (PeIN) is a precursor lesion of invasive HPV‐ associated SCC, whereas differentiated PeIN is a precursor lesion of HPV‐independent SCC. Block‐type positivity of p16 immunohistochemistry is the most practical daily utilised method to separate HPVassociated from HPVindependent penile SCC. If this is not feasible, the term SCC, not otherwise specified (NOS) is appropriate. Certain histologies that were previously classified as "subtypes" are now grouped, and coalesced as "patterns", under the rubric of usual type SCC and verrucous carcinoma (e.g. usual‐type SCC includes pseudohyperplastic and acantholytic/pseudoglandular carcinoma, and carcinoma cuniculatum is included as a pattern of verrucous carcinoma). If there is an additional component of the usual type of invasive SCC (formerly termed hybrid histology), the tumour would be a mixed carcinoma (e.g. carcinoma cuniculatum or verrucous carcinoma with usual invasive SCC); in such cases, reporting of the relative percentages in mixed tumours may be useful. The consistent use of uniform nomenclature and reporting of percentages will inform the refinement of future reporting classification schemes and guidelines/recommendations. The classification of scrotal tumours is provided for the first time in the fifth edition of the WHO Blue book, and it follows the schema of penile cancer classification for both precursor lesions and the common SCC of the scrotum. Basal cell carcinoma of the scrotum may have a variable clinical course and finds a separate mention. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
3. Putative Drivers of Aggressiveness in TCEB1-mutant Renal Cell Carcinoma: An Emerging Entity with Variable Clinical Course
- Author
-
DiNatale, RG, Gorelick, AN, Makarov, V, Blum, KA, Silagy, AW, Freeman, B, Chowell, D, Marcon, J, Mano, R, Sanchez, A, Attalla, K, Weng, S, Voss, M, Motzer, RJ, Russo, P, Coleman, JA, Reuter, VE, Chen, Y-B, Chan, TA, Reznik, E, Tickoo, SK, Hakimi, AA, DiNatale, RG, Gorelick, AN, Makarov, V, Blum, KA, Silagy, AW, Freeman, B, Chowell, D, Marcon, J, Mano, R, Sanchez, A, Attalla, K, Weng, S, Voss, M, Motzer, RJ, Russo, P, Coleman, JA, Reuter, VE, Chen, Y-B, Chan, TA, Reznik, E, Tickoo, SK, and Hakimi, AA
- Abstract
BACKGROUND: TCEB1-mutant renal cell carcinoma (RCC) is a rare variant of RCC with clear-cell features. Owing to its unique morphological and molecular features it has recently been proposed as a separate entity. Initial series suggested an indolent, early-stage phenotype. Here we expand our clinical cohort and describe a more detailed genomic analysis looking for potential drivers of aggressiveness. DESIGN, SETTING, AND PARTICIPANTS: We identified five new cases in our institutional sequencing cohort, four of whom were found to have high-stage disease (American Joint Committee on Cancer stage III/IV). Twelve previously reported cases were pooled for comparison purposes (Sato, The Cancer Genome Atlas, TRACERx Renal). OUTCOME MEASURES AND STATISTICAL ANALYSIS: We used our previously validated pipeline to analyze somatic mutations and copy number alterations (CNAs) in seven tumor samples with available data and estimated the number of cancer cells bearing each somatic mutation. The oncogenic potential of mutations was assessed using OncoKB and two other algorithms. Mann-Whitney U tests were used to evaluate differences in genomic markers between stage groups. RESULTS AND LIMITATIONS: All tumors showed biallelic inactivation of the TCEB1 gene according to a combination of somatic mutation and CNA analyses. Mutations were always found in residues involved in hydrophobic interactions with VHL. We found that high-stage tumors had additional oncogenic mutations (median 1, interquartile range [IQR] 1-1 vs 2, IQR 2-2; median difference 1, 95% confidence interval [CI] 1-1; p= 0.002) and showed whole-genome doubling events. They also seemed to have a higher burden of somatic CNAs (median fraction CNA genome 0.10, IQR 0.10-0.15 vs 0.63, IQR 0.58-0.68), however, this finding did not reach statistical significance (median difference 0.49, 95% CI 0.33-0.63; p=0.052). CONCLUSIONS: TCEB1-mutant RCC can show variable behavior ranging from very indolent to aggressive. Specific molecula
- Published
- 2021
4. The Clinicopathologic and Molecular Landscape of Clear Cell Papillary Renal Cell Carcinoma: Implications in Diagnosis and Management
- Author
-
Weng, S, DiNatale, RG, Silagy, A, Mano, R, Attalla, K, Kashani, M, Weiss, K, Benfante, NE, Winer, AG, Coleman, JA, Reuter, VE, Russo, P, Reznik, E, Tickoo, SK, Hakimi, AA, Weng, S, DiNatale, RG, Silagy, A, Mano, R, Attalla, K, Kashani, M, Weiss, K, Benfante, NE, Winer, AG, Coleman, JA, Reuter, VE, Russo, P, Reznik, E, Tickoo, SK, and Hakimi, AA
- Abstract
BACKGROUND: Clear cell papillary renal cell carcinoma (CCPRCC) is a recently described tumor entity. Several questions remain about its epidemiology, molecular features, and clinical behavior. OBJECTIVE: To comprehensively evaluate clinicopathologic and molecular features of CCPRCC, and compare it with more common kidney cancer subtypes. DESIGN, SETTING, AND PARTICIPANTS: We identified 89 CCPRCC patients and compared their clinicopathologic features with 1120 localized clear cell renal cell carcinoma (ccRCC) and 129 type 1 papillary renal cell carcinoma (pRCC) patients. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: Nonparametric statistical testing was used to compare relevant features between tumor types. Overall, cancer-specific survival (CSS) and metastasis-free survival estimates were calculated from initial diagnosis using the Kaplan-Meier method. Patients with ipsilateral multifocal disease were explored further. A subset of CCPRCC tumors underwent genomic analysis and were compared with other RCC subtypes. RESULTS AND LIMITATIONS: A higher proportion of female (45% vs 32%) and African-American (19% vs 3%) patients were observed in the CCPRCC cohort than in the ccRCC and pRCC cohorts. CCPRCC tumors also had increased odds of presenting with additional ipsilateral masses (odds ratio [OR]: 4.41 [confidence interval {CI}: 2.34, 8.15], p < 0.001) and bilateral disease (OR: 4.80 [CI: 2.40, 9.59], p < 0.001) compared with ccRCC tumors. On molecular analysis, CCPRCC tumors showed fewer somatic aberrations and a greater degree of mitochondrial DNA depletion. In multifocal CCPRCC tumors, histologic concordance among the different renal cell carcinoma masses was estimated at 44% (7/16), and none of the individuals presenting exclusively with CCPRCC tumors developed metastatic disease after 5 yr. In contrast, multifocal tumors with CCPRCC and other nonconcordant histologies were more likely to experience adverse outcomes (CSS, log rank p = 0.034). CONCLUSIONS: CCPRCC is
- Published
- 2021
5. Reporting and staging of testicular germ cell tumors: The International Society of Urological Pathology (ISUP) testicular cancer consultation conference recommendations
- Author
-
Verrill, C, Yilmaz, A, Srigley, JR, Amin, MB, Compérat, E, Egevad, L, Ulbright, TM, Tickoo, SK, Berney, DM, Epstein, JI, and Members of the ISUP Testicular Tumor Panel
- Subjects
Male ,Pathology ,medicine.medical_specialty ,Lymphovascular invasion ,030232 urology & nephrology ,Spermatic cord ,Pathology and Forensic Medicine ,Stromal Invasion ,03 medical and health sciences ,0302 clinical medicine ,Testicular Neoplasms ,Rete testis ,Humans ,Medicine ,Neoplasm Invasiveness ,Retroperitoneal Space ,Testicular cancer ,Neoplasm Staging ,business.industry ,Neoplasms, Germ Cell and Embryonal ,medicine.disease ,Dissection ,medicine.anatomical_structure ,Lymphatic Metastasis ,030220 oncology & carcinogenesis ,Pagetoid ,Lymph Node Excision ,Surgery ,Germ cell tumors ,Anatomy ,business ,Orchiectomy - Abstract
The International Society of Urological Pathology held a conference devoted to issues in testicular and penile pathology in Boston in March 2015, which included a presentation and discussion led by the testis microscopic features working group. This conference focused on controversies related to staging and reporting of testicular tumors and was preceded by an online survey of International Society of Urological Pathology members. The survey results were used to initiate discussions, but decisions were made by expert consensus rather than voting. A number of recommendations emerged from the conference, including that lymphovascular invasion should always be reported and no distinction need be made between lymphatic or blood invasion. If lymphovascular invasion is equivocal, then it should be regarded as negative to avoid triggering unnecessary therapy. Lymphovascular invasion in the spermatic cord is considered as category pT2, not pT3 unless future studies provide contrary evidence. At the time of gross dissection, a block should be taken just superior to the epididymis to define the base of the spermatic cord and direct invasion of tumor in this block indicates a category of pT3. Pagetoid involvement of the rete testis epithelium must be distinguished from rete testis stromal invasion, with only the latter being prognostically useful. Percentages of different tumor elements in mixed germ cell tumors should be reported. Although consensus was reached on many issues, there are still areas of practice that need further evidence on which to base firm recommendations.
- Published
- 2017
6. Phase II trial of sunitinib in patients with metastatic non-clear cell renal cell carcinoma.
- Author
-
Molina AM, Feldman DR, Ginsberg MS, Kroog G, Tickoo SK, Jia X, Georges M, Patil S, Baum MS, Reuter VE, and Motzer RJ
- Published
- 2012
7. Reappraisal of Morphological Differences between Renal Medullary Carcinoma, Collecting Duct Carcinoma, and Fumarate Hydratase-Deficient Renal Cell Carcinoma
- Author
-
Gabriella Nesi, Lakshmi P. Kunju, Gladell P. Paner, Fiona Maclean, Scott A. Tomlins, Steven C. Smith, Deepika Sirohi, Mahul B. Amin, Jonathan I. Epstein, Jesse K. McKenney, Anthony J. Gill, Adeboye O. Osunkoya, Pedram Argani, Mitual Amin, Kiril Trpkov, Ying-Bei Chen, Chisato Ohe, Abbas Agaimy, Victor E. Reuter, Michelle S. Hirsch, Eva Comperat, Priya Rao, Maurizio Colecchia, Maria M. Picken, Mariza de Peralta-Venturina, Mukul K. Divatia, Wolfram Jochum, Rohit Mehra, Isabela Werneck da Cunha, Liang Cheng, Ondřej Hes, Cristina Magi-Galluzzi, Luciana Schultz, Satish K. Tickoo, Paulo Guilherme de Oliveira Salles, Ohe, C, Smith, Sc, Sirohi, D, Divatia, M, de Peralta-Venturina, M, Paner, Gp, Agaimy, A, Amin, Mb, Argani, P, Chen, Yb, Cheng, L, Colecchia, M, Comperat, E, da Cunha, Iw, Epstein, Ji, Gill, Aj, Hes, O, Hirsch, M, Jochum, W, Kunju, Lp, Maclean, F, Magi-Galluzzi, C, Mckenney, Jk, Mehra, R, Nesi, G, Osunkoya, Ao, Picken, Mm, Rao, P, Reuter, Ve, Salles, Pgd, Schultz, L, Tickoo, Sk, Tomlins, Sa, and Trpkov, K
- Subjects
0301 basic medicine ,Male ,Pathology ,Biopsy ,DNA Mutational Analysis ,urologic and male genital diseases ,Fumarate Hydratase ,Collecting duct carcinoma ,0302 clinical medicine ,Renal cell carcinoma ,SMARCB1 ,Child ,Aged, 80 and over ,Kidney ,Kidney Medulla ,medicine.diagnostic_test ,Middle Aged ,Immunohistochemistry ,Kidney Neoplasms ,Europe ,medicine.anatomical_structure ,Phenotype ,030220 oncology & carcinogenesis ,Female ,Anatomy ,Brazil ,Adult ,medicine.medical_specialty ,Canada ,Adolescent ,Article ,Pathology and Forensic Medicine ,Renal medullary carcinoma ,Diagnosis, Differential ,03 medical and health sciences ,Young Adult ,Predictive Value of Tests ,medicine ,Carcinoma ,Biomarkers, Tumor ,Humans ,Genetic Predisposition to Disease ,Kidney Tubules, Collecting ,Carcinoma, Renal Cell ,Aged ,Retrospective Studies ,business.industry ,Australia ,medicine.disease ,United States ,030104 developmental biology ,Mutation ,Surgery ,Hereditary leiomyomatosis and renal cell carcinoma ,Neoplasm Grading ,business - Abstract
Renal Medullary Carcinomas (RMCs) and Collecting Duct Carcinomas (CDCs) are rare subsets of lethal high-stage, high-grade distal nephron-related adenocarcinomas with a predilection for the renal medullary region. Recent findings have established an emerging group of fumarate hydratase (FH)-deficient tumors related to hereditary leiomyomatosis and renal cell carcinoma syndrome (HLRCC-RCCs) within this morphologic spectrum. Recently-developed, reliable ancillary testing has enabled consistent separation between these tumor types. Here, we present the clinicopathologic features and differences in the morphological patterns between RMCs, CDCs and FH-deficient RCCs in consequence of these recent developments. This study included a total of 100 cases classified using contemporary criteria and ancillary tests. Thirty-three RMCs (SMARCB1/INI1-deficient, hemoglobinopathy), 38 CDCs (SMARCB1/INI1-retained) and 29 RCCs defined by the FH-deficient phenotype (FH-/2SC+ or FH±/2SC+ with FH mutation, regardless of HLRCC syndromic stigmata/ history) were selected. The spectrum of morphologic patterns was critically evaluated and the differences between the morphological patterns present in the three groups were analyzed statistically. Twenty five percent of cases initially diagnosed as CDC were reclassified as FH-deficient RCC based on our contemporary diagnostic approach. Among the different overlapping morphologic patterns, sieve-like/cribriform and reticular/yolk sac tumor-like patterns favored RMCs, whereas intracystic papillary and tubulocystic patterns favored FH-deficient RCC. Tubulopapillary pattern favored both CDCs and FH-deficient RCCs, and multinodular infiltrating papillary pattern favored CDCs. Infiltrating glandular and solid sheets/cords/nested patterns were not statistically different among the three groups. Viral inclusion-like large nucleoli considered as a hallmark of HLRCC-RCCs were observed significantly more frequently in FH-deficient RCCs. Despite the overlapping morphology found among these clinically aggressive infiltrating high-grade adenocarcinomas of the kidney, reproducible differences in morphology emerged between these categories after rigorous characterization. Finally, we recommend that definitive diagnosis of CDC should only be made if RMC and FH-deficient RCC are excluded.
- Published
- 2018
8. Chromophobe renal cell carcinoma: a comparative study of histological, immunohistochemical and ultrastructural features using high throughput tissue microarray
- Author
-
Claudio Doglioni, B Schumacher, A H Ormsby, S K Tickoo, Pheroze Tamboli, Neil A. Abrahams, J D Khoury, Gregory T. MacLennan, Abrahams, Na, Maclennan, Gt, Khoury, Jd, Ormsby, Ah, Tamboli, P, Doglioni, Claudio, Schumacher, B, and Tickoo, Sk
- Subjects
Pathology ,medicine.medical_specialty ,Histology ,Chromophobe Renal Cell Carcinoma ,Chromophobe cell ,Keratin-20 ,Biology ,Sensitivity and Specificity ,Pathology and Forensic Medicine ,Diagnosis, Differential ,Intermediate Filament Proteins ,Renal cell carcinoma ,medicine ,Adenoma, Oxyphilic ,Humans ,Vimentin ,Oncocytoma ,Single-Blind Method ,Prospective Studies ,Carcinoma, Renal Cell ,Retrospective Studies ,Observer Variation ,Tissue microarray ,Pathology, Clinical ,Keratin 20 ,Keratin-7 ,Mucin-1 ,General Medicine ,medicine.disease ,Immunohistochemistry ,Kidney Neoplasms ,Microscopy, Electron ,Parvalbumins ,Tissue Array Analysis ,Keratin 7 ,Keratins ,Neprilysin ,Clear cell - Abstract
Aims: In some cases distinction between chromophobe renal cell carcinoma (CRCC), oncocytoma and clear cell (conventional) renal cell carcinoma (eosinophilic variant) using routine light microscopy remains problematic. The present study investigates the level of agreement in the diagnosis of CRCC, as well as the histological features most frequently used for this diagnosis by two pathologists with a special interest in renal neoplasia. The sensitivity and specificity of immunohistochemical markers in cases with overlapping histological features in the diagnosis of CRCC were also studied. Electron microscopy was performed, as a diagnostic gold standard, on all of the cases. Methods and results: Thirty-two renal tumours with predominantly eosinophilic cytoplasm were reviewed in a blinded fashion by two pathologists. The diagnosis and morphological features used to render each diagnosis were tabulated. Validation of the utility of keratin 7 and 20, epithelial membrane antigen (EMA), vimentin, CD10, parvalbumin, RCC antigen, antimitochondrial antibody and Hale's colloidal iron was performed by the construction of a tissue microarray (TMA) master block. Based on histological criteria alone, overall agreement on the diagnosis of these tumours was reached in 69% of the cases, while there was total disagreement in 12%. In 59% of the cases, total agreement was reached in classifying the case as a CRCC based on histology alone. Kappa statistics for interobserver variability were calculated as only slight agreement (kappa = 0.3). The histological features most frequently associated with a diagnosis of CRCC were accentuated cell borders (87%) and a combination of hyperchromatic wrinkled nuclei (79%) and perinuclear halos (74%). The most sensitive and specific marker for CRCC was parvalbumin (sensitivity 0.91; specificity 1.0). The immunohistochemical profile of EMA+/ vimentin- was useful but had low specificity (sensitivity 0.75; specificity 0.4). CD10 had the highest sensitivity (1.0) but worst specificity (0.25) for CRCC. Keratin 7 had high sensitivity (0.83) but fairly low specificity (0.37) for CRCC. Hale's colloidal iron and the RCC antigen marker were not contributory. Finally, the antimitochondrial antibody was found to be fairly sensitive (0.83) for excluding CRCC. Conclusions: A small but significant proportion of renal tumours with cells having eosinophilic cytoplasm cannot be classified, even by experienced pathologists, based on histology alone. In these cases it is imperative to use markers with known sensitivity and specificity for the diagnosis of CRCC. ZR 0 Z8 1 ZS 1 ZB 48
- Published
- 2004
9. Acceptance of emerging renal oncocytic neoplasms: a survey of urologic pathologists.
- Author
-
Mohanty SK, Lobo A, Jha S, Sangoi AR, Akgul M, Trpkov K, Hes O, Mehra R, Hirsch MS, Moch H, Smith SC, Shah RB, Cheng L, Amin MB, Epstein JI, Parwani AV, Delahunt B, Desai S, Przybycin CG, Manini C, Luthringer DJ, Sirohi D, Jain D, Midha D, Jain E, Maclean F, Giannico GA, Paner GP, Martignoni G, Al-Ahmadie HA, McKenney J, Srigley JR, Lopez JI, Kunju LP, Browning L, Aron M, Picken MM, Tretiakova M, Zhou M, Sable M, Kuroda N, Pattnaik N, Gupta NS, Rao P, Fine SW, Mishra P, Adhya AK, Kulkarni BN, Dixit M, Baisakh MR, Arora S, Sancheti S, Menon S, Wobker SE, Tickoo SK, Kaushal S, Soni S, Kandukuri S, Sharma S, Mitra S, Reuter VE, Malik V, Rao V, Chen YB, and Williamson SR
- Subjects
- Humans, Surveys and Questionnaires, Biomarkers, Tumor analysis, Kidney Neoplasms pathology, Kidney Neoplasms diagnosis, Carcinoma, Renal Cell pathology, Carcinoma, Renal Cell diagnosis, Adenoma, Oxyphilic pathology, Adenoma, Oxyphilic diagnosis, Pathologists
- Abstract
Oncocytic renal neoplasms are a major source of diagnostic challenge in genitourinary pathology; however, they are typically nonaggressive in general, raising the question of whether distinguishing different subtypes, including emerging entities, is necessary. Emerging entities recently described include eosinophilic solid and cystic renal cell carcinoma (ESC RCC), low-grade oncocytic tumor (LOT), eosinophilic vacuolated tumor (EVT), and papillary renal neoplasm with reverse polarity (PRNRP). A survey was shared among 65 urologic pathologists using SurveyMonkey.com (Survey Monkey, Santa Clara, CA, USA). De-identified and anonymized respondent data were analyzed. Sixty-three participants completed the survey and contributed to the study. Participants were from Asia (n = 21; 35%), North America (n = 31; 52%), Europe (n = 6; 10%), and Australia (n = 2; 3%). Half encounter oncocytic renal neoplasms that are difficult to classify monthly or more frequently. Most (70%) indicated that there is enough evidence to consider ESC RCC as a distinct entity now, whereas there was less certainty for LOT (27%), EVT (29%), and PRNRP (37%). However, when combining the responses for sufficient evidence currently and likely in the future, LOT and EVT yielded > 70% and > 60% for PRNRP. Most (60%) would not render an outright diagnosis of oncocytoma on needle core biopsy. There was a dichotomy in the routine use of immunohistochemistry (IHC) in the evaluation of oncocytoma (yes = 52%; no = 48%). The most utilized IHC markers included keratin 7 and 20, KIT, AMACR, PAX8, CA9, melan A, succinate dehydrogenase (SDH)B, and fumarate hydratase (FH). Genetic techniques used included TSC1/TSC2/MTOR (67%) or TFE3 (74%) genes and pathways; however, the majority reported using these very rarely. Only 40% have encountered low-grade oncocytic renal neoplasms that are deficient for FH. Increasing experience with the spectrum of oncocytic renal neoplasms will likely yield further insights into the most appropriate work-up, classification, and clinical management for these entities., Competing Interests: Declarations Conflict of interest The authors declare no competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
10. Long-term Outcomes of Regressed or "Burnt Out" Primary Testicular Germ Cell Tumors.
- Author
-
Baky FJ, Liso N, Williams B, Reuter VE, Tickoo SK, Feldman DR, Funt SA, Carver BS, Sheinfeld J, and Matulewicz RS
- Subjects
- Humans, Male, Adult, Middle Aged, Time Factors, Retrospective Studies, Young Adult, Treatment Outcome, Neoplasm Staging, Testicular Neoplasms therapy, Testicular Neoplasms pathology, Testicular Neoplasms surgery, Testicular Neoplasms mortality, Neoplasms, Germ Cell and Embryonal therapy, Neoplasms, Germ Cell and Embryonal pathology, Neoplasms, Germ Cell and Embryonal surgery, Neoplasms, Germ Cell and Embryonal mortality, Orchiectomy
- Abstract
Objective: To review the presentation and long-term oncologic outcomes of patients with regressed ("burnt out") primary testicular germ cell tumors (GCT). Certain testicular GCT can present with complete regression of the primary tumor. It is not well established if this is associated with more aggressive disease or worse oncologic outcomes., Methods: We queried our prospectively maintained testicular cancer clinical database at a tertiary cancer center and identified patients without prior chemotherapy who had regressed primary GCT at radical orchiectomy from 1990 to 2023. All specimens were reviewed by a genitourinary pathologist at diagnosis. Long-term clinical outcomes were reported by Kaplan-Meier method., Results: Fifty-six patients met inclusion criteria; at diagnosis, 17 had no evidence of extra-testicular disease and 39 had advanced (clinical stage [CS] II+) GCT. All CSx (no viable disease or germ cell neoplasia in situ at orchiectomy, and no evidence of advanced disease) and CS0 patients were managed with surveillance and had 5-year recurrence-free survival (RFS) of 88% (95% CI: 39%, 98%). All patients with CS II+ disease underwent primary treatment with surgery (n = 5) or first-line chemotherapy (n = 34). Two- and 5-year RFS for patients with CSII+ disease was 94% (95% CI: 78%, 98%) and 90% (95% CI: 72%, 97%), respectively., Conclusion: Patients with regressed primary testicular GCT often present with advanced disease, possibly due to lack of early clinical signs from the primary tumor. Our analysis shows excellent long-term oncologic outcomes similar to those reported in the literature for patients with viable primary testicular GCT., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this article., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
11. Differential NEUROD1, ASCL1, and POU2F3 Expression Defines Molecular Subsets of Bladder Small Cell/Neuroendocrine Carcinoma With Prognostic Implications.
- Author
-
Akbulut D, Whiting K, Teo MY, Tallman JE, Ozcan GG, Basar M, Jia L, Rammal R, Chen JF, Sarungbam J, Chen YB, Gopalan A, Fine SW, Tickoo SK, Mehra R, Baine M, Bochner BH, Pietzak EJ, Bajorin DF, Rosenberg JE, Iyer G, Solit DB, Reuter VE, Rekhtman N, Ostrovnaya I, and Al-Ahmadie H
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Prognosis, Carcinoma, Small Cell pathology, Carcinoma, Small Cell metabolism, Carcinoma, Small Cell mortality, Carcinoma, Small Cell genetics, Tissue Array Analysis, POU Domain Factors genetics, POU Domain Factors metabolism, POU Domain Factors analysis, Adult, Aged, 80 and over, Immunohistochemistry, Disease-Free Survival, Basic Helix-Loop-Helix Transcription Factors analysis, Basic Helix-Loop-Helix Transcription Factors metabolism, Urinary Bladder Neoplasms pathology, Urinary Bladder Neoplasms mortality, Urinary Bladder Neoplasms metabolism, Biomarkers, Tumor analysis, Biomarkers, Tumor metabolism, Carcinoma, Neuroendocrine pathology, Carcinoma, Neuroendocrine metabolism, Carcinoma, Neuroendocrine mortality, Carcinoma, Neuroendocrine therapy
- 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., (Copyright © 2024 United States & Canadian Academy of Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Histopathologic and Molecular Characterization of IDH-Mutant Prostatic Adenocarcinoma.
- Author
-
Samueli B, Al-Ahmadie H, Chen YB, Gopalan A, Sarungbam J, Tickoo SK, Reuter VE, Fine SW, and Chen JF
- 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 IDH mutations have been described in prostatic adenocarcinoma (PCa). Recent publications have suggested that psammomatous calcifications in PCa are associated with IDH1 mutations. 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 IDH mutations at IDH1 R132 and IDH2 R140/R172, and other nonhotspot IDH mutations. 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 IDH1/2 mutations. A histologic review of 13 cases with IDH1 hotspot mutations and available material showed grade group 3 or higher disease. Immunohistochemistry was performed on cases with IDH1 hotspot 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 IDH1 hotspot mutations were identified from 2749 patients, and 9 cases carried nonhotspot IDH1/2 mutations. The combined cohorts of 23 PCa cases with clonal IDH1 hotspot mutations had no ETS fusions, SPOP hotspot 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 IDH mutations, there were 4 with TMPRSS2::ERG fusions, 6 with SPOP hotspot mutations, and 10 with AR amplifications/hotspot mutations; 8 were microsatellite instability high. Notably, two cases with IDH1 hotspot mutations had psammomatous calcifications. Our findings provide evidence that IDH1 hotspot 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., (Copyright © 2024 United States & Canadian Academy of Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. L1 Cell Adhesion Molecule (L1CAM) Expression and Molecular Alterations Distinguish Low-Grade Oncocytic Tumor From Eosinophilic Chromophobe Renal Cell Carcinoma.
- Author
-
Alghamdi M, Chen JF, Jungbluth A, Koutzaki S, Palmer MB, Al-Ahmadie HA, Fine SW, Gopalan A, Sarungbam J, Sirintrapun SJ, Tickoo SK, Reuter VE, and Chen YB
- Subjects
- Humans, Female, Male, Middle Aged, Aged, Adult, Diagnosis, Differential, Aged, 80 and over, Immunohistochemistry, Neoplasm Grading, Mutation, Kidney Neoplasms pathology, Kidney Neoplasms genetics, Kidney Neoplasms chemistry, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, Carcinoma, Renal Cell chemistry, Biomarkers, Tumor analysis, Biomarkers, Tumor genetics, Neural Cell Adhesion Molecule L1 genetics, Neural Cell Adhesion Molecule L1 analysis, Neural Cell Adhesion Molecule L1 metabolism, Adenoma, Oxyphilic pathology, Adenoma, Oxyphilic genetics
- Abstract
Renal low-grade oncocytic tumor (LOT) is a recently recognized renal cell neoplasm designated within the "other oncocytic tumors" category in the 2022 World Health Organization classification system. Although the clinicopathologic, immunohistochemical, and molecular features reported for LOT have been largely consistent, the data are relatively limited. The morphologic overlap between LOT and other low-grade oncocytic neoplasms, particularly eosinophilic chromophobe renal cell carcinoma (E-chRCC), remains a controversial area in renal tumor classification. To address this uncertainty, we characterized and compared large cohorts of LOT (n = 67) and E-chRCC (n = 69) and revealed notable differences between the 2 entities. Clinically, LOT predominantly affected women, whereas E-chRCC showed a male predilection. Histologically, although almost all LOTs were dominated by a small-nested pattern, E-chRCC mainly showed solid and tubular architectures. Molecular analysis revealed that 87% of LOT cases harbored mutations in the tuberous sclerosis complex (TSC)-mTOR complex 1 (mTORC1) pathway, most frequently in MTOR and RHEB genes; a subset of LOT cases had chromosomal 7 and 19q gains. In contrast, E-chRCC lacked mTORC1 mutations, and 60% of cases displayed chromosomal losses characteristic of chRCC. We also explored the cell of origin for LOT and identified L1 cell adhesion molecule (L1CAM), a collecting duct and connecting tubule principal cell marker, as a highly sensitive and specific ancillary test for differentiating LOT from E-chRCC. This distinctive L1CAM immunohistochemical labeling suggests the principal cells as the cell of origin for LOT, unlike the intercalated cell origin of E-chRCC and oncocytoma. The ultrastructural analysis of LOT showed normal-appearing mitochondria and intracytoplasmic lumina with microvilli, different from what has been described for chRCC. Our study further supports LOT as a unique entity with a benign clinical course. Based on the likely cell of origin and its clinicopathologic characteristics, we propose that changing the nomenclature of LOT to "Oncocytic Principal Cell Adenoma of the Kidney" may be a better way to define and describe this entity., (Copyright © 2024 United States & Canadian Academy of Pathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
14. Ganglioneuroblastoma intermixed: Clinicopathological implications of diagnosis at presentation and genomic correlations.
- Author
-
Nezami BG, Modak S, Cardenas FI, Sarungbam J, Sirintrapun SJ, Gopalan A, Chen Y, Al-Ahmadie H, Fine SW, Reuter VE, and Tickoo SK
- Subjects
- Humans, Infant, Retrospective Studies, Prognosis, Genomics, Neoplasm Staging, Ganglioneuroblastoma diagnosis, Ganglioneuroblastoma genetics, Ganglioneuroblastoma pathology, Neuroblastoma pathology
- Abstract
Background: Ganglioneuroblastoma intermixed (GNBI) is classified as "favorable" histology by International Neuroblastoma Pathology Classification system. However, the International Neuroblastoma Risk Group (INRG) stratifies patients using wider clinicopathological and cytogenetic/molecular parameters. While the diagnosis of GNBI is typically made on resected tumor, it may sometimes be rendered on initial biopsy. We studied GNBI noted at diagnosis to evaluate its correlation with INRG staging and other clinicopathological and molecular features., Methods: In this retrospective study, clinical, radiological, pathological, cytogenetic, and molecular information from patients with GNBI at diagnosis seen between 1995 and 2021 was analyzed. INRG staging was performed., Results: Of the 15,827 neuroblastoma specimens, GNBI was noted in 237 patients. Of these, 53 had the initial pathological diagnosis of GNBI; median follow-up 3.5 (range: 0.2-14) years. Disease was locoregional in 41 (77%, 16 stage L1 and 25 L2); none relapsed. Twelve (23%) had metastatic disease at presentation; six (50%) relapsed, and two died of disease. MYCN was amplified in two metastatic tumors. Six of 31 (19%) tumors tested had recurrent cytogenetic abnormalities and nonrecurrent somatic gene mutations in 10/23 (43%). The presence of any adverse molecular/cytogenetic findings was associated with metastatic disease (p < .05). For patients with localized GNBI undergoing both biopsy and resection, GNBI was diagnosed in both in 17/19 (90%)., Conclusions: Localized GNBI at diagnosis has excellent long-term clinical outcome even without cytotoxic therapy. For localized GNBI, a biopsy sample is adequate to make the diagnosis. When associated with metastasis at diagnosis, prognosis is poorer, possibly due to associated adverse biological features., (© 2023 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
15. Identification of immunotherapy and radioimmunotherapy targets on desmoplastic small round cell tumors.
- Author
-
Espinosa-Cotton M, Guo HF, Tickoo SK, and Cheung NV
- Abstract
Background: Development of successful antibody-based immunotherapeutic and radioimmunotherapeutic strategies rely on the identification of cell surface tumor-associated antigens (TAA) with restricted expression on normal tissues. Desmoplastic small round cell tumor (DSRCT) is a rare and generally neglected malignancy that primarily affects adolescent and young adult males. New therapies capable of treating disseminated disease are needed for DSRCT, which is often widespread at diagnosis., Methods: We used immunohistochemistry (IHC) on fresh frozen surgical specimens and patient-derived xenograft (PDX) tumors and flow cytometry on DSRCT cell lines to evaluate expression of TAAs in these tumors. In vitro cytotoxicity assays were used to evaluate the efficacy of T cell-engaging bispecific antibodies (T-BsAbs) directed at these targets. In vivo , we used an intraperitoneal xenograft mouse model of DSRCT to test T-BsAbs against several TAAs., Results: In DSRCT specimens we found widespread expression of B7-H3, EGFR, GD2, HER2, mesothelin, and polysialic acid, clinical targets for which specific antibody therapeutics are available. The expression of B7-H3, EGFR, HER2, and mesothelin was confirmed on the cell surface of DSRCT cell lines. In vitro cytotoxicity assays confirmed the efficacy of T cell-engaging bispecific antibodies (T-BsAbs) directed at these targets against DSRCT cells. Remarkably, a HER2xCD3 T-BsAb was capable of completely shrinking established tumors in an intraperitoneal mouse model of DSRCT., Conclusions: We propose that these TAAs should be further investigated in preclinical models as targets for immunotherapy and radioimmunotherapy with the hope of providing a rationale to extend these therapies to patients with advanced DSRCT., Competing Interests: N-KC reports receiving commercial research grants from Y-mAbs Therapeutics. N-KC is the inventor and owner of issued patents licensed by MSKCC to Y-mAbs Therapeutics, Biotec Pharmacon/Lallemand, and Abpro-labs. MSKCC and N-KC have financial interest in Y-mAbs. N-KC reports receiving stock options from Eureka Therapeutics. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest, (Copyright © 2023 Espinosa-Cotton, Guo, Tickoo and Cheung.)
- Published
- 2023
- Full Text
- View/download PDF
16. Prostate Adenocarcinoma Grade Group 1: Rationale for Retaining a Cancer Label in the 2022 World Health Organization Classification.
- Author
-
Netto GJ, Amin MB, Compérat EM, Gill AJ, Hartmann A, Moch H, Menon S, Raspollini MR, Rubin MA, Srigley JR, Hoon Tan P, Tickoo SK, Tsuzuki T, Turajlic S, Cree I, and Berney DM
- Subjects
- Male, Humans, Prostate pathology, Neoplasm Grading, Prostate-Specific Antigen, Prostatic Neoplasms pathology, Adenocarcinoma pathology
- Abstract
We present the rationale for keeping the "cancer" label for grade group 1 (GG1) prostate cancer. Maintaining GG1 as the lowest grade outweighs the potential benefits that a benign designation may bring. Patient and surgeon education on the vital role of active surveillance for GG1 cancers and avoidance of overtreatment should be the focus rather than such a drastic change in nomenclature., (Copyright © 2022 European Association of Urology. Published by Elsevier B.V. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Author Correction: FOXA1 repression drives lineage plasticity and immune heterogeneity in bladder cancers with squamous differentiation.
- Author
-
Warrick JI, Hu W, Yamashita H, Walter V, Shuman L, Craig JM, Gellert LL, Castro MAA, Robertson AG, Kuo F, Ostrovnaya I, Sarungbam J, Chen YB, Gopalan A, Sirintrapun SJ, Fine SW, Tickoo SK, Kim K, Thomas J, Karan N, Gao SP, Clinton TN, Lenis AT, Chan TA, Chen Z, Rao M, Hollman TJ, Li Y, Socci ND, Chavan S, Viale A, Mohibullah N, Bochner BH, Pietzak EJ, Teo MY, Iyer G, Rosenberg JE, Bajorin DF, Kaag M, Merrill SB, Joshi M, Adam R, Taylor JA 3rd, Clark PE, Raman JD, Reuter VE, Chen Y, Funt SA, Solit DB, DeGraff DJ, and Al-Ahmadie HA
- Published
- 2022
- Full Text
- View/download PDF
18. FOXA1 repression drives lineage plasticity and immune heterogeneity in bladder cancers with squamous differentiation.
- Author
-
Warrick JI, Hu W, Yamashita H, Walter V, Shuman L, Craig JM, Gellert LL, Castro MAA, Robertson AG, Kuo F, Ostrovnaya I, Sarungbam J, Chen YB, Gopalan A, Sirintrapun SJ, Fine SW, Tickoo SK, Kim K, Thomas J, Karan N, Gao SP, Clinton TN, Lenis AT, Chan TA, Chen Z, Rao M, Hollman TJ, Li Y, Socci ND, Chavan S, Viale A, Mohibullah N, Bochner BH, Pietzak EJ, Teo MY, Iyer G, Rosenberg JE, Bajorin DF, Kaag M, Merrill SB, Joshi M, Adam R, Taylor JA 3rd, Clark PE, Raman JD, Reuter VE, Chen Y, Funt SA, Solit DB, DeGraff DJ, and Al-Ahmadie HA
- Subjects
- Humans, Biomarkers, Tumor genetics, Hepatocyte Nuclear Factor 3-alpha genetics, Phylogeny, Cell Lineage, Carcinoma, Squamous Cell pathology, Carcinoma, Transitional Cell metabolism, Urinary Bladder Neoplasms pathology
- Abstract
Cancers arising from the bladder urothelium often exhibit lineage plasticity with regions of urothelial carcinoma adjacent to or admixed with regions of divergent histomorphology, most commonly squamous differentiation. To define the biologic basis for and clinical significance of this morphologic heterogeneity, here we perform integrated genomic analyses of mixed histology bladder cancers with separable regions of urothelial and squamous differentiation. We find that squamous differentiation is a marker of intratumoral genomic and immunologic heterogeneity in patients with bladder cancer and a biomarker of intrinsic immunotherapy resistance. Phylogenetic analysis confirms that in all cases the urothelial and squamous regions are derived from a common shared precursor. Despite the presence of marked genomic heterogeneity between co-existent urothelial and squamous differentiated regions, no recurrent genomic alteration exclusive to the urothelial or squamous morphologies is identified. Rather, lineage plasticity in bladder cancers with squamous differentiation is associated with loss of expression of FOXA1, GATA3, and PPARG, transcription factors critical for maintenance of urothelial cell identity. Of clinical significance, lineage plasticity and PD-L1 expression is coordinately dysregulated via FOXA1, with patients exhibiting morphologic heterogeneity pre-treatment significantly less likely to respond to immune checkpoint inhibitors., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
19. The 2022 World Health Organization Classification of Tumors of the Urinary System and Male Genital Organs-Part B: Prostate and Urinary Tract Tumors.
- Author
-
Netto GJ, Amin MB, Berney DM, Compérat EM, Gill AJ, Hartmann A, Menon S, Raspollini MR, Rubin MA, Srigley JR, Hoon Tan P, Tickoo SK, Tsuzuki T, Turajlic S, Cree I, and Moch H
- Subjects
- Humans, Male, Prognosis, Prostate pathology, World Health Organization, Urinary Tract pathology, Urologic Neoplasms pathology
- Abstract
The 2022 World Health Organization (WHO) classification of the urinary and male genital tumors was recently published by the International Agency for Research on Cancer. This fifth edition of the WHO "Blue Book" offers a comprehensive update on the terminology, epidemiology, pathogenesis, histopathology, diagnostic molecular pathology, and prognostic and predictive progress in genitourinary tumors. In this review, the editors of the fifth series volume on urologic and male genital neoplasms present a summary of the salient changes introduced to the classification of tumors of the prostate and the urinary tract., (Copyright © 2022. Published by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
20. The 2022 World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs-Part A: Renal, Penile, and Testicular Tumours.
- Author
-
Moch H, Amin MB, Berney DM, Compérat EM, Gill AJ, Hartmann A, Menon S, Raspollini MR, Rubin MA, Srigley JR, Hoon Tan P, Tickoo SK, Tsuzuki T, Turajlic S, Cree I, and Netto GJ
- Subjects
- Genitalia, Male pathology, Humans, Male, Receptor Protein-Tyrosine Kinases, World Health Organization, Carcinoma, Renal Cell pathology, Kidney Neoplasms diagnosis, Neoplasms, Germ Cell and Embryonal genetics, Neuroectodermal Tumors, Papillomavirus Infections, Testicular Neoplasms pathology
- Abstract
The fifth edition of the World Health Organization (WHO) classification of urogenital tumours (WHO "Blue Book"), published in 2022, contains significant revisions. This review summarises the most relevant changes for renal, penile, and testicular tumours. In keeping with other volumes in the fifth edition series, the WHO classification of urogenital tumours follows a hierarchical classification and lists tumours by site, category, family, and type. The section "essential and desirable diagnostic criteria" included in the WHO fifth edition represents morphologic diagnostic criteria, combined with immunohistochemistry and relevant molecular tests. The global introduction of massive parallel sequencing will result in a diagnostic shift from morphology to molecular analyses. Therefore, a molecular-driven renal tumour classification has been introduced, taking recent discoveries in renal tumour genomics into account. Such novel molecularly defined epithelial renal tumours include SMARCB1-deficient medullary renal cell carcinoma (RCC), TFEB-altered RCC, Alk-rearranged RCC, and ELOC-mutated RCC. Eosinophilic solid and cystic RCC is a novel morphologically defined RCC entity. The diverse morphologic patterns of penile squamous cell carcinomas are grouped as human papillomavirus (HPV) associated and HPV independent, and there is an attempt to simplify the morphologic classification. A new chapter with tumours of the scrotum has been introduced. The main nomenclature of testicular tumours is retained, including the use of the term "germ cell neoplasia in situ" (GCNIS) for the preneoplastic lesion of most germ cell tumours and division from those not derived from GCNIS. Nomenclature changes include replacement of the term "primitive neuroectodermal tumour" by "embryonic neuroectodermal tumour" to separate these tumours clearly from Ewing sarcoma. The term "carcinoid" has been changed to "neuroendocrine tumour", with most examples in the testis now classified as "prepubertal type testicular neuroendocrine tumour"., (Copyright © 2022 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
21. WHO 2022 landscape of papillary and chromophobe renal cell carcinoma.
- Author
-
Lobo J, Ohashi R, Amin MB, Berney DM, Compérat EM, Cree IA, Gill AJ, Hartmann A, Menon S, Netto GJ, Raspollini MR, Rubin MA, Tan PH, Tickoo SK, Tsuzuki T, Turajlic S, Zhou M, Srigley JR, and Moch H
- Subjects
- Diagnosis, Differential, Humans, Kidney pathology, Male, World Health Organization, Adenoma, Oxyphilic, Carcinoma, Renal Cell diagnosis, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology
- Abstract
The 5th edition of the WHO Classification of Tumours of the Urinary and Male Genital Systems contains relevant revisions and introduces a group of molecularly defined renal tumour subtypes. Herein we present the World Health Organization (WHO) 2022 perspectives on papillary and chromophobe renal cell carcinoma with emphasis on their evolving classification, differential diagnosis, and emerging entities. The WHO 2022 classification eliminated the type 1/2 papillary renal cell carcinoma (pRCC) subcategorization, given the recognition of frequent mixed tumour phenotypes and the existence of entities with a different molecular background within the type 2 pRCC category. Additionally, emerging entities such as biphasic squamoid alveolar RCC, biphasic hyalinising psammomatous RCC, papillary renal neoplasm with reverse polarity, and Warthin-like pRCC are included as part of the pRCC spectrum, while additional morphological and molecular data are being gathered. In addition to oncocytomas and chromophobe renal cell carcinoma (chRCC), a category of 'other oncocytic tumours' with oncocytoma/chRCC-like features has been introduced, including emerging entities, most with TSC/mTOR pathway alterations (eosinophilic vacuolated tumour and so-called 'low-grade' oncocytic tumour), deserving additional research. Eosinophilic solid and cystic RCC was accepted as a new and independent tumour entity. Finally, a highly reproducible and clinically relevant universal grading system for chRCC is still missing and is another niche of ongoing investigation. This review discusses these developments and highlights emerging morphological and molecular data relevant for the classification of renal cell carcinoma., (© 2022 John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
22. An introduction to the WHO 5th edition 2022 classification of testicular tumours.
- Author
-
Berney DM, Cree I, Rao V, Moch H, Srigley JR, Tsuzuki T, Amin MB, Comperat EM, Hartmann A, Menon S, Netto GJ, Rubin MA, Turajlic S, Raspollini MR, and Tickoo SK
- Subjects
- Humans, Male, World Health Organization, Carcinoid Tumor, Neoplasms, Germ Cell and Embryonal, Seminoma pathology, Sex Cord-Gonadal Stromal Tumors, Testicular Neoplasms pathology
- Abstract
The 5th edition of the World Health Organisation Blue Book was published recently and includes a comprehensive update on testicular tumours. This builds upon the work of the 4th edition, retaining its structure and main nomenclature, including the use of the term 'germ cell neoplasia in situ' (GCNIS) for the pre-invasive lesion of most germ cell tumours and division from those not derived from GCNIS. While there have been important developments in understanding the molecular underpinnings of testicular cancer, this updated classification paradigm and approach remains rooted in morphology. Nomenclature changes include replacement of the term 'primitive neuroectodermal tumour' by 'embryonic neuroectodermal tumour' based on the non-specificity of the former term and to separate these tumours clearly from Ewing sarcoma. Seminoma is placed in a germinoma family of tumours emphasising relation to those tumours at other sites. Criteria for the diagnosis of 'teratoma with somatic transformation' have been modified to not include variable field size assessments. The word 'carcinoid' has been changed to 'neuroendocrine tumour', with most examples in the testis now classified as 'prepubertal type testicular neuroendocrine tumour'. For sex cord-stromal tumours, the use of mitotic counts per high-power field has been changed to per mm2 for malignancy assessments, and the new entities, 'signet ring stromal tumour' and 'myoid gonadal stromal tumour', are defined. Well-differentiated papillary mesothelial tumour has now been defined as tumour type with a favourable prognosis. Sertoliform cystadenoma has been removed as an entity from testicular adnexal tumours and placed with Sertoli cell tumours., (© 2022 The Authors. Histopathology published by John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
23. Low grade oncocytic tumors of the kidney: a clinically relevant approach for the workup and accurate diagnosis.
- Author
-
Amin MB, McKenney JK, Martignoni G, Campbell SC, Pal S, and Tickoo SK
- Subjects
- Biomarkers, Tumor, Diagnosis, Differential, Humans, Immunohistochemistry, Kidney pathology, Adenoma, Oxyphilic diagnosis, Adenoma, Oxyphilic pathology, Carcinoma, Renal Cell diagnosis, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, Kidney Neoplasms diagnosis, Kidney Neoplasms pathology
- Abstract
Renal oncocytoma and chromophobe renal cell carcinoma were accepted as unique renal tumors in the late 1990s. Since their formal description, criteria for diagnosis have evolved and additional distinct tumor subtypes originally considered as one these two entities are now recognized. The last two decades have witnessed unprecedented interest in the spectrum of low grade oncocytic renal neoplasms in three specific areas: (1) histologic characterization of tumors with overlapping morphologic features between oncocytoma and chromophobe renal cell carcinoma; (2) description of potentially unique entities within this spectrum, such as eosinophilic vacuolated tumor and low-grade oncocytic tumor; and (3) better appreciation of the association between a subset of low grade oncocytic tumors and hereditary renal neoplasia. While this important work has been academically rewarding, the proposal of several histologic entities with overlapping morphologic and immunophenotypic features (which may require esoteric adjunctive immunohistochemical and/or molecular techniques for confirmation) has created frustration in the diagnostic pathology and urology community as information evolves regarding classification within this spectrum of renal neoplasia. Pathologists, including genitourinary subspecialists, are often uncertain as to the "best practice" diagnostic approach to such tumors. In this review, we present a practical clinically relevant algorithmic approach to classifying tumors within the low grade oncocytic family of renal neoplasia, including a proposal for compressing terminology for evolving categories where appropriate without sacrificing prognostic relevance., (© 2022. The Author(s), under exclusive licence to United States & Canadian Academy of Pathology.)
- Published
- 2022
- Full Text
- View/download PDF
24. What's new in WHO fifth edition - urinary tract.
- Author
-
Compérat E, Amin MB, Berney DM, Cree I, Menon S, Moch H, Netto GJ, Rao V, Raspollini MR, Rubin MA, Srigley JR, Tan PH, Tickoo SK, Turajlic S, and Tsuzuki T
- Subjects
- Humans, World Health Organization, Urinary Bladder Neoplasms pathology, Urinary Tract pathology, Urologic Neoplasms
- Abstract
The fifth edition of the WHO Blue Book on urological tumours, specifically in the bladder chapter, represents a refinement and update in the classification of bladder tumours building on the aggregate major changes made in previous editions. Progress in the molecular underpinnings of urothelial tumours, particularly with promising stratifiers for more precision-based treatment approaches, have been made. Special attention has been paid to burning questions in bladder pathology, such as grading, heterogeneous lesions, inverted tumours and substaging. The concept of neuroendocrine tumours will be explained precisely., (© 2022 John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
25. 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 MY, Guercio BJ, Arora A, Hao X, Regazzi AM, Donahue T, Herr HW, Goh AC, Cha EK, Pietzak E, Donat SM, Dalbagni G, Bochner BH, Olgac S, Sarungbam J, Sirintrapun SJ, Chen YB, Gopalan A, Fine SW, Tickoo SK, Reuter VE, Weigelt B, Schultheis AM, Funt SA, Bajorin DF, Solit DB, Iyer G, Ostrovnaya I, Rosenberg JE, and Al-Ahmadie H
- Subjects
- Chemotherapy, Adjuvant, Cystectomy, Genomics, Humans, Neoadjuvant Therapy, Retrospective Studies, Urinary Bladder pathology, Xeroderma Pigmentosum Group D Protein, Carcinoma, Small Cell pathology, Urinary Bladder Neoplasms drug therapy, Urinary Bladder Neoplasms genetics
- Abstract
Introduction: Small cell carcinoma of the bladder (SCCB) is a rare variant of bladder cancer with poor outcomes. We evaluated long-term outcomes of nonmetastatic (M0) and metastatic (M1) SCCB and correlated pathologic response with genomic alterations of patients treated with neoadjuvant chemotherapy (NAC)., Patients and Methods: Clinical history and pathology samples from SCCB patients diagnosed at our institution were reviewed., Results: One hundred and ninety-nine SCCB patients were identified. (M0: 147 [74%]; M1: 52 [26%]). Among M0 patients, 108 underwent radical cystectomy (RC) (NAC: 71; RC only: 23; adjuvant chemotherapy: 14); 14 received chemoradiotherapy; the rest received chemotherapy alone or no cancer-directed therapy. RC-only patients had a median follow-up of 9.1 years, and median disease-free survival (DFS) and overall survival (OS) were 1.1 and 1.2 years, respectively. NAC patients had pathologic response (
- Published
- 2022
- Full Text
- View/download PDF
26. Impact of Zone of Origin in Anterior Dominant Prostate Cancer: Long-Term Biochemical Recurrence-Free Survival in an Anatomically Well-Characterized Cohort.
- Author
-
Fine SW, Al-Ahmadie HA, Vertosick E, Vickers AJ, Chen YB, Gopalan A, Sarungbam J, Sirintrapun SJ, Tickoo SK, Eastham JA, Scardino PT, and Reuter VE
- Abstract
Introduction: Our goal was to determine whether zonal origin of anterior dominant prostate cancers is associated with clinical outcome among patients treated with radical prostatectomy., Methods: We investigated the clinical outcomes of 197 patients with previously well-characterized anterior dominant prostatic tumors on radical prostatectomy. Univariable Cox proportional hazards models were used to test for an association between anterior peripheral zone (PZ) or transition zone (TZ) tumor location and clinical outcomes., Results: Zonal origin of anterior dominant tumors: 97/197 (49%) anterior PZ, 70 (36%) TZ, 14 (7%) both zones and 16 (8%) indeterminate zone. Comparing anterior PZ and TZ tumors, there were no significant differences in Grade group, incidence of extraprostatic extension or surgical margin positivity rate. Overall, 19 (9.6%) patients experienced biochemical recurrence (BCR), including 10 with anterior PZ origin and 5 with TZ origin. Median followup time among those without BCR was 9.5 years (IQR 7.2, 12.7). BCR-free survival at 5 and 10 years was 91% and 89% for anterior PZ tumors, and 94% and 92% for TZ tumors, respectively. On univariate analysis, there was no evidence of a difference in time to BCR between anterior PZ and TZ tumor zone of origin (p=0.5)., Conclusions: In this anatomically well-characterized cohort of anterior dominant prostate cancers, long-term BCR-free survival was not significantly associated with zone of origin. Future studies utilizing zone of origin as a parameter should consider separating anterior and posterior PZ localization, as outcomes may differ.
- Published
- 2022
- Full Text
- View/download PDF
27. Neuroendocrine differentiation in the setting of prostatic carcinoma: contemporary assessment of a consecutive series.
- Author
-
Gopalan A, Al-Ahmadie H, Chen YB, Sarungbam J, Sirintrapun SJ, Tickoo SK, Reuter VE, and Fine SW
- Subjects
- Androgen Antagonists, Humans, Immunohistochemistry, Male, Prostate pathology, Carcinoma, Neuroendocrine pathology, Carcinoma, Small Cell pathology, Prostatic Neoplasms pathology
- Abstract
Aim: Clinicopathologic characterisation of a contemporary series of neuroendocrine (NE) differentiation in the setting of prostatic carcinoma (PCa) was examined., Methods and Results: We reviewed institutional databases for in-house cases with a history of PCa and histopathologic evidence of NE differentiation during the disease course. In all, 79 cases were identified: 32 primary and 47 metastases. Metastatic lesions were in liver (n = 15), lymph node (n = 9), bone (n = 6), lung (n = 3), brain (n = 1), and other sites (n = 13). In all, 63 of 76 (82%) cases with NE differentiation and available history were posttherapy: six postradiation therapy (RT), 24 post- androgen-deprivation therapy (ADT), and 33 post-RT + ADT. Morphologic assessment (n = 79): (i) 23 pure small-cell/high-grade NE carcinoma (HGNEC): 20/23 metastatic; (ii) 10 combined high-grade PCa and small-cell/HGNEC: 9/10 primary; (iii) 15 PCa with diffuse NE immunohistochemistry (IHC) marker positivity/differentiation, associated with nested to sheet-like growth of cells with abundant cytoplasm and prominent nucleoli, yet diffuse positivity for at least one prostatic and one NE IHC marker: all metastatic; (iv) 11 PCa with patchy NE differentiation, displaying more than single-cell positivity for NE IHC: five primary / six metastatic; (v) nine PCa with focal NE marker positive cells: four primary / five metastatic; (vi) 11 PCa with 'Paneth cell-like' change: all primary., Conclusions: In this contemporary series, the majority of NE differentiation in the setting of PCa was seen posttherapy. We highlight the tendencies of small-cell/HGNEC and PCa with diffuse NE differentiation by IHC to occur in metastatic settings, while morphologically combined high-grade PCa + small-cell/HGNEC and 'Paneth cell-like' change occur in primary disease., (© 2022 John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
28. Adult Wilms Tumor: Genetic Evidence of Origin of a Subset of Cases From Metanephric Adenoma.
- Author
-
Argani P, Tickoo SK, Matoso A, Pratilas CA, Mehra R, Tretiakova M, Sibony M, Meeker AK, Lin MT, Reuter VE, Epstein JI, Gagan J, and Palsgrove DN
- Subjects
- Adult, Child, Female, Humans, Male, Mutation, Proto-Oncogene Proteins B-raf genetics, Adenoma genetics, Adenoma pathology, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Wilms Tumor genetics, Wilms Tumor pathology
- Abstract
The genetics of nephroblastoma (Wilms tumor) occurring in adults is largely unknown, as studies have largely been limited to isolated case reports. We, therefore, studied 14 adult Wilms tumors for genetic alterations, using expanded targeted sequencing on 11 cases. The patients ranged from 17 to 46 years of age (mean and median, 31 y), and there were 8 males and 6 females. Five Wilms tumors harbored BRAF V600E mutations. All of these had better-differentiated areas identical to metanephric adenoma, as has previously been described. In 3 such cases, microdissection studies revealed that the BRAF V600E mutation was present in both the metanephric adenoma and Wilms tumor areas; however, additional genetic alterations (including TERT promoter mutations in 2 cases, ASLX1/ATR mutations in 1 other case) were limited to the Wilms tumor component. These findings suggest that the Wilms tumor developed from the metanephric adenoma. Other adult Wilms tumors harbored genetic alterations previously reported in the more common pediatric Wilms tumors, including WT1 mutations (2 cases), ASLX1 mutations (3 additional cases), NSD2 mutation (1 additional case), and 11p loss (3 cases). In summary, a significant subset of adult Wilms tumors (specifically those of epithelial type with differentiated areas) harbor targetable BRAF V600E mutations and appear to arise from metanephric adenomas as a consequence of additional acquired genetic alterations. Other adult Wilms tumors often harbor genetic alterations found in their more common pediatric counterparts, suggesting at least some similarities in their pathogenesis., Competing Interests: Conflicts of Interest and Source of Funding: Supported in part by the Dahan Fund (P.A.) and Joey’s Wings (P.A.). The authors have disclosed that they have no significant relationships with, or financial interest in, any commercial companies pertaining to this article., (Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. TERT Copy Number Alterations, Promoter Mutations and Rearrangements in Adrenocortical Carcinomas.
- Author
-
Gupta S, Won H, Chadalavada K, Nanjangud GJ, Chen YB, Al-Ahmadie HA, Fine SW, Sirintrapun SJ, Strong VE, Raj N, Lagunes DR, Vanderbilt CM, Berger MF, Ladanyi M, Dogan S, Tickoo SK, Reuter VE, and Gopalan A
- Subjects
- DNA Copy Number Variations, Humans, Mutation, Adrenal Cortex Neoplasms genetics, Adrenocortical Carcinoma genetics, Adrenocortical Carcinoma pathology, Telomerase genetics
- Abstract
Molecular characterization of adrenocortical carcinomas (ACC) by The Cancer Genome Atlas (TCGA) has highlighted a high prevalence of TERT alterations, which are associated with disease progression. Herein, 78 ACC were profiled using a combination of next generation sequencing (n = 76) and FISH (n = 9) to assess for TERT alterations. This data was combined with TCGA dataset (n = 91). A subset of borderline adrenocortical tumors (n = 5) and adrenocortical adenomas (n = 7) were also evaluated. The most common alteration involving the TERT gene involved gains/amplifications, seen in 22.2% (37/167) of cases. In contrast, "hotspot" promoter mutations (C > T promoter mutation at position -124, 7/167 cases, 4.2%) and promoter rearrangements (2/165, 1.2%) were rare. Recurrent co-alterations included 22q copy number losses seen in 24% (9/38) of cases. Although no significant differences were identified in cases with and without TERT alterations pertaining to age at presentation, tumor size, weight, laterality, mitotic index and Ki67 labeling, cases with TERT alterations showed worse outcomes. Metastatic behavior was seen in 70% (28/40) of cases with TERT alterations compared to 51.2% (65/127, p = 0.04) of cases that lacked these alterations. Two (of 5) borderline tumors showed amplifications and no TERT alterations were identified in 7 adenomas. In the borderline group, 0 (of 4) patients with available follow up had adverse outcomes. We found that TERT alterations in ACC predominantly involve gene amplifications, with a smaller subset harboring "hotspot" promoter mutations and rearrangements, and 70% of TERT-altered tumors are associated with metastases. Prospective studies are needed to validate the prognostic impact of these findings., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
30. Papillary renal cell carcinoma: a single institutional study of 199 cases addressing classification, clinicopathologic and molecular features, and treatment outcome.
- Author
-
Murugan P, Jia L, Dinatale RG, Assel M, Benfante N, Al-Ahmadie HA, Fine SW, Gopalan A, Sarungbam J, Sirintrapun SJ, Hakimi AA, Russo P, Chen YB, Tickoo SK, and Reuter VE
- Subjects
- DNA Copy Number Variations, Humans, Treatment Outcome, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell therapy, Kidney Neoplasms pathology
- 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., (© 2021. The Author(s), under exclusive licence to United States & Canadian Academy of Pathology.)
- Published
- 2022
- Full Text
- View/download PDF
31. Clinical and Genomic Characterization of Bladder Carcinomas With Glandular Phenotype.
- Author
-
Almassi N, Whiting K, Toubaji A, Lenis AT, Jordan EJ, Won H, Regazzi AM, Chen YB, Gopalan A, Sirintrapun SJ, Fine SW, Tickoo SK, Ostrovnaya I, Pietzak EJ, Cha EK, Goh AC, Donahue TF, Herr HW, Donat SM, Dalbagni G, Bochner BH, Teo MY, Funt SA, Rosenberg JE, Reuter VE, Bajorin DF, Solit DB, Al-Ahmadie H, and Iyer G
- Subjects
- Genomics methods, Humans, Phenotype, Retrospective Studies, Urinary Bladder pathology, Adenocarcinoma genetics, Carcinoma, Transitional Cell genetics, Colorectal Neoplasms pathology, Urinary Bladder Neoplasms genetics
- Abstract
Purpose: To compare oncologic outcomes and genomic alteration profiles in patients with bladder and urachal adenocarcinoma, urothelial carcinoma (UC) with glandular differentiation, and UC, not otherwise specified (NOS) undergoing surgical resection, with emphasis on response to systemic therapy., Methods: We identified patients with bladder cancer with glandular variants who underwent surgical resection at Memorial Sloan Kettering from 1995 to 2018 (surgical cohort) and/or patients who had tumor sequencing using a targeted next-generation sequencing platform (genomics cohort). Pathologic complete and partial response rates to neoadjuvant chemotherapy (NAC) and recurrence-free and cancer-specific survival were measured. Alteration frequencies between histologic subtypes were compared., Results: Thirty-seven patients with bladder adenocarcinoma, 46 with urachal adenocarcinoma, 84 with UC with glandular differentiation, and 1,049 with UC, NOS comprised the surgical cohort. Despite more advanced disease in patients with bladder and urachal adenocarcinoma, no significant differences in recurrence or cancer-specific survival by histology were observed after adjusting for stage. In patients with UC with glandular differentiation, NAC resulted in partial (≤ pT1N0) and complete (pT0N0) responses in 28% and 17%, respectively. Bladder and urachal adenocarcinoma genomic profiles resembled colorectal adenocarcinoma with frequent TP53 , KRAS , and PIK3CA alterations while the genomic profile of UC with glandular differentiation more closely resembled UC, NOS. Limitations include retrospective nature of analysis and small numbers of nonurothelial histology specimens., Conclusion: The genomic profile of bladder adenocarcinomas resembled colorectal adenocarcinomas, whereas UC with glandular differentiation more closely resembled UC, NOS. Differences in outcomes among patients with glandular bladder cancer variants undergoing surgical resection were largely driven by differences in stage. Cisplatin-based NAC demonstrated activity in UC with glandular differentiation, suggesting NAC should be considered for this histologic variant.
- Published
- 2022
- Full Text
- View/download PDF
32. Clinical utility of subclassifying positive surgical margins at radical prostatectomy.
- Author
-
Dason S, Vertosick EA, Udo K, Sjoberg DD, Vickers AJ, Al-Ahmadie H, Chen YB, Gopalan A, Joseph Sirintrapun S, Tickoo SK, Scardino PT, Eastham JA, Reuter VE, and Fine SW
- Subjects
- Humans, Male, Neoplasm Grading, Neoplasm Recurrence, Local pathology, Prostate pathology, Prostate-Specific Antigen, Prostatectomy, Margins of Excision, Prostatic Neoplasms pathology, Prostatic Neoplasms surgery
- Abstract
Objective: To determine whether subclassification of positive surgical margins (PSMs) increases predictive ability for biochemical recurrence (BCR) and aids clinical decision-making in patients undergoing radical prostatectomy., Patients and Methods: We studied 2147 patients with pT2 and pT3a prostate cancer with detailed surgical margin parameters and BCR status. We compared a base model, a linear predictor calculated from the Memorial Sloan Kettering Cancer Center postoperative nomogram (prostate-specific antigen, pathological tumour grade and stage), with the addition of surgical margin status to five additional models (base model plus surgical margin subclassifications) to evaluate enhancement in predictive accuracy. Decision curve analysis (DCA) was performed to determine the clinical utility of parameters that enhanced predictive accuracy., Results: Among 2147 men, 205 had PSMs, and 231 developed BCR. Discrimination for the base model with addition of surgical margin status was high (c-index = 0.801) and not meaningfully improved by adding surgical margin subclassification in the full cohort. In analyses considering only men with PSMs (N = 55 with BCR), adding surgical margin subclassification to the base model increased discrimination for total length of all PSMs - alone or with maximum Gleason grade at the margin (c-index improvement = 0.717 to 0.752 and 0.753, respectively). DCA demonstrated a modest benefit to clinical utility with the addition of these parameters., Conclusions: Specific subclassification parameters add predictive accuracy for BCR and may aid clinical utility in decision-making for patients with PSMs. These findings may be useful for patient counselling and future adjuvant therapy trial design., (© 2021 The Authors BJU International © 2021 BJU International.)
- Published
- 2022
- Full Text
- View/download PDF
33. Developmental chromatin programs determine oncogenic competence in melanoma.
- Author
-
Baggiolini A, Callahan SJ, Montal E, Weiss JM, Trieu T, Tagore MM, Tischfield SE, Walsh RM, Suresh S, Fan Y, Campbell NR, Perlee SC, Saurat N, Hunter MV, Simon-Vermot T, Huang TH, Ma Y, Hollmann T, Tickoo SK, Taylor BS, Khurana E, Koche RP, Studer L, and White RM
- Subjects
- ATPases Associated with Diverse Cellular Activities genetics, ATPases Associated with Diverse Cellular Activities metabolism, Animals, Animals, Genetically Modified, Chromatin genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, Humans, Melanocytes metabolism, Melanocytes pathology, Mice, Neoplasms, Experimental, Neoplastic Stem Cells pathology, Neural Crest metabolism, Pluripotent Stem Cells pathology, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf metabolism, SOXE Transcription Factors genetics, SOXE Transcription Factors metabolism, Transcription, Genetic, Zebrafish, Carcinogenesis genetics, Carcinogenesis pathology, Chromatin metabolism, Melanoma genetics, Melanoma pathology, Neural Crest pathology
- Abstract
Oncogenes only transform cells under certain cellular contexts, a phenomenon called oncogenic competence. Using a combination of a human pluripotent stem cell–derived cancer model along with zebrafish transgenesis, we demonstrate that the transforming ability of BRAF
V600E along with additional mutations depends on the intrinsic transcriptional program present in the cell of origin. In both systems, melanocytes are less responsive to mutations, whereas both neural crest and melanoblast populations are readily transformed. Profiling reveals that progenitors have higher expression of chromatin-modifying enzymes such as ATAD2, a melanoma competence factor that forms a complex with SOX10 and allows for expression of downstream oncogenic and neural crest programs. These data suggest that oncogenic competence is mediated by regulation of developmental chromatin factors, which then allow for proper response to those oncogenes.- Published
- 2021
- Full Text
- View/download PDF
34. TRIM63 is a sensitive and specific biomarker for MiT family aberration-associated renal cell carcinoma.
- Author
-
Wang XM, Zhang Y, Mannan R, Skala SL, Rangaswamy R, Chinnaiyan A, Su F, Cao X, Zelenka-Wang S, McMurry L, Xiao H, Spratt DE, Sangoi AR, Shao L, Betz BL, Brown N, Tickoo SK, McKenney JK, Argani P, Gupta S, Reuter VE, Chinnaiyan AM, Dhanasekaran SM, and Mehra R
- Subjects
- Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, Humans, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Microphthalmia-Associated Transcription Factor genetics, Muscle Proteins analysis, Oncogene Fusion, Sensitivity and Specificity, Translocation, Genetic, Tripartite Motif Proteins analysis, Ubiquitin-Protein Ligases analysis, Biomarkers, Tumor analysis, Carcinoma, Renal Cell diagnosis, Kidney Neoplasms diagnosis, Muscle Proteins metabolism, Tripartite Motif Proteins metabolism, Ubiquitin-Protein Ligases metabolism
- Abstract
Microphthalmia-associated transcription factor (MiT) family aberration-associated renal cell carcinoma (MiTF-RCC) is a subtype of renal cell carcinoma harboring recurrent chromosomal rearrangements involving TFE3 or TFEB genes. MiTF-RCC is morphologically diverse, can histologically resemble common RCC subtypes like clear cell RCC and papillary RCC, and often poses a diagnostic challenge in genitourinary clinical and pathology practice. To characterize the MiTF-RCC at the molecular level and identify biomarker signatures associated with MiTF-RCC, we analyzed RNAseq data from MiTF-RCC, other RCC subtypes and benign kidney. Upon identifying TRIM63 as a cancer-specific biomarker in MiTF-RCC, we evaluated its expression independently by RNA in situ hybridization (RNA-ISH) in whole tissue sections from 177 RCC cases. We specifically included 31 cytogenetically confirmed MiTF-RCC cases and 70 RCC cases suspicious for MiTF-RCC in terms of clinical and morphological features, to evaluate and compare TRIM63 RNA-ISH results with the results from TFE3/TFEB fluorescence in situ hybridization (FISH), which is the current clinical standard. We confirmed that TRIM63 mRNA was highly expressed in all classes of MiTF-RCC compared to other renal tumor categories, where it was mostly absent to low. While the TRIM63 RNA-ISH and TFE3/TFEB FISH results were largely concordant, importantly, TRIM63 RNA-ISH was strongly positive in TFE3 FISH false-negative cases with RBM10-TFE3 inversion. In conclusion, TRIM63 can serve as a diagnostic marker to distinguish MiTF-RCC from other renal tumor subtypes with overlapping morphology. We suggest a combination of TFE3/TFEB FISH and TRIM63 RNA-ISH assays to improve the accuracy and efficiency of MiTF-RCC diagnosis. Accurate diagnosis of MiTF-RCC and other RCC subtypes would enable effective targeted therapy and avoid poor therapeutic response due to tumor misclassification., (© 2021. The Author(s), under exclusive licence to United States & Canadian Academy of Pathology.)
- Published
- 2021
- Full Text
- View/download PDF
35. The Clinicopathologic and Molecular Landscape of Clear Cell Papillary Renal Cell Carcinoma: Implications in Diagnosis and Management.
- Author
-
Weng S, DiNatale RG, Silagy A, Mano R, Attalla K, Kashani M, Weiss K, Benfante NE, Winer AG, Coleman JA, Reuter VE, Russo P, Reznik E, Tickoo SK, and Hakimi AA
- Subjects
- Biopsy, Female, Humans, Retrospective Studies, Carcinoma, Renal Cell epidemiology, Carcinoma, Renal Cell genetics, Kidney Neoplasms genetics, Kidney Neoplasms therapy
- Abstract
Background: Clear cell papillary renal cell carcinoma (CCPRCC) is a recently described tumor entity. Several questions remain about its epidemiology, molecular features, and clinical behavior., Objective: To comprehensively evaluate clinicopathologic and molecular features of CCPRCC, and compare it with more common kidney cancer subtypes., Design, Setting, and Participants: We identified 89 CCPRCC patients and compared their clinicopathologic features with 1120 localized clear cell renal cell carcinoma (ccRCC) and 129 type 1 papillary renal cell carcinoma (pRCC) patients., Outcome Measurements and Statistical Analysis: Nonparametric statistical testing was used to compare relevant features between tumor types. Overall, cancer-specific survival (CSS) and metastasis-free survival estimates were calculated from initial diagnosis using the Kaplan-Meier method. Patients with ipsilateral multifocal disease were explored further. A subset of CCPRCC tumors underwent genomic analysis and were compared with other RCC subtypes., Results and Limitations: A higher proportion of female (45% vs 32%) and African-American (19% vs 3%) patients were observed in the CCPRCC cohort than in the ccRCC and pRCC cohorts. CCPRCC tumors also had increased odds of presenting with additional ipsilateral masses (odds ratio [OR]: 4.41 [confidence interval {CI}: 2.34, 8.15], p < 0.001) and bilateral disease (OR: 4.80 [CI: 2.40, 9.59], p < 0.001) compared with ccRCC tumors. On molecular analysis, CCPRCC tumors showed fewer somatic aberrations and a greater degree of mitochondrial DNA depletion. In multifocal CCPRCC tumors, histologic concordance among the different renal cell carcinoma masses was estimated at 44% (7/16), and none of the individuals presenting exclusively with CCPRCC tumors developed metastatic disease after 5 yr. In contrast, multifocal tumors with CCPRCC and other nonconcordant histologies were more likely to experience adverse outcomes (CSS, log rank p = 0.034)., Conclusions: CCPRCC is characterized by distinct molecular and epidemiologic features that could be used to refine current diagnostic approaches. Although their clinical course is generally indolent, multifocal CCPRCC tumors represent a unique diagnostic challenge. In this context, single-mass biopsies could miss concomitant aggressive disease, with a potential negative impact on patient outcomes. Furthermore, high discordance rates in multifocal CCPRCC tumors have important clinical implications in management., Patient Summary: We explored the molecular and clinical features of clear cell papillary renal cell carcinoma (CCPRCC) relative to other kidney cancer subtypes. While CCPRCC generally conveys a good prognosis, additional caution should be taken when it is diagnosed using biopsy if multiple kidney masses are present., (Copyright © 2020. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
36. Putative Drivers of Aggressiveness in TCEB1-mutant Renal Cell Carcinoma: An Emerging Entity with Variable Clinical Course.
- Author
-
DiNatale RG, Gorelick AN, Makarov V, Blum KA, Silagy AW, Freeman B, Chowell D, Marcon J, Mano R, Sanchez A, Attalla K, Weng S, Voss M, Motzer RJ, Russo P, Coleman JA, Reuter VE, Chen YB, Chan TA, Reznik E, Tickoo SK, and Hakimi AA
- Subjects
- Biomarkers, Tumor genetics, Carcinoma, Renal Cell pathology, DNA Copy Number Variations, Genomics, Humans, Kidney Neoplasms pathology, Sequence Analysis, DNA, Carcinoma, Renal Cell genetics, Kidney Neoplasms genetics
- Abstract
Background: TCEB1-mutant renal cell carcinoma (RCC) is a rare variant of RCC with clear-cell features. Owing to its unique morphological and molecular features it has recently been proposed as a separate entity. Initial series suggested an indolent, early-stage phenotype. Here we expand our clinical cohort and describe a more detailed genomic analysis looking for potential drivers of aggressiveness., Design, Setting, and Participants: We identified five new cases in our institutional sequencing cohort, four of whom were found to have high-stage disease (American Joint Committee on Cancer stage III/IV). Twelve previously reported cases were pooled for comparison purposes (Sato, The Cancer Genome Atlas, TRACERx Renal)., Outcome Measures and Statistical Analysis: We used our previously validated pipeline to analyze somatic mutations and copy number alterations (CNAs) in seven tumor samples with available data and estimated the number of cancer cells bearing each somatic mutation. The oncogenic potential of mutations was assessed using OncoKB and two other algorithms. Mann-Whitney U tests were used to evaluate differences in genomic markers between stage groups., Results and Limitations: All tumors showed biallelic inactivation of the TCEB1 gene according to a combination of somatic mutation and CNA analyses. Mutations were always found in residues involved in hydrophobic interactions with VHL. We found that high-stage tumors had additional oncogenic mutations (median 1, interquartile range [IQR] 1-1 vs 2, IQR 2-2; median difference 1, 95% confidence interval [CI] 1-1; p= 0.002) and showed whole-genome doubling events. They also seemed to have a higher burden of somatic CNAs (median fraction CNA genome 0.10, IQR 0.10-0.15 vs 0.63, IQR 0.58-0.68), however, this finding did not reach statistical significance (median difference 0.49, 95% CI 0.33-0.63; p=0.052)., Conclusions: TCEB1-mutant RCC can show variable behavior ranging from very indolent to aggressive. Specific molecular events leading to high genomic instability seem to be associated with aggressiveness. This study expands the clinical spectrum of TCEB1-mutant RCC., Patient Summary: We present four cases of aggressive TCEB1-mutant renal cell carcinoma, a rare type of kidney cancer. In-depth analysis of the genomes of these tumors revealed certain abnormalities that might explain this aggressive behavior., (Copyright © 2019 European Association of Urology. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
37. Adverse histology, homozygous loss of CDKN2A/B, and complex genomic alterations in locally advanced/metastatic renal mucinous tubular and spindle cell carcinoma.
- Author
-
Yang C, Cimera RS, Aryeequaye R, Jayakumaran G, Sarungbam J, Al-Ahmadie HA, Gopalan A, Sirintrapun SJ, Fine SW, Tickoo SK, Epstein JI, Reuter VE, Zhang Y, and Chen YB
- Subjects
- Aged, Female, Humans, Male, Middle Aged, Retrospective Studies, Adenocarcinoma, Mucinous genetics, Adenocarcinoma, Mucinous pathology, Cyclin-Dependent Kinase Inhibitor p15 genetics, Cyclin-Dependent Kinase Inhibitor p16 genetics, Kidney Neoplasms genetics, Kidney Neoplasms pathology
- Abstract
Mucinous tubular and spindle cell carcinoma (MTSCC) is a rare subtype of renal cell carcinoma with characteristic histologic features and chromosomal alterations. Although typically indolent, a small subset of cases has been reported to exhibit aggressive clinical behavior. We retrospectively identified 33 patients with MTSCC, consisting of 10 cases of locally advanced/metastatic MTSCC (pT3 or N1 or M1) and 23 kidney-confined MTSCC (pT1/T2) without disease recurrence or progression. Utilizing a single-nucleotide polymorphism array and a targeted next-generation sequencing platform, we examined genome-wide molecular alterations in 24 cases, including 11 available samples from 8 patients with locally advanced/metastatic MTSCC. Ten patients with locally advanced/metastatic MTSCC were 8 females (80%) and 2 males (20%). At nephrectomy, 7 of these 10 cases (70%) were pT3 or pN1 while the remaining 3 (30%) were pT1/T2. Eight patients (80%) developed metastases and common sites included lymph node (4, 40%), bone (4, 40%), and retroperitoneum (3, 30%). Four patients died of disease (40%) during follow-up. Locally advanced/metastatic MTSCCs shared typical MTSCC genomic profiles with loss of chromosomes 1, 4, 6, 8, 9, 13, 14, 15, and 22, while some exhibited additional complex genomic alterations, most frequently a relative gain of 1q (7/8). Homozygous loss of CDKN2A/B was observed in 3 (38%) locally advanced/metastatic MTSCCs. Tumor necrosis, solid nested/sheet pattern, irregular trabecular/single-file infiltration in a desmoplastic stroma, lymphovascular space invasion, and increased mitotic activity were associated with locally advanced/metastatic MTSCCs (all p < 0.05). Our findings reveal that MTSCCs with aggressive clinical behavior have progressed through clonal evolution; CDKN2A/B deletion and additional complex genomic abnormalities may contribute to this process. Recognizing the morphologic presentation of high-grade MTSCC and evaluating adverse histologic features seen in these tumors can help establish a definitive diagnosis and stratify patients for treatment and prognostication.
- Published
- 2021
- Full Text
- View/download PDF
38. CD274 ( PD-L1 ) Copy Number Changes (Gain) & Response to Immune Checkpoint Blockade Therapy in Carcinomas of the Urinary Tract.
- Author
-
Gupta S, Vanderbilt CM, Zhang Y, Tickoo SK, Fine SW, Gopalan A, Chen YB, Sirintrapun SJ, Teo MY, Funt SA, Iyer G, Rosenberg JE, Bajorin DF, Bochner BH, Pietzak EJ, Ross DS, Ladanyi M, Cheville JC, Solit DB, Reuter VE, and Al-Ahmadie HA
- 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-L1 copy 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-L1 amplification and 9 tumors with PD-L1 deletion. PD-L1 protein expression was the highest in PD-L1 amplified tumors. Of the 9 patients whose tumors harbored PD-L1 amplification, 6 received immunotherapy with 4 deriving clinical benefit, and two achieving durable response. Of the 9 patients whose tumors had PD-L1 copy number losses, 4 received immunotherapy with 3 experiencing disease progression., Conclusions: PD-L1 copy 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
39. Everolimus plus bevacizumab is an effective first-line treatment for patients with advanced papillary variant renal cell carcinoma: Final results from a phase II trial.
- Author
-
Feldman DR, Ged Y, Lee CH, Knezevic A, Molina AM, Chen YB, Chaim J, Coskey DT, Murray S, Tickoo SK, Reuter VE, Patil S, Xiao H, Aghalar J, Apollo AJ, Carlo MI, Motzer RJ, and Voss MH
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bevacizumab therapeutic use, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, DNA-Binding Proteins genetics, Everolimus therapeutic use, Female, Fumarate Hydratase genetics, High-Throughput Nucleotide Sequencing, Humans, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Male, Middle Aged, Mutation, Proto-Oncogene Proteins c-met genetics, Sequence Analysis, DNA, Survival Analysis, Transcription Factors genetics, Treatment Outcome, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Bevacizumab administration & dosage, Carcinoma, Renal Cell drug therapy, Everolimus administration & dosage, Kidney Neoplasms drug therapy
- Abstract
Background: We previously reported on a phase 2 study of everolimus plus bevacizumab across various nonclear cell renal cell carcinoma (nccRCC) histologies and observed encouraging activity among patients with papillary RCC (pRCC) and unclassified RCC (uRCC) with a major papillary component. We subsequently expanded the study to enroll additional patients with pRCC variants., Methods: Everolimus plus bevacizumab was administered at standard doses until disease progression or intolerance to therapy. The primary endpoint was the 6-month progression-free survival (PFS) rate; secondary endpoints included objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and safety. Correlative analyses included next-generation sequencing (NGS) from tumor and germline across >341 genes of interest., Results: In addition to 19 patients with pRCC variants in the original cohort, 20 patients with similar features were enrolled on the expansion cohort (uRCC with papillary features [n = 24], pRCC [n = 14], and translocation-associated RCC with papillary features [n = 1]). Among 37 evaluable patients, the 6-month PFS rate was 78%, the median PFS was 13.7 months (95% CI, 10.8-16.4 months), and the ORR was 35%. With a median follow-up of 17.6 months, the median OS was 33.9 months (95% CI, 23.3-71.9). Tolerance was consistent with prior reports for everolimus plus bevacizumab. NGS results (n = 33) identified responses in patients with a wide spectrum of genomic alterations, including ARID1A, FH, and MET mutations., Conclusion: The expansion cohort results confirm robust activity of everolimus plus bevacizumab in metastatic pRCC variants, supporting this regimen as a standard option for this patient population., (© 2020 American Cancer Society.)
- Published
- 2020
- Full Text
- View/download PDF
40. Sarcomatoid renal cell carcinoma: biology, natural history and management.
- Author
-
Blum KA, Gupta S, Tickoo SK, Chan TA, Russo P, Motzer RJ, Karam JA, and Hakimi AA
- Subjects
- B7-H1 Antigen metabolism, Carcinoma, Renal Cell metabolism, Carcinoma, Renal Cell therapy, Disease Management, Humans, Kidney Neoplasms metabolism, Kidney Neoplasms therapy, Lymphocytes, Tumor-Infiltrating, Prognosis, Programmed Cell Death 1 Receptor metabolism, Tumor Microenvironment, Carcinoma, Renal Cell pathology, Epithelial-Mesenchymal Transition, Immune Checkpoint Inhibitors therapeutic use, Kidney Neoplasms pathology, Nephrectomy
- Abstract
Sarcomatoid dedifferentiation is an uncommon feature that can occur in most histological subtypes of renal cell carcinomas (RCCs) and carries a decidedly poor prognosis. Historically, conventional treatments for sarcomatoid RCCs (sRCCs) have shown little efficacy, and median survival is commonly 6-13 months. Despite being first described in 1968, the mechanisms driving sarcomatoid dedifferentiation remain poorly understood, and information and treatment options available to physicians and patients are limited. When diagnosed at an early stage, surgical intervention remains the treatment of choice. However, preoperative identification through routine imaging or biopsy is unreliable and most patients present with advanced disease and systemic symptoms. For these patients, the role of cytoreductive nephrectomy is disputed. The expansion of immunotherapies approved for RCCs has generated a search for biomarkers that might be indicative of treatment response in sRCCs, although a proven effective systemic agent remains elusive. PDL1 expression is increased in sarcomatoid dedifferentiated renal tumours, which suggests that patients with sRCCs could benefit from PD1 and/or PDL1 immune checkpoint blockade therapy. Treatment outcomes for sarcomatoid tumours have remained relatively consistent compared with other RCCs, but further investigation of the tumour-immune cell microenvironment might yield insights into further therapeutic possibilities.
- Published
- 2020
- Full Text
- View/download PDF
41. Germ cell tumors and associated hematologic malignancies evolve from a common shared precursor.
- Author
-
Taylor J, Donoghue MT, Ho C, Petrova-Drus K, Al-Ahmadie HA, Funt SA, Zhang Y, Aypar U, Rao P, Chavan SS, Haddadin M, Tamari R, Giralt S, Tallman MS, Rampal RK, Baez P, Kappagantula R, Kosuri S, Dogan A, Tickoo SK, Reuter VE, Bosl GJ, Iacobuzio-Donahue CA, Solit DB, Taylor BS, Feldman DR, and Abdel-Wahab O
- Subjects
- Adolescent, Adult, Female, Humans, Middle Aged, Hematologic Neoplasms genetics, Hematologic Neoplasms metabolism, Hematologic Neoplasms pathology, Mutation, Neoplasms, Germ Cell and Embryonal genetics, Neoplasms, Germ Cell and Embryonal metabolism, Neoplasms, Germ Cell and Embryonal pathology, Neoplasms, Second Primary genetics, Neoplasms, Second Primary metabolism, Neoplasms, Second Primary pathology, Signal Transduction genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism
- Abstract
Germ cell tumors (GCTs) are the most common cancer in men between the ages of 15 and 40. Although most patients are cured, those with disease arising in the mediastinum have distinctly poor outcomes. One in every 17 patients with primary mediastinal nonseminomatous GCTs develop an incurable hematologic malignancy and prior data intriguingly suggest a clonal relationship exists between hematologic malignancies and GCTs in these cases. To date, however, the precise clonal relationship between GCTs and the diverse additional somatic malignancies arising in such individuals have not been determined. Here, we traced the clonal evolution and characterized the genetic features of each neoplasm from a cohort of 15 patients with GCTs and associated hematologic malignancies. We discovered that GCTs and hematologic malignancies developing in such individuals evolved from a common shared precursor, nearly all of which harbored allelically imbalanced p53 and/or RAS pathway mutations. Hematologic malignancies arising in this setting genetically resembled mediastinal GCTs rather than de novo myeloid neoplasms. Our findings argue that this scenario represents a unique clinical syndrome, distinct from de novo GCTs or hematologic malignancies, initiated by an ancestral precursor that gives rise to the parallel evolution of GCTs and blood cancers in these patients.
- Published
- 2020
- Full Text
- View/download PDF
42. RAS/MAPK Pathway Driver Alterations Are Significantly Associated With Oncogenic KIT Mutations in Germ-cell Tumors.
- Author
-
Mata DA, Yang SR, Ferguson DC, Liu Y, Sharma R, Benhamida JK, Al-Ahmadie HA, Chakravarty D, Solit DB, Tickoo SK, Gupta S, Arcila ME, Ladanyi M, Feldman DR, Reuter VE, and Vanderbilt CM
- Subjects
- Adolescent, Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Child, DNA Mutational Analysis statistics & numerical data, Female, Follow-Up Studies, Humans, MAP Kinase Kinase 1 antagonists & inhibitors, MAP Kinase Kinase 1 metabolism, MAP Kinase Signaling System drug effects, Male, Middle Aged, Mutation, Neoplasms, Germ Cell and Embryonal drug therapy, Neoplasms, Germ Cell and Embryonal pathology, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Proto-Oncogene Proteins c-kit antagonists & inhibitors, Retrospective Studies, Seminoma drug therapy, Seminoma pathology, Testicular Neoplasms drug therapy, Testicular Neoplasms pathology, Young Adult, ras Proteins metabolism, MAP Kinase Signaling System genetics, Neoplasms, Germ Cell and Embryonal genetics, Proto-Oncogene Proteins c-kit genetics, Seminoma genetics, Testicular Neoplasms genetics
- Abstract
Objective: To report the mutational profile and clinical outcomes of a cohort of patients with KIT-mutant seminomas and nonseminomatous germ-cell tumors (SGCT/NSGCTs)., Patients and Methods: Retrospective cohort study of all patients with KIT-mutant GCTs sequenced at Memorial Sloan Kettering between March 2014 and March 2020. Tumors were assessed with MSK-IMPACT, a DNA next-generation sequencing assay for targeted sequencing of up to 468 key cancer genes., Results: Among 568 patients with GCTs, 8.1% had somatic KIT mutations, including 28 seminomas and 18 mixed/NSGCTs. Exons 17 (67.3%), 11 (22.4%), and 13 (6.1%) were most commonly affected. KIT-mutant cases were enriched for oncogenic RAS/MAPK pathway alterations compared to KIT-wildtype cases (34.8% vs 19.2%, P = .02). Among KIT-mutant cases, concurrent mutations were noted in KRAS (21.7%), RRAS2 (11.8%), CBL (6.5%), NRAS (4.3%), MAP2K1 (2.2%), and RAC1 (2.2%). Mutations in KRAS, RRAS2, and NRAS were mutually exclusive. In all, 73.9% of patients developed metastases and 95.7% received chemotherapy. No patients received KIT-directed tyrosine kinase inhibitors (TKIs). Classification as a NSGCT rather than a SGCT was associated with an increased risk of death (hazard ratio 9.1, 95% confidence interval 1.1-78.4, P = .04) while the presence of a concurrent RAS/MAPK pathway alteration was not (hazard ratio 0.8, 95% confidence interval 0.1-4.3, P = .76)., Conclusion: Mitogenic driver alterations can co-occur with activating KIT mutations, which may explain the lack of efficacy of KIT-directed TKIs in prior trials. Novel KIT-directed TKIs that target exon 17 mutations may benefit chemotherapy-refractory patients with KIT-mutant GCTs without RAS/MAPK alterations. Dual MEK/KIT inhibitor therapy in KIT-mutant GCTs with concurrent RAS/MAPK alterations could also be a plausible therapeutic strategy., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
43. Immunohistochemistry-based assessment of androgen receptor status and the AR-null phenotype in metastatic castrate resistant prostate cancer.
- Author
-
Gupta S, Vanderbilt C, Abida W, Fine SW, Tickoo SK, Al-Ahmadie HA, Chen YB, Sirintrapun SJ, Chadalavada K, Nanjangud GJ, Bialik A, Morris MJ, Scher HI, Ladanyi M, Reuter VE, and Gopalan A
- Subjects
- Aged, Androgen Receptor Antagonists therapeutic use, Antineoplastic Agents, Hormonal therapeutic use, Biomarkers, Tumor genetics, Biopsy, DNA Copy Number Variations, Drug Resistance, Neoplasm genetics, Gene Amplification, Humans, Immunohistochemistry, Male, Middle Aged, Prostate pathology, Prostatic Neoplasms, Castration-Resistant genetics, Prostatic Neoplasms, Castration-Resistant pathology, Receptors, Androgen genetics, Retinoblastoma Binding Proteins analysis, Retinoblastoma Binding Proteins genetics, Tumor Suppressor Protein p53 analysis, Tumor Suppressor Protein p53 genetics, Ubiquitin-Protein Ligases analysis, Ubiquitin-Protein Ligases genetics, Androgen Receptor Antagonists pharmacology, Antineoplastic Agents, Hormonal pharmacology, Biomarkers, Tumor analysis, Prostatic Neoplasms, Castration-Resistant drug therapy, Receptors, Androgen analysis
- 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 TP53 and RB1 alterations. 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 AR copy 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 AR amplification and showed positive nuclear expression of AR by immunohistochemistry. Nine cases lacked AR copy 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 PTEN genes 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
44. Inverted urothelial papilloma and urothelial carcinoma with inverted growth are histologically and molecularly distinct entities.
- Author
-
Almassi N, Pietzak EJ, Sarungbam J, Tickoo SK, Reuter VE, Solit DB, and Al-Ahmadie HA
- Subjects
- Female, Gene Expression genetics, Humans, Male, Carcinoma, Transitional Cell pathology, Papilloma, Inverted pathology, Urinary Bladder Neoplasms pathology
- Published
- 2020
- Full Text
- View/download PDF
45. Morphologic and Immunohistochemical Assessment of CDH1 Loss of Function Alterations in Prostatic Adenocarcinoma.
- Author
-
Gupta S, Fine SW, Chang J, Tickoo SK, Chen YB, Al-Ahmadie HA, Sirintrapun SJ, Abida W, Ladanyi M, Reuter VE, and Gopalan A
- Subjects
- Adenocarcinoma metabolism, Aged, Antigens, CD metabolism, Biomarkers, Tumor metabolism, Cadherins metabolism, Humans, Immunohistochemistry, Male, Middle Aged, Neoplasm Grading, Prostatic Neoplasms metabolism, Adenocarcinoma genetics, Adenocarcinoma pathology, Antigens, CD genetics, Biomarkers, Tumor genetics, Cadherins genetics, Loss of Function Mutation, Prostatic Neoplasms genetics, Prostatic Neoplasms pathology
- Published
- 2020
- Full Text
- View/download PDF
46. Practice Patterns in Reporting Tertiary Grades at Radical Prostatectomy: Survey of a Large Group of Experienced Urologic Pathologists.
- Author
-
Fine SW, Meisels DL, Vickers AJ, Al-Ahmadie H, Chen YB, Gopalan A, Sirintrapun SJ, Tickoo SK, and Reuter VE
- Subjects
- Humans, Male, Neoplasm Grading, Neoplasm Recurrence, Local, Pathologists standards, Prostate pathology, Prostate surgery, Prostatectomy methods, Prostatic Neoplasms surgery, Urologic Neoplasms diagnosis, Urologic Neoplasms therapy, Pathologists statistics & numerical data, Practice Patterns, Physicians' statistics & numerical data, Prostatectomy statistics & numerical data, Prostatic Neoplasms pathology, Research Report, Surveys and Questionnaires statistics & numerical data
- Abstract
Context.—: In prostate cancer, "tertiary" higher-grade patterns (TPs) have been associated with biochemical recurrence after radical prostatectomy., Objective.—: To determine variation regarding definition and application of TPs., Design.—: Online survey regarding TPs in a range of grading scenarios circulated to 105 experienced urologic pathologists., Results.—: Among 95 respondents, 40 of 95 (42%) defined TPs as "third most common pattern" and 55 (58%) as "minor pattern/less than 5% of tumor." In a tumor with pattern 3 and less than 5% pattern 4, of the 95 respondents, 35 (37%) assigned 3 + 3 = 6 with TP4, while 56 (59%) assigned 3 + 4 = 7. In a tumor with pattern 4 and less than 5% pattern 5, of the 95 respondents, 51 (54%) assigned 4 + 4 = 8 with TP5, while 43 (45%) assigned 4 + 5 = 9. Six scenarios were presented in which the order of most common patterns was 3, 4, and 5 (Group 1) or 4, 3, and 5 (Group 2) with varying percentages. In both groups, when pattern 5 was less than 5%, we found that 98% and 93% of respondents would assign 3 + 4 = 7 or 4 + 3 = 7 with TP5. In scenarios with 15% or 25% pattern 5, most respondents (70% and 80%, respectively) would include pattern 5 as the secondary grade, that is, 3 + 5 = 8 (Group 1) or 4 + 5 = 9 (Group 2). For 85 of 95 (89%), a TP would not impact Grade Group assignment., Conclusions.—: This survey highlights substantial variation in practice patterns regarding definition and application of "tertiary" grading in radical prostatectomy specimens. High consistency was observed in 3 + 4 = 7/4 + 3 = 7 scenarios with truly minor pattern 5. These findings should inform future studies assessing the standardization and predictive value of "tertiary" patterns.
- Published
- 2020
- Full Text
- View/download PDF
47. The Role of Cytoreductive Nephrectomy for Sarcomatoid Renal Cell Carcinoma: A 29-Year Institutional Experience.
- Author
-
Silagy AW, Mano R, Blum KA, DiNatale RG, Marcon J, Tickoo SK, Reznik E, Coleman JA, Russo P, and Hakimi AA
- Subjects
- Aged, Cohort Studies, Female, Humans, Male, Middle Aged, Retrospective Studies, Time Factors, Treatment Outcome, Carcinoma, Renal Cell surgery, Cytoreduction Surgical Procedures, Kidney Neoplasms surgery, Nephrectomy methods
- Abstract
Objective: To assess which patients respond best following cytoreductive nephrectomy for renal cell carcinoma (RCC) with sarcomatoid dedifferentiation (sRCC) and whether outcomes are improving over time., Methods: We identified 562 patients with metastatic RCC treated between 1989 and 2018 with cytoreductive nephrectomy. We reviewed baseline clinical and pathologic characteristics, including the presence of sRCC, and metastatic sites at time of nephrectomy. The primary study endpoint was overall survival (OS). Univariate and multivariate Cox-regression analyses were used to identify significant predictors of OS., Results: The study cohort had 192 sRCC patients, with a median age of 59 years. Frequently involved metastatic locations were lung (n = 115), retroperitoneal nodes (n = 63), and axial skeleton (n = 43). Lung metastasis were more prevalent in clear cell histology (P = .0017) whereas nodal involvement was associated with nonclear cell subtypes (P = .0064). Median follow-up was 14 months. Estimated 2- and 5-year OS were 34.1% and 14.8%, respectively. On multivariate analysis, metastases to the liver (HR = 1.64; 95% CI 1.02-2.63; P = .04), lung (HR = 1.50; 95% CI 1.05-2.14; P = .03), retroperitoneal nodes (HR = 1.52; 95% CI 1.03-2.25; P = 0.04) and nonclear cell histology (HR = 1.61; 95% CI 1.10-2.35; P = .01) were associated with worse OS in the sRCC cohort., Conclusion: OS after cytoreductive nephrectomy for sRCC and non-sRCC is improving over time. In patients with sRCC, presentations with unifocal metastasis not involving the liver or lung, clear cell histology and node negative disease have better outcomes following cytoreductive nephrectomy and may yield greater benefit from the procedure., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
48. Molecular characterization of sarcomatoid clear cell renal cell carcinoma unveils new candidate oncogenic drivers.
- Author
-
Malouf GG, Flippot R, Dong Y, Dinatale RG, Chen YB, Su X, Compérat E, Rouprêt M, Mano R, Blum KA, Yao H, Mouawad R, Spano JP, Khayat D, Karam JA, Ho TH, Tickoo SK, Russo P, Hsieh JJ, Tannir NM, and Hakimi AA
- Subjects
- Adaptor Proteins, Signal Transducing genetics, Animals, Cell Nucleus genetics, Cell Nucleus metabolism, Cell Proliferation, Genetic Predisposition to Disease, Hippo Signaling Pathway, Humans, Laser Capture Microdissection, Male, Mice, Neoplasm Transplantation, Neurofibromin 2 genetics, Protein Serine-Threonine Kinases genetics, Sequence Analysis, DNA methods, Signal Transduction, Trans-Activators genetics, Transcription Factors genetics, Transcriptional Coactivator with PDZ-Binding Motif Proteins, Up-Regulation, YAP-Signaling Proteins, Carcinoma, Renal Cell genetics, Gene Regulatory Networks, Kidney Neoplasms genetics, Mutation
- Abstract
Sarcomatoid clear-cell renal cell carcinomas (sRCC) are associated with dismal prognosis. Genomic alterations associated with sarcomatoid dedifferentiation are poorly characterized. We sought to define the genomic landscape of sRCC and uncover potentially actionable therapeutic targets. We assessed the genomic landscape of sRCC using targeted panel sequencing including patients with microdissected sarcomatoid and epithelial components. Along with common genomic alterations associated with clear-cell histology, we found that Hippo was one of the most frequently altered pathways in these tumours. Hippo alterations were differentially enriched in sRCC compared to non-sRCC. Functional analysis showed that Hippo members mutations were associated with higher nuclear accumulation of YAP/TAZ, core effectors of the Hippo pathway. In a NF2-mutant sRCC model, YAP1 knockdown and NF2 reconstitution suppressed cell proliferation, tumour growth and invasion, both in vitro and in vivo. Overall, we show that Hippo pathway alterations are a feature of sRCC, and enable the exploration of the Hippo pathway as a novel potential therapeutic target.
- Published
- 2020
- Full Text
- View/download PDF
49. TFEB Expression Profiling in Renal Cell Carcinomas: Clinicopathologic Correlations.
- Author
-
Gupta S, Argani P, Jungbluth AA, Chen YB, Tickoo SK, Fine SW, Gopalan A, Al-Ahmadie HA, Sirintrapun SJ, Sanchez A, Hakimi AA, Mcfarlane T, Salazar PA, Williamson SR, Skala SL, Mehra R, Hes O, Antonescu CR, Ladanyi M, Arcila ME, and Reuter VE
- Subjects
- Adult, Aged, Aged, 80 and over, Carcinoma, Renal Cell diagnosis, Carcinoma, Renal Cell pathology, Child, Female, Follow-Up Studies, Gene Expression Profiling, Humans, Immunohistochemistry, In Situ Hybridization, Fluorescence, Kidney Neoplasms diagnosis, Kidney Neoplasms pathology, Male, Middle Aged, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Biomarkers, Tumor metabolism, Carcinoma, Renal Cell metabolism, Kidney Neoplasms metabolism
- Abstract
TFEB is overexpressed in TFEB-rearranged renal cell carcinomas as well as in renal tumors with amplifications of TFEB at 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 TFEB gene 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). TFEB status 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 TFEB mRNA 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 TFEB expression when normalized to B2M (mean: 168.9%, n=28), compared with non-TFEB-altered controls (mean: 7%, n=18, P=0.005). Interestingly, TFEB expression 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 TFEB expression (P=0.00004). Our results suggest that 6p21.1 amplified renal tumors show increased TFEB gene expression but not as much as t(6;11) renal tumors. These findings correlate with the less consistent/diffuse expression of downstream markers of TFEB activation (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 VEGFA and CCND3, or other genetic alterations.
- Published
- 2019
- Full Text
- View/download PDF
50. Chromophobe Renal Cell Carcinoma: Results From a Large Single-Institution Series.
- Author
-
Casuscelli J, Becerra MF, Seier K, Manley BJ, Benfante N, Redzematovic A, Stief CG, Hsieh JJ, Tickoo SK, Reuter VE, Coleman JA, Russo P, Ostrovnaya I, and Hakimi AA
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Carcinoma, Renal Cell surgery, Female, Follow-Up Studies, Humans, Kidney Neoplasms surgery, Male, Middle Aged, Nephrectomy, Prognosis, Retrospective Studies, Survival Rate, Young Adult, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology
- Abstract
Background: The purpose of the study was to evaluate clinical features and prognostic factors in a large single institutional cohort of chromophobe renal cell carcinoma (ChRCC) patients for identification of tumors with the highest metastatic potential., Patients and Methods: Clinicopathological parameters of all patients with ChRCC diagnosed and surgically treated at Memorial Sloan Kettering Cancer Center between 1990 and 2016 were identified and compared with patients treated for clear-cell renal cell carcinoma (ccRCC) in the same study period using Wilcoxon test for continuous variables and Fisher exact test for categorical variables. Recurrence-free survival (RFS) and overall survival (OS) were analyzed using the Kaplan-Meier method, log rank test, and Cox proportional hazards regression., Results: Four hundred ninety-six patients with ChRCC (10-year RFS, 91.7% and OS, 82.1%) and 3312 patients with ccRCC (10-year RFS, 79.4% and OS, 63.6%) were included in the analysis. Patients with ChRCC were younger (median 59 vs. 61 years; P = .0015), less frequently male (54.8% vs. 66.3%; P < .0001), showed more favorable T stages (T1-2 in 78% vs. 67%; P < .0001) and less frequent sarcomatoid differentiation (1.2 % vs. 4%; P = .0008) and showed lower rates of metastatic development compared with ccRCC patients. Larger tumor size, sarcomatoid differentiation, and higher T-stage are significantly associated with adverse RFS and OS in chromophobe tumors., Conclusion: ChRCC is more commonly diagnosed in female and younger patients and is associated with a more favorable clinical outcome and a lower propensity for metastatic development than ccRCC. Larger tumors and sarcomatoid differentiation of ChRCC might be considered as risk factors for metastatic development., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.