169 results on '"Chinnaiyan, Arul M"'
Search Results
2. Discovery of ZLC491 as a Potent, Selective, and Orally Bioavailable CDK12/13 PROTAC Degrader
- Author
-
Zhou, Licheng, Zhou, Kaijie, Chang, Yu, Yang, Jianzhang, Fan, Bohai, Su, Yuhan, Li, Zilu, Mannan, Rahul, Mahapatra, Somnath, Ding, Ming, Zhou, Fengtao, Huang, Weixue, Ren, Xiaomei, Xu, Jian, Wang, George Xiaoju, Zhang, Jinwei, Wang, Zhen, Chinnaiyan, Arul M., and Ding, Ke
- Abstract
Selective degradation of cyclin-dependent kinases 12 and 13 (CDK12/13) emerges as a new potential therapeutic approach for triple-negative breast cancer (TNBC) and other human cancers. While several proteolysis-targeting chimera (PROTAC) degraders of CDK12/13 were reported, none are orally bioavailable. Here, we report the discovery of ZLC491as a potent, selective, and orally bioavailable CDK12/13 PROTAC degrader. The compound effectively degraded CDK12 and CDK13 with DC50values of 32 and 28 nM, respectively, in TNBC MDA-MB-231 cells. Global proteomic assessment and mechanistic studies revealed that ZLC491selectively induced CDK12/13 degradation in a cereblon- and proteasome-dependent manner. Furthermore, the molecule efficiently suppressed transcription and expression of long genes, predominantly a subset of genes associated with DNA damage response, and significantly inhibited proliferation of multiple TNBC cell lines. Importantly, ZLC491achieved an oral bioavailability of 46.8% in rats and demonstrated potent in vivodegradative effects on CDK12/13 in an MDA-MB-231 xenografted mouse model.
- Published
- 2024
- Full Text
- View/download PDF
3. NSD2 is a requisite subunit of the AR/FOXA1 neo-enhanceosome in promoting prostate tumorigenesis
- Author
-
Parolia, Abhijit, Eyunni, Sanjana, Verma, Brijesh Kumar, Young, Eleanor, Liu, Yihan, Liu, Lianchao, George, James, Aras, Shweta, Das, Chandan Kanta, Mannan, Rahul, ur Rasool, Reyaz, Mitchell-Velasquez, Erick, Mahapatra, Somnath, Luo, Jie, Carson, Sandra E., Xiao, Lanbo, Gajjala, Prathibha R., Venkatesh, Sharan, Jaber, Mustapha, Wang, Xiaoju, He, Tongchen, Qiao, Yuanyuan, Pang, Matthew, Zhang, Yuping, Tien, Jean Ching-Yi, Louw, Micheala, Alhusayan, Mohammed, Cao, Xuhong, Su, Fengyun, Tavana, Omid, Hou, Caiyun, Wang, Zhen, Ding, Ke, Chinnaiyan, Arul M., and Asangani, Irfan A.
- Abstract
Androgen receptor (AR) is a ligand-responsive transcription factor that drives terminal differentiation of the prostatic luminal epithelia. By contrast, in tumors originating from these cells, AR chromatin occupancy is extensively reprogrammed to activate malignant phenotypes, the molecular mechanisms of which remain unknown. Here, we show that tumor-specific AR enhancers are critically reliant on H3K36 dimethyltransferase activity of NSD2. NSD2 expression is abnormally induced in prostate cancer, where its inactivation impairs AR transactivation potential by disrupting over 65% of its cistrome. NSD2-dependent AR sites distinctively harbor the chimeric FOXA1:AR half-motif, which exclusively comprise tumor-specific AR enhancer circuitries defined from patient specimens. NSD2 inactivation also engenders increased dependency on the NSD1 paralog, and a dual NSD1/2 PROTAC degrader is preferentially cytotoxic in AR-dependent prostate cancer models. Altogether, we characterize NSD2 as an essential AR neo-enhanceosome subunit that enables its oncogenic activity, and position NSD1/2 as viable co-targets in advanced prostate cancer.
- Published
- 2024
- Full Text
- View/download PDF
4. Development and Validation of an 18-Gene Urine Test for High-Grade Prostate Cancer
- Author
-
Tosoian, Jeffrey J., Zhang, Yuping, Xiao, Lanbo, Xie, Cassie, Samora, Nathan L., Niknafs, Yashar S., Chopra, Zoey, Siddiqui, Javed, Zheng, Heng, Herron, Grace, Vaishampayan, Neil, Robinson, Hunter S., Arivoli, Kumaran, Trock, Bruce J., Ross, Ashley E., Morgan, Todd M., Palapattu, Ganesh S., Salami, Simpa S., Kunju, Lakshmi P., Tomlins, Scott A., Sokoll, Lori J., Chan, Daniel W., Srivastava, Sudhir, Feng, Ziding, Sanda, Martin G., Zheng, Yingye, Wei, John T., and Chinnaiyan, Arul M.
- Abstract
IMPORTANCE: Benefits of prostate cancer (PCa) screening with prostate-specific antigen (PSA) alone are largely offset by excess negative biopsies and overdetection of indolent cancers resulting from the poor specificity of PSA for high-grade PCa (ie, grade group [GG] 2 or greater). OBJECTIVE: To develop a multiplex urinary panel for high-grade PCa and validate its external performance relative to current guideline-endorsed biomarkers. DESIGN, SETTING, AND PARTICIPANTS: RNA sequencing analysis of 58 724 genes identified 54 markers of PCa, including 17 markers uniquely overexpressed by high-grade cancers. Gene expression and clinical factors were modeled in a new urinary test for high-grade PCa (MyProstateScore 2.0 [MPS2]). Optimal models were developed in parallel without prostate volume (MPS2) and with prostate volume (MPS2+). The locked models underwent blinded external validation in a prospective National Cancer Institute trial cohort. Data were collected from January 2008 to December 2020, and data were analyzed from November 2022 to November 2023. EXPOSURE: Protocolized blood and urine collection and transrectal ultrasound-guided systematic prostate biopsy. MAIN OUTCOMES AND MEASURES: Multiple biomarker tests were assessed in the validation cohort, including serum PSA alone, the Prostate Cancer Prevention Trial risk calculator, and the Prostate Health Index (PHI) as well as derived multiplex 2-gene and 3-gene models, the original 2-gene MPS test, and the 18-gene MPS2 models. Under a testing approach with 95% sensitivity for PCa of GG 2 or greater, measures of diagnostic accuracy and clinical consequences of testing were calculated. Cancers of GG 3 or greater were assessed secondarily. RESULTS: Of 761 men included in the development cohort, the median (IQR) age was 63 (58-68) years, and the median (IQR) PSA level was 5.6 (4.6-7.2) ng/mL; of 743 men included in the validation cohort, the median (IQR) age was 62 (57-68) years, and the median (IQR) PSA level was 5.6 (4.1-8.0) ng/mL. In the validation cohort, 151 (20.3%) had high-grade PCa on biopsy. Area under the receiver operating characteristic curve values were 0.60 using PSA alone, 0.66 using the risk calculator, 0.77 using PHI, 0.76 using the derived multiplex 2-gene model, 0.72 using the derived multiplex 3-gene model, and 0.74 using the original MPS model compared with 0.81 using the MPS2 model and 0.82 using the MPS2+ model. At 95% sensitivity, the MPS2 model would have reduced unnecessary biopsies performed in the initial biopsy population (range for other tests, 15% to 30%; range for MPS2, 35% to 42%) and repeat biopsy population (range for other tests, 9% to 21%; range for MPS2, 46% to 51%). Across pertinent subgroups, the MPS2 models had negative predictive values of 95% to 99% for cancers of GG 2 or greater and of 99% for cancers of GG 3 or greater. CONCLUSIONS AND RELEVANCE: In this study, a new 18-gene PCa test had higher diagnostic accuracy for high-grade PCa relative to existing biomarker tests. Clinically, use of this test would have meaningfully reduced unnecessary biopsies performed while maintaining highly sensitive detection of high-grade cancers. These data support use of this new PCa biomarker test in patients with elevated PSA levels to reduce the potential harms of PCa screening while preserving its long-term benefits.
- Published
- 2024
- Full Text
- View/download PDF
5. Discovery of LLC0424 as a Potent and Selective in Vivo NSD2 PROTAC Degrader.
- Author
-
Liu, Lianchao, Parolia, Abhijit, Liu, Yihan, Hou, Caiyun, He, Tongchen, Qiao, Yuanyuan, Eyunni, Sanjana, Luo, Jie, Li, Chungen, Wang, Yongxing, Zhou, Fengtao, Huang, Weixue, Ren, Xiaomei, Wang, Zhen, Chinnaiyan, Arul M., and Ding, Ke
- Published
- 2024
- Full Text
- View/download PDF
6. Discovery of CBPD-409 as a Highly Potent, Selective, and Orally Efficacious CBP/p300 PROTAC Degrader for the Treatment of Advanced Prostate Cancer.
- Author
-
Chen, Zhixiang, Wang, Mi, Wu, Dimin, Zhao, Lijie, Metwally, Hoda, Jiang, Wei, Wang, Yu, Bai, Longchuan, McEachern, Donna, Luo, Jie, Wang, Meilin, Li, Qiuxia, Matvekas, Aleksas, Wen, Bo, Sun, Duxin, Chinnaiyan, Arul M., and Wang, Shaomeng
- Published
- 2024
- Full Text
- View/download PDF
7. Discovery of LLC0424 as a Potent and Selective in VivoNSD2 PROTAC Degrader
- Author
-
Liu, Lianchao, Parolia, Abhijit, Liu, Yihan, Hou, Caiyun, He, Tongchen, Qiao, Yuanyuan, Eyunni, Sanjana, Luo, Jie, Li, Chungen, Wang, Yongxing, Zhou, Fengtao, Huang, Weixue, Ren, Xiaomei, Wang, Zhen, Chinnaiyan, Arul M., and Ding, Ke
- Abstract
Nuclear receptor-binding SET domain-containing 2 (NSD2), a methyltransferase that primarily installs the dimethyl mark on lysine 36 of histone 3 (H3K36me2), has been recognized as a promising therapeutic target against cancer. However, existing NSD2 inhibitors suffer from low activity or inferior selectivity, and none of them can simultaneously remove the methyltransferase activity and chromatin binding function of NSD2. Herein we report the discovery of a novel NSD2 degrader LLC0424by leveraging the proteolysis-targeting chimera technology. LLC0424potently degraded NSD2 protein with a DC50value of 20 nM and a Dmaxvalue of 96% in acute lymphoblastic leukemia (ALL) RPMI-8402 cells. Mechanistic studies revealed LLC0424to selectively induce NSD2 degradation in a cereblon- and proteasome-dependent fashion. LLC0424also caused continuous downregulation of H3K36me2 and growth inhibition of ALL cell lines with NSD2 mutation. Importantly, intravenous or intraperitoneal injection of LLC0424showed potent NSD2 degradation in vivo.
- Published
- 2024
- Full Text
- View/download PDF
8. Discovery of CBPD-409 as a Highly Potent, Selective, and Orally Efficacious CBP/p300 PROTAC Degrader for the Treatment of Advanced Prostate Cancer
- Author
-
Chen, Zhixiang, Wang, Mi, Wu, Dimin, Zhao, Lijie, Metwally, Hoda, Jiang, Wei, Wang, Yu, Bai, Longchuan, McEachern, Donna, Luo, Jie, Wang, Meilin, Li, Qiuxia, Matvekas, Aleksas, Wen, Bo, Sun, Duxin, Chinnaiyan, Arul M., and Wang, Shaomeng
- Abstract
CBP/p300 are critical transcriptional coactivators of the androgen receptor (AR) and are promising cancer therapeutic targets. Herein, we report the discovery of highly potent, selective, and orally bioavailable CBP/p300 degraders using the PROTAC technology with CBPD-409 being the most promising compound. CBPD-409 induces robust CBP/p300 degradation with DC500.2–0.4 nM and displays strong antiproliferative effects with IC501.2–2.0 nM in the VCaP, LNCaP, and 22Rv1 AR+ prostate cancer cell lines. It has a favorable pharmacokinetic profile and achieves 50% of oral bioavailability in mice. A single oral administration of CBPD-409 at 1 mg/kg achieves >95% depletion of CBP/p300 proteins in the VCaP tumor tissue. CBPD-409 exhibits strong tumor growth inhibition and is much more potent and efficacious than two CBP/p300 inhibitors CCS1477 and GNE-049 and the AR antagonist Enzalutamide. CBPD-409 is a promising CBP/p300 degrader for further extensive evaluations for the treatment of advanced prostate cancer and other types of human cancers.
- Published
- 2024
- Full Text
- View/download PDF
9. Prospective comparison of restriction spectrum imaging and non-invasive biomarkers to predict upgrading on active surveillance prostate biopsy
- Author
-
Eng, Stefan E., Basasie, Benjamin, Lam, Alfonso, John Semmes, O., Troyer, Dean A., Clarke, Geoffrey D., Sunnapwar, Abhijit G., Leach, Robin J., Johnson-Pais, Teresa L., Sokoll, Lori J., Chan, Daniel W., Tosoian, Jeffrey J., Siddiqui, Javed, Chinnaiyan, Arul M., Thompson, Ian M., Boutros, Paul C., and Liss, Michael A.
- Abstract
Background: Protocol-based active surveillance (AS) biopsies have led to poor compliance. To move to risk-based protocols, more accurate imaging biomarkers are needed to predict upgrading on AS prostate biopsy. We compared restriction spectrum imaging (RSI-MRI) generated signal maps as a biomarker to other available non-invasive biomarkers to predict upgrading or reclassification on an AS biopsy. Methods: We prospectively enrolled men on prostate cancer AS undergoing repeat biopsy from January 2016 to June 2019 to obtain an MRI and biomarkers to predict upgrading. Subjects underwent a prostate multiparametric MRI and a short duration, diffusion-weighted enhanced MRI called RSI to generate a restricted signal map along with evaluation of 30 biomarkers (14 clinico-epidemiologic features, 9 molecular biomarkers, and 7 radiologic-associated features). Our primary outcome was upgrading or reclassification on subsequent AS prostate biopsy. Statistical analysis included operating characteristic improvement using AUROC and AUPRC. Results: The individual biomarker with the highest area under the receiver operator characteristic curve (AUC) was RSI-MRI (AUC = 0.84; 95% CI: 0.71–0.96). The best non-imaging biomarker was prostate volume-corrected Prostate Health Index density (PHI, AUC = 0.68; 95% CI: 0.53–0.82). Non-imaging biomarkers had a negligible effect on predicting upgrading at the next biopsy but did improve predictions of overall time to progression in AS. Conclusions: RSI-MRI, PIRADS, and PHI could improve the predictive ability to detect upgrading in AS. The strongest predictor of clinically significant prostate cancer on AS biopsy was RSI-MRI signal output.
- Published
- 2024
- Full Text
- View/download PDF
10. Circular RNA in cancer
- Author
-
Conn, Vanessa M., Chinnaiyan, Arul M., and Conn, Simon J.
- Abstract
Over the past decade, circular RNA (circRNA) research has evolved into a bona fide research field shedding light on the functional consequence of this unique family of RNA molecules in cancer. Although the method of formation and the abundance of circRNAs can differ from their cognate linear mRNA, the spectrum of interacting partners and their resultant cellular functions in oncogenesis are analogous. However, with 10 times more diversity in circRNA variants compared with linear RNA variants, combined with their hyperstability in the cell, circRNAs are equipped to influence every stage of oncogenesis. This is an opportune time to address the breadth of circRNA in cancer focused on their spatiotemporal expression, mutations in biogenesis factors and contemporary functions through each stage of cancer. In this Review, we highlight examples of functional circRNAs in specific cancers, which satisfy critical criteria, including their physical co-association with the target and circRNA abundance at stoichiometrically valid quantities. These considerations are essential to develop strategies for the therapeutic exploitation of circRNAs as biomarkers and targeted anticancer agents.
- Published
- 2024
- Full Text
- View/download PDF
11. Discovery of a First-in-Class Degrader for the Lipid Kinase PIKfyve.
- Author
-
Li, Chungen, Qiao, Yuanyuan, Jiang, Xia, Liu, Lianchao, Zheng, Yang, Qiu, Yudi, Cheng, Caleb, Zhou, Fengtao, Zhou, Yang, Huang, Weixue, Ren, Xiaomei, Wang, Yuzhuo, Wang, Zhen, Chinnaiyan, Arul M., and Ding, Ke
- Published
- 2023
- Full Text
- View/download PDF
12. Discovery of a Highly Potent and Selective Dual PROTAC Degrader of CDK12 and CDK13.
- Author
-
Yang, Jianzhang, Chang, Yu, Tien, Jean Ching-Yi, Wang, Zhen, Zhou, Yang, Zhang, Pujuan, Huang, Weixue, Vo, Josh, Apel, Ingrid J., Wang, Cynthia, Zeng, Victoria Zhixuan, Cheng, Yunhui, Li, Shuqin, Wang, George Xiaoju, Chinnaiyan, Arul M., and Ding, Ke
- Published
- 2023
- Full Text
- View/download PDF
13. MyProstateScore in men considering repeat biopsy: validation of a simple testing approach
- Author
-
Tosoian, Jeffrey J., Sessine, Michael S., Trock, Bruce J., Ross, Ashley E., Xie, Cassie, Zheng, Yingye, Samora, Nathan L., Siddiqui, Javed, Niknafs, Yashar, Chopra, Zoey, Tomlins, Scott, Kunju, Lakshmi P., Palapattu, Ganesh S., Morgan, Todd M., Wei, John T., Salami, Simpa S., and Chinnaiyan, Arul M.
- Abstract
Background: Men with persistent risk of Grade Group (GG) ≥ 2 cancer after a negative biopsy present a unique clinical challenge. The validated MyProstateScore test is clinically-available for pre-biopsy risk stratification. In biopsy-naïve patients, we recently validated a straightforward testing approach to rule-out GG ≥ 2 cancer with 98% negative predictive value (NPV) and 97% sensitivity. In the current study, we established a practical MPS-based testing approach in men with a previous negative biopsy being considered for repeat biopsy. Methods: Patients provided post-digital rectal examination urine prior to repeat biopsy. MyProstateScore was calculated using the validated, locked model including urinary PCA3 and TMPRSS2:ERG scores with serum PSA. In a clinically-appropriate primary (i.e., training) cohort, we identified a lower (rule-out) threshold approximating 90% sensitivity and an upper (rule-in) threshold approximating 80% specificity for GG ≥ 2 cancer. These thresholds were applied to an external validation cohort, and performance measures and clinical outcomes associated with their use were calculated. Results: MyProstateScore thresholds of 15 and 40 met pre-defined performance criteria in the primary cohort (422 patients; median PSA 6.4, IQR 4.3–9.1). In the 268-patient validation cohort, 25 men (9.3%) had GG ≥ 2 cancer on repeat biopsy. The rule-out threshold of 15 provided 100% NPV and sensitivity for GG ≥ 2 cancer and would have prevented 23% of unnecessary biopsies. Use of MyProstateScore >40 to rule-in biopsy would have prevented 67% of biopsies while maintaining 95% NPV. In the validation cohort, the prevalence of GG ≥ 2 cancer was 0% for MyProstateScore 0–15, 6.5% for MyProstateScore 15–40, and 19% for MyProstateScore >40. Conclusions: In patients who previously underwent a negative prostate biopsy, the MyProstateScore values of 15 and 40 yielded clinically-actionable rule-in and rule-out risk groups. Using this straightforward testing approach, MyProstateScore can meaningfully inform patients and physicians weighing the need for repeat biopsy.
- Published
- 2023
- Full Text
- View/download PDF
14. A transcriptomic model for homologous recombination deficiency in prostate cancer
- Author
-
Weiner, Adam B., Liu, Yang, McFarlane, Matthew, Bawa, Pushpinder S., Li, Eric V., Zhao, Xin, Li, Ziwen, Hammoud, Tanya, Hazime, Munna, Karnes, R. Jeffrey, Davicioni, Elai, Reichert, Zachery R., Chinnaiyan, Arul M., Lotan, Tamara L., Spratt, Daniel E., and Schaeffer, Edward M.
- Abstract
Background: Tumors with mutations associated with homologous recombination deficiency (HRD) are uncommon in prostate cancer (PCa) and variably responsive to PARP inhibition. To better identify tumors with HRD, we developed a transcriptomic signature for HRD in PCa (HRD-P). Methods: By using an established mutational signature, we created and validated HRD-P in six independent PCa cohorts (primary PCa, n= 8224; metastatic castration-resistant PCa [mCRPC], n= 328). Molecular and clinical features were compared between HRD-P+ tumors and those with single HR-gene mutations. Results: HRD-P+ tumors were more common than tumors with single HR-gene mutations in primary (201/491, 41% vs 32/491 6.5%) and mCRPC (126/328, 38% vs 82/328, 25%) cases, and HRD-P+ was more predictive of genomic instability suggestive of HRD. HRD-P+ was associated with a shorter time to recurrence following surgery and shorter overall survival in men with mCRPC. In a prospective trial of mCRPC treated with olaparib (n= 10), all three men with HRD-P+ experienced prolonged (>330 days) PSA progression-free survival. Conclusion: These results suggest transcriptomics can identify more patients that harbor phenotypic HRD than single HR-gene mutations and support further exploration of transcriptionally defined HRD tumors perhaps in conjunction with genomic markers for therapeutic application.
- Published
- 2022
- Full Text
- View/download PDF
15. Discovery of a Highly Potent and Selective Dual PROTAC Degrader of CDK12 and CDK13.
- Author
-
Yang, Jianzhang, Chang, Yu, Tien, Jean Ching-Yi, Wang, Zhen, Zhou, Yang, Zhang, Pujuan, Huang, Weixue, Vo, Josh, Apel, Ingrid J., Wang, Cynthia, Zeng, Victoria Zhixuan, Cheng, Yunhui, Li, Shuqin, Wang, George Xiaoju, Chinnaiyan, Arul M., and Ding, Ke
- Published
- 2022
- Full Text
- View/download PDF
16. Squamoid Eccrine Ductal Carcinoma Displays Ultraviolet Mutations and Intermediate Gene Expression Relative to Squamous Cell Carcinoma, Microcystic Adnexal Carcinoma, and Porocarcinoma
- Author
-
Harms, Paul W., Runge, Mason, Chan, May P., Liu, Chia-Jen, Qin, Zhaoping, Worden, Francis, Robinson, Dan R., Chinnaiyan, Arul M., Mclean, Scott A., Harms, Kelly L., Fullen, Douglas R., Patel, Rajiv M., Andea, Aleodor A., and Udager, Aaron M.
- Abstract
Squamoid eccrine ductal carcinoma is a rare infiltrative tumor with morphologic features intermediate between squamous cell carcinoma (SCC) and sweat gland carcinomas such as microcystic adnexal carcinoma. Although currently classified as a sweat gland carcinoma, it has been debated whether squamoid eccrine ductal carcinoma is better classified as a variant of SCC. Furthermore, therapeutic options for patients with advanced disease are lacking. Here, we describe clinicopathologic features of a cohort of 15 squamoid eccrine ductal carcinomas from 14 unique patients, with next-generation sequencing DNA profiling for 12 cases. UV signature mutations were the dominant signature in the majority of cases. TP53mutations were the most highly recurrent specific gene alteration, followed by mutations in NOTCHgenes. Recurrent mutations in driver oncogenes were not identified. By unsupervised comparison of global transcriptome profiles in squamoid eccrine ductal carcinoma (n = 7) to SCC (n = 10), porocarcinoma (n = 4), and microcystic adnexal carcinoma (n = 4), squamoid eccrine ductal carcinomas displayed an intermediate phenotype between SCC and sweat gland tumors. Squamoid eccrine ductal carcinoma displayed significantly higher expression of 364 genes (including certain eccrine markers) and significantly lower expression of 525 genes compared with other groups. Our findings support the classification of squamoid eccrine ductal carcinoma as a carcinoma with intermediate features between SCC and sweat gland carcinoma.
- Published
- 2024
- Full Text
- View/download PDF
17. CDK12 loss drives prostate cancer progression, transcription-replication conflicts, and synthetic lethality with paralog CDK13
- Author
-
Tien, Jean Ching-Yi, Luo, Jie, Chang, Yu, Zhang, Yuping, Cheng, Yunhui, Wang, Xiaoju, Yang, Jianzhang, Mannan, Rahul, Mahapatra, Somnath, Shah, Palak, Wang, Xiao-Ming, Todd, Abigail J., Eyunni, Sanjana, Cheng, Caleb, Rebernick, Ryan J., Xiao, Lanbo, Bao, Yi, Neiswender, James, Brough, Rachel, Pettitt, Stephen J., Cao, Xuhong, Miner, Stephanie J., Zhou, Licheng, Wu, Yi-Mi, Labanca, Estefania, Wang, Yuzhuo, Parolia, Abhijit, Cieslik, Marcin, Robinson, Dan R., Wang, Zhen, Feng, Felix Y., Chou, Jonathan, Lord, Christopher J., Ding, Ke, and Chinnaiyan, Arul M.
- Abstract
Biallelic loss of cyclin-dependent kinase 12 (CDK12) defines a metastatic castration-resistant prostate cancer (mCRPC) subtype. It remains unclear, however, whether CDK12loss drives prostate cancer (PCa) development or uncovers pharmacologic vulnerabilities. Here, we show Cdk12ablation in murine prostate epithelium is sufficient to induce preneoplastic lesions with lymphocytic infiltration. In allograft-based CRISPR screening, Cdk12loss associates positively with Trp53inactivation but negatively with Pteninactivation. Moreover, concurrent Cdk12/Trp53ablation promotes proliferation of prostate-derived organoids, while Cdk12knockout in Pten-null mice abrogates prostate tumor growth. In syngeneic systems, Cdk12/Trp53-null allografts exhibit luminal morphology and immune checkpoint blockade sensitivity. Mechanistically, Cdk12inactivation mediates genomic instability by inducing transcription-replication conflicts. Strikingly, CDK12-mutant organoids and patient-derived xenografts are sensitive to inhibition or degradation of the paralog kinase, CDK13. We therein establish CDK12as a bona fidetumor suppressor, mechanistically define how CDK12inactivation causes genomic instability, and advance a therapeutic strategy for CDK12-mutant mCRPC.
- Published
- 2024
- Full Text
- View/download PDF
18. Development of an orally bioavailable CDK12/13 degrader and induction of synthetic lethality with AKT pathway inhibition
- Author
-
Chang, Yu, Wang, Xiaoju, Yang, Jianzhang, Tien, Jean Ching-Yi, Mannan, Rahul, Cruz, Gabriel, Zhang, Yuping, Vo, Josh N., Magnuson, Brian, Mahapatra, Somnath, Cho, Hanbyul, Dhanasekaran, Saravana Mohan, Wang, Cynthia, Wang, Zhen, Zhou, Licheng, Zhou, Kaijie, Zhou, Yang, Zhang, Pujuan, Huang, Weixue, Xiao, Lanbo, Liu, Weihuang Raymond, Hamadeh, Rudana, Su, Fengyun, Wang, Rui, Miner, Stephanie J., Cao, Xuhong, Cheng, Yunhui, Mehra, Rohit, Ding, Ke, and Chinnaiyan, Arul M.
- Abstract
Cyclin-dependent kinases 12/13 play pivotal roles in orchestrating transcription elongation, DNA damage response, and maintenance of genomic stability. Biallelic CDK12loss has been documented in various malignancies. Here, we develop a selective CDK12/13 PROTAC degrader, YJ9069, which effectively inhibits proliferation in subsets of prostate cancer cells preferentially over benign immortalized cells. CDK12/13 degradation rapidly triggers gene-length-dependent transcriptional elongation defects, leading to DNA damage and cell-cycle arrest. In vivo, YJ9069 significantly suppresses prostate tumor growth. Modifications of YJ9069 yielded an orally bioavailable CDK12/13 degrader, YJ1206, which exhibits comparable efficacy with significantly less toxicity. To identify pathways synthetically lethal upon CDK12/13 degradation, phosphorylation pathway arrays were performed using cell lines treated with YJ1206. Interestingly, degradation or genetic knockdown of CDK12/13 led to activation of the AKT pathway. Targeting CDK12/13 for degradation, in conjunction with inhibiting the AKT pathway, resulted in a synthetic lethal effect in preclinical prostate cancer models.
- Published
- 2024
- Full Text
- View/download PDF
19. Viral Status Predicts the Patterns of Genome Methylation and Decitabine Response in Merkel Cell Carcinoma
- Author
-
Harms, Paul W., Verhaegen, Monique E., Vo, Josh N., Tien, Jean C., Pratt, Drew, Su, Fengyun, Dhanasekaran, Saravana M., Cao, Xuhong, Mangelberger, Doris, VanGoor, Julia, Choi, Jae Eun, Ma, Vincent T., Dlugosz, Andrzej A., and Chinnaiyan, Arul M.
- Abstract
Merkel cell carcinoma (MCC) is an aggressive cutaneous neuroendocrine carcinoma that is classified as Merkel cell polyomavirus-positive (virus positive [VP]) or Merkel cell polyomavirus-negative (virus negative [VN]). Epigenetic changes, such as DNA methylation, can alter gene expression and influence cancer progression. However, patterns of DNA methylation and the therapeutic efficacy of hypomethylating agents have not been fully explored in MCC. We characterized genome-wide DNA methylation in 16 MCC cell lines from both molecular subclasses in comparison with other cancer types and found that the overall profile of MCC is similar to that of small-cell lung carcinoma. Comparison of VP MCC with VN MCC revealed 2,260 differentially methylated positions. The hypomethylating agent decitabine upregulated the expression of antigen-presenting machinery in MCC cell lines and stimulated membrane expression of HLA-Ain VP and VN MCC xenograft tumors. Decitabine also induced prominent caspase- and large T antigen‒independent cell death in VP MCC, whereas VN MCC cell lines displayed decreased proliferation without increased cell death. In mouse xenografts, decitabine significantly decreased the size of VP tumors but not that of VN tumors. Our findings indicate that viral status predicts genomic methylation patterns in MCC and that decitabine may be therapeutically effective against MCC through antiproliferative effects, cell death, and increased immune recognition.
- Published
- 2022
- Full Text
- View/download PDF
20. Targeting SWI/SNF ATPases in enhancer-addicted prostate cancer
- Author
-
Xiao, Lanbo, Parolia, Abhijit, Qiao, Yuanyuan, Bawa, Pushpinder, Eyunni, Sanjana, Mannan, Rahul, Carson, Sandra E., Chang, Yu, Wang, Xiaoju, Zhang, Yuping, Vo, Josh N., Kregel, Steven, Simko, Stephanie A., Delekta, Andrew D., Jaber, Mustapha, Zheng, Heng, Apel, Ingrid J., McMurry, Lisa, Su, Fengyun, Wang, Rui, Zelenka-Wang, Sylvia, Sasmal, Sanjita, Khare, Leena, Mukherjee, Subhendu, Abbineni, Chandrasekhar, Aithal, Kiran, Bhakta, Mital S., Ghurye, Jay, Cao, Xuhong, Navone, Nora M., Nesvizhskii, Alexey I., Mehra, Rohit, Vaishampayan, Ulka, Blanchette, Marco, Wang, Yuzhuo, Samajdar, Susanta, Ramachandra, Murali, and Chinnaiyan, Arul M.
- Abstract
The switch/sucrose non-fermentable (SWI/SNF) complex has a crucial role in chromatin remodelling1and is altered in over 20% of cancers2,3. Here we developed a proteolysis-targeting chimera (PROTAC) degrader of the SWI/SNF ATPase subunits, SMARCA2 and SMARCA4, called AU-15330. Androgen receptor (AR)+forkhead box A1 (FOXA1)+prostate cancer cells are exquisitely sensitive to dual SMARCA2 and SMARCA4 degradation relative to normal and other cancer cell lines. SWI/SNF ATPase degradation rapidly compacts cis-regulatory elements bound by transcription factors that drive prostate cancer cell proliferation, namely AR, FOXA1, ERG and MYC, which dislodges them from chromatin, disables their core enhancer circuitry, and abolishes the downstream oncogenic gene programs. SWI/SNF ATPase degradation also disrupts super-enhancer and promoter looping interactions that wire supra-physiologic expression of the AR, FOXA1and MYConcogenes themselves. AU-15330 induces potent inhibition of tumour growth in xenograft models of prostate cancer and synergizes with the AR antagonist enzalutamide, even inducing disease remission in castration-resistant prostate cancer (CRPC) models without toxicity. Thus, impeding SWI/SNF-mediated enhancer accessibility represents a promising therapeutic approach for enhancer-addicted cancers.
- Published
- 2022
- Full Text
- View/download PDF
21. The Potential of Circular RNAs as Cancer Biomarkers.
- Author
-
Brown, Jason R. and Chinnaiyan, Arul M.
- Abstract
Circular RNA (circRNA) is a covalently closed RNA structure that has several proposed functions related to cancer development. Recently, cancer-specific and tissue-specific circRNAs have been identified by high-throughput sequencing and are curated in publicly available databases. CircRNAs have features that are ideal properties of biomarkers, including conservation, abundance, and stability in plasma, saliva, and urine. Many circRNAs with predictive and prognostic significance in cancer have been described, and functional mechanisms for some circRNAs have been suggested. CircRNA also has great potential as a noninvasive biomarker for early cancer detection, although further investigation is necessary before clinical application is feasible. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
22. TRIM63is a sensitive and specific biomarker for MiT family aberration-associated renal cell carcinoma
- Author
-
Wang, Xiao-Ming, Zhang, Yuping, Mannan, Rahul, Skala, Stephanie L., Rangaswamy, Roshni, Chinnaiyan, Anya, Su, Fengyun, Cao, Xuhong, Zelenka-Wang, Sylvia, McMurry, Lisa, Xiao, Hong, Spratt, Daniel E., Sangoi, Ankur R., Shao, Lina, Betz, Bryan L., Brown, Noah, Tickoo, Satish K., McKenney, Jesse K., Argani, Pedram, Gupta, Sounak, Reuter, Victor E., Chinnaiyan, Arul M., Dhanasekaran, Saravana M., and Mehra, Rohit
- 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 TFE3or TFEBgenes. 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 TRIM63as 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 TRIM63RNA-ISH results with the results from TFE3/TFEBfluorescence in situ hybridization (FISH), which is the current clinical standard. We confirmed that TRIM63mRNA was highly expressed in all classes of MiTF-RCC compared to other renal tumor categories, where it was mostly absent to low. While the TRIM63RNA-ISH and TFE3/TFEBFISH results were largely concordant, importantly, TRIM63RNA-ISH was strongly positive in TFE3FISH false-negative cases with RBM10-TFE3inversion. In conclusion, TRIM63can serve as a diagnostic marker to distinguish MiTF-RCC from other renal tumor subtypes with overlapping morphology. We suggest a combination of TFE3/TFEBFISH and TRIM63RNA-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.
- Published
- 2021
- Full Text
- View/download PDF
23. Comprehensive proteogenomic characterization of rare kidney tumors
- Author
-
Li, Ginny Xiaohe, Chen, Lijun, Hsiao, Yi, Mannan, Rahul, Zhang, Yuping, Luo, Jie, Petralia, Francesca, Cho, Hanbyul, Hosseini, Noshad, Leprevost, Felipe da Veiga, Calinawan, Anna, Li, Yize, Anand, Shankara, Dagar, Aniket, Geffen, Yifat, Kumar-Sinha, Chandan, Chugh, Seema, Le, Anne, Ponce, Sean, Guo, Shenghao, Zhang, Cissy, Schnaubelt, Michael, Al Deen, Nataly Naser, Chen, Feng, Caravan, Wagma, Houston, Andrew, Hopkins, Alex, Newton, Chelsea J., Wang, Xiaoming, Polasky, Daniel A., Haynes, Sarah, Yu, Fengchao, Jing, Xiaojun, Chen, Siqi, Robles, Ana I., Mesri, Mehdi, Thiagarajan, Mathangi, An, Eunkyung, Getz, Gad A., Linehan, W. Marston, Hostetter, Galen, Jewell, Scott D., Chan, Daniel W., Wang, Pei, Omenn, Gilbert S., Mehra, Rohit, Ricketts, Christopher J., Ding, Li, Chinnaiyan, Arul M., Cieslik, Marcin P., Dhanasekaran, Saravana M., Zhang, Hui, Nesvizhskii, Alexey I., Lazar, Alexander J., Paulovich, Amanda G., Antczak, Andrzej, Green, Anthony, Ma’ayan, Avi, Pruetz, Barb, Zhang, Bing, Reva, Boris, Druker, Brian J., Goldthwaite, Charles A., Birger, Chet, Mani, D.R., Chesla, David, Fenyö, David, Schadt, Eric E., Wilson, George, Kołodziejczak, Iga, John, Ivy, Hafron, Jason, Vo, Josh, Zaalishvili, Kakhaber, Ketchum, Karen A., Rodland, Karin D., Nyce, Kristen, Wiznerowicz, Maciej, Domagalski, Marcin J., Anurag, Meenakshi, Borucki, Melissa, Gillette, Michael A., Birrer, Michael J., Edwards, Nathan J., Vatanian, Negin, VanderKolk, Pamela, McGarvey, Peter B., Dhir, Rajiv, Thangudu, Ratna R., Crispen, Reese, Smith, Richard D., Payne, Samuel H., Cottingham, Sandra, Cai, Shuang, Carr, Steven A., Liu, Tao, Le, Toan, Ma, Weiping, Zhang, Xu, Lu, Yin, Shutack, Yvonne, and Zhang, Zhen
- Abstract
Non-clear cell renal cell carcinomas (non-ccRCCs) encompass diverse malignant and benign tumors. Refinement of differential diagnosis biomarkers, markers for early prognosis of aggressive disease, and therapeutic targets to complement immunotherapy are current clinical needs. Multi-omics analyses of 48 non-ccRCCs compared with 103 ccRCCs reveal proteogenomic, phosphorylation, glycosylation, and metabolic aberrations in RCC subtypes. RCCs with high genome instability display overexpression of IGF2BP3 and PYCR1. Integration of single-cell and bulk transcriptome data predicts diverse cell-of-origin and clarifies RCC subtype-specific proteogenomic signatures. Expression of biomarkers MAPRE3, ADGRF5, and GPNMB differentiates renal oncocytoma from chromophobe RCC, and PIGR and SOSTDC1 distinguish papillary RCC from MTSCC. This study expands our knowledge of proteogenomic signatures, biomarkers, and potential therapeutic targets in non-ccRCC.
- Published
- 2024
- Full Text
- View/download PDF
24. Assessment of Clinical Benefit of Integrative Genomic Profiling in Advanced Solid Tumors
- Author
-
Cobain, Erin F., Wu, Yi-Mi, Vats, Pankaj, Chugh, Rashmi, Worden, Francis, Smith, David C., Schuetze, Scott M., Zalupski, Mark M., Sahai, Vaibhav, Alva, Ajjai, Schott, Anne F., Caram, Megan E. V., Hayes, Daniel F., Stoffel, Elena M., Jacobs, Michelle F., Kumar-Sinha, Chandan, Cao, Xuhong, Wang, Rui, Lucas, David, Ning, Yu, Rabban, Erica, Bell, Janice, Camelo-Piragua, Sandra, Udager, Aaron M., Cieslik, Marcin, Lonigro, Robert J., Kunju, Lakshmi P., Robinson, Dan R., Talpaz, Moshe, and Chinnaiyan, Arul M.
- Abstract
IMPORTANCE: Use of next-generation sequencing (NGS) to identify clinically actionable genomic targets has been incorporated into routine clinical practice in the management of advanced solid tumors; however, the clinical utility of this testing remains uncertain. OBJECTIVE: To determine which patients derived the greatest degree of clinical benefit from NGS profiling. DESIGN, SETTING, AND PARTICIPANTS: Patients in this cohort study underwent fresh tumor biopsy and blood sample collection for genomic profiling of paired tumor and normal DNA (whole-exome or targeted-exome capture with analysis of 1700 genes) and tumor transcriptome (RNA) sequencing. Somatic and germline genomic alterations were annotated and classified according to degree of clinical actionability. Results were returned to treating oncologists. Data were collected from May 1, 2011, to February 28, 2018, and analyzed from May 1, 2011, to April 30, 2020. MAIN OUTCOMES AND MEASURES: Patients’ subsequent therapy and treatment response were extracted from the medical record to determine clinical benefit rate from NGS-directed therapy at 6 months and exceptional responses lasting 12 months or longer. RESULTS: During the study period, NGS was attempted on tumors from 1138 patients and was successful in 1015 (89.2%) (MET1000 cohort) (538 men [53.0%]; mean [SD] age, 57.7 [13.3] years). Potentially clinically actionable genomic alterations were discovered in 817 patients (80.5%). Of these, 132 patients (16.2%) received sequencing-directed therapy, and 49 had clinical benefit (37.1%). Exceptional responses were observed in 26 patients (19.7% of treated patients). Pathogenic germline variants (PGVs) were identified in 160 patients (15.8% of cohort), including 49 PGVs (4.8% of cohort) with therapeutic relevance. For 55 patients with carcinoma of unknown primary origin, NGS identified the primary site in 28 (50.9%), and sequencing-directed therapy in 13 patients resulted in clinical benefit in 7 instances (53.8%), including 5 exceptional responses. CONCLUSIONS AND RELEVANCE: The high rate of therapeutically relevant PGVs identified across diverse cancer types supports a recommendation for directed germline testing in all patients with advanced cancer. The high frequency of therapeutically relevant somatic and germline findings in patients with carcinoma of unknown primary origin and other rare cancers supports the use of comprehensive NGS profiling as a component of standard of care for these disease entities.
- Published
- 2021
- Full Text
- View/download PDF
25. Defining cancer growth beyond the mitotic index
- Author
-
Kumar-Sinha, Chandan and Chinnaiyan, Arul M.
- Abstract
Tumour growth involves anomalies in cell cycle genes and emergent phenotypes, but the tumour proliferation rate (or mitotic index) is defined by just one marker, Ki-67. A study now integrates expression patterns of several markers to generate spatio-temporal maps of cell proliferation in cancer tissues.
- Published
- 2022
- Full Text
- View/download PDF
26. Liver metastasis restrains immunotherapy efficacy via macrophage-mediated T cell elimination
- Author
-
Yu, Jiali, Green, Michael D., Li, Shasha, Sun, Yilun, Journey, Sara N., Choi, Jae Eun, Rizvi, Syed Monem, Qin, Angel, Waninger, Jessica J., Lang, Xueting, Chopra, Zoey, El Naqa, Issam, Zhou, Jiajia, Bian, Yingjie, Jiang, Long, Tezel, Alangoya, Skvarce, Jeremy, Achar, Rohan K., Sitto, Merna, Rosen, Benjamin S., Su, Fengyun, Narayanan, Sathiya P., Cao, Xuhong, Wei, Shuang, Szeliga, Wojciech, Vatan, Linda, Mayo, Charles, Morgan, Meredith A., Schonewolf, Caitlin A., Cuneo, Kyle, Kryczek, Ilona, Ma, Vincent T., Lao, Christopher D., Lawrence, Theodore S., Ramnath, Nithya, Wen, Fei, Chinnaiyan, Arul M., Cieslik, Marcin, Alva, Ajjai, and Zou, Weiping
- Abstract
Metastasis is the primary cause of cancer mortality, and cancer frequently metastasizes to the liver. It is not clear whether liver immune tolerance mechanisms contribute to cancer outcomes. We report that liver metastases diminish immunotherapy efficacy systemically in patients and preclinical models. Patients with liver metastases derive limited benefit from immunotherapy independent of other established biomarkers of response. In multiple mouse models, we show that liver metastases siphon activated CD8+T cells from systemic circulation. Within the liver, activated antigen-specific Fas+CD8+T cells undergo apoptosis following their interaction with FasL+CD11b+F4/80+monocyte-derived macrophages. Consequently, liver metastases create a systemic immune desert in preclinical models. Similarly, patients with liver metastases have reduced peripheral T cell numbers and diminished tumoral T cell diversity and function. In preclinical models, liver-directed radiotherapy eliminates immunosuppressive hepatic macrophages, increases hepatic T cell survival and reduces hepatic siphoning of T cells. Thus, liver metastases co-opt host peripheral tolerance mechanisms to cause acquired immunotherapy resistance through CD8+T cell deletion, and the combination of liver-directed radiotherapy and immunotherapy could promote systemic antitumor immunity.
- Published
- 2021
- Full Text
- View/download PDF
27. TRIM63is a sensitive and specific biomarker for MiT family aberration-associated renal cell carcinoma
- Author
-
Wang, Xiao-Ming, Zhang, Yuping, Mannan, Rahul, Skala, Stephanie L., Rangaswamy, Roshni, Chinnaiyan, Anya, Su, Fengyun, Cao, Xuhong, Zelenka-Wang, Sylvia, McMurry, Lisa, Xiao, Hong, Spratt, Daniel E., Sangoi, Ankur R., Shao, Lina, Betz, Bryan L., Brown, Noah, Tickoo, Satish K., McKenney, Jesse K., Argani, Pedram, Gupta, Sounak, Reuter, Victor E., Chinnaiyan, Arul M., Dhanasekaran, Saravana M., and Mehra, Rohit
- 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 TFE3or TFEBgenes. 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 TRIM63as 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 TRIM63RNA-ISH results with the results from TFE3/TFEBfluorescence in situ hybridization (FISH), which is the current clinical standard. We confirmed that TRIM63mRNA was highly expressed in all classes of MiTF-RCC compared to other renal tumor categories, where it was mostly absent to low. While the TRIM63RNA-ISH and TFE3/TFEBFISH results were largely concordant, importantly, TRIM63RNA-ISH was strongly positive in TFE3FISH false-negative cases with RBM10-TFE3inversion. In conclusion, TRIM63can serve as a diagnostic marker to distinguish MiTF-RCC from other renal tumor subtypes with overlapping morphology. We suggest a combination of TFE3/TFEBFISH and TRIM63RNA-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.
- Published
- 2021
- Full Text
- View/download PDF
28. Invasive squamous cell carcinomas and precursor lesions on UV-exposed epithelia demonstrate concordant genomic complexity in driver genes
- Author
-
Lazo de la Vega, Lorena, Bick, Nolan, Hu, Kevin, Rahrig, Samantha E., Silva, Camilla Duarte, Matayoshi, Suzana, Picciarelli, Patricia, Wang, Xiaoming, Sugar, Alan, Soong, Hunson Kaz, Mian, Shahzad I., Robinson, Dan R., Chinnaiyan, Arul M., Demirci, Hakan, Daniels, Anthony B., Worden, Francis, Eberhart, Charles G., Tomlins, Scott A., Rao, Rajesh C., and Harms, Paul W.
- Abstract
Although squamous cell carcinomas (SCC) are the most frequent human solid tumor at many anatomic sites, the driving molecular alterations underlying their progression from precursor lesions are poorly understood, especially in the context of photodamage. Therefore, we used high-depth, targeted next-generation sequencing (NGS) of RNA and DNA from routine tissue samples to characterize the progression of both well- (cutaneous) and poorly (ocular) studied SCCs. We assessed 56 formalin-fixed paraffin-embedded (FFPE) cutaneous lesions (n= 8 actinic keratosis, n= 30 carcinoma in situ [CIS], n= 18 invasive) and 43 FFPE ocular surface lesions (n= 2 conjunctival/corneal intraepithelial neoplasia, n= 20 CIS, n= 21 invasive), from institutions in the US and Brazil. An additional seven cases of advanced cutaneous SCC were profiled by hybrid capture-based NGS of >1500 genes. The cutaneous and ocular squamous neoplasms displayed a predominance of UV-signature mutations. Precursor lesions had highly similar somatic genomic landscapes to SCCs, including chromosomal gains of 3q involving SOX2, and highly recurrent mutations and/or loss of heterozygosity events affecting tumor suppressors TP53and CDKN2A. Additionally, we identify a novel molecular subclass of CIS with RB1mutations. Among TP53wild-type tumors, human papillomavirus transcript was detected in one matched pair of cutaneous CIS and SCC. Amplicon-based whole-transcriptome sequencing of select 20 cutaneous lesions demonstrated significant upregulation of pro-invasion genes in cutaneous SCCs relative to precursors, including MMP1, MMP3, MMP9, LAMC2, LGALS1, and TNFRSF12A. Together, ocular and cutaneous squamous neoplasms demonstrate similar alterations, supporting a common model for neoplasia in UV-exposed epithelia. Treatment modalities useful for cutaneous SCC may also be effective in ocular SCC given the genetic similarity between these tumor types. Importantly, in both systems, precursor lesions possess the full complement of major genetic changes seen in SCC, supporting non-genetic drivers of invasiveness.
- Published
- 2020
- Full Text
- View/download PDF
29. Invasive squamous cell carcinomas and precursor lesions on UV-exposed epithelia demonstrate concordant genomic complexity in driver genes
- Author
-
Lazo de la Vega, Lorena, Bick, Nolan, Hu, Kevin, Rahrig, Samantha E., Silva, Camilla Duarte, Matayoshi, Suzana, Picciarelli, Patricia, Wang, Xiaoming, Sugar, Alan, Soong, Hunson Kaz, Mian, Shahzad I., Robinson, Dan R., Chinnaiyan, Arul M., Demirci, Hakan, Daniels, Anthony B., Worden, Francis, Eberhart, Charles G., Tomlins, Scott A., Rao, Rajesh C., and Harms, Paul W.
- Abstract
Although squamous cell carcinomas (SCC) are the most frequent human solid tumor at many anatomic sites, the driving molecular alterations underlying their progression from precursor lesions are poorly understood, especially in the context of photodamage. Therefore, we used high-depth, targeted next-generation sequencing (NGS) of RNA and DNA from routine tissue samples to characterize the progression of both well- (cutaneous) and poorly (ocular) studied SCCs. We assessed 56 formalin-fixed paraffin-embedded (FFPE) cutaneous lesions (n= 8 actinic keratosis, n= 30 carcinoma in situ [CIS], n= 18 invasive) and 43 FFPE ocular surface lesions (n= 2 conjunctival/corneal intraepithelial neoplasia, n= 20 CIS, n= 21 invasive), from institutions in the US and Brazil. An additional seven cases of advanced cutaneous SCC were profiled by hybrid capture-based NGS of >1500 genes. The cutaneous and ocular squamous neoplasms displayed a predominance of UV-signature mutations. Precursor lesions had highly similar somatic genomic landscapes to SCCs, including chromosomal gains of 3q involving SOX2, and highly recurrent mutations and/or loss of heterozygosity events affecting tumor suppressors TP53and CDKN2A. Additionally, we identify a novel molecular subclass of CIS with RB1mutations. Among TP53wild-type tumors, human papillomavirus transcript was detected in one matched pair of cutaneous CIS and SCC. Amplicon-based whole-transcriptome sequencing of select 20 cutaneous lesions demonstrated significant upregulation of pro-invasion genes in cutaneous SCCs relative to precursors, including MMP1, MMP3, MMP9, LAMC2, LGALS1, and TNFRSF12A. Together, ocular and cutaneous squamous neoplasms demonstrate similar alterations, supporting a common model for neoplasia in UV-exposed epithelia. Treatment modalities useful for cutaneous SCC may also be effective in ocular SCC given the genetic similarity between these tumor types. Importantly, in both systems, precursor lesions possess the full complement of major genetic changes seen in SCC, supporting non-genetic drivers of invasiveness.
- Published
- 2020
- Full Text
- View/download PDF
30. Cancer SLC43A2 alters T cell methionine metabolism and histone methylation
- Author
-
Bian, Yingjie, Li, Wei, Kremer, Daniel M., Sajjakulnukit, Peter, Li, Shasha, Crespo, Joel, Nwosu, Zeribe C., Zhang, Li, Czerwonka, Arkadiusz, Pawłowska, Anna, Xia, Houjun, Li, Jing, Liao, Peng, Yu, Jiali, Vatan, Linda, Szeliga, Wojciech, Wei, Shuang, Grove, Sara, Liu, J. Rebecca, McLean, Karen, Cieslik, Marcin, Chinnaiyan, Arul M., Zgodziński, Witold, Wallner, Grzegorz, Wertel, Iwona, Okła, Karolina, Kryczek, Ilona, Lyssiotis, Costas A., and Zou, Weiping
- Abstract
Abnormal epigenetic patterns correlate with effector T cell malfunction in tumours1–4, but the cause of this link is unknown. Here we show that tumour cells disrupt methionine metabolism in CD8+T cells, thereby lowering intracellular levels of methionine and the methyl donor S-adenosylmethionine (SAM) and resulting in loss of dimethylation at lysine 79 of histone H3 (H3K79me2). Loss of H3K79me2 led to low expression of STAT5 and impaired T cell immunity. Mechanistically, tumour cells avidly consumed methionine and outcompeted T cells for methionine by expressing high levels of the methionine transporter SLC43A2. Genetic and biochemical inhibition of tumour SLC43A2 restored H3K79me2 in T cells, thereby boosting spontaneous and checkpoint-induced tumour immunity. Moreover, methionine supplementation improved the expression of H3K79me2 and STAT5 in T cells, and this was accompanied by increased T cell immunity in tumour-bearing mice and patients with colon cancer. Clinically, tumour SLC43A2 correlated negatively with T cell histone methylation and functional gene signatures. Our results identify a mechanistic connection between methionine metabolism, histone patterns, and T cell immunity in the tumour microenvironment. Thus, cancer methionine consumption is an immune evasion mechanism, and targeting cancer methionine signalling may provide an immunotherapeutic approach.
- Published
- 2020
- Full Text
- View/download PDF
31. The DNA methylation landscape of advanced prostate cancer
- Author
-
Zhao, Shuang G., Chen, William S., Li, Haolong, Foye, Adam, Zhang, Meng, Sjöström, Martin, Aggarwal, Rahul, Playdle, Denise, Liao, Arnold, Alumkal, Joshi J., Das, Rajdeep, Chou, Jonathan, Hua, Junjie T., Barnard, Travis J., Bailey, Adina M., Chow, Eric D., Perry, Marc D., Dang, Ha X., Yang, Rendong, Moussavi-Baygi, Ruhollah, Zhang, Li, Alshalalfa, Mohammed, Laura Chang, S., Houlahan, Kathleen E., Shiah, Yu-Jia, Beer, Tomasz M., Thomas, George, Chi, Kim N., Gleave, Martin, Zoubeidi, Amina, Reiter, Robert E., Rettig, Matthew B., Witte, Owen, Yvonne Kim, M., Fong, Lawrence, Spratt, Daniel E., Morgan, Todd M., Bose, Rohit, Huang, Franklin W., Li, Hui, Chesner, Lisa, Shenoy, Tanushree, Goodarzi, Hani, Asangani, Irfan A., Sandhu, Shahneen, Lang, Joshua M., Mahajan, Nupam P., Lara, Primo N., Evans, Christopher P., Febbo, Phillip, Batzoglou, Serafim, Knudsen, Karen E., He, Housheng H., Huang, Jiaoti, Zwart, Wilbert, Costello, Joseph F., Luo, Jianhua, Tomlins, Scott A., Wyatt, Alexander W., Dehm, Scott M., Ashworth, Alan, Gilbert, Luke A., Boutros, Paul C., Farh, Kyle, Chinnaiyan, Arul M., Maher, Christopher A., Small, Eric J., Quigley, David A., and Feng, Felix Y.
- Abstract
Although DNA methylation is a key regulator of gene expression, the comprehensive methylation landscape of metastatic cancer has never been defined. Through whole-genome bisulfite sequencing paired with deep whole-genome and transcriptome sequencing of 100 castration-resistant prostate metastases, we discovered alterations affecting driver genes that were detectable only with integrated whole-genome approaches. Notably, we observed that 22% of tumors exhibited a novel epigenomic subtype associated with hypermethylation and somatic mutations in TET2, DNMT3B, IDH1and BRAF. We also identified intergenic regions where methylation is associated with RNA expression of the oncogenic driver genes AR, MYCand ERG. Finally, we showed that differential methylation during progression preferentially occurs at somatic mutational hotspots and putative regulatory regions. This study is a large integrated study of whole-genome, whole-methylome and whole-transcriptome sequencing in metastatic cancer that provides a comprehensive overview of the important regulatory role of methylation in metastatic castration-resistant prostate cancer.
- Published
- 2020
- Full Text
- View/download PDF
32. PAX8 expression and TERTpromoter mutations in the nested variant of urothelial carcinoma: a clinicopathologic study with immunohistochemical and molecular correlates
- Author
-
Taylor, Alexander S., McKenney, Jesse K., Osunkoya, Adeboye O., Chan, May P., Al-Ahmadie, Hikmat A., Spratt, Daniel E., Fullen, Douglas R., Chinnaiyan, Arul M, Brown, Noah A., and Mehra, Rohit
- Abstract
The nested variant of urothelial carcinoma, a frequent mimic of benign lesions on limited specimens, has been associated with high-stage disease including metastases at presentation. While PAX8 immunohistochemistry has been noted to be infrequently present in urothelial carcinoma in general, it has not been studied specifically in a cohort of nested urothelial carcinomas. Furthermore, TERTpromoter mutation status is a potentially valuable biomarker for diagnosis of urothelial carcinoma and for noninvasive disease monitoring that has been observed in a majority of urothelial carcinoma and has previously been seen to be prevalent in multiple variant morphologies of urothelial carcinoma, including the nested variant. Twenty-five primary and three metastatic samples of nested urothelial carcinoma, along with 16 benign cases, were identified in a multicenter retrospective record review. PAX8 immunohistochemical stain was performed on all cases. In addition, TERTmutation analysis by allele-specific PCR was performed on 21 of the primary nested urothelial carcinoma cases and all benign cases. Positive PAX8 expression was identified in 52% (13 of 25) primary cases and 67% (2 of 3) metastatic cases of nested urothelial carcinoma; 50% (1 of 2) cases of large nested urothelial carcinoma were positive for PAX8. PAX8 expression was negative in the benign urothelium in all cases. TERTpromoter mutation was observed in 83% (15 of 18) nested urothelial carcinoma cases and in 6% (1 of 16) of the benign cases. Recognition of the prevalence of positive PAX8 staining in this clinically relevant variant of urothelial carcinoma is essential to avoiding inaccurate or delayed diagnosis during the diagnostic workup of bladder lesions suspicious for nested variant of urothelial carcinoma. Moreover, the prevalence of TERTpromoter mutations in nested urothelial carcinoma is similar to that of conventional urothelial carcinoma, further supporting its use as a biomarker that is stable across morphologic variants of urothelial carcinoma.
- Published
- 2020
- Full Text
- View/download PDF
33. PAX8 expression and TERTpromoter mutations in the nested variant of urothelial carcinoma: a clinicopathologic study with immunohistochemical and molecular correlates
- Author
-
Taylor, Alexander S., McKenney, Jesse K., Osunkoya, Adeboye O., Chan, May P., Al-Ahmadie, Hikmat A., Spratt, Daniel E., Fullen, Douglas R., Chinnaiyan, Arul M, Brown, Noah A., and Mehra, Rohit
- Abstract
The nested variant of urothelial carcinoma, a frequent mimic of benign lesions on limited specimens, has been associated with high-stage disease including metastases at presentation. While PAX8 immunohistochemistry has been noted to be infrequently present in urothelial carcinoma in general, it has not been studied specifically in a cohort of nested urothelial carcinomas. Furthermore, TERTpromoter mutation status is a potentially valuable biomarker for diagnosis of urothelial carcinoma and for noninvasive disease monitoring that has been observed in a majority of urothelial carcinoma and has previously been seen to be prevalent in multiple variant morphologies of urothelial carcinoma, including the nested variant. Twenty-five primary and three metastatic samples of nested urothelial carcinoma, along with 16 benign cases, were identified in a multicenter retrospective record review. PAX8 immunohistochemical stain was performed on all cases. In addition, TERTmutation analysis by allele-specific PCR was performed on 21 of the primary nested urothelial carcinoma cases and all benign cases. Positive PAX8 expression was identified in 52% (13 of 25) primary cases and 67% (2 of 3) metastatic cases of nested urothelial carcinoma; 50% (1 of 2) cases of large nested urothelial carcinoma were positive for PAX8. PAX8 expression was negative in the benign urothelium in all cases. TERTpromoter mutation was observed in 83% (15 of 18) nested urothelial carcinoma cases and in 6% (1 of 16) of the benign cases. Recognition of the prevalence of positive PAX8 staining in this clinically relevant variant of urothelial carcinoma is essential to avoiding inaccurate or delayed diagnosis during the diagnostic workup of bladder lesions suspicious for nested variant of urothelial carcinoma. Moreover, the prevalence of TERTpromoter mutations in nested urothelial carcinoma is similar to that of conventional urothelial carcinoma, further supporting its use as a biomarker that is stable across morphologic variants of urothelial carcinoma.
- Published
- 2020
- Full Text
- View/download PDF
34. Expression of L1 Cell Adhesion Molecule, a Nephronal Principal Cell Marker, in Nephrogenic Adenoma
- Author
-
Mannan, Rahul, Wang, Xiaoming, Mahapatra, Somnath, Wang, Susanna, Chinnaiyan, Anya K., Skala, Stephanie L., Zhang, Yuping, McMurray, Lisa M., Zelenka-Wang, Sylvia, Cao, Xuhong, Sangoi, Ankur R., Dadhania, Vipulkumar, Picken, Maria M., Menon, Santosh, Al-Ahmadie, Hikmat, Chinnaiyan, Arul M., Dhanasekaran, Saravana M., and Mehra, Rohit
- Abstract
Nephrogenic adenoma (NA) is a benign, reactive lesion seen predominantly in the urinary bladder and often associated with antecedent inflammation, instrumentation, or an operative history. Its histopathologic diversity can create diagnostic dilemmas and pathologists use morphologic evaluation along with available immunohistochemical (IHC) markers to navigate these challenges. IHC assays currently do not designate or specify NA’s potential putative cell of origin. Leveraging single-cell RNA-sequencing technology, we nominated a principal (P) cell–collecting duct marker, L1 cell adhesion molecule (L1CAM), as a potential biomarker for NA. IHC characterization revealed L1CAM to be positive in all 35 (100%) patient samples of NA; negative expression was seen in the benign urothelium, benign prostatic glands, urothelial carcinoma in situ, prostatic adenocarcinoma, majority of high-grade urothelial carcinoma, and metastatic urothelial carcinoma. In the study, we also used single-cell RNA sequencing to nominate a novel compendium of biomarkers specific for the proximal tubule, loop of Henle, and distal tubule (DT) (including P and intercalated cells), which can be used to perform nephronal mapping using RNA in situ hybridization and IHC technology. Employing this technique on NA we found enrichment of both the P-cell marker L1CAM and, the proximal tubule type-A and -B cell markers, PDZKI1P1and PIGR,respectively. The cell-type markers for the intercalated cell of DTs (LINC01187and FOXI1), and the loop of Henle (UMODand IRX5), were found to be uniformly absent in NA. Overall, our findings show that based on cell type–specific implications of L1CAM expression, the shared expression pattern of L1CAM between DT P cells and NA. L1CAM expression will be of potential value in assisting surgical pathologists toward a diagnosis of NA in challenging patient samples.
- Published
- 2024
- Full Text
- View/download PDF
35. Author Correction: Targeting SWI/SNF ATPases in enhancer-addicted prostate cancer
- Author
-
Xiao, Lanbo, Parolia, Abhijit, Qiao, Yuanyuan, Bawa, Pushpinder, Eyunni, Sanjana, Mannan, Rahul, Carson, Sandra E., Chang, Yu, Wang, Xiaoju, Zhang, Yuping, Vo, Josh N., Kregel, Steven, Simko, Stephanie A., Delekta, Andrew D., Jaber, Mustapha, Zheng, Heng, Apel, Ingrid J., McMurry, Lisa, Su, Fengyun, Wang, Rui, Zelenka-Wang, Sylvia, Sasmal, Sanjita, Khare, Leena, Mukherjee, Subhendu, Abbineni, Chandrasekhar, Aithal, Kiran, Bhakta, Mital S., Ghurye, Jay, Cao, Xuhong, Navone, Nora M., Nesvizhskii, Alexey I., Mehra, Rohit, Vaishampayan, Ulka, Blanchette, Marco, Wang, Yuzhuo, Samajdar, Susanta, Ramachandra, Murali, and Chinnaiyan, Arul M.
- Published
- 2024
- Full Text
- View/download PDF
36. A clonal expression biomarker associates with lung cancer mortality
- Author
-
Biswas, Dhruva, Birkbak, Nicolai J., Rosenthal, Rachel, Hiley, Crispin T., Lim, Emilia L., Papp, Krisztian, Boeing, Stefan, Krzystanek, Marcin, Djureinovic, Dijana, La Fleur, Linnea, Greco, Maria, Döme, Balázs, Fillinger, János, Brunnström, Hans, Wu, Yin, Moore, David A., Skrzypski, Marcin, Abbosh, Christopher, Litchfield, Kevin, Al Bakir, Maise, Watkins, Thomas B. K., Veeriah, Selvaraju, Wilson, Gareth A., Jamal-Hanjani, Mariam, Moldvay, Judit, Botling, Johan, Chinnaiyan, Arul M., Micke, Patrick, Hackshaw, Allan, Bartek, Jiri, Csabai, Istvan, Szallasi, Zoltan, Herrero, Javier, McGranahan, Nicholas, and Swanton, Charles
- Abstract
An aim of molecular biomarkers is to stratify patients with cancer into disease subtypes predictive of outcome, improving diagnostic precision beyond clinical descriptors such as tumor stage1. Transcriptomic intratumor heterogeneity (RNA-ITH) has been shown to confound existing expression-based biomarkers across multiple cancer types2–6. Here, we analyze multi-region whole-exome and RNA sequencing data for 156 tumor regions from 48 patients enrolled in the TRACERx study to explore and control for RNA-ITH in non-small cell lung cancer. We find that chromosomal instability is a major driver of RNA-ITH, and existing prognostic gene expression signatures are vulnerable to tumor sampling bias. To address this, we identify genes expressed homogeneously within individual tumors that encode expression modules of cancer cell proliferation and are often driven by DNA copy-number gains selected early in tumor evolution. Clonal transcriptomic biomarkers overcome tumor sampling bias, associate with survival independent of clinicopathological risk factors, and may provide a general strategy to refine biomarker design across cancer types.
- Published
- 2019
- Full Text
- View/download PDF
37. Distinct structural classes of activating FOXA1 alterations in advanced prostate cancer
- Author
-
Parolia, Abhijit, Cieslik, Marcin, Chu, Shih-Chun, Xiao, Lanbo, Ouchi, Takahiro, Zhang, Yuping, Wang, Xiaoju, Vats, Pankaj, Cao, Xuhong, Pitchiaya, Sethuramasundaram, Su, Fengyun, Wang, Rui, Feng, Felix Y., Wu, Yi-Mi, Lonigro, Robert J., Robinson, Dan R., and Chinnaiyan, Arul M.
- Abstract
Forkhead box A1 (FOXA1) is a pioneer transcription factor that is essential for the normal development of several endoderm-derived organs, including the prostate gland1,2. FOXA1 is frequently mutated in hormone-receptor-driven prostate, breast, bladder and salivary-gland tumours3–8. However, it is unclear how FOXA1 alterations affect the development of cancer, and FOXA1 has previously been ascribed both tumour-suppressive9–11and oncogenic12–14roles. Here we assemble an aggregate cohort of 1,546 prostate cancers and show that FOXA1 alterations fall into three structural classes that diverge in clinical incidence and genetic co-alteration profiles, with a collective prevalence of 35%. Class-1 activating mutations originate in early prostate cancer without alterations in ETS or SPOP, selectively recur within the wing-2 region of the DNA-binding forkhead domain, enable enhanced chromatin mobility and binding frequency, and strongly transactivate a luminal androgen-receptor program of prostate oncogenesis. By contrast, class-2 activating mutations are acquired in metastatic prostate cancers, truncate the C-terminal domain of FOXA1, enable dominant chromatin binding by increasing DNA affinity and—through TLE3 inactivation—promote metastasis driven by the WNT pathway. Finally, class-3 genomic rearrangements are enriched in metastatic prostate cancers, consist of duplications and translocations within the FOXA1locus, and structurally reposition a conserved regulatory element—herein denoted FOXA1 mastermind (FOXMIND)—to drive overexpression of FOXA1or other oncogenes. Our study reaffirms the central role of FOXA1 in mediating oncogenesis driven by the androgen receptor, and provides mechanistic insights into how the classes of FOXA1 alteration promote the initiation and/or metastatic progression of prostate cancer. These results have direct implications for understanding the pathobiology of other hormone-receptor-driven cancers and rationalize the co-targeting of FOXA1 activity in therapeutic strategies.
- Published
- 2019
- Full Text
- View/download PDF
38. Utility of ERG Immunohistochemistry for Evaluation of Lymphovascular Invasion in Testicular Germ Cell Tumors: A Retrospective Pilot Study
- Author
-
Udager, Aaron M., McHugh, Jonathan B., Morgan, Todd M., Spratt, Daniel E., Chinnaiyan, Arul M., and Mehra, Rohit
- Abstract
Lymphovascular invasion (LVI) of testicular germ cell tumors (GCT) is an important stage-determining variable in the evaluation of radical orchiectomy specimens. ERG endothelial cell expression, as detected by immunohistochemistry (IHC), robustly highlights lymphovascular spaces, and thus, we sought to assess the utility of ERG IHC for evaluation of GCT LVI. Hematoxylin and eosin (H&E) slides from a retrospective cohort of 25 GCT radical orchiectomy specimens (emanating from a parent cohort of 159 radical orchiectomy GCT cases identified between 2003 and 2013) were reviewed, and sections with foci of positive or equivocal LVI were identified. ERG IHC was performed on sections off the surface of corresponding paraffin tissue blocks. All foci were then rescored as positive, equivocal, or negative for LVI based on ERG endothelial cell expression. Twenty-three and 13 foci were positive or equivocal for LVI by H&E staining, respectively. Among the H&E positive LVI foci, 20 (87%) were ERG IHC positive, whereas of the H&E equivocal LVI foci, 5 (38%) were ERG IHC positive, 3 (23%) were ERG IHC negative, and 2 (15%) were ERG IHC equivocal; all other foci were lost for evaluation. Overall, ERG IHC helped resolve the LVI status of 61% of foci deemed equivocal for LVI by H&E staining only. Although ERG IHC is useful in confirming definitive LVI status in a subset of GCT cases, the overall clinical impact of ERG IHC is limited for H&E equivocal LVI foci in this specific retrospective patient cohort. Overall, in carefully selected clinical scenarios, these data suggest a supportive role for ERG IHC in evaluation of GCT LVI in radical orchiectomy specimens.
- Published
- 2019
- Full Text
- View/download PDF
39. VSTM2AOverexpression Is a Sensitive and Specific Biomarker for Mucinous Tubular and Spindle Cell Carcinoma (MTSCC) of the Kidney
- Author
-
Wang, Lisha, Zhang, Yuping, Chen, Ying-Bei, Skala, Stephanie L., Al-Ahmadie, Hikmat A., Wang, Xiaoming, Cao, Xuhong, Veeneman, Brendan A., Chen, Jin, Cieślik, Marcin, Qiao, Yuanyuan, Su, Fengyun, Vats, Pankaj, Siddiqui, Javed, Xiao, Hong, Sadimin, Evita T., Epstein, Jonathan I., Zhou, Ming, Sangoi, Ankur R., Trpkov, Kiril, Osunkoya, Adeboye O., Giannico, Giovanna A., McKenney, Jesse K., Argani, Pedram, Tickoo, Satish K., Reuter, Victor E., Chinnaiyan, Arul M., Dhanasekaran, Saravana M., and Mehra, Rohit
- Abstract
Supplemental Digital Content is available in the text.Our recent study revealed recurrent chromosomal losses and somatic mutations of genes in the Hippo pathway in mucinous tubular and spindle cell carcinoma (MTSCC). Here, we performed an integrative analysis of 907 renal cell carcinoma (RCC) samples (combined from The Cancer Genome Atlas and in-house studies) and the Knepper data set of microdissected rat nephrons. We identified VSTM2Aand IRX5as novel cancer-specific and lineage-specific biomarkers in MTSCC. We then assessed their expression by RNA in situ hybridization (ISH) in 113 tumors, including 33 MTSCC, 40 type 1 papillary RCC, 8 type 2 papillary RCC, 2 unclassified RCC, 15 clear cell RCC, and 15 chromophobe RCC. Sensitivity and specificity were calculated as the area under the receiver operating characteristics curve (AUC). All MTSCC tumors demonstrated moderate to high expression of VSTM2A(mean ISH score=255). VSTM2Agene expression assessed by RNA sequencing strongly correlated with VSTM2AISH score (r2=0.81, P=0.00016). The majority of non-MTSCC tumors demonstrated negative or low expression of VSTM2A. IRX5, nominated as a lineage-specific biomarker, showed moderate to high expression in MTSCC tumors (mean ISH score=140). IRX5gene expression assessed by RNA sequencing strongly correlated with IRX5ISH score (r2=0.69, P=0.00291). VSTM2A(AUC: 99.2%) demonstrated better diagnostic efficacy than IRX5(AUC: 87.5%), and may thus serve as a potential diagnostic marker to distinguish tumors with overlapping histology. Furthermore, our results suggest MTSCC displays an overlapping phenotypic expression pattern with the loop of Henle region of normal nephrons.
- Published
- 2018
- Full Text
- View/download PDF
40. Deep learning integrates histopathology and proteogenomics at a pan-cancer level
- Author
-
Wang, Joshua M., Hong, Runyu, Demicco, Elizabeth G., Tan, Jimin, Lazcano, Rossana, Moreira, Andre L., Li, Yize, Calinawan, Anna, Razavian, Narges, Schraink, Tobias, Gillette, Michael A., Omenn, Gilbert S., An, Eunkyung, Rodriguez, Henry, Tsirigos, Aristotelis, Ruggles, Kelly V., Ding, Li, Robles, Ana I., Mani, D.R., Rodland, Karin D., Lazar, Alexander J., Liu, Wenke, Fenyö, David, Aguet, François, Akiyama, Yo, Anand, Shankara, Anurag, Meenakshi, Babur, Özgün, Bavarva, Jasmin, Birger, Chet, Birrer, Michael J., Cantley, Lewis C., Cao, Song, Carr, Steven A., Ceccarelli, Michele, Chan, Daniel W., Chinnaiyan, Arul M., Cho, Hanbyul, Chowdhury, Shrabanti, Cieslik, Marcin P., Clauser, Karl R., Colaprico, Antonio, Zhou, Daniel Cui, da Veiga Leprevost, Felipe, Day, Corbin, Dhanasekaran, Saravana M., Domagalski, Marcin J., Dou, Yongchao, Druker, Brian J., Edwards, Nathan, Ellis, Matthew J., Selvan, Myvizhi Esai, Foltz, Steven M., Francis, Alicia, Geffen, Yifat, Getz, Gad, Gonzalez Robles, Tania J., Gosline, Sara J.C., Gümüş, Zeynep H., Heiman, David I., Hiltke, Tara, Hostetter, Galen, Hu, Yingwei, Huang, Chen, Huntsman, Emily, Iavarone, Antonio, Jaehnig, Eric J., Jewell, Scott D., Ji, Jiayi, Jiang, Wen, Johnson, Jared L., Katsnelson, Lizabeth, Ketchum, Karen A., Kolodziejczak, Iga, Krug, Karsten, Kumar-Sinha, Chandan, Lei, Jonathan T., Liang, Wen-Wei, Liao, Yuxing, Lindgren, Caleb M., Liu, Tao, Ma, Weiping, Rodrigues, Fernanda Martins, McKerrow, Wilson, Mesri, Mehdi, Nesvizhskii, Alexey I., Newton, Chelsea J., Oldroyd, Robert, Paulovich, Amanda G., Payne, Samuel H., Petralia, Francesca, Pugliese, Pietro, Reva, Boris, Rykunov, Dmitry, Satpathy, Shankha, Savage, Sara R., Schadt, Eric E., Schnaubelt, Michael, Schürer, Stephan, Shi, Zhiao, Smith, Richard D., Song, Xiaoyu, Song, Yizhe, Stathias, Vasileios, Storrs, Erik P., Terekhanova, Nadezhda V., Thangudu, Ratna R., Thiagarajan, Mathangi, Tignor, Nicole, Wang, Liang-Bo, Wang, Pei, Wang, Ying, Wen, Bo, Wiznerowicz, Maciej, Wu, Yige, Wyczalkowski, Matthew A., Yao, Lijun, Yaron, Tomer M., Yi, Xinpei, Zhang, Bing, Zhang, Hui, Zhang, Qing, Zhang, Xu, and Zhang, Zhen
- Abstract
We introduce a pioneering approach that integrates pathology imaging with transcriptomics and proteomics to identify predictive histology features associated with critical clinical outcomes in cancer. We utilize 2,755 H&E-stained histopathological slides from 657 patients across 6 cancer types from CPTAC. Our models effectively recapitulate distinctions readily made by human pathologists: tumor vs. normal (AUROC = 0.995) and tissue-of-origin (AUROC = 0.979). We further investigate predictive power on tasks not normally performed from H&E alone, including TP53 prediction and pathologic stage. Importantly, we describe predictive morphologies not previously utilized in a clinical setting. The incorporation of transcriptomics and proteomics identifies pathway-level signatures and cellular processes driving predictive histology features. Model generalizability and interpretability is confirmed using TCGA. We propose a classification system for these tasks, and suggest potential clinical applications for this integrated human and machine learning approach. A publicly available web-based platform implements these models.
- Published
- 2023
- Full Text
- View/download PDF
41. Comprehensive Evaluation of Programmed Death-Ligand 1 Expression in Primary and Metastatic Prostate Cancer
- Author
-
Haffner, Michael C., Guner, Gunes, Taheri, Diana, Netto, George J., Palsgrove, Doreen N., Zheng, Qizhi, Guedes, Liana Benevides, Kim, Kunhwa, Tsai, Harrison, Esopi, David M., Lotan, Tamara L., Sharma, Rajni, Meeker, Alan K., Chinnaiyan, Arul M., Nelson, William G., Yegnasubramanian, Srinivasan, Luo, Jun, Mehra, Rohit, Antonarakis, Emmanuel S., Drake, Charles G., and De Marzo, Angelo M.
- Abstract
Antibodies targeting the programmed cell death protein 1/programmed death-ligand 1 (PD-L1) interaction have shown clinical activity in multiple cancer types. PD-L1 protein expression is a clinically validated predictive biomarker of response for such therapies. Prior studies evaluating the expression of PD-L1 in primary prostate cancers have reported highly variable rates of PD-L1 positivity. In addition, limited data exist on PD-L1 expression in metastatic castrate-resistant prostate cancer (mCRPC). Here, we determined PD-L1 protein expression by immunohistochemistry using a validated PD-L1–specific antibody (SP263) in a large and representative cohort of primary prostate cancers and prostate cancer metastases. The study included 539 primary prostate cancers comprising 508 acinar adenocarcinomas, 24 prostatic duct adenocarcinomas, 7 small-cell carcinomas, and a total of 57 cases of mCRPC. PD-L1 positivity was low in primary acinar adenocarcinoma, with only 7.7% of cases showing detectable PD-L1 staining. Increased levels of PD-L1 expression were noted in 42.9% of small-cell carcinomas. In mCRPC, 31.6% of cases showed PD-L1–specific immunoreactivity. In conclusion, in this comprehensive evaluation of PD-L1 expression in prostate cancer, PD-L1 expression is rare in primary prostate cancers, but increased rates of PD-L1 positivity were observed in mCRPC. These results will be important for the future clinical development of programmed cell death protein 1/PD-L1–targeting therapies in prostate cancer.
- Published
- 2018
- Full Text
- View/download PDF
42. Cancer transcriptome profiling at the juncture of clinical translation
- Author
-
Cieślik, Marcin and Chinnaiyan, Arul M.
- Abstract
Methodological breakthroughs over the past four decades have repeatedly revolutionized transcriptome profiling. Using RNA sequencing (RNA-seq), it has now become possible to sequence and quantify the transcriptional outputs of individual cells or thousands of samples. These transcriptomes provide a link between cellular phenotypes and their molecular underpinnings, such as mutations. In the context of cancer, this link represents an opportunity to dissect the complexity and heterogeneity of tumours and to discover new biomarkers or therapeutic strategies. Here, we review the rationale, methodology and translational impact of transcriptome profiling in cancer.
- Published
- 2018
- Full Text
- View/download PDF
43. Detection of 6 TFEB-amplified renal cell carcinomas and 25 renal cell carcinomas with MITF translocations: systematic morphologic analysis of 85 cases evaluated by clinical TFE3 and TFEB FISH assays
- Author
-
Skala, Stephanie L, Xiao, Hong, Udager, Aaron M, Dhanasekaran, Saravana M, Shukla, Sudhanshu, Zhang, Yang, Landau, Carrie, Shao, Lina, Roulston, Diane, Wang, Lisha, Siddiqui, Javed, Cao, Xuhong, Magi-Galluzzi, Cristina, Zhang, Miao, Osunkoya, Adeboye O, Smith, Steven C, McKenney, Jesse K, Betz, Bryan L, Myers, Jeffrey L, Chinnaiyan, Arul M, Tomlins, Scott A, and Mehra, Rohit
- Abstract
Renal cell carcinomas with MITF aberrations demonstrate a wide morphologic spectrum, highlighting the need to consider these entities within the differential diagnosis of renal tumors encountered in clinical practice. Herein, we describe our experience with application of clinical fluorescence in situ hybridization (FISH) assays for detection of TFE3 and TFEB gene aberrations from 85 consecutive renal cell carcinoma cases submitted to our genitourinary FISH service. Results from 170 FISH assays performed on these tumors were correlated with available clinicopathologic findings. Ninety-eight percent of renal tumors submitted for FISH evaluation were from adult patients. Thirty-one (37%) tumors were confirmed to demonstrate MITF aberrations (21 TFE3 translocation, 4 TFEB translocation, and 6 TFEB amplification cases). Overall, renal cell carcinomas with MITF aberrations demonstrated morphologic features overlapping with clear cell, papillary, or clear cell papillary renal cell carcinomas. Renal cell carcinomas with MITF aberrations were significantly more likely to demonstrate dual (eosinophilic and clear) cytoplasmic tones (P=0.030), biphasic TFEB translocation renal cell carcinoma-like morphology (P=0.002), psammomatous calcifications (P=0.002), and nuclear pseudoinclusions (P=0.001) than renal cell carcinomas without MITF aberrations. Notably, 7/9 (78%) renal cell carcinomas exhibiting subnuclear clearing and linear nuclear array (6 of which showed high World Health Organization/International Society of Urological Pathology nucleolar grade) demonstrated TFE3 translocation, an association that was statistically significant when compared with renal cell carcinomas without MITF aberrations (P=0.009). In this cohort comprising consecutive cases, TFEB-amplified renal cell carcinomas were more commonly identified than renal cell carcinomas with TFEB translocations, and four (67%) of these previously unreported TFEB-amplified renal cell carcinomas demonstrated oncocytic and papillary features with a high World Health Organization/International Society of Urological Pathology nucleolar grade. In summary, TFE3 and TFEB FISH evaluation aids in identification and accurate classification of renal cell carcinomas with MITF aberrations, including TFEB-amplified renal cell carcinoma, which may demonstrate aggressive behavior.
- Published
- 2018
- Full Text
- View/download PDF
44. Detection of 6 TFEB-amplified renal cell carcinomas and 25 renal cell carcinomas with MITF translocations: systematic morphologic analysis of 85 cases evaluated by clinical TFE3and TFEBFISH assays
- Author
-
Skala, Stephanie L, Xiao, Hong, Udager, Aaron M, Dhanasekaran, Saravana M, Shukla, Sudhanshu, Zhang, Yang, Landau, Carrie, Shao, Lina, Roulston, Diane, Wang, Lisha, Siddiqui, Javed, Cao, Xuhong, Magi-Galluzzi, Cristina, Zhang, Miao, Osunkoya, Adeboye O, Smith, Steven C, McKenney, Jesse K, Betz, Bryan L, Myers, Jeffrey L, Chinnaiyan, Arul M, Tomlins, Scott A, and Mehra, Rohit
- Abstract
Renal cell carcinomas with MITF aberrations demonstrate a wide morphologic spectrum, highlighting the need to consider these entities within the differential diagnosis of renal tumors encountered in clinical practice. Herein, we describe our experience with application of clinical fluorescence in situhybridization (FISH) assays for detection of TFE3and TFEBgene aberrations from 85 consecutive renal cell carcinoma cases submitted to our genitourinary FISH service. Results from 170 FISH assays performed on these tumors were correlated with available clinicopathologic findings. Ninety-eight percent of renal tumors submitted for FISH evaluation were from adult patients. Thirty-one (37%) tumors were confirmed to demonstrate MITF aberrations (21 TFE3translocation, 4 TFEBtranslocation, and 6 TFEBamplification cases). Overall, renal cell carcinomas with MITF aberrations demonstrated morphologic features overlapping with clear cell, papillary, or clear cell papillary renal cell carcinomas. Renal cell carcinomas with MITF aberrations were significantly more likely to demonstrate dual (eosinophilic and clear) cytoplasmic tones (P=0.030), biphasic TFEBtranslocation renal cell carcinoma-like morphology (P=0.002), psammomatous calcifications (P=0.002), and nuclear pseudoinclusions (P=0.001) than renal cell carcinomas without MITF aberrations. Notably, 7/9 (78%) renal cell carcinomas exhibiting subnuclear clearing and linear nuclear array (6 of which showed high World Health Organization/International Society of Urological Pathology nucleolar grade) demonstrated TFE3translocation, an association that was statistically significant when compared with renal cell carcinomas without MITF aberrations (P=0.009). In this cohort comprising consecutive cases, TFEB-amplified renal cell carcinomas were more commonly identified than renal cell carcinomas with TFEBtranslocations, and four (67%) of these previously unreported TFEB-amplified renal cell carcinomas demonstrated oncocytic and papillary features with a high World Health Organization/International Society of Urological Pathology nucleolar grade. In summary, TFE3and TFEBFISH evaluation aids in identification and accurate classification of renal cell carcinomas with MITF aberrations, including TFEB-amplified renal cell carcinoma, which may demonstrate aggressive behavior.
- Published
- 2018
- Full Text
- View/download PDF
45. Associations of Luminal and Basal Subtyping of Prostate Cancer With Prognosis and Response to Androgen Deprivation Therapy
- Author
-
Zhao, Shuang G., Chang, S. Laura, Erho, Nicholas, Yu, Menggang, Lehrer, Jonathan, Alshalalfa, Mohammed, Speers, Corey, Cooperberg, Matthew R., Kim, Won, Ryan, Charles J., Den, Robert B., Freedland, Stephen J., Posadas, Edwin, Sandler, Howard, Klein, Eric A., Black, Peter, Seiler, Roland, Tomlins, Scott A., Chinnaiyan, Arul M., Jenkins, Robert B., Davicioni, Elai, Ross, Ashley E., Schaeffer, Edward M., Nguyen, Paul L., Carroll, Peter R., Karnes, R. Jeffrey, Spratt, Daniel E., and Feng, Felix Y.
- Abstract
IMPORTANCE: There is a clear need for a molecular subtyping approach in prostate cancer to identify clinically distinct subgroups that benefit from specific therapies. OBJECTIVES: To identify prostate cancer subtypes based on luminal and basal lineage and to determine associations with clinical outcomes and response to treatment. DESIGN, SETTING, AND PARTICIPANTS: The PAM50 classifier was used to subtype 1567 retrospectively collected (median follow-up, 10 years) and 2215 prospectively collected prostate cancer samples into luminal- and basal-like subtypes. MAIN OUTCOMES AND MEASURES: Metastasis, biochemical recurrence, overall survival, prostate cancer–specific survival, associations with biological pathways, and clinicopathologic variables were the main outcomes. RESULTS: Among the 3782 samples, the PAM50 classifier consistently segregated prostate cancer into 3 subtypes in both the retrospective and prospective cohorts: luminal A (retrospective, 538 [34.3%]; prospective, 737 [33.3%]), luminal B (retrospective, 447 [28.5%]; prospective, 723 [32.6%]), and basal (retrospective, 582 [37.1%]; prospective, 755 [34.1%]). Known luminal lineage markers, such as NKX3.1 and KRT18, were enriched in luminal-like cancers, and the basal lineage CD49f signature was enriched in basal-like cancers, demonstrating the connection between these subtypes and established prostate cancer biology. In the retrospective cohort, luminal B prostate cancers exhibited the poorest clinical prognoses on both univariable and multivariable analyses accounting for standard clinicopathologic prognostic factors (10-year biochemical recurrence-free survival [bRFS], 29%; distant metastasis-free survival [DMFS], 53%; prostate cancer-specific survival [PCSS], 78%; overall survival [OS], 69%), followed by basal prostate cancers (10-year bRFS, 39%; DMFS, 73%; PCSS, 86%; OS, 80%) and luminal A prostate cancers (10-year bRFS, 41%; DMFS, 73%; PCSS, 89%; OS, 82%). Although both luminal-like subtypes were associated with increased androgen receptor expression and signaling, only luminal B prostate cancers were significantly associated with postoperative response to androgen deprivation therapy (ADT) in a subset analysis in our retrospective cohorts (n = 315) matching patients based on clinicopathologic variables (luminal B 10-year metastasis: treated, 33% vs untreated, 55%; nonluminal B 10-year metastasis: treated, 37% vs untreated, 21%; P = .006 for interaction). CONCLUSIONS AND RELEVANCE: Luminal- and basal-like prostate cancers demonstrate divergent clinical behavior, and patients with luminal B tumors respond better to postoperative ADT than do patients with non–luminal B tumors. These findings contribute novel insight into prostate cancer biology, providing a potential clinical tool to personalize ADT treatment for prostate cancer by predicting which men may benefit from ADT after surgery.
- Published
- 2017
- Full Text
- View/download PDF
46. Prognostic Value of Percent Gleason Grade 4 at Prostate Biopsy in Predicting Prostatectomy Pathology and Recurrence.
- Author
-
Cole, Adam I., Morgan, Todd M., Spratt, Daniel E., Palapattu, Ganesh S., He, Chang, Tomlins, Scott A., Weizer, Alon Z., Feng, Felix Y., Wu, Angela, Siddiqui, Javed, Chinnaiyan, Arul M., Montgomery, Jeffrey S., Kunju, Lakshmi P., Miller, David C., Hollenbeck, Brent K., Wei, John T., and Mehra, Rohit
- Subjects
PROSTATE cancer treatment ,PROSTATECTOMY ,GLEASON grading system ,PROSTATE biopsy ,CANCER relapse ,PROSTATE cancer prognosis - Abstract
Purpose The importance of primary Gleason grade among men with Gleason score 7 disease has been well-defined. However, this dichotomization may oversimplify the continuous spectrum of absolute percent Gleason grade 4 disease (G4%). In this study we report the prognostic value of G4% in cancer related outcomes of men undergoing radical prostatectomy. Materials and Methods Patients who underwent radical prostatectomy for clinically localized Gleason 6-8 prostate cancer from 2005 to 2013 were included in the study. G4% was determined as biopsy tumor length containing Gleason pattern 4/total tumor length, which performed better than alternative quantifications of pattern 4 involvement. G4% was correlated with time to biochemical recurrence and presence of adverse radical prostatectomy pathology, defined as primary Gleason 4 or pT3 or greater, by multivariable Cox and logistic regressions. Results Of 1,691 patients 517 (30.6%) had adverse pathological features and 86 (5.6%) experienced biochemical recurrence. On multivariable analyses G4% was a significant predictor of adverse pathology (OR 1.04, 95% CI 1.03–1.05) and time to biochemical recurrence (HR 1.02, CI 1.01–1.03). G4% was also a significant independent predictor of adverse pathology in subsets of patients with Gleason score 7 (OR 1.05, 95% CI 1.03–1.06), 3+4 (OR 1.06, 95% CI 1.04–1.08) and 4+3 cancer (OR 1.05, 95% CI 1.03–1.06). We found a significantly increased risk of adverse pathology at potentially meaningful G4% thresholds (1% to 10% vs 20% to 30%). Conclusions The incremental percentage of Gleason grade 4 disease in biopsy specimens is an important predictor of adverse pathology and biochemical recurrence across the entire range of G4% disease. Accounting for G4% can improve risk assessment even among those patients with Gleason 3+4 or 4+3 cancer and may help inform patient counseling. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
47. Role of BET proteins in castration-resistant prostate cancer.
- Author
-
Fernandez-Salas, Ester, Wang, Shaomeng, and Chinnaiyan, Arul M
- Subjects
PROSTATE cancer treatment ,CASTRATION ,GENETIC transcription ,DRUG efficacy ,TARGETED drug delivery - Abstract
Castration resistant prostate cancer (CRPC) is a deadly disease with few therapeutic options once patients become resistant to second generation drugs targeting the AR-transcriptional program. The BET-BRD readers of chromatin are key regulators of AR-, ERG-, and c-Myc-mediated transcription in CRPC. BET-BRD inhibitors have demonstrated pre-clinical efficacy in models of CRPC and are currently being evaluated in several clinical trials. These novel drugs have the potential to transform the way we treat CRPC in the near future. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
48. Integrative clinical genomics of metastatic cancer
- Author
-
Robinson, Dan R., Wu, Yi-Mi, Lonigro, Robert J., Vats, Pankaj, Cobain, Erin, Everett, Jessica, Cao, Xuhong, Rabban, Erica, Kumar-Sinha, Chandan, Raymond, Victoria, Schuetze, Scott, Alva, Ajjai, Siddiqui, Javed, Chugh, Rashmi, Worden, Francis, Zalupski, Mark M., Innis, Jeffrey, Mody, Rajen J., Tomlins, Scott A., Lucas, David, Baker, Laurence H., Ramnath, Nithya, Schott, Ann F., Hayes, Daniel F., Vijai, Joseph, Offit, Kenneth, Stoffel, Elena M., Roberts, J. Scott, Smith, David C., Kunju, Lakshmi P., Talpaz, Moshe, Cieślik, Marcin, and Chinnaiyan, Arul M.
- Abstract
Metastasis is the primary cause of cancer-related deaths. Although The Cancer Genome Atlas has sequenced primary tumour types obtained from surgical resections, much less comprehensive molecular analysis is available from clinically acquired metastatic cancers. Here we perform whole-exome and -transcriptome sequencing of 500 adult patients with metastatic solid tumours of diverse lineage and biopsy site. The most prevalent genes somatically altered in metastatic cancer included TP53, CDKN2A, PTEN, PIK3CA, and RB1. Putative pathogenic germline variants were present in 12.2% of cases of which 75% were related to defects in DNA repair. RNA sequencing complemented DNA sequencing to identify gene fusions, pathway activation, and immune profiling. Our results show that integrative sequence analysis provides a clinically relevant, multi-dimensional view of the complex molecular landscape and microenvironment of metastatic cancers.
- Published
- 2017
- Full Text
- View/download PDF
49. Immunohistochemical Characterization of Fumarate Hydratase (FH) and Succinate Dehydrogenase (SDH) in Cutaneous Leiomyomas for Detection of Familial Cancer Syndromes
- Author
-
Carter, Cody S., Skala, Stephanie L., Chinnaiyan, Arul M., McHugh, Jonathan B., Siddiqui, Javed, Cao, Xuhong, Dhanasekaran, Saravana M., Fullen, Douglas R., Lagstein, Amir, Chan, May P., and Mehra, Rohit
- Abstract
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is caused by germline mutations in the FHgene, and is associated with increased incidence of leiomyomas and a potentially aggressive variant of renal cell carcinoma (HLRCC-associated RCC). Absent immunohistochemical expression of fumarate hydratase (FH) has previously been used to diagnose HLRCC-associated RCC, but immunohistochemical staining of leiomyomas is not standard practice. We performed immunohistochemistry (IHC) on whole sections from consecutive cutaneous leiomyomas from our archives to evaluate for both FH and succinate dehydrogenase B expression, in addition to clinicopathologic data collection and review of all hematoxylin and eosin–stained slides for blinded morphologic evaluation of features reported to be seen in HLRCC-associated uterine leiomyomas. Ninety-six cutaneous leiomyomas from 87 patients were identified; 12 of these specimens were from 7 patients with documented HLRCC. FH expression by IHC was absent in 9 specimens and retained in 85 specimens; 2 cases were equivocal with minimal FH expression. Seven of the 9 absent expression specimens were from patients with HLRCC, as were both of the equivocal specimens. The overall sensitivity and specificity of absent FH expression in leiomyomas for detection of patients with HLRCC were 70.0% and 97.6%, respectively. Inclusion of cases classified as equivocal increased sensitivity to 75.0%. Succinate dehydrogenase B expression was retained in 95 specimens and equivocal in 1 specimen. None of the evaluated morphologic features showed any association with leiomyomas in HLRCC. Loss of FH immunohistochemical expression in cutaneous leiomyomas is a sensitive and specific marker for detection of HLRCC, thus suggesting a role for prospective FH IHC in patients with these tumors to screen for HLRCC.
- Published
- 2017
- Full Text
- View/download PDF
50. Genome-Wide STAT3 Binding Analysis after Histone Deacetylase Inhibition Reveals Novel Target Genes in Dendritic Cells
- Author
-
Sun, Yaping, Iyer, Matthew, McEachin, Richard, Zhao, Meng, Wu, Yi-Mi, Cao, Xuhong, Oravecz-Wilson, Katherine, Zajac, Cynthia, Mathewson, Nathan, Wu, Shin-Rong Julia, Rossi, Corinne, Toubai, Tomomi, Qin, Zhaohui S., Chinnaiyan, Arul M., and Reddy, Pavan
- Abstract
STAT3 is a master transcriptional regulator that plays an important role in the induction of both immune activation and immune tolerance in dendritic cells (DCs). The transcriptional targets of STAT3 in promoting DC activation are becoming increasingly understood; however, the mechanisms underpinning its role in causing DC suppression remain largely unknown. To determine the functional gene targets of STAT3, we compared the genome-wide binding of STAT3 using ChIP sequencing coupled with gene expression microarrays to determine STAT3-dependent gene regulation in DCs after histone deacetylase (HDAC) inhibition. HDAC inhibition boosted the ability of STAT3 to bind to distinct DNA targets and regulate gene expression. Among the top 500 STAT3 binding sites, the frequency of canonical motifs was significantly higher than that of noncanonical motifs. Functional analysis revealed that after treatment with an HDAC inhibitor, the upregulated STAT3 target genes were those that were primarily the negative regulators of proinflammatory cytokines and those in the IL-10 signaling pathway. The downregulated STAT3-dependent targets were those involved in immune effector processes and antigen processing/presentation. The expression and functional relevance of these genes were validated. Specifically, functional studies confirmed that the upregulation of IL-10Ra by STAT3 contributed to the suppressive function of DCs following HDAC inhibition.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.