254 results on '"Hsieh JJ"'
Search Results
2. Who Should Grasp Decision-making Authority, Human Or Intelligent System?
- Author
-
Cui, Xiaocong, primary, Keil, Mark, additional, Chen, Liwei, additional, and Hsieh, JJ Po-An, additional
- Published
- 2021
- Full Text
- View/download PDF
3. Attaining Individual Creativity and Performance in Multi-Disciplinary and Geographically-Distributed it Project Teams: The Role of Transactive Memory Systems
- Author
-
Hsieh, JJ Po-An, primary
- Published
- 2021
- Full Text
- View/download PDF
4. How Does Employee Infusion Use of CRM Systems Drive Customer Satisfaction? Mechanism Differences between Face-To-Face and Virtual Channels
- Author
-
Chen, Liwei, primary, Hsieh, JJ Po-An, additional, Rai, Arun, additional, and Xu, Sean, additional
- Published
- 2021
- Full Text
- View/download PDF
5. Distance Makes the Heart Grow Fonder: How AI Implementation Influences Human Relationships.
- Author
-
Monds, Cazembe, Hsieh, JJ Po-An, and Chen, Liwei
- Abstract
As artificial intelligence (AI) systems are being integrated into our social and professional lives, there is a clear need for more research that explores how AI affects human relationships along the service-profit chain through its use for organizational processes. We explore how supervisor and employee interactions, important foundational aspects of the service-profit chain, are impacted with the introduction of AI systems, and how this change in turn influences customer perceptions of trust in service employees. We also examine the moderating effects of employee-perceived service climate and a difference in supervisor and employee education, and how these affect the relationship between supervisor and employee interaction difference and customer trust in employees. Using multiple waves and multiple sources of data, our findings posit that both employee-perceived service climate and supervisor and employee education differences moderate the relationship between supervisor and employee interaction difference and customer trust. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. The Cancer Genome Atlas Comprehensive Molecular Characterization of Renal Cell Carcinoma
- Author
-
Ricketts, CJ, de Cubas, AA, Fan, H, Smith, CC, Lang, M, Reznik, E, Bowlby, R, Gibb, EA, Akbani, R, Beroukhim, R, Bottaro, DP, Choueiri, TK, Gibbs, RA, Godwin, AK, Haake, S, Hakimi, AA, Henske, EP, Hsieh, JJ, Ho, TH, Kanchi, RS, Krishnan, B, Kwaitkowski, DJ, Lui, W, Merino, MJ, Mills, GB, Myers, J, Nickerson, ML, Reuter, VE, Schmidt, LS, Shelley, CS, Shen, H, Shuch, B, Signoretti, S, Srinivasan, R, Tamboli, P, Thomas, G, Vincent, BG, Vocke, CD, Wheeler, DA, Yang, L, Kim, WT, Robertson, AG, Spellman, PT, Rathmell, WK, Linehan, WM, Ricketts, CJ, de Cubas, AA, Fan, H, Smith, CC, Lang, M, Reznik, E, Bowlby, R, Gibb, EA, Akbani, R, Beroukhim, R, Bottaro, DP, Choueiri, TK, Gibbs, RA, Godwin, AK, Haake, S, Hakimi, AA, Henske, EP, Hsieh, JJ, Ho, TH, Kanchi, RS, Krishnan, B, Kwaitkowski, DJ, Lui, W, Merino, MJ, Mills, GB, Myers, J, Nickerson, ML, Reuter, VE, Schmidt, LS, Shelley, CS, Shen, H, Shuch, B, Signoretti, S, Srinivasan, R, Tamboli, P, Thomas, G, Vincent, BG, Vocke, CD, Wheeler, DA, Yang, L, Kim, WT, Robertson, AG, Spellman, PT, Rathmell, WK, and Linehan, WM
- Abstract
Renal cell carcinoma (RCC) is not a single disease, but several histologically defined cancers with different genetic drivers, clinical courses, and therapeutic responses. The current study evaluated 843 RCC from the three major histologic subtypes, including 488 clear cell RCC, 274 papillary RCC, and 81 chromophobe RCC. Comprehensive genomic and phenotypic analysis of the RCC subtypes reveals distinctive features of each subtype that provide the foundation for the development of subtype-specific therapeutic and management strategies for patients affected with these cancers. Somatic alteration of BAP1, PBRM1, and PTEN and altered metabolic pathways correlated with subtype-specific decreased survival, while CDKN2A alteration, increased DNA hypermethylation, and increases in the immune-related Th2 gene expression signature correlated with decreased survival within all major histologic subtypes. CIMP-RCC demonstrated an increased immune signature, and a uniform and distinct metabolic expression pattern identified a subset of metabolically divergent (MD) ChRCC that associated with extremely poor survival.
- Published
- 2018
7. Converting Expertise Dissimilarity to Creativity: Impacts of Team TMS and Geographical Dispersion
- Author
-
He, Wei, primary, Schroeder, Andreas, additional, Fang, Yulin, additional, and Hsieh, JJ Po-An, additional
- Published
- 2016
- Full Text
- View/download PDF
8. Genomic investigation of etiologic heterogeneity: Methodologic challenges
- Author
-
Begg, CB, Seshan, VE, Zabor, EC, Furberg, H, Arora, A, Shen, R, Maranchie, JK, Nielsen, ME, Rathmell, WK, Signoretti, S, Tamboli, P, Karam, JA, Choueiri, TK, Hakimi, AA, Hsieh, JJ, Begg, CB, Seshan, VE, Zabor, EC, Furberg, H, Arora, A, Shen, R, Maranchie, JK, Nielsen, ME, Rathmell, WK, Signoretti, S, Tamboli, P, Karam, JA, Choueiri, TK, Hakimi, AA, and Hsieh, JJ
- Abstract
Background: The etiologic heterogeneity of cancer has traditionally been investigated by comparing risk factor frequencies within candidate sub-types, defined for example by histology or by distinct tumor markers of interest. Increasingly tumors are being profiled for molecular features much more extensively. This greatly expands the opportunities for defining distinct sub-types. In this article we describe an exploratory analysis of the etiologic heterogeneity of clear cell kidney cancer. Data are available on the primary known risk factors for kidney cancer, while the tumors are characterized on a genome-wide basis using expression, methylation, copy number and mutational profiles. Methods: We use a novel clustering strategy to identify sub-types. This is accomplished independently for the expression, methylation and copy number profiles. The goals are to identify tumor sub-types that are etiologically distinct, to identify the risk factors that define specific sub-types, and to endeavor to characterize the key genes that appear to represent the principal features of the distinct sub-types. Results: The analysis reveals strong evidence that gender represents an important factor that distinguishes disease sub-types. The sub-types defined using expression data and methylation data demonstrate considerable congruence and are also clearly correlated with mutations in important cancer genes. These sub-types are also strongly correlated with survival. The complexity of the data presents many analytical challenges including, prominently, the risk of false discovery. Conclusions: Genomic profiling of tumors offers the opportunity to identify etiologically distinct sub-types, paving the way for a more refined understanding of cancer etiology.
- Published
- 2014
9. Leverage Points for Addressing Digital Inequality: An Extended Theory of Planned Behavior Perspective
- Author
-
Hsieh, JJ Po-An
- Abstract
Digital inequality, or the disparity in the access and use of information and communication technologies (ICT), is one of the most critical issues in the knowledge economy. This inequality prevents under-privileged people from exploring digital opportunities to enhance their life quality. Governments, business, and the public have devoted tremendous resources to address this issue, but the results are inconclusive. Theoretical understanding, complemented with theory-based empirical assessment of the phenomenon, is essential to inform effective policy-making and interventions. This dissertation explored the key factors that lead to the inequality in the access and use of ICT, particularly the high-speed Internet, between the privileged and under-privileged. I applied a belief-based perspective to understand how distinctive beliefs concerning ICT acceptance differentially influence under-privileged and privileged people?s innovation decision and behavior at different stages of the implementation process. A theoretical model that drew upon the Theory of Planned Behavior, Motivation Theory, Social Learning Theory, Diffusion of Innovation, and Trust was developed to explain how cognitive, social, behavioral, and institutional factors inform digital inequality as a whole. The conceptual model and forwarded hypotheses in the dissertation were empirically tested using data collected from a large-scale field survey. The survey investigated the adoption and usage behavior of residents in the city of LaGrange, Georgia where the city government, aiming to address digital inequality, provided high-speed Internet connection and devices to residents at no cost. A complementary case study was subsequently conducted to examine a multi-stage process model in which various barriers and facilitators may prevent or promote the progress of individuals? ICT innovation. The results of this research reveal valuable insights into the differential patterns of ICT access and usage, and the key factors that cause them, for under-privileged and privileged people. The findings, in turn, suggest a segmentation and stepwise technology implementation strategy for people with different backgrounds and at different stages of their innovation processes. This dissertation makes several notable contributions for both researchers and practitioners. First, the dissertation contributes a holistic and theoretically grounded perspective that extends beyond the technology-centered view in most digital inequality studies. It also highlights the multifaceted nature of the phenomenon. As such, this research meets the challenge set forward by notable researchers to develop theoretical models capable of revealing the complexity embedded in this issue. Second, the dissertation presents a unifying theory reflected upon adoption and diffusion of innovation. Testing theories in the context of digital inequality extends and complements our existing knowledge about these related fields. Most importantly, the empirical findings derived from the rich data set identity powerful leverage points for stimulating the adoption and use of ICT among the under-privileged. With such insights, practitioners, particularly policy-makers and service providers, can formulate effective interventions to address the problem of digital inequality.
- Published
- 2006
- Full Text
- View/download PDF
10. Expertise dissimilarity and creativity: The contingent roles of tacit and explicit knowledge sharing.
- Author
-
Huang, Xu, primary, Hsieh, JJ Po-An, additional, and He, Wei, additional
- Published
- 2014
- Full Text
- View/download PDF
11. The Credibility Crisis in IS: A Global Stakeholder Perspective
- Author
-
Looney, Clayton Arlen, primary, Firth, David, additional, Koch, Hope, additional, Cecez-Kecmanovic, Dubravka, additional, Hsieh, JJ Po-An, additional, Soh, Christina, additional, Valacich, Joseph S., additional, and Whitley, Edgar A., additional
- Published
- 2014
- Full Text
- View/download PDF
12. The contingent effect of personal IT innovativeness and IT self-efficacy on innovative use of complex IT
- Author
-
Wang, Wei, primary, Li, Xixi, additional, and Hsieh, JJ Po-An, additional
- Published
- 2013
- Full Text
- View/download PDF
13. Exploring the Zone of Tolerance for Internal Customers in IT-Enabled Call Centers
- Author
-
Hsieh, JJ Po-An, primary, Sharma, Piyush, additional, Rai, Arun, additional, and Parasuraman, A., additional
- Published
- 2013
- Full Text
- View/download PDF
14. Understanding User Satisfaction With Instant Messaging: An Empirical Survey Study
- Author
-
Wang, Wei, primary, Hsieh, JJ Po-An, additional, and Song, Baoxiang, additional
- Published
- 2012
- Full Text
- View/download PDF
15. Innovate with Complex Information Technologies: A Theoretical Model and Empirical Examination
- Author
-
Wang, Wei, primary, Butler, John E., additional, Hsieh, Jj Po-An, additional, and Hsu, Sheng-Hsun, additional
- Published
- 2008
- Full Text
- View/download PDF
16. Production of multiple cytokines and induction of cachexia in athymic nude mice by a new anaplastic thyroid carcinoma cell line
- Author
-
Chang, JW, primary, Yeh, KY, additional, Shen, YC, additional, Hsieh, JJ, additional, Chuang, CK, additional, Liao, SK, additional, Tsai, LH, additional, and Wang, CH, additional
- Published
- 2003
- Full Text
- View/download PDF
17. Public IT Policies in Less Developed Countries: A Critical Assessment of the Literature and a Reference Framework for Future Work
- Author
-
Checchi, Ricardo M., primary, Hsieh, JJ Po-An, additional, and Straub, Detmar, additional
- Published
- 2003
- Full Text
- View/download PDF
18. Clinical implications of high MET gene dosage in non-small cell lung cancer patients without previous tyrosine kinase inhibitor treatment.
- Author
-
Chen YT, Chang JW, Liu HP, Yu TF, Chiu YT, Hsieh JJ, Chen YR, Wu HD, Huang SF, Chen, Ya-Ting, Chang, John Wen-Cheng, Liu, Hui-Ping, Yu, Tsung-Fu, Chiu, Yu-Ting, Hsieh, Jia-Juan, Chen, Ying-Tsong, Chen, Yi-Rong, Wu, Hong-Dar Isaac, and Huang, Shiu-Feng
- Published
- 2011
- Full Text
- View/download PDF
19. FXR deletion attenuates intestinal barrier dysfunction in murine acute intestinal inflammation.
- Author
-
O'Guinn ML, Handler DA, Hsieh JJ, Mallicote MU, Feliciano K, and Gayer CP
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Male, Inflammation metabolism, Inflammation genetics, Tight Junctions metabolism, Tight Junctions pathology, Disease Models, Animal, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, Cytoplasmic and Nuclear genetics, Receptors, Cytoplasmic and Nuclear deficiency, Mice, Knockout, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Lipopolysaccharides toxicity
- Abstract
Accumulating literature suggests that the farnesoid-X receptor (FXR), a nuclear bile acid receptor best known for its role in bile acid homeostasis, is also a potent context-dependent regulator of inflammation. FXR may thus be relevant to several intestinal disease states including inflammatory bowel disease, necrotizing enterocolitis, and sepsis. In this study, we tested the effects of FXR deletion on acute murine intestinal inflammation. We found that FXR knockout (KO) mice were protected from intestinal injury and barrier dysfunction induced by lipopolysaccharide (LPS) injection, dithizone (DI)/ Klebsiella , and cecal ligation/puncture models. In the LPS model, RNA sequencing and qPCR analysis showed that this protection correlated with substantial reduction in LPS-induced proinflammatory gene expression, including lower tissue levels of Il1a , Il1b , and Tnf . Examining functional effects on the epithelium, we found that LPS-induced tight junctional disruption as assessed by internalization of ZO-1 and occludin was ameliorated in FXR KO animals. Taken together, these data suggest a role for FXR in the intestinal barrier during inflammatory injury. NEW & NOTEWORTHY Intestinal barrier failure is a hallmark in gut-origin sepsis. We demonstrate that the intestinal barriers of farnesoid-X receptor (FXR) knockout (KO) animals are protected from inflammatory insult using multiple models of acute intestinal inflammation. This protection is due to decreased inflammatory cytokine production and maintenance of tight junctional architecture seen within the KO animals. This is the first report of FXR deletion being protective to the intestinal barrier.
- Published
- 2024
- Full Text
- View/download PDF
20. Lenvatinib plus Pembrolizumab Following Immune Checkpoint Inhibitor Treatment in Patients with Metastatic Clear Cell Renal Cell Carcinoma: Results from Study 111/KEYNOTE-146.
- Author
-
Lee CH, Yogesh Shah A, Rao A, Taylor MH, Di Simone C, Hsieh JJ, Pinto A, Gironés Sarrió R, Lee Cohn A, Asim Bilen M, Gunnestad Ribe S, Krohn Tennøe Ø, Richards D, Sweis RF, Courtright J, Heinrich D, Perini R, Kubiak P, Bock D, Okpara CE, and Motzer RJ
- Published
- 2024
- Full Text
- View/download PDF
21. The Cancer Genome Atlas Comprehensive Molecular Characterization of Renal Cell Carcinoma.
- Author
-
Ricketts CJ, De Cubas AA, Fan H, Smith CC, Lang M, Reznik E, Bowlby R, Gibb EA, Akbani R, Beroukhim R, Bottaro DP, Choueiri TK, Gibbs RA, Godwin AK, Haake S, Hakimi AA, Henske EP, Hsieh JJ, Ho TH, Kanchi RS, Krishnan B, Kwiatkowski DJ, Liu W, Merino MJ, Mills GB, Myers J, Nickerson ML, Reuter VE, Schmidt LS, Shelley CS, Shen H, Shuch B, Signoretti S, Srinivasan R, Tamboli P, Thomas G, Vincent BG, Vocke CD, Wheeler DA, Yang L, Kim WY, Robertson AG, Spellman PT, Rathmell WK, and Linehan WM
- Published
- 2024
- Full Text
- View/download PDF
22. Gut microbiota and clinical response to immune checkpoint inhibitor therapy in patients with advanced cancer.
- Author
-
Chang JW, Hsieh JJ, Tsai CY, Chiu HY, Lin YF, Wu CE, Shen YC, Hou MM, Chang CY, Chen JA, Chen CL, Chiu CT, Yeh YM, and Chiu CH
- Abstract
Background: There is currently no well-accepted consensus on the association between gut microbiota and the response to treatment of immune checkpoint inhibitors (ICIs) in patients with advanced cancer., Methods: Fecal samples were collected before ICI treatment. Gut microbiota was analyzed using 16 S ribosomal RNA sequencing. We investigated the relationship between the α-diversity of fecal microbiota and patients' clinical outcomes. Microbiota profiles from patients and healthy controls were determined. Pre-treatment serum was examined by cytokine array., Results: We analyzed 74 patients, including 42 with melanoma, 8 with kidney cancer, 13 with lung cancer, and 11 with other cancers. Combination therapy of anti-PD1 and anti-CTLA-4 was used in 14 patients, and monotherapy in the rest. Clinical benefit was observed in 35 (47.3 %) cases, including 2 complete responses, 16 partial responses, and 17 stable diseases according to RECIST criteria. No significant difference in α-diversity was found between the benefiter and non-benefiter groups. However, patients with α-diversity within the range of our healthy control had a significantly longer median overall survival (18.9 months), compared to the abnormal group (8.2 months) (p = 0.041, hazard ratio = 0.546) for all patients. The microbiota composition of the benefiters was similar to that of healthy individuals. Furthermore, specific bacteria, such as Prevotella copri and Faecalibacterium prausnitzii, were associated with a favorable outcome. We also observed that serum IL-18 before treatment was significantly lower in the benefiters, compared to non-benefiters., Conclusions: The α-diversity of gut microbiota is positively correlated with more prolonged overall survival in cancer patients following ICI therapy., Competing Interests: Declaration of competing interest The authors have no financial or ethical conflicts of interest to report., (© 2024 The Authors. Published by Elsevier B.V. on behalf of Chang Gung University. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).)
- Published
- 2024
- Full Text
- View/download PDF
23. Genomic and tumour microenvironmental biomarkers of immune checkpoint inhibitor response in advanced Taiwanese melanoma.
- Author
-
Chang JW, Huang CJ, Huang WK, Wang YC, Hsieh JJ, Chang YY, Huang YL, Wu CL, Wang YH, Chen SJ, Tan KT, Chen CP, and Wu CE
- Abstract
Objective: Genomic biomarkers predicting immune checkpoint inhibitor (ICI) treatment outcomes for Asian metastatic melanoma have been rarely reported. This study presents data on next-generation sequencing (NGS) and tumour microenvironment biomarkers in 33 cases., Methods: Thirty-three patients with advanced melanoma, who underwent ICI treatment at the Chang Gung Memorial Hospital in Taiwan, were recruited. The study evaluated clinical outcomes, including response rate, disease control rate, progression-free survival (PFS) rate and overall survival (OS) rate. Archived tissue samples from 33 cases were subjected to NGS by ACTOnco, and ACTTME was employed in 25 cases., Results: The most prevalent driver mutations were BRAF mutations (24.2%), followed by NRAS (15.2%), KIT (12.1%), KRAS (9.1%) and NF1 (9.1%) mutations. Acral/mucosal melanomas exhibited distinct mutation patterns compared to non-acral melanomas. Tumour mutational burden estimated using ACTOnco was not associated with ICI efficacy. Notably, genetic alterations in the p53 pathway ( CDKNA2 loss, MDM2 gain/amplification and TP53 mutation) accounted for 36.4% and were significantly associated with unfavourable PFS (median PFS 2.7 months vs. 3.9 months, P = 0.0394). Moreover, 26 genes were identified as differentially expressed genes that were upregulated in patients with clinical benefits compared to those without benefits. Four genes, GZMH , GZMK , AIM2 and CTLA4 , were found to be associated with both PFS and OS., Conclusion: Genetic alterations in the p53 pathway may be critical in Asian patients with melanoma undergoing ICI treatment. Further investigation is required to explore this mechanism and validate these findings., Competing Interests: KTT was an employee of ACT Genomics Co., Ltd. CLW, YHW and SJC are employees of ACT Genomics Co., Ltd. The other authors declare that the research was conducted without commercial or financial relationships that could be construed as potential conflicts of interest., (© 2023 The Authors. Clinical & Translational Immunology published by John Wiley & Sons Australia, Ltd on behalf of Australian and New Zealand Society for Immunology, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
24. Author Correction: Epigenetic and transcriptomic characterization reveals progression markers and essential pathways in clear cell renal cell carcinoma.
- Author
-
Wu Y, Terekhanova NV, Caravan W, Naser Al Deen N, Lal P, Chen S, Mo CK, Cao S, Li Y, Karpova A, Liu R, Zhao Y, Shinkle A, Strunilin I, Weimholt C, Sato K, Yao L, Serasanambati M, Yang X, Wyczalkowski M, Zhu H, Zhou DC, Jayasinghe RG, Mendez D, Wendl MC, Clark D, Newton C, Ruan Y, Reimers MA, Pachynski RK, Kinsinger C, Jewell S, Chan DW, Zhang H, Chaudhuri AA, Chheda MG, Humphreys BD, Mesri M, Rodriguez H, Hsieh JJ, Ding L, and Chen F
- Published
- 2023
- Full Text
- View/download PDF
25. Epigenetic and transcriptomic characterization reveals progression markers and essential pathways in clear cell renal cell carcinoma.
- Author
-
Wu Y, Terekhanova NV, Caravan W, Naser Al Deen N, Lal P, Chen S, Mo CK, Cao S, Li Y, Karpova A, Liu R, Zhao Y, Shinkle A, Strunilin I, Weimholt C, Sato K, Yao L, Serasanambati M, Yang X, Wyczalkowski M, Zhu H, Zhou DC, Jayasinghe RG, Mendez D, Wendl MC, Clark D, Newton C, Ruan Y, Reimers MA, Pachynski RK, Kinsinger C, Jewell S, Chan DW, Zhang H, Chaudhuri AA, Chheda MG, Humphreys BD, Mesri M, Rodriguez H, Hsieh JJ, Ding L, and Chen F
- Subjects
- Humans, Transcriptome, Epigenesis, Genetic, Tumor Suppressor Proteins genetics, Gene Expression Regulation, Neoplastic, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology
- Abstract
Identifying tumor-cell-specific markers and elucidating their epigenetic regulation and spatial heterogeneity provides mechanistic insights into cancer etiology. Here, we perform snRNA-seq and snATAC-seq in 34 and 28 human clear cell renal cell carcinoma (ccRCC) specimens, respectively, with matched bulk proteogenomics data. By identifying 20 tumor-specific markers through a multi-omics tiered approach, we reveal an association between higher ceruloplasmin (CP) expression and reduced survival. CP knockdown, combined with spatial transcriptomics, suggests a role for CP in regulating hyalinized stroma and tumor-stroma interactions in ccRCC. Intratumoral heterogeneity analysis portrays tumor cell-intrinsic inflammation and epithelial-mesenchymal transition (EMT) as two distinguishing features of tumor subpopulations. Finally, BAP1 mutations are associated with widespread reduction of chromatin accessibility, while PBRM1 mutations generally increase accessibility, with the former affecting five times more accessible peaks than the latter. These integrated analyses reveal the cellular architecture of ccRCC, providing insights into key markers and pathways in ccRCC tumorigenesis., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
26. SETD2 regulates chromatin accessibility and transcription to suppress lung tumorigenesis.
- Author
-
Xie Y, Sahin M, Wakamatsu T, Inoue-Yamauchi A, Zhao W, Han S, Nargund AM, Yang S, Lyu Y, Hsieh JJ, Leslie CS, and Cheng EH
- Subjects
- Animals, Mice, Carcinogenesis genetics, Cell Transformation, Neoplastic, Lung metabolism, Proto-Oncogene Proteins p21(ras) genetics, Chromatin, Histone-Lysine N-Methyltransferase genetics, Lung Neoplasms genetics, Lung Neoplasms pathology
- Abstract
SETD2, a H3K36 trimethyltransferase, is the most frequently mutated epigenetic modifier in lung adenocarcinoma, with a mutation frequency of approximately 9%. However, how SETD2 loss of function promotes tumorigenesis remains unclear. Using conditional Setd2-KO mice, we demonstrated that Setd2 deficiency accelerated the initiation of KrasG12D-driven lung tumorigenesis, increased tumor burden, and significantly reduced mouse survival. An integrated chromatin accessibility and transcriptome analysis revealed a potentially novel tumor suppressor model of SETD2 in which SETD2 loss activates intronic enhancers to drive oncogenic transcriptional output, including the KRAS transcriptional signature and PRC2-repressed targets, through regulation of chromatin accessibility and histone chaperone recruitment. Importantly, SETD2 loss sensitized KRAS-mutant lung cancer to inhibition of histone chaperones, the FACT complex, or transcriptional elongation both in vitro and in vivo. Overall, our studies not only provide insight into how SETD2 loss shapes the epigenetic and transcriptional landscape to promote tumorigenesis, but they also identify potential therapeutic strategies for SETD2 mutant cancers.
- Published
- 2023
- Full Text
- View/download PDF
27. Deep Crypt Secretory Cell Differentiation in the Colonic Epithelium Is Regulated by Sprouty2 and Interleukin 13.
- Author
-
Schumacher MA, Liu CY, Katada K, Thai MH, Hsieh JJ, Hansten BJ, Waddell A, Rosen MJ, and Frey MR
- Subjects
- Animals, Mice, Cell Differentiation, Epithelium, Lymphocytes, Mice, Knockout, Pancreatitis-Associated Proteins, Proteins, Immunity, Innate, Interleukin-13
- Abstract
Background & Aims: Deep crypt secretory (DCS) cells are a critical component of the colonic stem cell niche. However, the regulatory mechanisms controlling DCS cell numbers and function are not well understood. Sprouty2 is an inflammation-responsive regulator of intracellular signaling that influences colonic secretory cell numbers in colitis via an epithelial-stromal interleukin (IL)33/IL13 signaling loop. Here, we tested the hypothesis that IL13, induced by epithelial Sprouty2 down-regulation, promotes DCS cell differentiation and function., Methods: Distal colons from mice with an intestinal epithelial-specific Sprouty2 deletion (Spry2
ΔIE ) and littermate controls were analyzed by in situ hybridization for Reg4+ DCS cells. Single-cell RNA sequencing and immunostaining were used to identify DCS cell-derived host defense peptides (HDPs) and localization of IL13 and IL13 receptor; bulk RNA sequencing and quantitative polymerase chain reaction were used to quantify changes in expression of identified HDPs. Cytokine-treated colonoids were assessed for DCS cells. A requirement for an IL33/IL13 signaling loop in the regulation of DCS cells was assessed in vivo using IL13 null mice., Results: Reg4+ DCS cell numbers were increased 2-fold in distal colons of Spry2ΔIE mice with a concomitant overall increase in DCS cell marker expression (Reg4, Spink4, and Agr2). Single-cell transcriptomics showed the HDP Retnlb/Resistin Like Beta (RELMβ) is highly enriched in DCS cells. Retnlb/RELMβ expression was increased in Spry2ΔIE colons. IL13, but not IL33, induced Reg4 and Retnlb expression in colonic epithelial organoids, and IL33-mediated expansion of the DCS cell population in vivo was dependent on IL13, which was expressed predominantly by type II innate lymphoid cells in the colonic mucosa., Conclusions: Sprouty2 limits colonic DCS cell differentiation through suppression of IL13 signaling. At homeostasis, DCS cells are marked by high levels of the HDP RELMβ. Loss of epithelial Sprouty2 activates type II innate lymphoid cells to release IL13, promoting expansion of the DCS cell population and increased colonic RELMβ levels., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
28. Tumor-associated macrophages expressing the transcription factor IRF8 promote T cell exhaustion in cancer.
- Author
-
Nixon BG, Kuo F, Ji L, Liu M, Capistrano K, Do M, Franklin RA, Wu X, Kansler ER, Srivastava RM, Purohit TA, Sanchez A, Vuong L, Krishna C, Wang X, Morse Iii HC, Hsieh JJ, Chan TA, Murphy KM, Moon JJ, Hakimi AA, and Li MO
- Subjects
- Humans, Animals, Mice, Tumor-Associated Macrophages, Interferon Regulatory Factors genetics, Interferon Regulatory Factors metabolism, T-Lymphocytes, Cytotoxic, Dendritic Cells, Carcinoma, Renal Cell, Kidney Neoplasms
- Abstract
Tumors are populated by antigen-presenting cells (APCs) including macrophage subsets with distinct origins and functions. Here, we examined how cancer impacts mononuclear phagocytic APCs in a murine model of breast cancer. Tumors induced the expansion of monocyte-derived tumor-associated macrophages (TAMs) and the activation of type 1 dendritic cells (DC1s), both of which expressed and required the transcription factor interferon regulatory factor-8 (IRF8). Although DC1s mediated cytotoxic T lymphocyte (CTL) priming in tumor-draining lymph nodes, TAMs promoted CTL exhaustion in the tumor, and IRF8 was required for TAMs' ability to present cancer cell antigens. TAM-specific IRF8 deletion prevented exhaustion of cancer-cell-reactive CTLs and suppressed tumor growth. Tumors from patients with immune-infiltrated renal cell carcinoma had abundant TAMs that expressed IRF8 and were enriched for an IRF8 gene expression signature. Furthermore, the TAM-IRF8 signature co-segregated with CTL exhaustion signatures across multiple cancer types. Thus, CTL exhaustion is promoted by TAMs via IRF8., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
29. Multiregional single-cell proteogenomic analysis of ccRCC reveals cytokine drivers of intratumor spatial heterogeneity.
- Author
-
Miheecheva N, Postovalova E, Lyu Y, Ramachandran A, Bagaev A, Svekolkin V, Galkin I, Zyrin V, Maximov V, Lozinsky Y, Isaev S, Ovcharov P, Shamsutdinova D, Cheng EH, Nomie K, Brown JH, Tsiper M, Ataullakhanov R, Fowler N, and Hsieh JJ
- Subjects
- Cytokines genetics, Genetic Heterogeneity, Humans, Single-Cell Analysis, Tumor Microenvironment genetics, Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell pathology, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Proteogenomics
- Abstract
Intratumor heterogeneity (ITH) represents a major challenge for anticancer therapies. An integrated, multidimensional, multiregional approach dissecting ITH of the clear cell renal cell carcinoma (ccRCC) tumor microenvironment (TME) is employed at the single-cell level with mass cytometry (CyTOF), multiplex immunofluorescence (MxIF), and single-nucleus RNA sequencing (snRNA-seq) and at the bulk level with whole-exome sequencing (WES), RNA-seq, and methylation profiling. Multiregional analyses reveal unexpected conservation of immune composition within each individual patient, with profound differences among patients, presenting patient-specific tumor immune microenvironment signatures despite underlying genetic heterogeneity from clonal evolution. Spatial proteogenomic TME analysis using MxIF identifies 14 distinct cellular neighborhoods and, conversely, demonstrated architectural heterogeneity among different tumor regions. Tumor-expressed cytokines are identified as key determinants of the TME and correlate with clinical outcome. Overall, this work signifies that spatial ITH occurs in ccRCC, which may drive clinical heterogeneity and warrants further interrogation to improve patient outcomes., Competing Interests: Declaration of interests J.H. is a medical oncologist in the Division of Oncology at Washington University School of Medicine in St. Louis and has received consulting fees and research funding from BostonGene. N.F. is the chief medical officer of BostonGene Corp. and a professor at The University of Texas MD Anderson Cancer Center. This research was funded by BostonGene Corp., and all BostonGene authors were employees thereof at the time the study was performed., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
30. Precise reconstruction of the TME using bulk RNA-seq and a machine learning algorithm trained on artificial transcriptomes.
- Author
-
Zaitsev A, Chelushkin M, Dyikanov D, Cheremushkin I, Shpak B, Nomie K, Zyrin V, Nuzhdina E, Lozinsky Y, Zotova A, Degryse S, Kotlov N, Baisangurov A, Shatsky V, Afenteva D, Kuznetsov A, Paul SR, Davies DL, Reeves PM, Lanuti M, Goldberg MF, Tazearslan C, Chasse M, Wang I, Abdou M, Aslanian SM, Andrewes S, Hsieh JJ, Ramachandran A, Lyu Y, Galkin I, Svekolkin V, Cerchietti L, Poznansky MC, Ataullakhanov R, Fowler N, and Bagaev A
- Subjects
- Algorithms, CD8-Positive T-Lymphocytes, Humans, Machine Learning, RNA-Seq, Sequence Analysis, RNA, Tumor Microenvironment genetics, Neoplasms genetics, Transcriptome
- Abstract
Cellular deconvolution algorithms virtually reconstruct tissue composition by analyzing the gene expression of complex tissues. We present the decision tree machine learning algorithm, Kassandra, trained on a broad collection of >9,400 tissue and blood sorted cell RNA profiles incorporated into millions of artificial transcriptomes to accurately reconstruct the tumor microenvironment (TME). Bioinformatics correction for technical and biological variability, aberrant cancer cell expression inclusion, and accurate quantification and normalization of transcript expression increased Kassandra stability and robustness. Performance was validated on 4,000 H&E slides and 1,000 tissues by comparison with cytometric, immunohistochemical, or single-cell RNA-seq measurements. Kassandra accurately deconvolved TME elements, showing the role of these populations in tumor pathogenesis and other biological processes. Digital TME reconstruction revealed that the presence of PD-1-positive CD8
+ T cells strongly correlated with immunotherapy response and increased the predictive potential of established biomarkers, indicating that Kassandra could potentially be utilized in future clinical applications., Competing Interests: Declaration of interests N.F. is the Chief Medical Officer of BostonGene, Corp. and a professor at the University of Texas MD Anderson Cancer Center. A. Zaitsev, M. Chelushkin, V.Z., B.S., D.D., E. Nuzhdina, A. Bagaev, and R.A. are inventors on patent applications related to Kassandra. All other authors declare no competing interests., (Copyright © 2022 Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
31. Telaglenastat plus Everolimus in Advanced Renal Cell Carcinoma: A Randomized, Double-Blinded, Placebo-Controlled, Phase II ENTRATA Trial.
- Author
-
Lee CH, Motzer R, Emamekhoo H, Matrana M, Percent I, Hsieh JJ, Hussain A, Vaishampayan U, Liu S, McCune S, Patel V, Shaheen M, Bendell J, Fan AC, Gartrell BA, Goodman OB, Nikolinakos PG, Kalebasty AR, Zakharia Y, Zhang Z, Parmar H, Akella L, Orford K, and Tannir NM
- Subjects
- Angiogenesis Inhibitors therapeutic use, Everolimus, Glutaminase therapeutic use, Glutamine, Humans, Protein Kinase Inhibitors adverse effects, Sirolimus adverse effects, Carcinoma, Renal Cell drug therapy, Carcinoma, Renal Cell pathology, Kidney Neoplasms pathology
- Abstract
Purpose: Glutaminase is a key enzyme, which supports elevated dependency of tumors on glutamine-dependent biosynthesis of metabolic intermediates. Dual targeting of glucose and glutamine metabolism by the mTOR inhibitor everolimus plus the oral glutaminase inhibitor telaglenastat showed preclinical synergistic anticancer effects, which translated to encouraging safety and efficacy findings in a phase I trial of 2L+ renal cell carcinoma (RCC). This study evaluated telaglenastat plus everolimus (TelaE) versus placebo plus everolimus (PboE) in patients with advanced/metastatic RCC (mRCC) in the 3L+ setting (NCT03163667)., Patients and Methods: Eligible patients with mRCC, previously treated with at least two prior lines of therapy [including ≥1 VEGFR-targeted tyrosine kinase inhibitor (TKI)] were randomized 2:1 to receive E, plus Tela or Pbo, until disease progression or unacceptable toxicity. Primary endpoint was investigator-assessed progression-free survival (PFS; one-sided α <0.2)., Results: Sixty-nine patients were randomized (46 TelaE, 23 PboE). Patients had a median three prior lines of therapy, including TKIs (100%) and checkpoint inhibitors (88%). At median follow-up of 7.5 months, median PFS was 3.8 months for TelaE versus 1.9 months for PboE [HR, 0.64; 95% confidence interval (CI), 0.34-1.20; one-sided P = 0.079]. One TelaE patient had a partial response and 26 had stable disease (SD). Eleven patients on PboE had SD. Treatment-emergent adverse events included fatigue, anemia, cough, dyspnea, elevated serum creatinine, and diarrhea; grade 3 to 4 events occurred in 74% TelaE patients versus 61% PboE., Conclusions: TelaE was well tolerated and improved PFS versus PboE in patients with mRCC previously treated with TKIs and checkpoint inhibitors., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
32. Atypical Auditory Perception Caused by Environmental Stimuli in Autism Spectrum Disorder: A Systematic Approach to the Evaluation of Self-Reports.
- Author
-
Hsieh JJ, Nagai Y, Kumagaya SI, Ayaya S, and Asada M
- Abstract
Recent studies have revealed that atypical sensory perception is common in individuals with autism spectrum disorder (ASD) and is considered a potential cause of social difficulties. Self-reports by individuals with ASD have provided great insights into atypical perception from the first-person point of view and indicated its dependence on the environment. This study aimed to investigate the patterns and environmental causes of atypical auditory perception in individuals with ASD. Qualitative data from subject reports are inappropriate for statistical analysis, and reporting subjective sensory experiences is not easy for every individual. To cope with such challenges, we employed audio signal processing methods to simulate the potential patterns of atypical auditory perception. The participants in our experiment were able to select and adjust the strength of the processing methods to manipulate the sounds in the videos to match their experiences. Thus, the strength of atypical perception was recorded quantitatively and then analyzed to assess its correlation with the audio-visual stimuli contained in the videos the participants observed. In total, 22 participants with ASD and 22 typically developed (TD) participants were recruited for the experiment. The results revealed several common patterns of atypical auditory perception: Louder sounds perceived in a quiet environment, noise perception induced by intense and unsteady audio-visual stimuli, and echo perception correlated with movement and variation in sound level. The ASD group reported atypical perceptions more frequently than the control group. However, similar environmental causes were shared by the ASD and TD groups. The results help us infer the potential neural and physiological mechanisms of sensory processing in ASD., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Hsieh, Nagai, Kumagaya, Ayaya and Asada.)
- Published
- 2022
- Full Text
- View/download PDF
33. SETD2 loss perturbs the kidney cancer epigenetic landscape to promote metastasis and engenders actionable dependencies on histone chaperone complexes.
- Author
-
Xie Y, Sahin M, Sinha S, Wang Y, Nargund AM, Lyu Y, Han S, Dong Y, Hsieh JJ, Leslie CS, and Cheng EH
- Subjects
- Animals, Carcinogenesis genetics, Cell Cycle Proteins genetics, Epigenesis, Genetic, Female, Histone Chaperones genetics, Histone-Lysine N-Methyltransferase genetics, Histones genetics, Humans, Male, Mice, Molecular Chaperones genetics, Carcinoma, Renal Cell genetics, Histone-Lysine N-Methyltransferase metabolism, Kidney Neoplasms genetics
- Abstract
SETD2 is a histone H3 lysine 36 (H3K36) trimethyltransferase that is mutated with high prevalence (13%) in clear cell renal cell carcinoma (ccRCC). Genomic profiling of primary ccRCC tumors reveals a positive correlation between SETD2 mutations and metastasis. However, whether and how SETD2 loss promotes metastasis remains unclear. In this study, we used a SETD2-mutant (SETD2
MT ) metastatic ccRCC human-derived cell line and xenograft models and showed that H3K36me3 restoration greatly reduced distant metastases of ccRCC in mice in a matrix metalloproteinase 1 (MMP1)-dependent manner. An integrated multiomics analysis using assay for transposase-accessible chromatin using sequencing (ATAC-seq), chromatin immunoprecipitation-sequencing (ChIP-seq) and RNA sequencing (RNA-seq) established a tumor suppressor model in which loss of SETD2-mediated H3K36me3 activates enhancers to drive oncogenic transcriptional output through regulation of chromatin accessibility. Furthermore, we uncovered mechanism-based therapeutic strategies for SETD2-deficient cancer through the targeting of specific histone chaperone complexes, including ASF1A/ASF1B and SPT16. Overall, SETD2 loss creates a permissive epigenetic landscape for cooperating oncogenic drivers to amplify transcriptional output, providing unique therapeutic opportunities., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2022
- Full Text
- View/download PDF
34. Diagnostic Utility of RNA-Seq for Evaluation of PD-L1 Expression in Clear Cell Renal Cell Carcinoma.
- Author
-
Sorokina M, Stupichev D, Lyu Y, Ramachandran A, Miheecheva N, Brown JH, Nomie K, Postovalova E, Bagaev A, Tsiper M, and Hsieh JJ
- Subjects
- B7-H1 Antigen genetics, Humans, Immunohistochemistry, RNA-Seq, Tumor Microenvironment, Carcinoma, Renal Cell diagnosis, Carcinoma, Renal Cell genetics, Kidney Neoplasms diagnosis, Kidney Neoplasms genetics
- Abstract
Background: Although there are immune checkpoint inhibitors (ICIs) available for the treatment of renal cell carcinoma (RCC), the utility of PD-L1 detection by immunohistochemistry (IHC) as a predictive biomarker in clear cell RCC (ccRCC) remains controversial. Nevertheless, alternative methods for PD-L1 detection, such as RNA sequencing (RNA-Seq), may be clinically useful in ccRCC; therefore, we sought to determine the ability of RNA-Seq to accurately and sensitively detect PD-L1 expression across different ccRCC clinical samples in comparison with IHC., Patients and Methods: Patients with ccRCC (n=127) who received treatment from Washington University in St. Louis between 2018 and 2020 were identified. Tumors from these patients were analyzed using RNA-Seq and IHC., Results: PD-L1 detection by RNA-Seq strongly correlated with IHC (P < .001), which was further validated using two independent datasets. Furthermore, RNA-Seq analysis identified an immune-enriched (higher PD-L1 positivity) and an immune-desert (lower PD-L1 positivity) microenvironment of ccRCC, which also correlated with IHC (P < .00001)., Conclusion: The results demonstrate the ability of RNA-Seq to detect PD-L1 in various ccRCC clinical samples compared to IHC. Ultimately, these findings suggest that PD-L1 detection by RNA-Seq can be further developed to determine the clinical utility of this methodology in ccRCC., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
35. Targeting Aurora B kinase prevents and overcomes resistance to EGFR inhibitors in lung cancer by enhancing BIM- and PUMA-mediated apoptosis.
- Author
-
Tanaka K, Yu HA, Yang S, Han S, Selcuklu SD, Kim K, Ramani S, Ganesan YT, Moyer A, Sinha S, Xie Y, Ishizawa K, Osmanbeyoglu HU, Lyu Y, Roper N, Guha U, Rudin CM, Kris MG, Hsieh JJ, and Cheng EH
- Subjects
- Apoptosis Regulatory Proteins metabolism, Aurora Kinase B antagonists & inhibitors, Bcl-2-Like Protein 11 metabolism, Carcinoma, Non-Small-Cell Lung drug therapy, Cell Line, Tumor, Drug Screening Assays, Antitumor, Drug Synergism, Epithelial-Mesenchymal Transition drug effects, Gene Expression Regulation, Neoplastic drug effects, High-Throughput Screening Assays, Humans, Lung Neoplasms drug therapy, Proto-Oncogene Proteins metabolism, Small Molecule Libraries pharmacology, Acrylamides pharmacology, Aniline Compounds pharmacology, Carcinoma, Non-Small-Cell Lung metabolism, Drug Resistance, Neoplasm drug effects, Lung Neoplasms metabolism, Protein Kinase Inhibitors pharmacology
- Abstract
The clinical success of EGFR inhibitors in EGFR-mutant lung cancer is limited by the eventual development of acquired resistance. We hypothesize that enhancing apoptosis through combination therapies can eradicate cancer cells and reduce the emergence of drug-tolerant persisters. Through high-throughput screening of a custom library of ∼1,000 compounds, we discover Aurora B kinase inhibitors as potent enhancers of osimertinib-induced apoptosis. Mechanistically, Aurora B inhibition stabilizes BIM through reduced Ser87 phosphorylation, and transactivates PUMA through FOXO1/3. Importantly, osimertinib resistance caused by epithelial-mesenchymal transition (EMT) activates the ATR-CHK1-Aurora B signaling cascade and thereby engenders hypersensitivity to respective kinase inhibitors by activating BIM-mediated mitotic catastrophe. Combined inhibition of EGFR and Aurora B not only efficiently eliminates cancer cells but also overcomes resistance beyond EMT., Competing Interests: Declaration of interests H.A.Y. has consulted for AstraZeneca, Daiichi, Janssen, and Blueprint Medicine; she has received research funding from AstraZeneca, Daiichi, Janssen, Pfizer, Novartis, Cullinan, and Lilly. C.M.R. has consulted for AbbVie, Amgen, AstraZeneca, Epizyme, Genentech/Roche, Ipsen, Jazz, Lilly, and Syros; he serves on the scientific advisory boards of Bridge Medicines, Earli, and Harpoon Therapeutics. M.G.K. has consulted for AstraZeneca, Daiichi-Sankyo, Janssen, Novartis, Pfizer, Regeneron, and Sanofi/Genzyme. U.G. has a clinical trial agreement with AstraZeneca and received research funding from AstraZeneca, Esanex, and Aurigene; he is a current employee of Bristol Myers Squibb. J.J.H. has consulted for Eisai and BostonGene; he has received clinical trial funding from Bristol Myers Squibb, Merck, AstraZeneca, Exelixis, Calithera, and SillaJen; he has received research funding from Merck, BostonGene, and TScan., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
36. Taspase1 orchestrates fetal liver hematopoietic stem cell and vertebrae fates by cleaving TFIIA.
- Author
-
Niizuma H, Searleman AC, Takeda S, Armstrong SA, Park CY, Cheng EH, and Hsieh JJ
- Subjects
- Animals, Embryo, Mammalian, Hematopoietic Stem Cells, Histone Code genetics, Histone-Lysine N-Methyltransferase metabolism, Mice, Mice, Knockout, Mutation, Myeloid-Lymphoid Leukemia Protein metabolism, RNA Cleavage, Stem Cell Transplantation, Abnormalities, Multiple genetics, Endopeptidases genetics, Endopeptidases metabolism, Fetal Development physiology, Transcription Factor TFIIA genetics
- Abstract
Taspase1, a highly conserved threonine protease encoded by TASP1, cleaves nuclear histone-modifying factors and basal transcription regulators to orchestrate diverse transcription programs. Hereditary loss-of-function mutation of TASP1 has recently been reported in humans as resulting in an anomaly complex syndrome, which manifests with hematological, facial, and skeletal abnormalities. Here, we demonstrate that Taspase1-mediated cleavage of TFIIAα-β, rather than of MLL1 or MLL2, in mouse embryos was required for proper fetal liver hematopoiesis and correct segmental identities of the axial skeleton. Homozygous genetic deletion of Taspase1 disrupted embryonic hematopoietic stem cell self-renewal and quiescence states and axial skeleton fates. Strikingly, mice carrying knockin noncleavable mutations of TFIIAα-β, a well-characterized basal transcription factor, displayed more pronounced fetal liver and axial skeleton defects than those with noncleavable MLL1 and MLL2, 2 trithorax group histone H3 trimethyl transferases. Our study offers molecular insights into a syndrome in humans that results from loss of TASP1 and describes an unexpected role of TFIIAα-β cleavage in embryonic cell fate decisions.
- Published
- 2021
- Full Text
- View/download PDF
37. Structural insights into the function of the catalytically active human Taspase1.
- Author
-
Nagaratnam N, Delker SL, Jernigan R, Edwards TE, Snider J, Thifault D, Williams D, Nannenga BL, Stofega M, Sambucetti L, Hsieh JJ, Flint AJ, Fromme P, and Martin-Garcia JM
- Subjects
- Cloning, Molecular, Crystallography, X-Ray, Dynamic Light Scattering, Endopeptidases genetics, Enzyme Activation, Humans, Models, Molecular, Protein Domains, Protein Structure, Secondary, Endopeptidases chemistry, Endopeptidases metabolism
- Abstract
Taspase1 is an Ntn-hydrolase overexpressed in primary human cancers, coordinating cancer cell proliferation, invasion, and metastasis. Loss of Taspase1 activity disrupts proliferation of human cancer cells in vitro and in mouse models of glioblastoma. Taspase1 is synthesized as an inactive proenzyme, becoming active upon intramolecular cleavage. The activation process changes the conformation of a long fragment at the C-terminus of the α subunit, for which no full-length structural information exists and whose function is poorly understood. We present a cloning strategy to generate a circularly permuted form of Taspase1 to determine the crystallographic structure of active Taspase1. We discovered that this region forms a long helix and is indispensable for the catalytic activity of Taspase1. Our study highlights the importance of this element for the enzymatic activity of Ntn-hydrolases, suggesting that it could be a potential target for the design of inhibitors with potential to be developed into anticancer therapeutics., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
38. A Single-arm, Multicenter, Phase 2 Study of Lenvatinib Plus Everolimus in Patients with Advanced Non-Clear Cell Renal Cell Carcinoma.
- Author
-
Hutson TE, Michaelson MD, Kuzel TM, Agarwal N, Molina AM, Hsieh JJ, Vaishampayan UN, Xie R, Bapat U, Ye W, Jain RK, and Fishman MN
- Subjects
- Everolimus adverse effects, Humans, Phenylurea Compounds, Quinolines, Carcinoma, Renal Cell drug therapy, Kidney Neoplasms drug therapy
- Abstract
Background: Non-clear cell renal cell carcinoma (nccRCC) accounts for ≤20% of RCC cases. Lenvatinib (a multitargeted tyrosine kinase inhibitor) in combination with everolimus (an mTOR inhibitor) is approved for the treatment of advanced RCC after one prior antiangiogenic therapy., Objective: To determine the safety and efficacy of lenvatinib plus everolimus as a first-line treatment for patients with advanced nccRCC., Design, Setting, and Participants: This open-label, single-arm, multicenter, phase 2 study enrolled patients with unresectable advanced or metastatic nccRCC and no prior anticancer therapy for advanced disease., Intervention: Lenvatinib (18 mg) plus everolimus (5 mg) orally once daily., Outcome Measurements and Statistical Analysis: The primary endpoint was the objective response rate (ORR) as assessed by investigators according to Response Evaluation Criteria in Solid Tumors version 1.1. Secondary endpoints included progression-free survival (PFS), overall survival (OS), and safety assessments. The 95% confidence intervals (CIs) for ORRs were calculated using the two-sided Clopper-Pearson method. Median PFS and median OS were estimated using the Kaplan-Meier product-limit method and their 95% CIs were estimated via a generalized Brookmeyer and Crowley method., Results and Limitations: The study (start date: February 20, 2017) enrolled 31 patients with nccRCC (papillary, n = 20; chromophobe, n = 9; unclassified, n = 2). At the data cutoff date (July 17, 2019), the best overall response was a partial response (eight patients: papillary, n = 3; chromophobe, n = 4; unclassified, n = 1) for an overall ORR of 26% (95% CI 12-45). Median PFS was 9.2 mo (95% CI 5.5-not estimable), and median OS was 15.6 mo (95% CI 9.2-not estimable). The most common treatment-emergent adverse events were fatigue (71%), diarrhea (58%), decreased appetite (55%), nausea (55%), and vomiting (52%). Limitations include the small sample size and single-arm design., Conclusions: Lenvatinib plus everolimus showed promising anticancer activity in patients with advanced nccRCC with an ORR of 26% and is worthy of further study. The safety profile was consistent with the established profile of the study-drug combination., Patient Summary: We examined the combination of lenvatinib plus everolimus as the first therapy for 31 patients who had advanced nccRCC. We found that this treatment seemed effective, because most patients had a decrease in tumor size and manageable treatment-related side effects., Clinical Registration: This trial is registered at ClinicalTrials.Gov as NCT02915783., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
39. Lenvatinib plus pembrolizumab in patients with either treatment-naive or previously treated metastatic renal cell carcinoma (Study 111/KEYNOTE-146): a phase 1b/2 study.
- Author
-
Lee CH, Shah AY, Rasco D, Rao A, Taylor MH, Di Simone C, Hsieh JJ, Pinto A, Shaffer DR, Girones Sarrio R, Cohn AL, Vogelzang NJ, Bilen MA, Gunnestad Ribe S, Goksel M, Tennøe ØK, Richards D, Sweis RF, Courtright J, Heinrich D, Jain S, Wu J, Schmidt EV, Perini RF, Kubiak P, Okpara CE, Smith AD, and Motzer RJ
- Subjects
- Aged, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, Carcinoma, Renal Cell immunology, Carcinoma, Renal Cell secondary, Europe, Female, Humans, Immune Checkpoint Inhibitors adverse effects, Kidney Neoplasms immunology, Kidney Neoplasms pathology, Male, Middle Aged, Phenylurea Compounds adverse effects, Protein Kinase Inhibitors adverse effects, Quinolines adverse effects, Time Factors, Treatment Outcome, United States, Antibodies, Monoclonal, Humanized therapeutic use, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Renal Cell drug therapy, Immune Checkpoint Inhibitors therapeutic use, Kidney Neoplasms drug therapy, Phenylurea Compounds therapeutic use, Protein Kinase Inhibitors therapeutic use, Quinolines therapeutic use
- Abstract
Background: Despite advances in the first-line treatment of metastatic renal cell carcinoma (RCC), there is an unmet need for options to address disease progression during or after treatment with immune checkpoint inhibitors (ICIs). Pembrolizumab and lenvatinib are active as monotherapies in RCC; thus, we aimed to evaluate the combination of lenvatinib plus pembrolizumab in these patients., Methods: We report results of the metastatic RCC cohort from an open-label phase 1b/2 study of lenvatinib plus pembrolizumab in patients aged at least 18 years with selected solid tumours and an Eastern Cooperative Oncology Group performance status of 0-1. Oral lenvatinib at 20 mg was given once daily along with intravenous pembrolizumab at 200 mg once every 3 weeks. Patients remained on study drug treatment until disease progression, development of unacceptable toxicity, or withdrawal of consent. Efficacy was analysed in patients with clear cell metastatic RCC receiving study drug by previous therapy grouping: treatment naive, previously treated ICI naive (previously treated with at least one line of therapy but not with an anti-PD-1 or anti-PD-L1 ICI), and ICI pretreated (ie, anti-PD-1 or anti-PD-L1) patients. Safety was analysed in all enrolled and treated patients. The primary endpoint was the objective response rate at week 24 per immune-related Response Evaluation Criteria In Solid Tumors (irRECIST) by investigator assessment. This trial is registered with ClinicalTrials.gov (NCT02501096) and with the EU Clinical Trials Register (EudraCT2017-000300-26), and is closed to new participants., Findings: Between July 21, 2015, and Oct 16, 2019, 145 patients were enrolled in the study. Two patients had non-clear cell RCC and were excluded from the efficacy analysis (one in the treatment-naive group and one in the ICI-pretreated group); thus, the population evaluated for efficacy comprised 143 patients (n=22 in the treatment-naive group, n=17 in the previously treated ICI-naive group, and n=104 in the ICI-pretreated group). All 145 enrolled patients were included in the safety analysis. The median follow-up was 19·8 months (IQR 14·3-28·4). The number of patients with an objective response at week 24 by irRECIST was 16 (72·7%, 95% CI 49·8-89·3) of 22 treatment-naive patients, seven (41·2%, 18·4-67·1) of 17 previously treated ICI-naive patients, and 58 (55·8%, 45·7-65·5) of 104 ICI-pretreated patients. Of 145 patients, 82 (57%) had grade 3 treatment-related adverse events and ten (7%) had grade 4 treatment-related adverse events. The most common grade 3 treatment-related adverse event was hypertension (30 [21%] of 145 patients). Treatment-related serious adverse events occurred in 36 (25%) patients, and there were three treatment-related deaths (upper gastrointestinal haemorrhage, sudden death, and pneumonia)., Interpretation: Lenvatinib plus pembrolizumab showed encouraging antitumour activity and a manageable safety profile and might be an option for post-ICI treatment of metastatic RCC., Funding: Eisai and Merck Sharp & Dohme., Competing Interests: Declaration of interests C-HL received support for this Article from Eisai and Merck; grants or contracts to their institution from Eisai and Merck; consulting fees from Eisai and Merck; for attending meetings and travel from Eisai; and fees for participation in a scientific advisory committee for Merck; consulting fees from Amgen, Bristol Myers Squibb, Exelixis, Pfizer, and EMD Serono; honoraria from AiCME, Intellisphere, and Research to Practice; research funds to the institute from Bristol Myers Squibb, Calithera, Eisai, Eli Lilly, Exelixis, Merck, and Pfizer. AYS participated on an advisory board for Eisai, Exelixis, Pfizer, and Roche; and received research funding from Bristol Myers Squibb, Eisai, and EMD Serono. DR received support for this Article from Eisai. AR received grants or contracts to their institution from Eisai and Merck; consulting fees to their institution from Eli Lilly; payment or honoraria for lectures, presentations, speakers' bureaus, manuscript writing, or educational events from Bayer, Cardinal Health, Eli Lilly, and Sanofi; and research funding to their institution from Clovis Oncology, Eli Lilly, Pfizer/Astellas, and Seattle Genetics/Astellas. MHT received clinical research funding for the present work to their institution from Eisai; and honoraria for participation in advisory boards and speakers' bureaus from Eisai. JJH received grants or contracts from Merck; consulting fees from BostonGene and Eisai; support for attending meetings or travel, or both from Elsevier; and stock or stock options from BostonGene. AP received payment or honoraria for lectures, presentations, speakers' bureaus, manuscript writing, or educational events from Bristol Myers Squibb, Ipsen, and Pfizer; and support for attending meetings or travel, or both, from Bristol Myers Squibb and Pfizer. RGS received payment or honoraria for lectures, presentations, speakers' bureaus, manuscript writing, or educational events from Bristol Myers Squibb, Janssen, and Roche; support for attending meetings or travel, or both, from MSD-Merck and Pfizer; and participation on a data safety monitoring board or advisory board from Bristol Myers Squibb. ALC received consulting fees from Amgen; and payment for expert testimony from the Department of Justice. NJV received support for this Article, including provision of study patients, payments to institution, medical writing, and manuscript writing charges from Eisai and Merck; consulting fees from Eisai and Merck; and payment for legal testimony from Merck. MAB received grants or contracts to their institution from Advanced Accelerator Applications, a Novartis Company, AstraZeneca, Bayer, Bristol Myers Squibb, Genentech/Roche, Genome & Company, Incyte, Nektar, Peloton Therapeutics, Pfizer, Seattle Genetics, Tricon Pharmaceuticals, and Xencor; honoraria for participation in a data safety monitoring board or advisory board for AstraZeneca, Bayer, Bristol Myers Squibb, Calithera Biosciences, Eisai, Exelixis, Genomic Health, Janssen, Nektar, Pfizer, and Sanofi. ØKT received payment or honoraria for lectures, presentations, speakers' bureaus, manuscript writing or educational events from Astellas and Bristol Myers Squibb; and fees for participation on a data safety monitoring board or advisory board for Bayer. RFS received grants or contracts to their institution from AbbVie, Aduro, Bayer, Bristol Myers Squibb, CytomX, Eisai, Eli Lilly, Genentech/Roche, Immunocore, Merck, Mirati, Moderna, Novartis, and QED therapeutics; consulting fees from Aduro, Astellas, AstraZeneca, Bristol Myers Squibb, Eisai, EMD Serono, Exelixis, Janssen, Mirati, Pfizer, Puma, and Seattle Genetics; payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Astellas, Bristol Myers Squibb, Eisai, Exelixis, Pfizer, and Seattle Genetics; support for attending meetings or travel, or both, from AstraZeneca, Bristol Myers Squibb, Eisai, Exelixis, Mirati; and patents pending, PCT/US2020/031357 on neoantigens in cancer. DH received support for this Article to their institution from Eisai; consulting fees from Astellas, AstraZeneca, Bayer, Eisai, Ipsen, Janssen-Cilag, and Roche; payment or honoraria for lectures, presentations, speakers bureaus, manuscript writing or educational events from Advanced Accelerator Applications, a Novartis Company, Astellas, Bayer, Bristol Myers Squibb, Ipsen, Janssen-Cilag, Novartis, and Sanofi; and participation on an advisory board for Astellas, AstraZeneca, Bayer, Eisai, Ipsen, Janssen-Cilag, and Roche; SJ has stock or stock options in Merck. EVS and RFP are employed by, and have stock or stock options in, Merck. JW and PK are employed by Eisai. CEO and ADS are employed by Eisai Europe. RJM received support for the present Article from Eisai and Merck; grants or contracts from Bristol Myers Squibb, Genentech, Novartis, Pfizer, and Roche; consulting fees from AstraZeneca, Aveo Pharmaceuticals, EMD Serono, Exelixis, Genentech, Incyte, Eli Lilly, Novartis, Pfizer, and Roche; and support for attending meetings or travel, or both, from Bristol Myers Squibb. CDS, DRS, SGR, MG, DR, and JC declare no competing interests., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
40. Single-cell Spatial Proteomic Revelations on the Multiparametric MRI Heterogeneity of Clinically Significant Prostate Cancer.
- Author
-
Pachynski RK, Kim EH, Miheecheva N, Kotlov N, Ramachandran A, Postovalova E, Galkin I, Svekolkin V, Lyu Y, Zou Q, Cao D, Gaut J, Ippolito JE, Bagaev A, Bruttan M, Gancharova O, Nomie K, Tsiper M, Andriole GL, Ataullakhanov R, and Hsieh JJ
- Subjects
- Artificial Intelligence, Ecosystem, Humans, Male, Proteomics, Multiparametric Magnetic Resonance Imaging, Prostatic Neoplasms diagnostic imaging, Prostatic Neoplasms genetics, Prostatic Neoplasms surgery
- Abstract
Purpose: Multiparametric MRI (mpMRI) has become an indispensable radiographic tool in diagnosing prostate cancer. However, mpMRI fails to visualize approximately 15% of clinically significant prostate cancer (csPCa). The molecular, cellular, and spatial underpinnings of such radiographic heterogeneity in csPCa are unclear., Experimental Design: We examined tumor tissues from clinically matched patients with mpMRI-invisible and mpMRI-visible csPCa who underwent radical prostatectomy. Multiplex immunofluorescence single-cell spatial imaging and gene expression profiling were performed. Artificial intelligence-based analytic algorithms were developed to examine the tumor ecosystem and integrate with corresponding transcriptomics., Results: More complex and compact epithelial tumor architectures were found in mpMRI-visible than in mpMRI-invisible prostate cancer tumors. In contrast, similar stromal patterns were detected between mpMRI-invisible prostate cancer and normal prostate tissues. Furthermore, quantification of immune cell composition and tumor-immune interactions demonstrated a lack of immune cell infiltration in the malignant but not in the adjacent nonmalignant tissue compartments, irrespective of mpMRI visibility. No significant difference in immune profiles was detected between mpMRI-visible and mpMRI-invisible prostate cancer within our patient cohort, whereas expression profiling identified a 24-gene stromal signature enriched in mpMRI-invisible prostate cancer. Prostate cancer with strong stromal signature exhibited a favorable survival outcome within The Cancer Genome Atlas prostate cancer cohort. Notably, five recurrences in the 8 mpMRI-visible patients with csPCa and no recurrence in the 8 clinically matched patients with mpMRI-invisible csPCa occurred during the 5-year follow-up post-prostatectomy., Conclusions: Our study identified distinct molecular, cellular, and structural characteristics associated with mpMRI-visible csPCa, whereas mpMRI-invisible tumors were similar to normal prostate tissue, likely contributing to mpMRI invisibility., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
41. Resource-efficient pooled sequencing expands translational impact in solid tumors.
- Author
-
DiNatale RG, Mano R, Makarov V, Rusk N, Drill E, Winer A, Sankin A, Yoo A, Freeman BA, Hsieh JJ, Chen YB, Coleman JA, Berger M, Ostrovnaya I, Chan TA, Russo P, Reznik E, and Hakimi AA
- Abstract
Intratumoral genetic heterogeneity (ITH) poses a significant challenge to utilizing sequencing for decision making in the management of cancer. Although sequencing of multiple tumor regions can address the pitfalls of ITH, it does so at a significant increase in cost and resource utilization. We propose a pooled multiregional sequencing strategy, whereby DNA aliquots from multiple tumor regions are mixed prior to sequencing, as a cost-effective strategy to boost translational value by addressing ITH while preserving valuable residual tissue for secondary analysis. Focusing on kidney cancer, we demonstrate that DNA pooling from as few as two regions significantly increases mutation detection while reducing clonality misattribution. This leads to an increased fraction of patients identified with therapeutically actionable mutations, improved patient risk stratification, and improved inference of evolutionary trajectories with an accuracy comparable to bona fide multiregional sequencing. The same approach applied to non-small-cell lung cancer data substantially improves tumor mutational burden (TMB) detection. Our findings demonstrate that pooled DNA sequencing strategies are a cost-effective alternative to address intrinsic genetic heterogeneity in clinical settings., Competing Interests: The rest of the authors have no conflicts to disclose.
- Published
- 2021
- Full Text
- View/download PDF
42. The 2021 Variant Histology Issue.
- Author
-
Hsieh JJ
- Published
- 2021
- Full Text
- View/download PDF
43. Nivolumab plus Cabozantinib versus Sunitinib for Advanced Renal-Cell Carcinoma.
- Author
-
Choueiri TK, Powles T, Burotto M, Escudier B, Bourlon MT, Zurawski B, Oyervides Juárez VM, Hsieh JJ, Basso U, Shah AY, Suárez C, Hamzaj A, Goh JC, Barrios C, Richardet M, Porta C, Kowalyszyn R, Feregrino JP, Żołnierek J, Pook D, Kessler ER, Tomita Y, Mizuno R, Bedke J, Zhang J, Maurer MA, Simsek B, Ejzykowicz F, Schwab GM, Apolo AB, and Motzer RJ
- Subjects
- Adult, Aged, Aged, 80 and over, Anilides adverse effects, Antineoplastic Agents adverse effects, Antineoplastic Combined Chemotherapy Protocols adverse effects, B7-H1 Antigen antagonists & inhibitors, Carcinoma, Renal Cell mortality, Female, Humans, Intention to Treat Analysis, Kidney Neoplasms mortality, Male, Middle Aged, Nivolumab adverse effects, Progression-Free Survival, Proportional Hazards Models, Pyridines adverse effects, Quality of Life, Receptor Protein-Tyrosine Kinases antagonists & inhibitors, Sunitinib adverse effects, Survival Analysis, Anilides administration & dosage, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Carcinoma, Renal Cell drug therapy, Kidney Neoplasms drug therapy, Nivolumab administration & dosage, Pyridines administration & dosage, Sunitinib therapeutic use
- Abstract
Background: The efficacy and safety of nivolumab plus cabozantinib as compared with those of sunitinib in the treatment of previously untreated advanced renal-cell carcinoma are not known., Methods: In this phase 3, randomized, open-label trial, we randomly assigned adults with previously untreated clear-cell, advanced renal-cell carcinoma to receive either nivolumab (240 mg every 2 weeks) plus cabozantinib (40 mg once daily) or sunitinib (50 mg once daily for 4 weeks of each 6-week cycle). The primary end point was progression-free survival, as determined by blinded independent central review. Secondary end points included overall survival, objective response as determined by independent review, and safety. Health-related quality of life was an exploratory end point., Results: Overall, 651 patients were assigned to receive nivolumab plus cabozantinib (323 patients) or sunitinib (328 patients). At a median follow-up of 18.1 months for overall survival, the median progression-free survival was 16.6 months (95% confidence interval [CI], 12.5 to 24.9) with nivolumab plus cabozantinib and 8.3 months (95% CI, 7.0 to 9.7) with sunitinib (hazard ratio for disease progression or death, 0.51; 95% CI, 0.41 to 0.64; P<0.001). The probability of overall survival at 12 months was 85.7% (95% CI, 81.3 to 89.1) with nivolumab plus cabozantinib and 75.6% (95% CI, 70.5 to 80.0) with sunitinib (hazard ratio for death, 0.60; 98.89% CI, 0.40 to 0.89; P = 0.001). An objective response occurred in 55.7% of the patients receiving nivolumab plus cabozantinib and in 27.1% of those receiving sunitinib (P<0.001). Efficacy benefits with nivolumab plus cabozantinib were consistent across subgroups. Adverse events of any cause of grade 3 or higher occurred in 75.3% of the 320 patients receiving nivolumab plus cabozantinib and in 70.6% of the 320 patients receiving sunitinib. Overall, 19.7% of the patients in the combination group discontinued at least one of the trial drugs owing to adverse events, and 5.6% discontinued both. Patients reported better health-related quality of life with nivolumab plus cabozantinib than with sunitinib., Conclusions: Nivolumab plus cabozantinib had significant benefits over sunitinib with respect to progression-free survival, overall survival, and likelihood of response in patients with previously untreated advanced renal-cell carcinoma. (Funded by Bristol Myers Squibb and others; CheckMate 9ER ClinicalTrials.gov number, NCT03141177.)., (Copyright © 2021 Massachusetts Medical Society.)
- Published
- 2021
- Full Text
- View/download PDF
44. Sprouty2 limits intestinal tuft and goblet cell numbers through GSK3β-mediated restriction of epithelial IL-33.
- Author
-
Schumacher MA, Hsieh JJ, Liu CY, Appel KL, Waddell A, Almohazey D, Katada K, Bernard JK, Bucar EB, Gadeock S, Maselli KM, Washington MK, Grikscheit TC, Warburton D, Rosen MJ, and Frey MR
- Subjects
- Animals, Cell Count, Cell Differentiation drug effects, Cell Proliferation drug effects, Child, Colitis chemically induced, Colitis metabolism, Colitis pathology, Colon drug effects, Colon metabolism, Colon pathology, Female, Gene Expression Regulation, Glycogen Synthase Kinase 3 beta metabolism, Goblet Cells drug effects, Goblet Cells metabolism, Goblet Cells pathology, HT29 Cells, Homeostasis drug effects, Humans, Interleukin-33 metabolism, Intestinal Mucosa drug effects, Intestinal Mucosa metabolism, Intestinal Mucosa pathology, Male, Membrane Proteins metabolism, Mice, Mice, Inbred C57BL, Mice, Knockout, Microfilament Proteins genetics, Microfilament Proteins metabolism, Phosphatidylinositol 3-Kinases genetics, Phosphatidylinositol 3-Kinases metabolism, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-akt genetics, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Sodium Dodecyl Sulfate administration & dosage, Colitis genetics, Glycogen Synthase Kinase 3 beta genetics, Homeostasis genetics, Interleukin-33 genetics, Membrane Proteins genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Dynamic regulation of intestinal cell differentiation is crucial for both homeostasis and the response to injury or inflammation. Sprouty2, an intracellular signaling regulator, controls pathways including PI3K and MAPKs that are implicated in differentiation and are dysregulated in inflammatory bowel disease. Here, we ask whether Sprouty2 controls secretory cell differentiation and the response to colitis. We report that colonic epithelial Sprouty2 deletion leads to expanded tuft and goblet cell populations. Sprouty2 loss induces PI3K/Akt signaling, leading to GSK3β inhibition and epithelial interleukin (IL)-33 expression. In vivo, this results in increased stromal IL-13+ cells. IL-13 in turn induces tuft and goblet cell expansion in vitro and in vivo. Sprouty2 is downregulated by acute inflammation; this appears to be a protective response, as VillinCre;Sprouty2
F/F mice are resistant to DSS colitis. In contrast, Sprouty2 is elevated in chronic colitis and in colons of inflammatory bowel disease patients, suggesting that this protective epithelial-stromal signaling mechanism is lost in disease.- Published
- 2021
- Full Text
- View/download PDF
45. The 2021 COVID-19 Artificial Intelligence Issue.
- Author
-
Hsieh JJ
- Subjects
- COVID-19 epidemiology, COVID-19 transmission, Global Burden of Disease, Humans, Medical Oncology instrumentation, Medical Oncology methods, Personal Protective Equipment, Physical Distancing, SARS-CoV-2, Smartphone, Telemedicine instrumentation, Telemedicine methods, Time-to-Treatment, Urogenital Neoplasms diagnosis, Urogenital Neoplasms mortality, Wireless Technology, Artificial Intelligence, COVID-19 prevention & control, Contact Tracing methods, Pandemics prevention & control, Urogenital Neoplasms therapy
- Published
- 2021
- Full Text
- View/download PDF
46. Genomic Landscapes of Acral Melanomas in East Asia.
- Author
-
Chang JW, Hsieh JJ, Wu CE, Lim AH, Ng CC, Teh BT, and Chan JY
- Subjects
- Adult, Aged, Aged, 80 and over, Asia, Eastern, Female, Humans, Male, Melanoma pathology, Middle Aged, Skin Neoplasms pathology, Melanoma, Cutaneous Malignant, Genomics methods, Melanoma genetics, Skin Neoplasms genetics
- Abstract
Background/aim: Acral melanomas (AM) represent a rare subgroup of melanomas with poor clinical outcomes and are enriched in Asian populations. Recent advances in next generation sequencing have provided opportunities to apply precision medicine to AM., Patients and Methods: Here, we present a series of 13 patients with melanomas from Taiwan and Singapore, including 8 patients with AM profiled using whole exome sequencing and summarize the recent studies on the genomic landscape of AM., Results: We identified mutually exclusive mutations in BRAF, NRAS, HRAS, NF1 and KIT in 6 AM cases. In addition, recurrent copy number gains in CCND1 and CDK4, as well as recurrent deletions in CDKN2A/CDKN2B, ATM and RAD51 were observed, supporting the potential use of CDK4/6 or PARP inhibitors in the treatment of these patients., Conclusion: The genomic landscape of AM provides an important resource for applying novel targeted therapies in this rare disease., (Copyright© 2021, International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
47. Laparoscopic cytoreductive nephrectomy is associated with significantly improved survival compared with open cytoreductive nephrectomy or targeted therapy alone.
- Author
-
Zhao K, Kim EH, Vetter JM, Hsieh JJ, Venkatesh R, Bhayani SB, and Figenshau RS
- Abstract
The aim of the present study was to compare the survival outcomes for patients with metastatic renal cell carcinoma (mRCC) who underwent laparoscopic cytoreductive nephrectomy (CN) vs. open CN vs. targeted therapy (TT) alone at our institution. A retrospective chart review was performed at our institution for patients who underwent CN prior to TT (laparoscopic, n=48; open, n=48) or who were deemed unfit for surgery and received TT alone (n=36), between January 2007 and December 2012. Kaplan-Meier estimated survival and Cox proportional hazards analyses were performed. Laparoscopic CN was associated with significantly longer survival compared with open CN or TT alone (median survival 24 vs. <12 months, respectively; P<0.01). On multivariate analysis, laparoscopic CN was an independent predictor of survival [hazard ratio (HR)=0.48, P<0.01), controlling for preoperative risk factors, while survival was similar between open CN and TT alone (HR=0.85, P=0.54). In our experience, laparoscopic CN appears to be a significant predictor of survival in mRCC. Selection bias of the surgeon for patients with improved survival may account for clinical variables that were otherwise difficult to quantify. For patients who were not candidates for laparoscopic CN, open CN did not confer a survival benefit over TT alone, while it was associated with increased morbidity., (Copyright © 2020, Spandidos Publications.)
- Published
- 2020
- Full Text
- View/download PDF
48. The 2020 nccRCC Immunotherapy Issue.
- Author
-
Hsieh JJ
- Subjects
- Humans, Immunotherapy, Carcinoma, Renal Cell, Kidney Neoplasms
- Published
- 2020
- Full Text
- View/download PDF
49. TGF-β suppresses type 2 immunity to cancer.
- Author
-
Liu M, Kuo F, Capistrano KJ, Kang D, Nixon BG, Shi W, Chou C, Do MH, Stamatiades EG, Gao S, Li S, Chen Y, Hsieh JJ, Hakimi AA, Taniuchi I, Chan TA, and Li MO
- Subjects
- Animals, CD8-Positive T-Lymphocytes immunology, Cell Death drug effects, Cell Hypoxia, Cell Line, Disease Progression, Female, Gene Expression Regulation immunology, Humans, Interferon-gamma immunology, Interleukin-4 immunology, Male, Mice, Mice, Inbred C57BL, Neoplasms blood supply, Neoplasms metabolism, Receptor, Transforming Growth Factor-beta Type II deficiency, Signal Transduction drug effects, Stromal Cells cytology, Stromal Cells immunology, Th2 Cells metabolism, Transforming Growth Factor beta antagonists & inhibitors, Neoplasms immunology, Neoplasms pathology, Signal Transduction immunology, Th2 Cells immunology, Transforming Growth Factor beta immunology
- Abstract
The immune system uses two distinct defence strategies against infections: microbe-directed pathogen destruction characterized by type 1 immunity
1 , and host-directed pathogen containment exemplified by type 2 immunity in induction of tissue repair2 . Similar to infectious diseases, cancer progresses with self-propagating cancer cells inflicting host-tissue damage. The immunological mechanisms of cancer cell destruction are well defined3-5 , but whether immune-mediated cancer cell containment can be induced remains poorly understood. Here we show that depletion of transforming growth factor-β receptor 2 (TGFBR2) in CD4+ T cells, but not CD8+ T cells, halts cancer progression as a result of tissue healing and remodelling of the blood vasculature, causing cancer cell hypoxia and death in distant avascular regions. Notably, the host-directed protective response is dependent on the T helper 2 cytokine interleukin-4 (IL-4), but not the T helper 1 cytokine interferon-γ (IFN-γ). Thus, type 2 immunity can be mobilized as an effective tissue-level defence mechanism against cancer.- Published
- 2020
- Full Text
- View/download PDF
50. The 2020 Prostate Cancer Issue.
- Author
-
Hsieh JJ
- Subjects
- Humans, Male, Prostate-Specific Antigen, Prostatic Neoplasms
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.