34 results on '"Aditi Qamra"'
Search Results
2. A myeloid program associated with COVID-19 severity is decreased by therapeutic blockade of IL-6 signaling
- Author
-
Jason A. Hackney, Haridha Shivram, Jason Vander Heiden, Chris Overall, Luz Orozco, Xia Gao, Eugene Kim, Nathan West, Aditi Qamra, Diana Chang, Arindam Chakrabarti, David F. Choy, Alexis J. Combes, Tristan Courau, Gabriela K. Fragiadakis, Arjun Arkal Rao, Arja Ray, Jessica Tsui, Kenneth Hu, Nicholas F. Kuhn, Matthew F. Krummel, David J. Erle, Kirsten Kangelaris, Aartik Sarma, Zoe Lyon, Carolyn S. Calfee, Prescott G. Woodruff, Rajani Ghale, Eran Mick, Ashley Byrne, Beth Shoshana Zha, Charles Langelier, Carolyn M. Hendrickson, Monique G.P. van der Wijst, George C. Hartoularos, Tianna Grant, Raymund Bueno, David S. Lee, John R. Greenland, Yang Sun, Richard Perez, Anton Ogorodnikov, Alyssa Ward, Chun Jimmie Ye, Thiru Ramalingam, Jacqueline M. McBride, Fang Cai, Anastasia Teterina, Min Bao, Larry Tsai, Ivan O. Rosas, Aviv Regev, Sharookh B. Kapadia, Rebecca N. Bauer, Carrie M. Rosenberger, Yumiko Abe-Jones, Michael Adkisson, K. Mark Ansel, Saurabh Asthana, Alexander Beagle, Sharvari Bhide, Cathy Cai, Saharai Caldera, Maria Calvo, Sidney A. Carrillo, Suzanna Chak, Stephanie Christenson, Zachary Collins, Spyros Darmanis, Angela Detweiler, Catherine DeVoe, Walter Eckalbar, Jeremy Giberson, Ana Gonzalez, Gracie Gordon, Paula Hayakawa Serpa, Alejandra Jauregui, Chayse Jones, Serena Ke, Divya Kushnoor, Tasha Lea, Deanna Lee, Aleksandra Leligdowicz, Yale Liu, Salman Mahboob, Lenka Maliskova, Michael Matthay, Elizabeth McCarthy, Priscila Muñoz-Sandoval, Norma Neff, Viet Nguyen, Nishita Nigam, Randy Parada, Maira Phelps, Logan Pierce, Priya Prasad, Sadeed Rashid, Gabriella Reeder, Nicklaus Rodriguez, Bushra Samad, Andrew Schroeder, Cole Shaw, Alan Shen, Austin Sigman, Pratik Sinha, Matthew Spitzer, Sara Sunshine, Kevin Tang, Luz Torres Altamirano, Alexandra Tsitsiklis, Erden Tumurbaatar, Vaibhav Upadhyay, Alexander Whatley, Andrew Willmore, Michael Wilson, Juliane Winkler, Kristine Wong, Kimberly Yee, Michelle Yu, Mingyue Zhou, and Wandi S. Zhu
- Subjects
Clinical genetics ,Molecular medicine ,Immune response ,Science - Abstract
Summary: Altered myeloid inflammation and lymphopenia are hallmarks of severe infections. We identified the upregulated EN-RAGE gene program in airway and blood myeloid cells from patients with acute lung injury from SARS-CoV-2 or other causes across 7 cohorts. This program was associated with greater clinical severity and predicted future mechanical ventilation and death. EN-RAGEhi myeloid cells express features consistent with suppressor cell functionality, including low HLA-DR and high PD-L1. Sustained EN-RAGE program expression in airway and blood myeloid cells correlated with clinical severity and increasing expression of T cell dysfunction markers. IL-6 upregulated many EN-RAGE program genes in monocytes in vitro. IL-6 signaling blockade by tocilizumab in a placebo-controlled clinical trial led to rapid normalization of EN-RAGE and T cell gene expression. This identifies IL-6 as a key driver of myeloid dysregulation associated with worse clinical outcomes in COVID-19 patients and provides insights into shared pathophysiological mechanisms in non-COVID-19 ARDS.
- Published
- 2023
- Full Text
- View/download PDF
3. Transcriptomic and proteomic assessment of tocilizumab response in a randomized controlled trial of patients hospitalized with COVID-19
- Author
-
Haridha Shivram, Jason A. Hackney, Carrie M. Rosenberger, Anastasia Teterina, Aditi Qamra, Olusegun Onabajo, Jacqueline McBride, Fang Cai, Min Bao, Larry Tsai, Aviv Regev, Ivan O. Rosas, and Rebecca N. Bauer
- Subjects
Pharmaceutical science ,Virology ,Omics ,Science - Abstract
Summary: High interleukin (IL)-6 levels are associated with greater COVID-19 severity. IL-6 receptor blockade by tocilizumab (anti-IL6R; Actemra) is used globally for the treatment of severe COVID-19, yet a molecular understanding of the therapeutic benefit remains unclear. We characterized the immune profile and identified cellular and molecular pathways modified by tocilizumab in peripheral blood samples from patients enrolled in the COVACTA study, a phase 3, randomized, double-blind, placebo-controlled trial of the efficacy and safety of tocilizumab in hospitalized patients with severe COVID-19. We identified markers of inflammation, lymphopenia, myeloid dysregulation, and organ injury that predict disease severity and clinical outcomes. Proteomic analysis confirmed a pharmacodynamic effect for tocilizumab and identified novel pharmacodynamic biomarkers. Transcriptomic analysis revealed that tocilizumab treatment leads to faster resolution of lymphopenia and myeloid dysregulation associated with severe COVID-19, indicating greater anti-inflammatory activity relative to placebo and potentially leading to faster recovery in patients hospitalized with COVID-19.
- Published
- 2023
- Full Text
- View/download PDF
4. CRISPRi enables isoform-specific loss-of-function screens and identification of gastric cancer-specific isoform dependencies
- Author
-
Rebecca Davies, Ling Liu, Sheng Taotao, Natasha Tuano, Richa Chaturvedi, Kie Kyon Huang, Catherine Itman, Amit Mandoli, Aditi Qamra, Changyuan Hu, David Powell, Roger J. Daly, Patrick Tan, and Joseph Rosenbluh
- Subjects
Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Introduction Genes contain multiple promoters that can drive the expression of various transcript isoforms. Although transcript isoforms from the same gene could have diverse and non-overlapping functions, current loss-of-function methodologies are not able to differentiate between isoform-specific phenotypes. Results Here, we show that CRISPR interference (CRISPRi) can be adopted for targeting specific promoters within a gene, enabling isoform-specific loss-of-function genetic screens. We use this strategy to test functional dependencies of 820 transcript isoforms that are gained in gastric cancer (GC). We identify a subset of GC-gained transcript isoform dependencies, and of these, we validate CIT kinase as a novel GC dependency. We further show that some genes express isoforms with opposite functions. Specifically, we find that the tumour suppressor ZFHX3 expresses an isoform that has a paradoxical oncogenic role that correlates with poor patient outcome. Conclusions Our work finds isoform-specific phenotypes that would not be identified using current loss-of-function approaches that are not designed to target specific transcript isoforms.
- Published
- 2021
- Full Text
- View/download PDF
5. Telomere dysfunction cooperates with epigenetic alterations to impair murine embryonic stem cell fate commitment
- Author
-
Mélanie Criqui, Aditi Qamra, Tsz Wai Chu, Monika Sharma, Julissa Tsao, Danielle A Henry, Dalia Barsyte-Lovejoy, Cheryl H Arrowsmith, Neil Winegarden, Mathieu Lupien, and Lea Harrington
- Subjects
telomerase reverse transcriptase ,telomeres ,epigenetics ,cell differentiation ,mouse ,embryonic stem cells ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The precise relationship between epigenetic alterations and telomere dysfunction is still an extant question. Previously, we showed that eroded telomeres lead to differentiation instability in murine embryonic stem cells (mESCs) via DNA hypomethylation at pluripotency-factor promoters. Here, we uncovered that telomerase reverse transcriptase null (Tert-/-) mESCs exhibit genome-wide alterations in chromatin accessibility and gene expression during differentiation. These changes were accompanied by an increase of H3K27me3 globally, an altered chromatin landscape at the Pou5f1/Oct4 promoter, and a refractory response to differentiation cues. Inhibition of the Polycomb Repressive Complex 2 (PRC2), an H3K27 tri-methyltransferase, exacerbated the impairment in differentiation and pluripotency gene repression in Tert-/- mESCs but not wild-type mESCs, whereas inhibition of H3K27me3 demethylation led to a partial rescue of the Tert-/- phenotype. These data reveal a new interdependent relationship between H3K27me3 and telomere integrity in stem cell lineage commitment that may have implications in aging and cancer.
- Published
- 2020
- Full Text
- View/download PDF
6. HoxC5 and miR-615-3p target newly evolved genomic regions to repress hTERT and inhibit tumorigenesis
- Author
-
TingDong Yan, Wen Fong Ooi, Aditi Qamra, Alice Cheung, DongLiang Ma, Gopinath Meenakshi Sundaram, Chang Xu, Manjie Xing, LaiFong Poon, Jing Wang, Yan Ping Loh, Jess Hui Jie Ho, Joscelyn Jun Quan Ng, Muhammad Khairul Ramlee, Luay Aswad, Steve G. Rozen, Sujoy Ghosh, Frederic A. Bard, Prabha Sampath, Vinay Tergaonkar, James O. J. Davies, Jim R. Hughes, Eyleen Goh, Xuezhi Bi, Melissa Jane Fullwood, Patrick Tan, and Shang Li
- Subjects
Science - Abstract
The expression of telomerase catalytic subunit hTERT is frequently upregulated in many cancers. Here, the authors show HoxC5 and miR-615-3p can negatively regulate hTERT to impede tumorigenesis by targeting the newly evolved cis-regulatory genomic elements of hTERT.
- Published
- 2018
- Full Text
- View/download PDF
7. Author Correction: CRISPRi enables isoform-specific loss-of-function screens and identification of gastric cancer-specific isoform dependencies
- Author
-
Rebecca Davies, Ling Liu, Sheng Taotao, Natasha Tuano, Richa Chaturvedi, Kie Kyon Huang, Catherine Itman, Amit Mandoli, Aditi Qamra, Changyuan Hu, David Powell, Roger J. Daly, Patrick Tan, and Joseph Rosenbluh
- Subjects
Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
An amendment to this paper has been published and can be accessed via the original article.
- Published
- 2021
- Full Text
- View/download PDF
8. Epigenomic profiling of primary gastric adenocarcinoma reveals super-enhancer heterogeneity
- Author
-
Wen Fong Ooi, Manjie Xing, Chang Xu, Xiaosai Yao, Muhammad Khairul Ramlee, Mei Chee Lim, Fan Cao, Kevin Lim, Deepak Babu, Lai-Fong Poon, Joyce Lin Suling, Aditi Qamra, Astrid Irwanto, James Qu Zhengzhong, Tannistha Nandi, Ai Ping Lee-Lim, Yang Sun Chan, Su Ting Tay, Ming Hui Lee, James O. J. Davies, Wai Keong Wong, Khee Chee Soo, Weng Hoong Chan, Hock Soo Ong, Pierce Chow, Chow Yin Wong, Sun Young Rha, Jianjun Liu, Axel M. Hillmer, Jim R. Hughes, Steve Rozen, Bin Tean Teh, Melissa Jane Fullwood, Shang Li, and Patrick Tan
- Subjects
Science - Abstract
Gene expression is regulated by enhancers and super-enhancers, which can be identified by chromatin profiling. Here, the authors surveyed gastric cancer samples and cell lines to identify enhancer elements exhibiting somatic alterations.
- Published
- 2016
- Full Text
- View/download PDF
9. Chronic hypoxia favours adoption to a castration-resistant cell state in prostate cancer
- Author
-
Sarina Cameron, Genevieve Deblois, James R. Hawley, Aditi Qamra, Stanley Zhou, Seyed Ali Madani Tonekaboni, Alexander Murison, Romy Van Vliet, Juan Liu, Jason W. Locasale, and Mathieu Lupien
- Subjects
Cancer Research ,Genetics ,Molecular Biology - Abstract
Predicting and treating recurrence in intermediate-risk prostate cancer patients remains a challenge despite having identified genomic instability (1) and hypoxia (2,3) as risk factors. This underlies challenges in assigning the functional impact of these risk factors to mechanisms promoting prostate cancer progression. Here we show chronic hypoxia (CH), as observed in prostate tumours (4), leads to the adoption of an androgen-independent state in prostate cancer cells. Specifically, CH results in prostate cancer cells adopting transcriptional and metabolic alterations typical of castration-resistant prostate cancer cells. These changes include increased expression of transmembrane transporters for the methionine cycle and related pathways leading to increased abundance of metabolites and expression of enzymes related to glycolysis. Targeting the Glucose Transporter 1 (GLUT1) identified a dependency on glycolysis in androgen-independent cells. Overall, we identified a therapeutically targetable weakness in chronic hypoxia and androgen-independent prostate cancer. These findings may offer additional strategies for treatment development against hypoxic prostate cancer.
- Published
- 2023
10. Tables S1-S16 from VHL Deficiency Drives Enhancer Activation of Oncogenes in Clear Cell Renal Cell Carcinoma
- Author
-
Patrick Tan, Bin Tean Teh, Jim R. Hughes, Gertrud Steger, Alexander Lezhava, David Lawrence Silver, Puay Hoon Tan, Kenneth Tou En Chang, Shang Li, Christopher Wai Sam Cheng, Iain Bee Huat Tan, Qiang Yu, Steven G. Rozen, Bryan C. Tan, Gary Loh, Jian Yuan Goh, James O. J. Davies, Swe Swe Myint, Chang Xu, Aditi Qamra, Tannistha Nandi, Joyce Suling Lin, Cassandra Zhengxuan He, Michelle Shu Wen Ng, Peiyong Guan, Ai Ping Lee-Lim, Jing Han Hong, Yue Ning Lam, Giovani Wijaya, Su Ting Tay, James Zhengzhong Qu, Yang Sun Chan, Manjie Xing, Dachuan Huang, Zhimei Li, Wen Fong Ooi, Joanna Koh, Kevin Junliang Lim, Jing Tan, and Xiaosai Yao
- Abstract
Supplementary tables
- Published
- 2023
11. Figure S1-9; Supplementary methods from VHL Deficiency Drives Enhancer Activation of Oncogenes in Clear Cell Renal Cell Carcinoma
- Author
-
Patrick Tan, Bin Tean Teh, Jim R. Hughes, Gertrud Steger, Alexander Lezhava, David Lawrence Silver, Puay Hoon Tan, Kenneth Tou En Chang, Shang Li, Christopher Wai Sam Cheng, Iain Bee Huat Tan, Qiang Yu, Steven G. Rozen, Bryan C. Tan, Gary Loh, Jian Yuan Goh, James O. J. Davies, Swe Swe Myint, Chang Xu, Aditi Qamra, Tannistha Nandi, Joyce Suling Lin, Cassandra Zhengxuan He, Michelle Shu Wen Ng, Peiyong Guan, Ai Ping Lee-Lim, Jing Han Hong, Yue Ning Lam, Giovani Wijaya, Su Ting Tay, James Zhengzhong Qu, Yang Sun Chan, Manjie Xing, Dachuan Huang, Zhimei Li, Wen Fong Ooi, Joanna Koh, Kevin Junliang Lim, Jing Tan, and Xiaosai Yao
- Abstract
Supplementary methods. Supplementary Figures 1-9
- Published
- 2023
12. Supplementary Text, Supplementary Figures 1 through 13, Supplementary Methods from Epigenomic Promoter Alterations Amplify Gene Isoform and Immunogenic Diversity in Gastric Adenocarcinoma
- Author
-
Patrick Tan, John Connolly, Jeeyun Lee, Dennis Kappei, Bin Tean Teh, Steven G. Rozen, Wai Keong Wong, Kyoung-Mee Kim, Khee Chee Soo, Hock Soo Ong, Weng Hoong Chan, Pierce K.H. Chow, Wei Peng Yong, Khay Guan Yeoh, Sun Young Rha, Hassan Ashktorab, Duane Smoot, Apinya Jusakul, Alvin Ng, Cedric Chuan Young Ng, Erna Gondo Santoso, Angie Tan Lay Keng, Su Ting Tay, Minghui Lee, Xuewen Ong, Xiaosai Yao, Tannistha Nandi, Joyce Suling Lin, Wen Fong Ooi, Qianqao Tang, Ai Ping Lee Lim, Yan Shan Leong, Chang Xu, Shenli Zhang, Jeffrey Jun Ting Kwok, Nisha Padmanabhan, Manjie Xing, and Aditi Qamra
- Abstract
Supplementary text S1. Supplementary Figure 1: Chromatin Profiles of Primary GC. Supplementary Figure 2: Epithelial features of GC promoters. Supplementary Figure 3: GC Somatic Promoter Features. Supplementary Figure 4: Association of Somatic Promoters with Gene Expression in GC and Other Tumor Types. Supplementary Figure 5: Changes in DNA methylation at CpG island containing promoters. Supplementary Figure 6: Expression distribution of alternative and canonical isoforms. Supplementary Figure 7: Characterization of RASA3 Isoform. Supplementary Figure 8: Characterization of MET Isoforms. Supplementary Figure 9: Immunogenicity of N-terminal peptides. Supplementary Figure 10: Immunogenicity Assay and Nanostring Profiling. Supplementary Figure 11: Functional Assessment of Peptide Immunogenicity. Supplementary Figure 12: EZH2 Inhibition. Supplementary Figure 13: Unannotated somatic promoters.
- Published
- 2023
13. Abstract PD10-05: Activity of atezolizumab (atezo) plus paclitaxel (pac) in metastatic triple-negative breast cancer (mTNBC) according to Burstein molecular subtype: Analysis of the IMpassion131 trial
- Author
-
Fabrice André, Regula Deurloo, Aditi Qamra, David Cameron, Joseph Gligorov, Andreas Schneeweiss, Carlos Barrios, Binghe Xu, Ching-Wei Chang, Luciana Molinero, Shilpen Patel, Andrea Liptrot, Leilani Morales, David Miles, and Joyce O’Shaughnessy
- Subjects
Cancer Research ,Oncology - Abstract
Background: In contrast to the IMpassion130 trial evaluating atezo + nab-pac [Schmid, NEJM 2018], the randomized phase 3 IMpassion131 trial (NCT03125902) did not demonstrate significantly improved progression-free survival (PFS; primary endpoint) and showed no improvement in overall survival (OS; secondary endpoint) with the addition of atezo to pac as first-line therapy for mTNBC in either the PD-L1+ or the intention-to-treat (ITT) population [Miles, Ann Oncol 2021]. In IMpassion130, enhanced PFS and OS improvement with atezo + nab-pac were seen in the basal-like immune-activated (BLIA) subtype, whereas potential resistance to atezo + nab-pac was observed in the luminal androgen receptor (LAR) subtype [Emens, ASCO 2021]. LAR may be more prevalent in Asian populations [Ding, Oncotarget 2019], which represented one-third of patients enrolled in IMpassion131 and could have influenced the overall result. PFS outcomes were numerically worse in Asian vs non-Asian subgroups in IMpassion131 [Miles, Ann Oncol 2021]. To investigate potential reasons for observing different effects in IMpassion130 and IMpassion131, we explored the prevalence and impact on clinical outcomes of TNBC molecular subtypes and race in IMpassion131. Patients and Methods: Patients with mTNBC (no prior systemic therapy or ≥12 months since [neo]adjuvant chemotherapy) were randomized 2:1 to atezolizumab 840 mg or placebo (days 1, 15), both with paclitaxel 90 mg/m2 (days 1, 8, 15), every 28 days until disease progression or unacceptable toxicity, stratified by tumor PD-L1 status, prior taxane, liver metastases, and geographic region. Molecular subtypes according to Burstein classification were determined by RNA sequencing (Illumina TruSeq RNA Access) of baseline tumor samples. Prevalence of Burstein molecular subtypes [Burstein, Clin Cancer Res 2015] was assessed in Asian (of whom 69% were enrolled in China) and non-Asian populations; clinical outcomes were assessed according to molecular subtype and race using Cox proportional hazards analysis. All analyses were performed using the final data cut-off (Sep 4, 2020; median follow-up duration: 14.4 months). Results: The biomarker-evaluable population (BEP; n=471) was representative of the ITT population (n=651) with respect to baseline characteristics and PFS hazard ratio (HR; 0.75 [95% CI 0.61-0.92] vs 0.81 [95% CI 0.68-0.96], respectively). Distribution of molecular subtypes in the BEP was: 30% BLIA, 41% basal-like immune suppressed (BLIS), 24% LAR, 5% mesenchymal (MES), similar to IMpassion130. Among the BLIA samples, 82% were PD-L1+ and 18% PD-L1-; corresponding percentages were 41% vs 59% for BLIS, 31% vs 69% for LAR, and 32% vs 68% for MES. Compared with non-Asian patients, the Asian subgroup (n=117) included more LAR (31% vs 22%) and fewer MES (1% vs 6%) samples; this was particularly pronounced in the cohort enrolled in China (n=79; 37% LAR, 0% MES). PFS was improved with atezo + pac in the BLIA subtype (HR 0.66, 95% CI 0.45-0.97). None of the Burstein subgroups derived OS benefit from atezo + pac. Findings were similar irrespective of PD-L1 status. Direction of effect for PFS and OS favored the placebo + pac arm in the LAR Asian subgroup (n=30). Conclusion: In these exploratory analyses, the distribution of molecular subtypes and enhanced effect of atezo + pac in the BLIA subtype are consistent with findings from IMpassion130. The lack of improved efficacy with the combination of atezo + pac in the IMpassion131 trial cannot be explained by overrepresentation of a Burstein subtype less sensitive to atezo in the trial population. Citation Format: Fabrice André, Regula Deurloo, Aditi Qamra, David Cameron, Joseph Gligorov, Andreas Schneeweiss, Carlos Barrios, Binghe Xu, Ching-Wei Chang, Luciana Molinero, Shilpen Patel, Andrea Liptrot, Leilani Morales, David Miles, Joyce O’Shaughnessy. Activity of atezolizumab (atezo) plus paclitaxel (pac) in metastatic triple-negative breast cancer (mTNBC) according to Burstein molecular subtype: Analysis of the IMpassion131 trial [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr PD10-05.
- Published
- 2022
14. Tocilizumab treatment leads to early resolution of lymphopenia and myeloid dysregulation in patients hospitalized with COVID-19
- Author
-
Haridha Shivram, Jason A. Hackney, Carrie M Rosenberger, Anastasia Teterina, Aditi Qamra, Olusegun Onabajo, Jacqueline McBride, Fang Cai, Min Bao, Larry Tsai, Aviv Regev, Ivan O. Rosas, and Rebecca N. Bauer
- Abstract
High interleukin (IL)-6 levels are associated with more severe clinical manifestations in patients hospitalized with COVID-19, but the complex role of IL-6 in antiviral and inflammatory processes has made it difficult to decipher its involvement in the disease. IL-6 receptor blockade by tocilizumab (anti-IL6R; Actemra) is used globally for the treatment of severe COVID-19, yet a molecular understanding of the therapeutic benefit remains unclear. We characterized the immune profile and identified cellular and molecular pathways directly modified by tocilizumab in peripheral blood samples collected from patients enrolled in the COVACTA study, a phase 3, randomized, double-blind, placebo-controlled trial that assessed the efficacy and safety of tocilizumab in hospitalized patients with severe COVID-19 pneumonia. We identified factors predicting disease severity and clinical outcomes, including markers of inflammation, lymphopenia, myeloid dysregulation, and organ injury. Proteomic analysis confirmed a pharmacodynamic effect for tocilizumab in addition to identifying novel pharmacodynamic biomarkers. Transcriptomic analysis revealed that tocilizumab treatment leads to faster resolution of lymphopenia and myeloid dysregulation associated with severe COVID-19, indicating greater anti-inflammatory activity relative to standard of care and potentially leading to faster recovery in patients hospitalized with COVID-19.One sentence summaryInterleukin-6 receptor blockade with tocilizumab accelerated resolution of myeloid dysfunction and lymphopenia in patients hospitalized with COVID-19
- Published
- 2022
15. Epigenetic promoter alterations in GI tumour immune-editing and resistance to immune checkpoint inhibition
- Author
-
Edward Cha, Su Pin Choo, Sun Young Rha, Kei Muro, Anand D. Jeyasekharan, Arne Fabritius, Joe Yeong, Manjie Xing, Filippo Pietrantonio, Angie Lay-Keng Tan, Aditi Qamra, Zul Fazreen Adam Isa, Kalpana Ramnarayanan, Jimmy Bok Yan So, Mark De Simone, Yukiya Narita, Radhika Patnala, Monica Niger, David Tai, Jonathan Göke, Luciana Molinero, Jeeyun Lee, Kie-Kyon Huang, Deniz Demircioğlu, Yu Amanda Guo, Meghna Das Thakur, Wei Peng Yong, Qingfeng Chen, Patrick Tan, Marcella Fassò, Zhisheng Her, Vikrant Kumar, Xuewen Ong, Weiwei Zhai, Cedric Chuan Young Ng, Jia Qi Lim, Anders Jacobsen Skanderup, and Raghav Sundar
- Subjects
Epigenomics ,medicine.medical_treatment ,Gastroenterology ,Cancer ,Immunotherapy ,Biology ,medicine.disease ,Immune checkpoint ,Epigenesis, Genetic ,Mice ,Immune system ,Stomach Neoplasms ,In vivo ,Hepatocellular carcinoma ,Tumor Microenvironment ,Cancer research ,medicine ,Animals ,Humans ,sense organs ,Epigenetics ,Immune Checkpoint Inhibitors ,Gastrointestinal Neoplasms - Abstract
ObjectivesEpigenomic alterations in cancer interact with the immune microenvironment to dictate tumour evolution and therapeutic response. We aimed to study the regulation of the tumour immune microenvironment through epigenetic alternate promoter use in gastric cancer and to expand our findings to other gastrointestinal tumours.DesignAlternate promoter burden (APB) was quantified using a novel bioinformatic algorithm (proActiv) to infer promoter activity from short-read RNA sequencing and samples categorised into APBhigh, APBint and APBlow. Single-cell RNA sequencing was performed to analyse the intratumour immune microenvironment. A humanised mouse cancer in vivo model was used to explore dynamic temporal interactions between tumour kinetics, alternate promoter usage and the human immune system. Multiple cohorts of gastrointestinal tumours treated with immunotherapy were assessed for correlation between APB and treatment outcomes.ResultsAPBhigh gastric cancer tumours expressed decreased levels of T-cell cytolytic activity and exhibited signatures of immune depletion. Single-cell RNAsequencing analysis confirmed distinct immunological populations and lower T-cell proportions in APBhigh tumours. Functional in vivo studies using ‘humanised mice’ harbouring an active human immune system revealed distinct temporal relationships between APB and tumour growth, with APBhigh tumours having almost no human T-cell infiltration. Analysis of immunotherapy-treated patients with GI cancer confirmed resistance of APBhigh tumours to immune checkpoint inhibition. APBhigh gastric cancer exhibited significantly poorer progression-free survival compared with APBlow (median 55 days vs 121 days, HR 0.40, 95% CI 0.18 to 0.93, p=0.032).ConclusionThese findings demonstrate an association between alternate promoter use and the tumour microenvironment, leading to immune evasion and immunotherapy resistance.
- Published
- 2021
16. Abstract 5705: Digital pathology based prognostic & predictive biomarkers in metastatic non-small cell lung cancer
- Author
-
Aditi Qamra, Minu K. Srivastava, Eloisa Fuentes, Ben Trotter, Raymond Biju, Guillaume Chhor, James Cowan, Steven Gendreau, Webster Lincoln, Lisa McGinnis, Luciana Molinero, Namrata S. Patil, Amber Schedlbauer, Katja Schulze, Adam Stanford-Moore, Laura Chambre, Ilan Wapinski, David S. Shames, Hartmut Koeppen, Stephanie Hennek, Jane Fridlyand, Jennifer M. Giltnane, and Assaf Amitai
- Subjects
Cancer Research ,Oncology - Abstract
Background: In recent years, a relationship between the tumor microenvironment (TME) and patient response to targeted cancer immunotherapy has been suggested. We applied machine-learning algorithms on H&E stained tissue to study the TME in metastatic non-small cell lung cancer (NSCLC) patients. Our goal was to identify digital pathology (DP) features associated with outcome under combination treatment or monotherapy with atezolizumab (atezo), an anti-PD-L1 therapy, and relate those features to other data modalities. We analyzed patient data from two phase 3 clinical trials, OAK (docetaxel versus atezo in 2L+ NSCLC) and IMpower150 (bevacizumab, carboplatin, and paclitaxel (BCP) versus BCP+atezo (ABCP) in advanced 1L non-squamous NSCLC). Methods: As part of our effort to build a DP-based tumor-immune microenvironment atlas, digitized H&E images were registered onto the PathAI research platform. Over 200K annotations from 90 pathologists were used to train convolutional neural networks (CNNs) that classify slide-level human-interpretable features (HIFs) of cells and tissue structures from images and deployed on images from OAK and IMpower150. HIFs and PD-L1 status were associated with outcome in all samples in each arm in OAK and results were validated in IMpower150, using Cox proportional hazard models. Bulk RNAseq was run using samples extracted from the same area as the H&E slide. Results: We identified a composite feature capturing the ratio of immune cells to fibroblasts in the stroma predictive of both overall survival (OS) (HR=0.74 p=0.0046) and progression-free survival (PFS) (HR=0.87 p=0.14). While patients primarily benefit from atezo if they are PD-L1 high, we found that even PD-L1 negative patients benefited from atezo when enriched for this feature (22C3 PD-L1 assay: OS HR=0.59 p=0.015, PFS HR=0.8 p=0.25; SP142 PD-L1 assay: OS HR=0.74 p=0.12, PFS HR=0.88 p=0.45). We thus recognized a DP feature that was predictive for positive outcome with atezo treatment, independent of PD-L1 levels. This association was then validated in IMpower150 comparing ABCP to BCP, both overall (OS HR=0.69 p=0.012) and in PD-L1 negative patients (SP263 assay OS HR=0.56 p=0.034). Integrating with RNAseq, patients enriched for this DP feature showed similar enrichment for B and T gene signatures and depletion in CAF-related gene signatures, thus showing the harmonization of TME between different data modalities. Conclusions: Using a deep learning-based assay for quantifying pathology features of the TME from H&E images in two NSCLC trials, we identified a novel biomarker predictive of outcome to PD-L1 targeting therapy, even in PD-L1 low & negative patients. Importantly, our work shows how different data modalities (DP, gene expression) can be integrated to further our understanding of the TME. Citation Format: Aditi Qamra, Minu K. Srivastava, Eloisa Fuentes, Ben Trotter, Raymond Biju, Guillaume Chhor, James Cowan, Steven Gendreau, Webster Lincoln, Lisa McGinnis, Luciana Molinero, Namrata S. Patil, Amber Schedlbauer, Katja Schulze, Adam Stanford-Moore, Laura Chambre, Ilan Wapinski, David S. Shames, Hartmut Koeppen, Stephanie Hennek, Jane Fridlyand, Jennifer M. Giltnane, Assaf Amitai. Digital pathology based prognostic & predictive biomarkers in metastatic non-small cell lung cancer. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 5705.
- Published
- 2023
17. DNA epigenetic signature predictive of benefit from neoadjuvant chemotherapy in oesophageal adenocarcinoma
- Author
-
Steven G. Rozen, Heike I. Grabsch, Alvin Wei Tian Ng, William H. Allum, Wen Fong Ooi, Jimmy Bok Yan So, Wei Peng Yong, Patrick Tan, Ruth E Langley, Raghav Sundar, Vivien Koh, Ming Hui Lee, Lindsay C. Hewitt, David Cunningham, Matthew Nankivell, Nisha Padmanabhan, Taotao Sheng, Shenli Zhang, Aditi Qamra, Hermioni Zouridis, Imran Inam, RS: GROW - R2 - Basic and Translational Cancer Biology, Promovendi ODB, and Pathologie
- Subjects
Male ,0301 basic medicine ,Oncology ,Cancer Research ,Esophageal Neoplasms ,SURGERY ,medicine.medical_treatment ,Oesophageal adenocarcinoma ,Epigenesis, Genetic ,chemistry.chemical_compound ,0302 clinical medicine ,Antineoplastic Combined Chemotherapy Protocols ,Randomized Controlled Trials as Topic ,Aged, 80 and over ,DNA methylation ,METHYLATION ,Middle Aged ,Prognosis ,OPEN-LABEL ,Neoadjuvant Therapy ,Survival Rate ,Predictive biomarker ,030220 oncology & carcinogenesis ,Cohort ,Female ,Fluorouracil ,Adult ,medicine.medical_specialty ,RESECTION ,CAPECITABINE ,Adenocarcinoma ,03 medical and health sciences ,Internal medicine ,medicine ,Humans ,Chemotherapy ,Epigenetics ,Epigenetic signature ,Aged ,Neoplasm Staging ,Proportional Hazards Models ,business.industry ,PERIOPERATIVE CHEMOTHERAPY ,Cancer ,medicine.disease ,030104 developmental biology ,chemistry ,Cisplatin ,Neoplasm Grading ,business ,DNA ,GASTRIC-CANCER - Abstract
Background: DNA methylation signatures describing distinct histological subtypes of oesophageal cancer have been reported. We studied DNA methylation in samples from the MRC OE02 phase III trial, which randomised patients with resectable oesophageal cancer to surgery alone (S) or neoadjuvant chemotherapy followed by surgery (CS).Aim: The aim of the study was to identify epigenetic signatures predictive of chemotherapy benefit in patients with oesophageal adenocarcinoma (OAC) from the OE02 trial and validate the findings in an independent cohort.Methods: DNA methylation was analysed using the Illumina GoldenGate platform on surgically resected OAC specimens from patients in the OE02 trial. Cox proportional hazard analysis was performed to select probes predictive of survival in the CS arm. Non-negative matrix factorisation was used to perform clustering and delineate DNA methylation signatures. The findings were validated in an independent cohort of patients with gastroesophageal adenocarcinoma treated with neoadjuvant chemotherapy.Results: A total of 229 patients with OAC were analysed from the OE02 trial (118 in the CS arm and 111 in the S arm). There was no difference in DNA methylation status between the CS and S arms. A metagene signature was created by dichotomising samples into two clusters. In cluster 1, patients in the CS arm had significant overall survival (OS) benefit (median OS CS: 931 days vs. S: 536 days [HR: 1.54, P = 0.031]). In cluster 2, patients in the CS arm had similar (or worse) OS compared with patients in the S arm (CS: 348 days vs. S: 472 days [HR: 0.70, P = 0.1], and test of interaction was significant (p = 0.005). In the validation cohort (n = 13), there was no difference in DNA methylation status in paired pre- and post-treatment samples. When the epigenetic signature was applied, cluster 1 samples had better OS (median OS, cluster 1: 1174 days vs. cluster 2: 392 days, HR: 3.47, p = 0.059)Conclusions: This is the first and largest study of DNA methylation in patients with OAC uniformly treated in a randomised phase III trial. We identified an epigenetic signature that may serve as a predictive biomarker for chemotherapy benefit in OAC. (C) 2019 Elsevier Ltd. All rights reserved.
- Published
- 2019
18. Prognostic and Predictive Biomarkers in Patients With Coronavirus Disease 2019 Treated With Tocilizumab in a Randomized Controlled Trial
- Author
-
Fang Cai, Ivan O. Rosas, Jordi Carratalà, David Summers, Jennifer Tom, Celia J F Lin, Paolo Bonfanti, Jacqueline McBride, Bruno François, William Stubbings, Montserrat Carrasco-Triguero, Charles Edouard Luyt, Kevin G. Blyth, Cor H van der Leest, Farrah Kheradmand, Lothar Wiese, Nidhi Rohatgi, Carrie M. Rosenberger, Larry W. Tsai, Thomas Benfield, Min Bao, Aditi Qamra, Derrick Haslem, Tom, J, Bao, M, Tsai, L, Qamra, A, Summers, D, Carrasco-Triguero, M, Mcbride, J, Rosenberger, C, Lin, C, Stubbings, W, Blyth, K, Carratalà, J, François, B, Benfield, T, Haslem, D, Bonfanti, P, van der Leest, C, Rohatgi, N, Wiese, L, Luyt, C, Kheradmand, F, Rosas, I, and Cai, F
- Subjects
medicine.medical_specialty ,Randomization ,medicine.medical_treatment ,Critical Care and Intensive Care Medicine ,Placebo ,law.invention ,chemistry.chemical_compound ,coronavirus disease 2019 ,tocilizumab ,Tocilizumab ,Randomized controlled trial ,law ,Internal medicine ,medicine ,Mechanical ventilation ,biology ,business.industry ,interleukin-6 ,ferritin ,Biochemical markers ,biomarkers ,COVID-19 ,medicine.disease ,Ferritin ,Pneumonia ,chemistry ,Marcadors bioquímics ,biology.protein ,biomarker ,Observational study ,business ,severe acute respiratory syndrome coronavirus 2 - Abstract
OBJECTIVES: To explore candidate prognostic and predictive biomarkers identified in retrospective observational studies (interleukin-6, C-reactive protein, lactate dehydrogenase, ferritin, lymphocytes, monocytes, neutrophils, d-dimer, and platelets) in patients with coronavirus disease 2019 pneumonia after treatment with tocilizumab, an anti-interleukin-6 receptor antibody, using data from the COVACTA trial in patients hospitalized with severe coronavirus disease 2019 pneumonia. DESIGN: Exploratory analysis from a multicenter, randomized, double-blind, placebo-controlled, phase 3 trial. SETTING: Hospitals in North America and Europe. PATIENTS: Adults hospitalized with severe coronavirus disease 2019 pneumonia receiving standard care. INTERVENTION: Randomly assigned 2:1 to IV tocilizumab 8 mg/kg or placebo. MEASUREMENTS AND MAIN RESULTS: Candidate biomarkers were measured in 295 patients in the tocilizumab arm and 142 patients in the placebo arm. Efficacy outcomes assessed were clinical status on a seven-category ordinal scale (1, discharge; 7, death), mortality, time to hospital discharge, and mechanical ventilation (if not receiving it at randomization) through day 28. Prognostic and predictive biomarkers were evaluated continuously with proportional odds, binomial or Fine-Gray models, and additional sensitivity analyses. Modeling in the placebo arm showed all candidate biomarkers except lactate dehydrogenase and d-dimer were strongly prognostic for day 28 clinical outcomes of mortality, mechanical ventilation, clinical status, and time to hospital discharge. Modeling in the tocilizumab arm showed a predictive value of ferritin for day 28 clinical outcomes of mortality (predictive interaction, p = 0.03), mechanical ventilation (predictive interaction, p = 0.01), and clinical status (predictive interaction, p = 0.02) compared with placebo. CONCLUSIONS: Multiple biomarkers prognostic for clinical outcomes were confirmed in COVACTA. Ferritin was identified as a predictive biomarker for the effects of tocilizumab in the COVACTA patient population; high ferritin levels were associated with better clinical outcomes for tocilizumab compared with placebo at day 28.
- Published
- 2021
19. HNF4α pathway mapping identifies wild-type IDH1 as a targetable metabolic node in gastric cancer
- Author
-
Manjie Xing, Bin Tean Teh, Jing Tan, Xuewen Ong, Shang Li, Angie Lay Keng Tan, Patrick Tan, Wen Fong Ooi, Tingdong Yan, Zul Fazreen Adam Isa, Zhimei Li, Hassan Ashktorab, Duane T. Smoot, Aditi Qamra, Kakoli Das, Chang Xu, Shyh-Chang Ng, Ming Hui Lee, Kie Kyon Huang, Benjamin Yan-Jiang Chua, Shamaine Wei Ting Ho, Tannistha Nandi, Xiaosai Yao, Su Ting Tay, and Joyce Suling Lin
- Subjects
Transcriptome ,IDH1 ,Metabolomics ,Isocitrate dehydrogenase ,Gene expression ,Gastroenterology ,medicine ,Cancer research ,Cancer ,Biology ,medicine.disease ,Gene ,Chromatin immunoprecipitation - Abstract
ObjectiveGastric cancer (GC) is a leading cause of cancer mortality. Previous studies have shown that hepatocyte nuclear factor-4α (HNF4α) is specifically overexpressed in GC and functionally required for GC development. In this study, we investigated, on a genome-wide scale, target genes of HNF4α and oncogenic pathways driven by HNF4α and HNF4α target genes.DesignWe performed HNF4α chromatin immunoprecipitation followed by sequencing across multiple GC cell lines, integrating HNF4α occupancy data with (epi)genomic and transcriptome data of primary GCs to define HNF4α target genes of in vitro and in vivo relevance. To investigate mechanistic roles of HNF4α and HNF4α targets, we performed cancer metabolic measurements, drug treatments and functional assays including murine xenograft experiments.ResultsGene expression analysis across 19 tumour types revealed HNF4α to be specifically upregulated in GCs. Unbiased pathway analysis revealed organic acid metabolism as the top HNF4α-regulated pathway, orthogonally supported by metabolomic analysis. Isocitrate dehydrogenase 1 (IDH1) emerged as a convergent HNF4α direct target gene regulating GC metabolism. We show that wild-type IDH1 is essential for GC cell survival, and that certain GC cells can be targeted by IDH1 inhibitors.ConclusionsOur results highlight a role for HNF4α in sustaining GC oncogenic metabolism, through the regulation of IDH1. Drugs targeting wild-type IDH1 may thus have clinical utility in GCs exhibiting HNF4α overexpression, expanding the role of IDH1 in cancer beyond IDH1/2 mutated malignancies.
- Published
- 2019
20. Epigenomic promoter alterations predict for benefit from immune checkpoint inhibition in metastatic gastric cancer
- Author
-
Patrick Tan, Raghav Sundar, J.W. Lee, Seungtae Kim, Won Ki Kang, Angie Lay-Keng Tan, Kyoung-Mee Kim, Aditi Qamra, and Kie Kyon Huang
- Subjects
Epigenomics ,0301 basic medicine ,Cell cycle checkpoint ,Biopsy ,T-Lymphocytes ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,Pembrolizumab ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Stomach Neoplasms ,Neoplasms ,Gastrointestinal Tumors ,medicine ,Humans ,Immunologic Factors ,Progression-free survival ,Neoplasm Metastasis ,Promoter Regions, Genetic ,Base Sequence ,business.industry ,gastric cancer ,Original Articles ,Hematology ,Immunotherapy ,Progression-Free Survival ,epigenetic alternate promoter ,Immune checkpoint ,Early Gastric Cancer ,Nivolumab ,030104 developmental biology ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,immune checkpoint inhibition ,pembrolizumab ,Transcription Initiation Site ,business - Abstract
Background Utilization of alternative transcription start sites through alterations in epigenetic promoter regions causes reduced expression of immunogenic N-terminal peptides, which may facilitate immune evasion in early gastric cancer. We hypothesized that tumors with high alternate promoter utilization would be resistant to immune checkpoint inhibition in metastatic gastric cancer. Patients and methods Two cohorts of patients with metastatic gastric cancer treated with immunotherapy were analyzed. The first cohort (N = 24) included patients treated with either nivolumab or pembrolizumab. Alternate promoter utilization was measured using the NanoString® (NanoString Technologies, Seattle, WA, USA) platform on archival tissue samples. The second cohort was a phase II clinical trial of patients uniformly treated with pembrolizumab (N = 37). Fresh tumor biopsies were obtained, and transcriptomic analysis was carried out on RNAseq data. Alternate promoter utilization was correlated to T-cell cytolytic activity, objective response rate and survival. Results In the first cohort 8 of 24 (33%) tumors were identified to have high alternate promoter utilization (APhigh), and this was used to define the APhigh tertile of the second cohort (13 APhigh of 37). APhigh tumors exhibited decreased markers of T-cell cytolytic activity and lower response rates (8% versus 42%, P = 0.03). Median progression-free survival was lower in the APhigh group (55 versus 180 days, P = 0.0076). In multivariate analysis, alternative promoter utilization was an independent predictor of immunotherapy survival [hazard ratio 0.29, 95% confidence interval 0.099–0.85, P = 0.024). Analyzing tumoral evolution through paired pre-treatment and post-treatment biopsies, we observed consistent shifts in alternative promoter utilization rate associated with clinical response. Conclusion A substantial proportion of metastatic gastric cancers utilize alternate promoters as a mechanism of immune evasion, and these tumors may be resistant to anti-PD1 immune checkpoint inhibition. Alternate promoter utilization is thus a potential mechanism of resistance to immune checkpoint inhibition, and a novel predictive biomarker for immunotherapy. Trial Registration ClinicalTrials.gov Identifier: NCT#02589496
- Published
- 2019
21. Accessibility Over Transposable Elements Reveals Genetic Determinants of Stemness Properties in Normal and Leukemic Hematopoiesis
- Author
-
Jean C.Y. Wang, Amanda Mitchell, John E. Dick, Giacomo Grillo, Naoya Takayama, Seyed Ali Madani Tonekaboni, Christopher Arlidge, Aditi Qamra, Andrea Arruda, Bettina Nadorp, Mathieu Lupien, Alex Murison, and Mark D. Minden
- Subjects
Leukemia ,Haematopoiesis ,LYL1 ,CTCF ,FLI1 ,medicine ,Myeloid leukemia ,Computational biology ,Stem cell ,Biology ,medicine.disease ,Chromatin - Abstract
Despite most acute myeloid leukemia (AML) patients achieving complete remission after induction chemotherapy, two thirds of patients will relapse with fatal disease within 5 years. AML is organized as a cellular hierarchy sustained by leukemia stem cells (LSC) at the apex, with LSC properties directly linked to tumor progression, therapy failure and disease relapse 1–5. Despite the central role of LSC in poor patient outcomes, little is known of the genetic determinants of their stemness properties 6–8. Although much AML research focuses on mutational processes and their impact on gene expression programs, the genetic determinants of cell state properties including stemness expand beyond mutations, relying on the genetic architecture captured in the chromatin of each cell 9–11. As LSCs share many functional and molecular properties with normal hematopoietic stem cells (HSC), we identified genetic determinants of primitive populations enriched for LSCs and HSCs in comparison with their downstream mature progeny by investigating their chromatin accessibility. Our work reveals how distinct transposable element (TE) subfamilies are used in primitive versus mature populations, functioning as docking sites for stem cell-associated regulators of genome topology, including CTCF, or lineage-specific transcription regulators in primitive and mature populations, respectively. We further show how TE subfamilies accessible in LSCs define docking sites for several oncogenic drivers in AML, namely FLI1, LYL1 and MEIS1. Using chromatin accessibility profiles from a cohort of AML patients, we further show the clinical utility of our TE accessibility-based LSCTE121 scoring scheme to identify patients with high rates of relapse. Collectively, our work reveals how different accessible TE subfamilies serve as genetic determinants of stemness properties in normal and leukemic hematopoietic stem cells.
- Published
- 2021
22. Prognostic and predictive biomarkers in patients with COVID-19 treated with tocilizumab in a randomised controlled trial
- Author
-
Cor H van der Leest, Charles Edouard Luyt, Celia J F Lin, Jacqueline McBride, Bruno François, Kevin G. Blyth, Lothar Wiese, Fang Cai, Ivan O. Rosas, Nidhi Rohatgi, David Summers, Montserrat Carrasco-Triguero, Jordi Carratalà, Paolo Bonfanti, Jennifer Tom, Larry Tsai, Carrie M. Rosenberger, Min Bao, Thomas Benfield, Farrah Kheradmand, William Stubbings, Aditi Qamra, and Derrick Haslem
- Subjects
Mechanical ventilation ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Context (language use) ,Subgroup analysis ,Placebo ,law.invention ,chemistry.chemical_compound ,Tocilizumab ,Randomized controlled trial ,chemistry ,law ,Internal medicine ,Clinical endpoint ,medicine ,Biomarker (medicine) ,business - Abstract
SummaryBackgroundRetrospective observational studies suggest that interleukin-6 (IL-6), C-reactive protein (CRP), lactate dehydrogenase (LDH), ferritin, lymphocytes, monocytes, neutrophils, D-dimer, and platelets are associated with disease progression, treatment outcomes, or both, in patients with COVID-19 pneumonia. We explored these candidate prognostic and predictive biomarkers with efficacy outcomes after treatment with tocilizumab, an anti–IL-6 receptor antibody using data from the COVACTA trial for patients hospitalised with severe COVID-19 pneumonia.MethodsCandidate biomarkers were measured in 295 patients in the tocilizumab arm and 142 patients in the placebo arm. Efficacy outcomes assessed were clinical status on a seven-category ordinal scale (1, discharge; 7, death), mortality, time to hospital discharge, and mechanical ventilation (if not receiving it at randomisation) through day 28. Prognostic and predictive biomarkers were evaluated continuously with proportional odds, binomial or Fine-Gray models, and additional sensitivity analyses.FindingsModelling in the placebo arm showed all candidate biomarkers except LDH and D-dimer were strongly prognostic for day 28 clinical outcomes of mortality, mechanical ventilation, clinical status, and time to hospital discharge. Modelling in the tocilizumab arm showed a predictive value of ferritin for day 28 clinical outcomes of mortality (predictive interaction p=0.03), mechanical ventilation (predictive interaction p=0.01), and clinical status (predictive interaction p=0.02) compared with placebo.InterpretationMultiple biomarkers prognostic for clinical outcomes were confirmed in COVACTA. Ferritin was identified as a predictive biomarker for the effects of tocilizumab in the COVACTA patient population; high ferritin levels were associated with better clinical outcomes for tocilizumab compared with placebo at day 28.RESEARCH IN CONTEXTEvidence before this studyThe efficacy and safety of the anti–interleukin-6 receptor antibody tocilizumab in the treatment of patients hospitalised with COVID-19 pneumonia was investigated in COVACTA, a double-blind, randomised, placebo-controlled trial. The primary endpoint of improved clinical status on a seven-category ordinal scale (1, discharged/ready for discharge; 7, death) at day 28 was not met in this trial. Among the secondary endpoints, no difference in mortality at day 28 was observed, but time to hospital discharge was shorter in the tocilizumab group. Subgroup analysis suggested there might be a treatment benefit in patients grouped according to their ordinal scale category at baseline.We searched PubMed on September 14, 2020, using the terms “tocilizumab AND (COVID-19 OR coronavirus) AND biomarker” with no language or date restrictions. The search retrieved 18 articles, four of which identified laboratory measures as potential biomarkers in patients who received tocilizumab for the treatment of COVID-19 pneumonia. The biomarkers reported in these studies include interleukin-6, C-reactive protein, ferritin, fibrinogen, liver transaminases, lymphocytes, platelets, and D-dimer. However, these previous studies were single-centre, retrospective, observational studies. Larger, prospective, controlled trials are needed to investigate potential prognostic and predictive biomarkers to assess the outcomes and response to treatments for COVID-19.Added value of this studyThis exploratory analysis of data from COVACTA demonstrated interleukin-6, C-reactive protein, ferritin, neutrophils (percentage and absolute count), neutrophil-to-lymphocyte ratio, lymphocytes (percentage and absolute count), monocytes (percentage), and platelets as strong prognostic biomarkers in patients hospitalised with severe COVID-19 pneumonia. More important, ferritin showed predictive value for tocilizumab treatment effects on day 28 clinical outcomes of mortality, mechanical ventilation (among the subgroup of patients not receiving mechanical ventilation at randomisation), and clinical status compared with placebo.Implications of all the available evidenceIn patients with elevated levels of ferritin at baseline, tocilizumab decreased the probability of death, mechanical ventilation, and worsening clinical status at day 28 compared with placebo, suggesting that ferritin might be useful as a predictive biomarker of efficacy outcomes for tocilizumab in patients with severe COVID-19 pneumonia.
- Published
- 2020
23. Telomere dysfunction cooperates with epigenetic alterations to impair murine embryonic stem cell fate commitment
- Author
-
Lea Harrington, Tsz Wai Chu, Julissa Tsao, Neil Winegarden, Cheryl H. Arrowsmith, Monika Sharma, Mathieu Lupien, Danielle A Henry, Aditi Qamra, Dalia Barsyte-Lovejoy, and Mélanie Criqui
- Subjects
0301 basic medicine ,QH301-705.5 ,Science ,Cellular differentiation ,macromolecular substances ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Epigenesis, Genetic ,Histones ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Telomerase reverse transcriptase ,Epigenetics ,Biology (General) ,Cellular Senescence ,mouse ,epigenetics ,General Immunology and Microbiology ,General Neuroscience ,telomerase reverse transcriptase ,General Medicine ,Telomere ,embryonic stem cells ,Chromosomes and Gene Expression ,telomeres ,Embryonic stem cell ,Chromatin ,Cell biology ,cell differentiation ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,Medicine ,Stem cell ,PRC2 ,Research Article - Abstract
The precise relationship between epigenetic alterations and telomere dysfunction is still an extant question. Previously, we showed that eroded telomeres lead to differentiation instability in murine embryonic stem cells (mESCs) via DNA hypomethylation at pluripotency-factor promoters. Here, we uncovered that telomerase reverse transcriptase null (Tert-/-) mESCs exhibit genome-wide alterations in chromatin accessibility and gene expression during differentiation. These changes were accompanied by an increase of H3K27me3 globally, an altered chromatin landscape at the Pou5f1/Oct4 promoter, and a refractory response to differentiation cues. Inhibition of the Polycomb Repressive Complex 2 (PRC2), an H3K27 tri-methyltransferase, exacerbated the impairment in differentiation and pluripotency gene repression in Tert-/- mESCs but not wild-type mESCs, whereas inhibition of H3K27me3 demethylation led to a partial rescue of the Tert-/- phenotype. These data reveal a new interdependent relationship between H3K27me3 and telomere integrity in stem cell lineage commitment that may have implications in aging and cancer.
- Published
- 2020
24. Author response: Telomere dysfunction cooperates with epigenetic alterations to impair murine embryonic stem cell fate commitment
- Author
-
Lea Harrington, Tsz Wai Chu, Aditi Qamra, Dalia Barsyte-Lovejoy, Neil Winegarden, Cheryl H. Arrowsmith, Mathieu Lupien, Mélanie Criqui, Monika Sharma, Danielle A Henry, and Julissa Tsao
- Subjects
Telomere dysfunction ,Epigenetics ,Biology ,Embryonic stem cell ,Cell biology - Published
- 2020
25. Genomic and epigenomic EBF1 alterations modulate TERT expression in gastric cancer
- Author
-
Zul Fazreen Adam Isa, Mandoli Amit, Aditi Qamra, Mei Mei Chang, Nisha Padmanabhan, Kakoli Das, Jing Wang, Mohana Ray, Angie Lay Keng Tan, Giovani Claresta Wijaya, Michael A. Beer, Shamaine Wei Ting Ho, Xuewen Ong, Patrick Tan, Ming Hui Lee, Jing Tan, Kie Kyon Huang, Bin Tean Teh, Chukwuemeka George Anene-Nzelu, Taotao Sheng, Zhimei Li, Heike I. Grabsch, Polly Poon, Su Ting Tay, Shenli Zhang, Shang Li, Tannistha Nandi, Jing Quan Lim, Xiaosai Yao, Po Hsien Lee, Wen Fong Ooi, Kevin P. White, Roger Foo, Tingdong Yan, Ley Moy Ng, Gregorio E. Fazzi, Steven G. Rozen, Jeanie Wu, Yu Amanda Guo, Manjie Xing, Kevin Lim, Lijia Ma, Yue Ning Lam, Joyce Suling Lin, Anders Jacobsen Skanderup, Chang Xu, Pathologie, and RS: GROW - R2 - Basic and Translational Cancer Biology
- Subjects
0301 basic medicine ,Epigenomics ,Somatic cell ,GROUP PROTEIN EZH2 ,Repressor ,PROGRESSION ,CAPECITABINE ,Biology ,TELOMERASE ACTIVITY ,Response Elements ,DNA methyltransferase ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,0302 clinical medicine ,Stomach Neoplasms ,Cell Line, Tumor ,medicine ,Humans ,Transcription factor ,Telomerase ,RECOGNITION ,Cancer ,General Medicine ,PROMOTER MUTATIONS ,CHEMOTHERAPY ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,TRANSCRIPTION FACTORS ,030104 developmental biology ,DIFFERENTIATION ,030220 oncology & carcinogenesis ,B-CELL FACTOR-1 ,Mutation ,Cancer research ,biology.protein ,Trans-Activators ,Histone deacetylase activity ,PRC2 ,Research Article - Abstract
Transcriptional reactivation of telomerase catalytic subunit (TERT) is a frequent hallmark of cancer, occurring in 90% of human malignancies. However, specific mechanisms driving TERT reactivation remain obscure for many tumor types and in particular gastric cancer (GC), a leading cause of global cancer mortality. Here, through comprehensive genomic and epigenomic analysis of primary GCs and GC cell lines, we identified the transcription factor early B cell factor 1 (EBF1) as a TERT transcriptional repressor and inactivation of EBF1 function as a major cause of TERT upregulation. Abolishment of EBF1 function occurs through 3 distinct (epi)genomic mechanisms. First, EBF1 is epigenetically silenced via DNA methyltransferase, polycomb-repressive complex 2 (PRC2), and histone deacetylase activity in GCs. Second, recurrent, somatic, and heterozygous EBF1 DNA-binding domain mutations result in the production of dominant-negative EBF1 isoforms. Third, more rarely, genomic deletions and rearrangements proximal to the TERT promoter remobilize or abolish EBF1-binding sites, derepressing TERT and leading to high TERT expression. EBF1 is also functionally required for various malignant phenotypes in vitro and in vivo, highlighting its importance for GC development. These results indicate that multimodal genomic and epigenomic alterations underpin TERT reactivation in GC, converging on transcriptional repressors such as EBF1.
- Published
- 2018
26. HoxC5 and miR-615-3p target newly evolved genomic regions to repress hTERT and inhibit tumorigenesis
- Author
-
Jing Wang, Melissa J. Fullwood, Lai-Fong Poon, Steve Rozen, Xuezhi Bi, Prabha Sampath, Chang Xu, Yan Ping Loh, James O.J. Davies, Patrick Tan, Dongliang Ma, Alice Cheung, Manjie Xing, Muhammad Khairul Ramlee, Sujoy Ghosh, Aditi Qamra, Wen Fong Ooi, Eyleen L. K. Goh, Frederic Bard, Shang Li, Joscelyn Jun Quan Ng, Jim R. Hughes, Gopinath M Sundaram, Jess Hui Jie Ho, Tingdong Yan, Luay Aswad, Vinay Tergaonkar, and School of Biological Sciences
- Subjects
0301 basic medicine ,Science ,cells ,Cellular differentiation ,General Physics and Astronomy ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Mice ,Cell Line, Tumor ,Neoplasms ,microRNA ,Animals ,Humans ,Telomerase reverse transcriptase ,lcsh:Science ,Promoter Regions, Genetic ,Psychological repression ,Transcription factor ,neoplasms ,3' Untranslated Regions ,Telomerase ,Homeodomain Proteins ,Multidisciplinary ,Three prime untranslated region ,Cell Differentiation ,General Chemistry ,Hep G2 Cells ,Upstream Enhancer ,Cell biology ,enzymes and coenzymes (carbohydrates) ,MicroRNAs ,030104 developmental biology ,Cell Transformation, Neoplastic ,Enhancer Elements, Genetic ,HEK293 Cells ,Tumorigenesis ,embryonic structures ,Genomic ,MCF-7 Cells ,Homeobox ,lcsh:Q ,biological phenomena, cell phenomena, and immunity ,5' Untranslated Regions ,HeLa Cells - Abstract
The repression of telomerase activity during cellular differentiation promotes replicative aging and functions as a physiological barrier for tumorigenesis in long-lived mammals, including humans. However, the underlying mechanisms remain largely unclear. Here we describe how miR-615-3p represses hTERT expression. mir-615-3p is located in an intron of the HOXC5 gene, a member of the highly conserved homeobox family of transcription factors controlling embryogenesis and development. Unexpectedly, we found that HoxC5 also represses hTERT expression by disrupting the long-range interaction between hTERT promoter and its distal enhancer. The 3′UTR of hTERT and its upstream enhancer region are well conserved in long-lived primates. Both mir-615-3p and HOXC5 are activated upon differentiation, which constitute a feed-forward loop that coordinates transcriptional and post-transcriptional repression of hTERT during cellular differentiation. Deregulation of HOXC5 and mir-615-3p expression may contribute to the activation of hTERT in human cancers., The expression of telomerase catalytic subunit hTERT is frequently upregulated in many cancers. Here, the authors show HoxC5 and miR-615-3p can negatively regulate hTERT to impede tumorigenesis by targeting the newly evolved cis-regulatory genomic elements of hTERT.
- Published
- 2018
27. VHL deficiency drives enhancer activation of oncogenes in clear cell renal cell carcinoma
- Author
-
Tannistha Nandi, Xiaosai Yao, Jim R. Hughes, Manjie Xing, Jian Yuan Goh, Kenneth Tou En Chang, Steven G. Rozen, Dachuan Huang, Zhimei Li, Qiang Yu, Iain Beehuat Tan, Kevin Lim, Wen Fong Ooi, Jing Tan, Chang Xu, James O.J. Davies, Cassandra Zhengxuan He, Peiyong Guan, Joanna Koh, Su Ting Tay, Christopher Cheng, James Z.Z. Qu, Shang Li, Bin Tean Teh, Bryan C. Tan, Gertrud Steger, Puay Hoon Tan, Alexander Lezhava, Patrick Tan, Yue Ning Lam, Swe Swe Myint, Joyce Suling Lin, Gary Loh, Michelle Shu Wen Ng, Yang Sun Chan, David L. Silver, Jing Han Hong, Aditi Qamra, Ai Ping Lee-Lim, and Giovani Claresta Wijaya
- Subjects
0301 basic medicine ,Regulation of gene expression ,Genetics ,Histone Acetyltransferase p300 ,Biology ,medicine.disease ,medicine.disease_cause ,Chromatin ,law.invention ,03 medical and health sciences ,Clear cell renal cell carcinoma ,030104 developmental biology ,Oncology ,law ,Cancer research ,medicine ,Suppressor ,Enhancer ,Carcinogenesis ,Epigenomics - Abstract
Protein-coding mutations in clear cell renal cell carcinoma (ccRCC) have been extensively characterized, frequently involving inactivation of the von Hippel–Lindau (VHL) tumor suppressor. Roles for noncoding cis-regulatory aberrations in ccRCC tumorigenesis, however, remain unclear. Analyzing 10 primary tumor/normal pairs and 9 cell lines across 79 chromatin profiles, we observed pervasive enhancer malfunction in ccRCC, with cognate enhancer-target genes associated with tissue-specific aspects of malignancy. Superenhancer profiling identified ZNF395 as a ccRCC-specific and VHL-regulated master regulator whose depletion causes near-complete tumor elimination in vitro and in vivo. VHL loss predominantly drives enhancer/superenhancer deregulation more so than promoters, with acquisition of active enhancer marks (H3K27ac, H3K4me1) near ccRCC hallmark genes. Mechanistically, VHL loss stabilizes HIF2α–HIF1β heterodimer binding at enhancers, subsequently recruiting histone acetyltransferase p300 without overtly affecting preexisting promoter–enhancer interactions. Subtype-specific driver mutations such as VHL may thus propagate unique pathogenic dependencies in ccRCC by modulating epigenomic landscapes and cancer gene expression. Significance: Comprehensive epigenomic profiling of ccRCC establishes a compendium of somatically altered cis-regulatory elements, uncovering new potential targets including ZNF395, a ccRCC master regulator. Loss of VHL, a ccRCC signature event, causes pervasive enhancer malfunction, with binding of enhancer-centric HIF2α and recruitment of histone acetyltransferase p300 at preexisting lineage-specific promoter–enhancer complexes. Cancer Discov; 7(11); 1284–305. ©2017 AACR. See related commentary by Ricketts and Linehan, p. 1221. This article is highlighted in the In This Issue feature, p. 1201
- Published
- 2017
28. Epigenetic alternate promoter utilization and association with PD-L1 expression in Epstein–Barr virus positive gastric cancer
- Author
-
Aditi Qamra, Raghav Sundar, Shenli Zhang, Cedric Chuan Young Ng, Angie Lay Keng Tan, Kyoung-Mee Kim, Patrick Tan, Bin Tean Teh, and Jeeyun Lee
- Subjects
Cancer Research ,business.industry ,Cancer ,Epstein-Barr Virus Positive ,medicine.disease ,Immune checkpoint ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,Pd l1 expression ,Epigenetics ,business ,030215 immunology - Abstract
e15509 Background: We recently elicited the role of epigenetic promoter alterations as a mechanism of immune-evasion and primary resistance to immune checkpoint inhibition in gastric cancer. High prevalence of epigenetic modifications are known to occur in Epstein-Barr virus associated gastric cancer (EBVaGC). EBVaGC has high response rates to anti-PD-1 immune checkpoint inhibitors and is associated with high levels of PD-L1 expression. However, not all EBVaGC express PD-L1 and mechanisms that mediate these phenomena are unknown. Methods: We performed NanoString profiling and PD-L1 immunohistochemistry (using Dako PD-L1 IHC 22C3) on tissue from gastric cancer patients undergoing primary tumor resections at Samsung Medical Centre, South Korea. NanoString panel was designed for 90 recurrent somatic alternate promoter-related genes, and immune-related genes including PD-L1. EBV status was determined using EBV-encoded RNA in situ hybridization and categorized as EBVaGC and EBV-negative. Samples in the top-quartile of alternate promoter utilization were classified as APhigh and the remaining APlow. Results: A total of 272 samples (EBVaGC n = 79; EBV-negative n = 193) were included in this study. EBVaGC had significantly higher PD-L1 expression (p < 0.001) compared to EBV-negative samples. APhigh group (n = 67) consisted of 61 EBV-negative and 6 EBVaGC samples. EBVaGC APhigh tumors had significantly lower PD-L1 transcript expression compared to EBVaGC APlow tumors (p = 0.011, Wilcoxon-rank sum). Similar correlation was also found with PD-L1 IHC combined positive score (CPS)(median CPS score 1 vs 8, p = 0.047). There was a trend towards poorer survival for EBVaGC APhigh tumors (vs EBVaGC APlow; HR 0.23, 95% CI: 0.046 – 1.23, p = 0.087). EBV-negative APhigh tumors also had lower PD-L1 expression (vs EBV-negative APlow; p = 0.046, Wilcoxon-rank sum). Conclusions: Increased utilization of epigenetic alternate promoter isoforms correlates with lower transcriptomic and protein expression of PD-L1 in EBVaGC. Here we describe a potential mechanism of immune-evasion to explain low immune-infiltration and PD-L1 expression that occurs in a group of EBVaGC that is traditionally considered highly immunogenic.
- Published
- 2019
29. DNA methylation signature predictive of benefit from neoadjuvant chemotherapy in esophageal adenocarcinoma: Results from the MRC OEO2 phase III trial
- Author
-
Taotao Sheng, Ming Hui Lee, Matthew Nankivell, Patrick Tan, Raghav Sundar, Wen Fong Ooi, Shenli Zhang, William H. Allum, Ruth E Langley, Nisha Padmanabhan, David Cunningham, Aditi Qamra, Hermioni Zouridis, Heike I. Grabsch, and Alvin Wei Tian Ng
- Subjects
Oncology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Esophageal adenocarcinoma ,03 medical and health sciences ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Internal medicine ,DNA methylation ,medicine ,business ,030215 immunology - Abstract
43 Background: Platinum and 5-Fluorouracil (5FU) neoadjuvant chemotherapy followed by surgery is one of the standard approaches for patients with resectable EAC. To date, there are no predictive biomarkers of chemotherapy benefit. We hypothesize that DNA methylation of genes in key biologic and oncogenic pathways predict for chemotherapy benefit in EAC. Methods: In the OE02 trial, 802 patients with resectable esophageal carcinoma were randomised to surgery alone (S) versus two cycles of cisplatin and 5FU chemotherapy followed by surgery (CS). DNA was extracted from 213 EAC resection specimens (110 from the (CS) arm, 103 from the (S) arm). DNA methylation was analyzed at 1505 CpG sites within 807 genes using the Illumina GoldenGate platform. Cox proportional hazard analysis was performed to identify predictive markers of survival in (CS) arm; non-negative matrix factorization (NMF) was used to delineate methylation signatures. Results: Methylation status of 1505 CpG sites had no statistical difference between the (CS) and (S) arms. In the (CS) arm, 87 (5.7%) CpG sites were initially identified as promising candidates in univariate analysis (p < 0.05 cutoff). NMF generated a 4 CpG site signature which divided patients into poor risk and good risk. Genes involved in the signature include RUNX1T1, CCND2, MST1R and MMP14. Survival was significantly different between poor risk and good risk in (CS) arm (HR 0.32, 95% CI: 0.21 to 0.52, p < 0.0001). No difference in survival was detected in the surgery arm (HR 1.12, 95% CI: 0.76 to 1.80, p = 0.48), suggesting the signature served as a predictive and not prognostic biomarker. Methylation signature remained an independent predictor of survival in multivariate analysis with clinicopathologic factors (along with age and vascular invasion). Conclusions: Chemotherapy does not appear to change methylation status of EAC. Hypermethylation of RUNX1T1, CCND2 and hypomethylation of MST1R and MMP14 leads to significantly decreased benefit from chemotherapy in EA. We describe an epigenetic signature which may serve as a predictive biomarker for chemotherapy benefit using data form the largest bank of DNA methylation in EA reported to date.
- Published
- 2019
30. Epigenomic profiling of primary gastric adenocarcinoma reveals super-enhancer heterogeneity
- Author
-
Chang Xu, Weng Hoong Chan, Patrick Tan, Mei Chee Lim, Muhammad Khairul Ramlee, Shang Li, James Qu Zhengzhong, Joyce Lin Suling, Aditi Qamra, James O.J. Davies, Ai Ping Lee-Lim, Melissa J. Fullwood, Steve Rozen, Wen Fong Ooi, Ming Hui Lee, Khee Chee Soo, Tannistha Nandi, Yang Sun Chan, Chow Yin Wong, Su Ting Tay, Jianjun Liu, Pierce K. H. Chow, Kevin Lim, Deepak Babu, Sun Young Rha, Axel M. Hillmer, Bin Tean Teh, Jim R. Hughes, Lai Fong Poon, Manjie Xing, Fan Cao, Xiaosai Yao, Astrid Irwanto, Wai-Keong Wong, and Hock Soo Ong
- Subjects
0301 basic medicine ,Genetics ,Multidisciplinary ,Somatic cell ,Science ,General Physics and Astronomy ,General Chemistry ,Biology ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,3. Good health ,Chromatin ,03 medical and health sciences ,030104 developmental biology ,Super-enhancer ,medicine ,Cancer research ,Epigenetics ,Enhancer ,Carcinogenesis ,Transcription factor ,Epigenomics - Abstract
Regulatory enhancer elements in solid tumours remain poorly characterized. Here we apply micro-scale chromatin profiling to survey the distal enhancer landscape of primary gastric adenocarcinoma (GC), a leading cause of global cancer mortality. Integrating 110 epigenomic profiles from primary GCs, normal gastric tissues and cell lines, we highlight 36,973 predicted enhancers and 3,759 predicted super-enhancers respectively. Cell-line-defined super-enhancers can be subclassified by their somatic alteration status into somatic gain, loss and unaltered categories, each displaying distinct epigenetic, transcriptional and pathway enrichments. Somatic gain super-enhancers are associated with complex chromatin interaction profiles, expression patterns correlated with patient outcome and dense co-occupancy of the transcription factors CDX2 and HNF4α. Somatic super-enhancers are also enriched in genetic risk SNPs associated with cancer predisposition. Our results reveal a genome-wide reprogramming of the GC enhancer and super-enhancer landscape during tumorigenesis, contributing to dysregulated local and regional cancer gene expression., Gene expression is regulated by enhancers and super-enhancers, which can be identified by chromatin profiling. Here, the authors surveyed gastric cancer samples and cell lines to identify enhancer elements exhibiting somatic alterations.
- Published
- 2016
31. Transcriptional analysis of immune genes in Epstein-Barr virus-associated gastric cancer and association with clinical outcomes
- Author
-
Jeeyun Lee, Raghav Sundar, Angie Lay Keng Tan, Patrick Tan, Kyoung-Mee Kim, Aditi Qamra, Cedric Chuan Young Ng, Shenli Zhang, and Bin Tean Teh
- Subjects
Male ,0301 basic medicine ,Oncology ,Epstein-Barr Virus Infections ,Herpesvirus 4, Human ,Cancer Research ,Lymphocyte ,Immune checkpoint inhibitors ,Programmed Cell Death 1 Receptor ,medicine.disease_cause ,B7-H1 Antigen ,Cohort Studies ,0302 clinical medicine ,Surgical oncology ,hemic and lymphatic diseases ,Transcriptional analysis ,IRGs ,biology ,digestive, oral, and skin physiology ,Gastroenterology ,General Medicine ,Middle Aged ,Advanced gastric cancer ,Immunohistochemistry ,Primary tumor ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Adult ,medicine.medical_specialty ,CD8 Antigens ,Disease-Free Survival ,Virus ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Immune system ,Stomach Neoplasms ,Internal medicine ,PD-L1 ,Biomarkers, Tumor ,medicine ,Humans ,Immune gene ,Aged ,Perforin ,business.industry ,Gene Expression Profiling ,Cancer ,medicine.disease ,Epstein–Barr virus ,digestive system diseases ,030104 developmental biology ,biology.protein ,Cancer research ,business - Abstract
Epstein–Barr virus-associated gastric cancer (EBVaGC) has traditionally been associated with high expression of PD-L1 and immune infiltration. Correlations between PD-L1 and other immune-related gene (IRG) expressions in EBVaGC have not been previously described. We performed NanoString® transcriptomic profiling and PD-L1 immunohistochemistry (IHC) (using the FDA approved Dako PD-L1 IHC 22C3) on EBVaGC samples from gastric cancer patients undergoing primary tumor resections at Samsung Medical Centre, South Korea. For controls, EBV-negative samples from the previously reported Asian Cancer Research Group (EBVnegACRG) cohort were used. Genes tested included PD-L1 and other IRGs related to intra-tumoral cytolytic activity, cytokines and immune checkpoints. Samples with PD-L1 expression > 34th percentile were defined as PD-L1high and the remaining as PD-L1low. We identified 71 cases of EBVaGC and 193 EBV-negative ACRG samples as controls. EBVaGC showed higher expression of all queried immune genes compared to EBVnegACRG samples (p
- Published
- 2018
32. ADAR-Mediated RNA Editing Predicts Progression and Prognosis of Gastric Cancer
- Author
-
Bin Tean Teh, Steve Rozen, Daniel G. Tenen, Jeeyun Lee, Jimmy Bok Yan So, Sun Yong Rha, Kar Tong Tan, Aditi Qamra, Patrick Tan, Yujing Liu, HuiQi Hong, Henry Yang, Jaymie Siqi Lin, Tim Chan, Leilei Chen, Lihua Qi, Yangyang Song, Vanessa Hui En Ng, Feng Zhu, Su Ting Tay, Jing Guo, and Khay Guan Yeoh
- Subjects
0301 basic medicine ,Male ,Adenosine Deaminase ,Sialoglycoproteins ,Biology ,Epigenesis, Genetic ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,Stomach Neoplasms ,Humans ,Codon ,Gene ,Genetics ,Messenger RNA ,Hepatology ,Sequence Analysis, RNA ,Gastroenterology ,RNA ,RNA-Binding Proteins ,Middle Aged ,Prognosis ,RNA silencing ,030104 developmental biology ,chemistry ,RNA editing ,ADAR ,Disease Progression ,Female ,RNA Editing ,DNA - Abstract
Backgroud & Aims Gastric cancer (GC) is the third leading cause of global cancer mortality. Adenosine-to-inosine RNA editing is a recently described novel epigenetic mechanism involving sequence alterations at the RNA but not DNA level, primarily mediated by ADAR (adenosine deaminase that act on RNA) enzymes. Emerging evidence suggests a role for RNA editing and ADARs in cancer, however, the relationship between RNA editing and GC development and progression remains unknown. Methods In this study, we leveraged on the next-generation sequencing transcriptomics to demarcate the GC RNA editing landscape and the role of ADARs in this deadly malignancy. Results Relative to normal gastric tissues, almost all GCs displayed a clear RNA misediting phenotype with ADAR1/2 dysregulation arising from the genomic gain and loss of the ADAR1 and ADAR2 gene in primary GCs, respectively. Clinically, patients with GCs exhibiting ADAR1/2 imbalance demonstrated extremely poor prognoses in multiple independent cohorts. Functionally, we demonstrate in vitro and in vivo that ADAR-mediated RNA misediting is closely associated with GC pathogenesis, with ADAR1 and ADAR2 playing reciprocal oncogenic and tumor suppressive roles through their catalytic deaminase domains, respectively. Using an exemplary target gene PODXL (podocalyxin-like), we demonstrate that the ADAR2-regulated recoding editing at codon 241 (His to Arg) confers a loss-of-function phenotype that neutralizes the tumorigenic ability of the unedited PODXL . Conclusions Our study highlights a major role for RNA editing in GC disease and progression, an observation potentially missed by previous next-generation sequencing analyses of GC focused on DNA alterations alone. Our findings also suggest new GC therapeutic opportunities through ADAR1 enzymatic inhibition or the potential restoration of ADAR2 activity.
- Published
- 2015
33. Abstract 3559: Functional characterization of HNF4α in gastric cancer
- Author
-
Chang Xu, Aditi Qamra, Wen Fong Ooi, and Patrick Tan
- Subjects
Cancer Research ,Oncology ,business.industry ,Cancer research ,Medicine ,Cancer ,business ,medicine.disease - Abstract
Gastric cancer (GC) is a leading cause of cancer mortality worldwide and particularly common in East Asia. Transcription factors are frequently amplified or overexpressed in GC and might contribute to tumorgenesis, however the underlying mechanisms largely remain unknown. Our previous data indicates that HNF4α is a common target of three amplified transcription factors (GATA4, GATA6 and KLF5) in gastric cancer. HNF4α had also been found to be related to diverse aspects of tumorigenesis. In liver and colon cancers, there are reports indicating that the targets of HNF4α are mainly involved in intermediary metabolism, however, its tissue-specific downstream targets and pathways have not been systemically studied yet. Therefore, a comprehensive functional characterization of HNF4α is required to identify its tissue-specific role in gastric cancer. In this study, we analyzed the expression features of HNF4α in GC, and found the expression level of HNF4α is significantly higher in early stage tumor and in CIN and MSI compared to GS and EBV subtypes of GC according to TCGA classification method. In addition, we performed the HNF4α ChIP in GC cell lines, and the results revealed a number of HNF4α binding sites in GC. Comparison with the published ChIP-seq data of Caco2 and HepG2 cell lines indicated the existence of GC-exclusive targets. Furthermore, functional annotation analysis revealed that the GC-exclusive targets are significantly related to the negative regulation of cellular metabolic process compared to the common targets. These preliminary data suggests that HNF4α might play a tissue specific role in GC, especially in metabolic related processes. Through further target validation and biological function studies, this study hopes to systemically investigate of HNF4α’s role in GC tumorigenesis, especially in metabolism related pathways. Citation Format: Chang Xu, Wen Fong Ooi, Aditi Qamra, Patrick Tan. Functional characterization of HNF4α in gastric cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3559. doi:10.1158/1538-7445.AM2017-3559
- Published
- 2017
34. Nanoscale chromatin profiling of gastric adenocarcinoma reveals cancer-associated cryptic promoters and somatically acquired regulatory elements
- Author
-
Masafumi Muratani, Suling Joyce Lin, Steve Rozen, Wai-Keong Wong, Ming Hui Lee, Khee Chee Soo, Wen Fong Ooi, Hock Soo Ong, Hui-Hoon Chua, Huck-Hui Ng, Manjie Xing, Weng Hoong Chan, Liang Kee Goh, Niantao Deng, Bin Tean Teh, Su Ting Tay, Simeen Malik, Chang Xu, London L.P.J. Ooi, Aditi Qamra, Luke Lin Chuen Tan, Jeanie Wu, Patrick Tan, Shenli Zhang, Qiang Yu, and Pierce Kah-How Chow
- Subjects
General Physics and Astronomy ,Adenocarcinoma ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Histones ,Stomach Neoplasms ,Humans ,Nanotechnology ,Regulatory Elements, Transcriptional ,Promoter Regions, Genetic ,Enhancer ,Gene ,Alleles ,ChIA-PET ,Genetics ,Multidisciplinary ,biology ,Promoter ,DNA ,DNA, Neoplasm ,General Chemistry ,DNA Methylation ,DNA Fingerprinting ,Chromatin ,Histone ,Case-Control Studies ,Mutation ,DNA methylation ,biology.protein ,Transcription Initiation Site - Abstract
Chromatin alterations are fundamental hallmarks of cancer. To study chromatin alterations in primary gastric adenocarcinomas, we perform nanoscale chromatin immunoprecipitation sequencing of multiple histone modifications in five gastric cancers and matched normal tissues. We identify hundreds of somatically altered promoters and predicted enhancers. Many cancer-associated promoters localize to genomic sites lacking previously annotated transcription start sites (cryptic promoters), driving expression of nearby genes involved in gastrointestinal cancer, embryonic development and tissue specification. Cancer-associated promoters overlap with embryonic stem cell regions targeted by polycomb repressive complex 2, exhibiting promoter bivalency and DNA methylation loss. We identify somatically acquired elements exhibiting germline allelic biases and non-coding somatic mutations creating new promoters. Our findings demonstrate the feasibility of profiling chromatin from solid tumours with limited tissue to identify regulatory elements, transcriptional patterns and regulatory genetic variants associated with cancer.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.