1
|
McDermott M, Mehta R, Roussos Torres ET, MacLean AL. Modeling the dynamics of EMT reveals genes associated with pan-cancer intermediate states and plasticity. NPJ Syst Biol Appl 2025; 11:31. [PMID: 40210876 PMCID: PMC11986130 DOI: 10.1038/s41540-025-00512-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/28/2025] [Indexed: 04/12/2025] Open
Abstract
Epithelial-mesenchymal transition (EMT) is a cell state transition co-opted by cancer that drives metastasis via stable intermediate states. Here we study EMT dynamics to identify marker genes of highly metastatic intermediate cells via mathematical modeling with single-cell RNA sequencing (scRNA-seq) data. Across multiple tumor types and stimuli, we identified genes consistently upregulated in EMT intermediate states, many previously unrecognized as EMT markers. Bayesian parameter inference of a simple EMT mathematical model revealed tumor-specific transition rates, providing a framework to quantify EMT progression. Consensus analysis of differential expression, RNA velocity, and model-derived dynamics highlighted SFN and NRG1 as key regulators of intermediate EMT. Independent validation confirmed SFN as an intermediate state marker. Our approach integrates modeling and inference to identify genes associated with EMT dynamics, offering biomarkers and therapeutic targets to modulate tumor-promoting cell state transitions driven by EMT.
Collapse
Affiliation(s)
- MeiLu McDermott
- Department of Quantitative and Computational Biology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Riddhee Mehta
- Department of Quantitative and Computational Biology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA
| | - Evanthia T Roussos Torres
- Department of Medicine, Division of Medical Oncology, Keck School of Medicine, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Adam L MacLean
- Department of Quantitative and Computational Biology, Dornsife College of Letters, Arts and Sciences, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Tseng YC, Liu PF, Chen YR, Yang WH, Chang CC, Chang HW, Lee CH, Goan YG, Shu CW. Elevated neuregulin‑1 expression modulates tumor malignancy and autophagy in esophageal squamous cell carcinoma. Int J Mol Med 2025; 55:62. [PMID: 39950316 PMCID: PMC11878479 DOI: 10.3892/ijmm.2025.5503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/24/2025] [Indexed: 03/06/2025] Open
Abstract
The 5‑year survival rate of patients with esophageal squamous cell carcinoma (ESCC) is <20%, highlighting the need for the development of novel therapeutic targets. Neuregulin‑1 (NRG1), a transmembrane protein involved in cell proliferation and survival signaling, has unclear biological functions and clinical value in ESCC. The present study investigated the association between NRG1 expression and ESCC by analyzing data from both patients with ESCC and The Cancer Genome Atlas database. Reverse transcription‑quantitative PCR and immunohistochemistry staining were used to determine the levels of gene and protein in the tissue. The findings revealed that NRG1 gene and protein levels were significantly higher in tumor tissues compared with the normal tissues. Elevated expression of NRG1 was associated with poor outcomes, particularly in patients with advanced ESCC. Silencing NRG1 decreased both its mRNA and protein levels, disrupting key signaling pathways, such as phosphorylated (p‑)AKT and cellular rapidly accelerated fibrosarcoma (p‑cRAF), which led to decreased cancer cell proliferation, migration and tumor sphere formation, along with increased cell death. High expression levels of NRG1 and cRAF were significantly associated with poor prognosis. Additionally, silencing NRG1 promoted autophagosome and autolysosome formation, decreasing LC3B levels. The use of the autophagy inhibitor chloroquine significantly enhanced cell death induced by NRG1 silencing, suggesting that autophagy functions as a survival mechanism in ESCC cells in which NRG1 is silenced. Furthermore, high co‑expression of NRG1 and LC3B was associated with a worse prognosis. On the whole, the present study demonstrated that targeting NRG1 with autophagy inhibitors may serve as a potential therapeutic strategy for ESCC.
Collapse
Affiliation(s)
- Yen-Chiang Tseng
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 81341, Taiwan, R.O.C
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 300025, Taiwan, R.O.C
- Department of Pharmacy and Master Program, Tajen University, Pingtung 907391, Taiwan, R.O.C
| | - Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80756, Taiwan, R.O.C
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80756, Taiwan, R.O.C
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80756, Taiwan, R.O.C
| | - Yu-Ru Chen
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Wen-Hsin Yang
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| | - Chia-Che Chang
- Department of Oncology, Zuoying Armed Forces General Hospital, Kaohsiung 81320, Taiwan, R.O.C
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80756, Taiwan, R.O.C
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80756, Taiwan, R.O.C
| | - Cheng-Hsin Lee
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80756, Taiwan, R.O.C
| | - Yih-Gang Goan
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung 81341, Taiwan, R.O.C
- Division of Thoracic Surgery, Department of Surgery, Pingtung Veterans General Hospital, Pingtung 91245, Taiwan, R.O.C
| | - Chih-Wen Shu
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80756, Taiwan, R.O.C
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
- Innovation Center for Drug Development and Optimization, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, R.O.C
| |
Collapse
|
3
|
Diao H, Zhao F, Wu M, Zhang Y, Tao Q, Chen S, Lin D. LncRNA Expression Profiles in C6 Ceramide Treatment Reveal lnc_025370 as a Promoter in Canine Mammary Carcinoma CHMp Cells Progression. Curr Issues Mol Biol 2024; 46:14190-14203. [PMID: 39727977 DOI: 10.3390/cimb46120849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
Canine mammary carcinomas (CMCs) represent the most prevalent form of cancer in female dogs, characterized by a high incidence and mortality rate. C6 ceramide is recognized for its multifaceted anti-cancer properties, yet its specific influence on CMCs remains to be elucidated. Long noncoding RNAs (lncRNAs), now recognized as functional "dark matter" in precision oncology, are particularly intriguing, with 44% of canine lncRNAs exhibiting tissue-specific expression. In this study, we performed a thorough analysis of lncRNA expression profiles to uncover the mechanisms behind C6 ceramide's anti-cancer activity in CHMp cells. Our findings reveal that C6 ceramide notably inhibits the proliferation of CHMp cells. RNA sequencing identified 4522 lncRNAs with expression changes following C6 ceramide treatment, of which 2936 were upregulated and 1586 were downregulated. Further investigation into Lnc_025370 showed that it is predominantly nuclear-localized and is significantly downregulated by C6 ceramide treatment. Functional studies discovered that overexpression of Lnc_025370 enhances the growth and metastatic capabilities of CHMp cells, which is associated with an increase in NRG1, and concurrently diminishes the anti-cancer effectiveness of C6 ceramide in vitro. Mouse xenograft models also showed that Lnc_025370 overexpression promotes tumor growth and Ki67 expression. Together, our results suggest that Lnc_025370 acts as a pivotal target mediator of C6 ceramide's anti-cancer effects, facilitating the malignant progression of CHMp cells.
Collapse
Affiliation(s)
- Hongxiu Diao
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Fangying Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Meijin Wu
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Yan Zhang
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Qianting Tao
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Shichao Chen
- Joint Laboratory of Animal Pathogen Prevention and Control of Fujian-Nepal, College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
- Fujian Province Joint Laboratory of Animal Pathogen Prevention and Control of the "Belt and Road", College of Animal Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Degui Lin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
4
|
Puchała Ł, Gonkowski S, Rytel L, Wojtkiewicz J, Grzegorzewski WJ. Distribution and neurochemical characterisation of neurons containing neuregulin 1 in the enteric nervous system within the porcine small intestine. J Vet Res 2024; 68:623-632. [PMID: 39776680 PMCID: PMC11702255 DOI: 10.2478/jvetres-2024-0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction The enteric nervous system (ENS) in the wall of the gastrointestinal tract is complex and comprises many neurons, which are differentiated in terms of structure, function and neurochemistry. Neuregulin 1 (NRG 1) is one of the neuronal factors synthesised in the ENS about the distribution and functions of which relatively little is known. The present study is the first description of the distribution of NRG 1 in the ENS in various segments of the porcine small intestine. Material and Methods Fragments were excised from the duodenum, jejunum and ileum of five euthanised Piétrain × Duroc sows, 18-20 kg in weight and eight weeks of age. Paraformaldehyde-fixed and dehydrated tissue was sectioned and double-labelling immunofluorescence was performed using Alexa Fluor-conjugated secondary antibodies to visualise neuregulin 1 and its colocalisation with vasoactive intestinal polypeptide (VIP), galanin (GAL), and the neuronal isoform of nitric oxide synthase (nNOS) in the myenteric and inner and outer submucosal plexuses, with PGP 9.5 serving as a pan-neuronal marker. Results Neuregulin 1 was observed in all enteric plexuses in each segment of the small intestine. The percentage of NRG 1-positive neurons ranged from 8.38 ± 0.55% of all neurons in the jejunal inner submucous plexus to 21.52 ± 0.98% in the duodenal myenteric plexus. Cells which were NRG 1-positive also contained VIP, GAL and nNOS in all segments of the small intestine to a degree which varied by small intestine segment and enteric plexus type. Conclusion The results indicate that NRG 1-positive neurons are present in the ENS of the porcine small intestine and differ significantly neurochemically, which may suggest a multifaceted role for NRG-1 in the controlling of the small intestine activity.
Collapse
Affiliation(s)
| | | | - Liliana Rytel
- Department of Internal Diseases with Clinic, Faculty of Veterinary Medicine, University of Warmia and Mazury, 10-719Olsztyn, Poland
| | - Joanna Wojtkiewicz
- Department of Physiology and Pathophysiology, School of Medicine, University of Warmia and Mazury, 10-082Olsztyn, Poland
| | | |
Collapse
|
5
|
Zhou YT, Chu JH, Zhao SH, Li GL, Fu ZY, Zhang SJ, Gao XH, Ma W, Shen K, Gao Y, Li W, Yin YM, Zhao C. Quantitative systems pharmacology modeling of HER2-positive metastatic breast cancer for translational efficacy evaluation and combination assessment across therapeutic modalities. Acta Pharmacol Sin 2024; 45:1287-1304. [PMID: 38360930 PMCID: PMC11130324 DOI: 10.1038/s41401-024-01232-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/23/2024] [Indexed: 02/17/2024]
Abstract
HER2-positive (HER2+) metastatic breast cancer (mBC) is highly aggressive and a major threat to human health. Despite the significant improvement in patients' prognosis given the drug development efforts during the past several decades, many clinical questions still remain to be addressed such as efficacy when combining different therapeutic modalities, best treatment sequences, interindividual variability as well as resistance and potential coping strategies. To better answer these questions, we developed a mechanistic quantitative systems pharmacology model of the pathophysiology of HER2+ mBC that was extensively calibrated and validated against multiscale data to quantitatively predict and characterize the signal transduction and preclinical tumor growth kinetics under different therapeutic interventions. Focusing on the second-line treatment for HER2+ mBC, e.g., antibody-drug conjugates (ADC), small molecule inhibitors/TKI and chemotherapy, the model accurately predicted the efficacy of various drug combinations and dosing regimens at the in vitro and in vivo levels. Sensitivity analyses and subsequent heterogeneous phenotype simulations revealed important insights into the design of new drug combinations to effectively overcome various resistance scenarios in HER2+ mBC treatments. In addition, the model predicted a better efficacy of the new TKI plus ADC combination which can potentially reduce drug dosage and toxicity, while it also shed light on the optimal treatment ordering of ADC versus TKI plus capecitabine regimens, and these findings were validated by new in vivo experiments. Our model is the first that mechanistically integrates multiple key drug modalities in HER2+ mBC research and it can serve as a high-throughput computational platform to guide future model-informed drug development and clinical translation.
Collapse
Affiliation(s)
- Ya-Ting Zhou
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jia-Hui Chu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Shu-Han Zhao
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ge-Li Li
- Gusu School, Nanjing Medical University, Suzhou, 215000, China
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Zi-Yi Fu
- Department of Breast Disease Research Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Su-Jie Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xue-Hu Gao
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Jiangsu Hengrui Medicine Co. Ltd, Shanghai, 200245, China
| | - Wen Ma
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Kai Shen
- Jiangsu Hengrui Medicine Co. Ltd, Shanghai, 200245, China
| | - Yuan Gao
- QSPMed Technologies, Nanjing, 210000, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yong-Mei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Chen Zhao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| |
Collapse
|
6
|
Hou G, Niu T, Jia A, Zhang Y, Chen X, Wei H, Jia Y, Xu Y, Li Y, Wang P, Chatterjee A. NRG1 promotes tumorigenesis and metastasis and afatinib treatment efficiency is enhanced by NRG1 inhibition in esophageal squamous cell carcinoma. Biochem Pharmacol 2023; 218:115920. [PMID: 37989416 DOI: 10.1016/j.bcp.2023.115920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly aggressive tumor with significant heterogeneity in incidence and outcomes. The role of Neuregulin 1 (NRG1) in ESCC and its contribution to aggressiveness remain unknown. This study aims to investigate the functions and molecular mechanisms of NRG1 in ESCC as well as the treatment strategy for ESCC with overexpression of NRG1. We firstly demonstrated the upregulation of NRG1 and a negative correlation trend between patients' overall survival (OS) and the expression level of NRG1 in esophageal cancer. And then we found NRG1 promoted cell proliferation, migration, inhibited apoptosis, and accelerated tumorigenesis and metastasis in ESCC using cell lines and xenograft models. Furthermore, we discovered that NRG1 activated the NF-κB/MMP9 signaling pathway, contributing to the metastatic phenotype in ESCC. Finally, we show that afatinib (FDA approved cancer growth blocker) could inhibit ESCC with overexpressed NRG1 and down-regulation of NRG1 along with afatinib treatment provides higher efficient strategy. This study uncovers the critical role and molecular mechanism of NRG1 in ESCC tumorigenesis and metastasis, suggesting its potential as a novel biomarker for ESCC treatment.
Collapse
Affiliation(s)
- Guiqin Hou
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand.
| | - Tengda Niu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ang Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yingying Zhang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xunan Chen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Huiyun Wei
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yilin Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yichao Xu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Li
- Center of Advanced Analysis & Gene Sequencing, Zhengzhou University, Zhengzhou 450001, China
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Aniruddha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, PO Box 56, Dunedin 9054, New Zealand; School of Health Sciences and Technology, UPES, Dehradun, India.
| |
Collapse
|
7
|
Hirabayashi M, Georges D, Clifford GM, de Martel C. Estimating the Global Burden of Epstein-Barr Virus-Associated Gastric Cancer: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol 2023; 21:922-930.e21. [PMID: 35963539 DOI: 10.1016/j.cgh.2022.07.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Evidence suggests that a fraction of new gastric cancer cases may be etiologically associated with Epstein-Barr virus (EBV), a known carcinogenic agent. We aimed to systematically explore the proportion of EBV-positive gastric cancer. METHODS We did a systematic review (PROSPERO CRD42020164473) from January 1990 to August 2021. For each country and geographical region with available data, pooled prevalence and corresponding 95% confidence intervals (CIs) of EBV in gastric tumors were calculated for 3 subtypes of gastric adenocarcinoma (conventional adenocarcinoma, lymphoepithelioma-like gastric carcinoma, and remnant/stump carcinoma). For conventional adenocarcinoma, prevalence ratios (PRs) were presented for sex, Lauren's classification, gastric cancer stage, and anatomical location of the stomach. RESULTS In 220 eligible studies including over 68,000 cases of conventional gastric adenocarcinoma, EBV prevalence in tumor cells was 7.5% (95% CI, 6.9%-8.1%) and was higher in men compared with women (PR, 2.1; 95% CI, 1.9-2.4), in diffuse type compared with intestinal type (PR, 1.3; 95% CI, 1.1-1.5), and in the proximal region compared with the distal region (PR, 2.5; 95% CI, 2.0-3.1). There was no difference in EBV prevalence by gastric cancer stage. EBV prevalence was 75.9% (95% CI, 62.8%-85.5%) among lymphoepithelioma-like gastric carcinoma and 26.3% (95% CI, 22.2%-32.0%) among remnant or stump carcinoma. CONCLUSIONS Assuming a causal association between EBV and gastric cancer, our findings, when applied to the GLOBOCAN 2020 gastric cancer incidence, suggest that primary prevention such as the development of an effective EBV vaccine might prevent 81,000 EBV-associated gastric cancer cases worldwide annually.
Collapse
Affiliation(s)
- Mayo Hirabayashi
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Damien Georges
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Gary M Clifford
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France
| | - Catherine de Martel
- Early Detection, Prevention and Infections Branch, International Agency for Research on Cancer, Lyon, France.
| |
Collapse
|
8
|
Long-term platinum-based drug accumulation in cancer-associated fibroblasts promotes colorectal cancer progression and resistance to therapy. Nat Commun 2023; 14:746. [PMID: 36765091 PMCID: PMC9918738 DOI: 10.1038/s41467-023-36334-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 01/27/2023] [Indexed: 02/12/2023] Open
Abstract
A substantial proportion of cancer patients do not benefit from platinum-based chemotherapy (CT) due to the emergence of drug resistance. Here, we apply elemental imaging to the mapping of CT biodistribution after therapy in residual colorectal cancer and achieve a comprehensive analysis of the genetic program induced by oxaliplatin-based CT in the tumor microenvironment. We show that oxaliplatin is largely retained by cancer-associated fibroblasts (CAFs) long time after the treatment ceased. We determine that CT accumulation in CAFs intensifies TGF-beta activity, leading to the production of multiple factors enhancing cancer aggressiveness. We establish periostin as a stromal marker of chemotherapeutic activity intrinsically upregulated in consensus molecular subtype 4 (CMS4) tumors and highly expressed before and/or after treatment in patients unresponsive to therapy. Collectively, our study underscores the ability of CT-retaining CAFs to support cancer progression and resistance to treatment.
Collapse
|
9
|
Epigenetic regulation of Neuregulin 1 promotes breast cancer progression associated to hyperglycemia. Nat Commun 2023; 14:439. [PMID: 36707514 PMCID: PMC9883495 DOI: 10.1038/s41467-023-36179-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 01/18/2023] [Indexed: 01/28/2023] Open
Abstract
Hyperglycemia is a risk factor for breast cancer-related morbidity and mortality. Hyperglycemia induces Neuregulin 1 (Nrg1) overexpression in breast cancer, which subsequently promotes tumor progression. However, molecular mechanisms underlying hyperglycemia-induced Nrg1 overexpression remain poorly understood. Here, we show that hyperglycemia causes active histone modifications at the Nrg1 enhancer, forming enhanceosome complexes where recombination signal binding protein for immunoglobulin kappa J region (RBPJ), E1A binding protein p300 (P300), and SET domain containing 1 A (SETD1A) are recruited to upregulate Nrg1 expression. Deletions in RBPJ-binding sites causes hyperglycemia-controlled Nrg1 levels to be downregulated, resulting in decreased tumor growth in vitro and in vivo. Mice with modest-temporary hyperglycemia, induced by low-dose short-exposure streptozotocin, display accelerated tumor growth and lapatinib resistance, whereas combining lapatinib with N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S42 phenylglycine t-butyl ester (DAPT) ameliorates tumor growth under these modest hyperglycemic conditions by inhibiting NOTCH and EGFR superfamilies. NOTCH activity is correlated with NRG1 levels, and high NRG1 levels predicts poor outcomes, particularly in HER2-positive breast cancer patients. Our findings highlight the hyperglycemia-linked epigenetic modulation of NRG1 as a potential therapeutic strategy for treating breast cancer patients with diabetes.
Collapse
|
10
|
Zhang C, Mei W, Zeng C. Oncogenic Neuregulin 1 gene (NRG1) fusions in cancer: A potential new therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2022; 1877:188707. [PMID: 35247506 DOI: 10.1016/j.bbcan.2022.188707] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 10/19/2022]
Abstract
It is widely established that chromosomal rearrangements induce oncogenesis in solid tumors. However, discovering chromosomal rearrangements that are targetable and actionable remains a difficulty. Targeting gene fusion or chromosomal rearrangement seems to be a powerful strategy to address malignancies characterized by gene rearrangement. Oncogenic NRG1 fusions are relatively rare drivers that infrequently occur across most tumor types. NRG1 fusions exhibit unique biological properties and are difficult to identify owing to their large intronic regions. NRG1 fusions can be detected using a variety of techniques, including fluorescence in situ hybridization, immunohistochemistry, or next-generation sequencing (NGS), with NGS-based RNA sequencing being the most sensitive. Previous studies have shown that NRG1 fusion protein induces tumorigenesis, and numerous therapies targeting the ErbB signaling pathway, such as ErbB kinase inhibitors and monoclonal antibodies, have initially demonstrated encouraging anticancer efficacy in malignant tumors carrying NRG1 fusions. In this review, we present the characteristics and prevalence of NRG1 fusions in solid tumors. Additionally, we discuss the laboratory approaches for diagnosing NRG1 gene fusions. More importantly, we outline promising strategies for treating malignancies with NRG1 fusion.
Collapse
Affiliation(s)
- Congwang Zhang
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China
| | - Wuxuan Mei
- Clinical Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, China
| | - Changchun Zeng
- Department of Medical Laboratory, Shenzhen Longhua District Central Hospital, Guangdong Medical University, Shenzhen 518110, China.
| |
Collapse
|
11
|
Chen D, Ye Y, Guo S, Yao K. Progress in the Research and Targeted Therapy of ErbB/HER Receptors in Urothelial Bladder Cancer. Front Mol Biosci 2022; 8:800945. [PMID: 35004854 PMCID: PMC8735837 DOI: 10.3389/fmolb.2021.800945] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 11/30/2021] [Indexed: 11/19/2022] Open
Abstract
Bladder cancer is a lethal malignancy and a majority of bladder cancer arise from urothelial cells. Infiltration and metastasis are barriers for the radical cystectomy to achieve favored outcome and are the main cause of death. Systemic therapy, including chemotherapy, targeted therapy, and immunotherapy, is fundamental for these patients. erbB/HER receptors are found to be overexpressed in a subgroup of urothelial carcinoma, targeting erbB/HER receptors in these patients was found to be an efficient way in the era of genetic testing. To evaluate the role of erbB/HER receptors in bladder cancer, we reviewed the literature and ongoing clinical trials as regards to this topic to unveil the context of erbB/HER receptors in bladder cancer, which probably help to solidate the theoretical basis and might instruct further research.
Collapse
Affiliation(s)
- Dong Chen
- Department of Urology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yunlin Ye
- Department of Urology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shengjie Guo
- Department of Urology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kao Yao
- Department of Urology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
12
|
Hu L, Fan M, Shi S, Song X, Wang F, He H, Qi B. Dual target inhibitors based on EGFR: Promising anticancer agents for the treatment of cancers (2017-). Eur J Med Chem 2022; 227:113963. [PMID: 34749202 DOI: 10.1016/j.ejmech.2021.113963] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 02/06/2023]
Abstract
The EGFR family play a significant role in cell signal transduction and their overexpression is implicated in the pathogenesis of numerous human solid cancers. Inhibition of the EGFR-mediated signaling pathways by EGFR inhibitors is a widely used strategy for the treatment of cancers. In most cases, the EGFR inhibitors used in clinic were only effective when the cancer cells harbored specific activating EGFR mutations which appeared to preserve the ligand-dependency of receptor activation but altered the pattern of downstream signaling pathways. Moreover, cancer is a kind of multifactorial disease, and therefore manipulating a single target may result in treatment failure. Although drug combinations for the treatment of cancers proved to be successful, the use of two or more drugs concurrently still was a challenge in clinical therapy owing to various dose-limiting toxicities and drug-drug interactions caused by pharmacokinetic profiles changed. Therefore, a single drug targeting two or multiple targets could serve as an effective strategy for the treatment of cancers. In recent, drugs with diverse pharmacological effects have been shown to be more advantageous than combination therapies due to their lower incidences of side effects and more resilient therapies. Accordingly, dual target-single-agent strategy has become a popular field for cancer treatment, and researchers became more and more interest in the development of novel dual-target drugs in recent years. In this review, we briefly introduce the EGFR family proteins and synergisms between EGFR and other anticancer targets, and summarizes the development of potential dual target inhibitors based on wild-type and/or mutant EGFR for the treatment of solid cancers in the past five years. Additionally, the rational design and SARs of these dual target agents are also presented in detailed, which will lay a significant foundation for the further development of novel EGFR-based dual inhibitors with excellent druggability.
Collapse
Affiliation(s)
- Liping Hu
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Mengmeng Fan
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Shengmin Shi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Xiaomeng Song
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Fei Wang
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China
| | - Huan He
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| | - Baohui Qi
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, Guangdong Province, China.
| |
Collapse
|
13
|
Rosas D, Raez LE, Russo A, Rolfo C. Neuregulin 1 Gene ( NRG1). A Potentially New Targetable Alteration for the Treatment of Lung Cancer. Cancers (Basel) 2021; 13:cancers13205038. [PMID: 34680187 PMCID: PMC8534274 DOI: 10.3390/cancers13205038] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Treatment in oncology has and will keep evolving into an agnostic approach where therapies are guided more towards the identification and targeting of genetic abnormalities and less by organ of origin of the cancer, as has been done for decades. With every genetic abnormality being identified as a target, the pharmaceutical development of medications targeting these genes has grown, leading to better survival rates, quality of life and a bigger interest in finding new targets. Lung cancer is one of the best examples where targetable genetic abnormalities have led to substantial survival differences compared to patients undergoing empirical conventional chemotherapy. Translocations in the neuregulin 1 gene (NRG1) are one of many gene fusions that are becoming clinically significant, and it has the potential to become a targetable gene with ongoing clinical trials already in Europe and the US. This review aims to portray the importance and latest developments regarding this new fusion in lung cancer treatment. Abstract Oncogenic gene fusions are hybrid genes that result from structural DNA rearrangements, leading to unregulated cell proliferation by different mechanisms in a wide variety of cancer. This has led to the development of directed therapies to antagonize a variety of mechanisms that lead to cell growth or proliferation. Multiple oncogene fusions are currently targeted in lung cancer treatment, such as those involving ALK, RET, NTRK and ROS1 among many others. Neuregulin (NRG) gene fusion has been described in the development of normal tissue as well as in a variety of diseases, such as schizophrenia, Hirschsprung’s disease, atrial fibrillation and, most recently, the development of various types of solid tumors, such as renal, gastric, pancreatic, breast, colorectal and, more recently, lung cancer. The mechanism for this is that the NRG1 chimeric ligand leads to aberrant activation of ERBB2 signaling via PI3K-AKT and MAPK cellular cascades, leading to cell division and proliferation. Details regarding the incidence of these gene rearrangements are lacking. Limited case reports and case series have evaluated their clinicopathologic features and prognostic significance in the lung cancer population. Taking this into account, NRG1 could become a targetable alteration in selected patients. This review highlights how the knowledge of new molecular mechanisms of NRG1 fusion may help in gaining new insights into the molecular status of lung cancer patients and unveil a novel targetable molecular marker.
Collapse
Affiliation(s)
- Daniel Rosas
- The Internal Medicine Department, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Correspondence:
| | - Luis E. Raez
- Thoracic Oncology Program, Memorial Cancer Institute/Memorial Health Care System, Florida International University (FIU), Miami, FL 33021, USA;
| | | | - Christian Rolfo
- Clinical Research and Center for Thoracic Oncology, The Tisch Cancer Institute, Mount Sinai Health System & Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
14
|
MiR-200b Suppresses Gastric Cancer Cell Migration and Invasion by Inhibiting NRG1 through ERBB2/ERBB3 Signaling. JOURNAL OF ONCOLOGY 2021; 2021:4470778. [PMID: 34531912 PMCID: PMC8440071 DOI: 10.1155/2021/4470778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/20/2021] [Indexed: 12/09/2022]
Abstract
Purpose Accumulating evidence indicates that miRNAs (miRs) play crucial roles in the modulation of tumors development. However, the accurately mechanisms have not been entirely clarified. In this study, we aimed to explore the role of miR-200b in the development of gastric cancer (GC). Methods Western blot and RT-PCR were applied to detect epithelial-mesenchymal transition (EMT) marker expression and mRNA expression. Transwell assay was used for measuring the metastasis and invasiveness of GC cells. TargetScan system, luciferase reporter assay, and rescue experiments were applied for validating the direct target of miR-200b. Results MiR-200b was prominently decreased in GC tissues and cells, and its downregulation was an indicator of poor prognosis of GC patients. Reexpression of miR-200b suppressed EMT along with GC cell migration and invasion. Neuregulin 1 (NRG1) was validated as the target of miR-200b, and it rescued miR-200b inhibitory effect on GC progression. In GC tissues, the correlation of miR-200b with NRG1 was inverse. Conclusion MiR-200b suppressed EMT-related migration and invasion of GC through the ERBB2/ERBB3 signaling pathway via targeting NRG1.
Collapse
|
15
|
Gan HK, Millward M, Jalving M, Garrido-Laguna I, Lickliter JD, Schellens JHM, Lolkema MP, Van Herpen CLM, Hug B, Tang L, O'Connor-Semmes R, Gagnon R, Ellis C, Ganji G, Matheny C, Drilon A. A Phase I, First-in-Human Study of GSK2849330, an Anti-HER3 Monoclonal Antibody, in HER3-Expressing Solid Tumors. Oncologist 2021; 26:e1844-e1853. [PMID: 34132450 PMCID: PMC8488777 DOI: 10.1002/onco.13860] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 05/14/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND GSK2849330, an anti-HER3 monoclonal antibody that blocks HER3/Neuregulin 1 (NRG1) signaling in cancer cells, is engineered for enhanced antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity. This phase I, first-in-human, open-label study assessed the safety, pharmacokinetics (PK), pharmacodynamics, and preliminary activity of GSK2849330 in patients with HER3-expressing advanced solid tumors. PATIENTS AND METHODS Patients with various tumor types were prospectively selected for HER3 expression by immunohistochemistry; a subset was also screened for NRG1 mRNA expression. In the dose-escalation phase, patients received GSK2849330 1.4-30 mg/kg every 2 weeks, or 3 mg/kg or 30 mg/kg weekly, intravenously (IV). In the dose-expansion phase, patients received 30 mg/kg GSK2849330 IV weekly. RESULTS Twenty-nine patients with HER3-expressing cancers, of whom two expressed NRG1, received GSK2849330 (dose escalation: n = 18, dose expansion: n = 11). GSK2849330 was well tolerated. No dose-limiting toxicities were observed. The highest dose, of 30 mg/kg weekly, expected to provide full target engagement, was selected for dose expansion. Treatment-emergent adverse events (AEs) were mostly grade 1 or 2. The most common AEs were diarrhea (66%), fatigue (62%), and decreased appetite (31%). Dose-proportional plasma exposures were achieved, with evidence of HER3 inhibition in paired tissue biopsies. Of 29 patients, only 1 confirmed partial response, lasting 19 months, was noted in a patient with CD74-NRG1-rearranged non-small cell lung cancer (NSCLC). CONCLUSION GSK2849330 demonstrated a favorable safety profile, dose-proportional PK, and evidence of target engagement, but limited antitumor activity in HER3-expressing cancers. The exceptional response seen in a patient with CD74-NRG1-rearranged NSCLC suggests further exploration in NRG1-fusion-positive cancers. IMPLICATIONS FOR PRACTICE This first-in-human study confirms that GSK2849330 is well tolerated. Importantly, across a variety of HER3-expressing advanced tumors, prospective selection by HER3/NRG1 expression alone was insufficient to identify patients who could benefit from treatment with this antibody-dependent cell-mediated cytotoxicity- and complement-dependent cytotoxicity-enhanced anti-HER3 antibody. The only confirmed durable response achieved was in a patient with CD74-NRG1-rearranged lung cancer. This highlights the potential utility of screening for NRG1 fusions prospectively across tumor types to enrich potential responders to anti-HER3 agents in ongoing trials.
Collapse
Affiliation(s)
- Hui K Gan
- Department of Medical Oncology, Austin Health and Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia.,School of Medicine, Latrobe University School of Cancer Medicine, Melbourne, Victoria, Australia.,Department of Medicine, Melbourne University, Melbourne, Victoria, Australia
| | - Michael Millward
- Linear Clinical Research and University of Western Australia, Perth, Western Australia, Australia
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Centre Groningen, Groningen, The Netherlands
| | - Ignacio Garrido-Laguna
- Department of Internal Medicine, Oncology Division, University of Utah School of Medicine, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | | | - Jan H M Schellens
- Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Martijn P Lolkema
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Carla L M Van Herpen
- Radboud University Medical Center, Radboud University, Nijmegen, The Netherlands
| | - Bruce Hug
- GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Lihua Tang
- Independent Consultant, North Carolina, USA
| | - Robin O'Connor-Semmes
- Clinical Pharmacology, Modeling and Simulation, Parexel International, Durham, North Carolina, USA
| | | | | | | | | | - Alexander Drilon
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
16
|
Cheng LC, Kao TJ, Phan NN, Chiao CC, Yen MC, Chen CF, Hung JH, Jiang JZ, Sun Z, Wang CY, Hsu HP. Novel signaling pathways regulate SARS-CoV and SARS-CoV-2 infectious disease. Medicine (Baltimore) 2021; 100:e24321. [PMID: 33607766 PMCID: PMC7899890 DOI: 10.1097/md.0000000000024321] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/21/2020] [Indexed: 01/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 induces severe infection, and it is responsible for a worldwide disease outbreak starting in late 2019. Currently, there are no effective medications against coronavirus. In the present study, we utilized a holistic bioinformatics approach to study gene signatures of SARS-CoV- and SARS-CoV-2-infected Calu-3 lung adenocarcinoma cells. Through the Gene Ontology platform, we determined that several cytokine genes were up-regulated after SARS-CoV-2 infection, including TNF, IL6, CSF2, IFNL1, IL-17C, CXCL10, and CXCL11. Differentially regulated pathways were detected by the Kyoto Encyclopedia of Genes and Genomes, gene ontology, and Hallmark platform, including chemokines, cytokines, cytokine receptors, cytokine metabolism, inflammation, immune responses, and cellular responses to the virus. A Venn diagram was utilized to illustrate common overlapping genes from SARS-CoV- and SARS-CoV-2-infected datasets. An Ingenuity pathway analysis discovered an enrichment of tumor necrosis factor- (TNF-) and interleukin (IL)-17-related signaling in a gene set enrichment analysis. Downstream networks were predicted by the Database for Annotation, Visualization, and Integrated Discovery platform also revealed that TNF and TNF receptor 2 signaling elicited leukocyte recruitment, activation, and survival of host cells after coronavirus infection. Our discovery provides essential evidence for transcript regulation and downstream signaling of SARS-CoV and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Li-Chin Cheng
- Division of Colorectal Surgery, Department of Surgery, Chi-Mei Medical Center
| | - Tzu-Jen Kao
- The PhD Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes
- TMU Research Center of Neuroscience, Taipei Medical University, Taipei, Taiwan
| | - Nam Nhut Phan
- NTT Institute of Hi-Technology, Nguyen Tat Thanh (NTT) University, Ho Chi Minh City, Vietnam
| | - Chung-Chieh Chiao
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University
| | - Meng-Chi Yen
- Department of Emergency Medicine, Kaohsiung Medical University Hospital
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung
| | - Chien-Fu Chen
- School of Chinese Medicine for Post-Baccalaureate, I-Shou University
| | - Jui-Hsiang Hung
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Jia-Zhen Jiang
- Emergency Department, Huashan Hospital North, Fudan University, Shanghai, People's Republic of China
| | - Zhengda Sun
- Kaiser Permanente, Northern California Regional Laboratories, the Permanente Medical Group, Berkeley, CA, USA
| | - Chih-Yang Wang
- PhD Program for Cancer Molecular Biology and Drug Discovery
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei
| | - Hui-Ping Hsu
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
17
|
Xu X, Lu Y, Wu Y, Wang M, Wang X, Wang H, Chen B, Li Y. A signature of seven immune-related genes predicts overall survival in male gastric cancer patients. Cancer Cell Int 2021; 21:117. [PMID: 33602220 PMCID: PMC7891008 DOI: 10.1186/s12935-021-01823-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 01/06/2021] [Accepted: 02/09/2021] [Indexed: 12/24/2022] Open
Abstract
Background Gastric cancer (GC) has a high mortality rate and is one of the most fatal malignant tumours. Male sex has been proven as an independent risk factor for GC. This study aimed to identify immune-related genes (IRGs) associated with the prognosis of male GC. Methods RNA sequencing and clinical data were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed IRGs between male GC and normal tissues were identified by integrated bioinformatics analysis. Univariate and multivariate Cox regression analyses were applied to screen survival-associated IRGs. Then, GC patients were separated into high- and low-risk groups based on the median risk score. Furthermore, a nomogram was constructed based on the TCGA dataset. The prognostic value of the risk signature model was evaluated by Kaplan-Meier curve, receiver operating characteristic (ROC), Harrell’s concordance index and calibration curves. In addition, the gene expression dataset from the Gene Expression Omnibus (GEO) was also downloaded for external validation. The relative proportions of 22 types of infiltrating immune cells in each male GC sample were evaluated using CIBERSORT. Results
A total of 276 differentially expressed IRGs were screened, including 189 up-regulated and 87 down-regulated genes. Subsequently, a seven-IRGs signature (LCN12, CCL21, RNASE2, CGB5, NRG4, AGTR1 and NPR3) was identified to be significantly associated with the overall survival (OS) of male GC patients. Survival analysis indicated that patients in the high-risk group exhibited a poor clinical outcome. The results of multivariate analysis revealed that the risk score was an independent prognostic factor. The established nomogram could be used to evaluate the prognosis of individual male GC patients. Further analysis showed that the prognostic model had excellent predictive performance in both TCGA and validated cohorts. Besides, the results of tumour-infiltrating immune cell analysis indicated that the seven-IRGs signature could reflect the status of the tumour immune microenvironment. Conclusions Our study developed a novel seven-IRGs risk signature for individualized survival prediction of male GC patients.
Collapse
Affiliation(s)
- Xin Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China.,Anhui Medical University, Hefei, 230022, China
| | - Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China.,Anhui Medical University, Hefei, 230022, China
| | - Youliang Wu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China.,Anhui Medical University, Hefei, 230022, China
| | - Mingliang Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China.,Anhui Medical University, Hefei, 230022, China
| | - Xiaodong Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China.,Anhui Medical University, Hefei, 230022, China
| | - Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Bo Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Avenue, Hefei, 230022, Anhui, China.
| |
Collapse
|
18
|
Abstract
Neuregulins, members of the largest subclass of growth factors of the epidermal growth factor family, mediate a myriad of cellular functions including survival, proliferation, and differentiation in normal tissues through binding to receptor tyrosine kinases of the ErbB family. However, aberrant neuregulin signaling in the tumor microenvironment is increasingly recognized as a key player in initiation and malignant progression of human cancers. In this chapter, we focus on the role of neuregulin signaling in the hallmarks of cancer, including cancer initiation and development, metastasis, as well as therapeutic resistance. Moreover, role of neuregulin signaling in the regulation of tumor microenvironment and targeting of neuregulin signaling in cancer from the therapeutic perspective are also briefly discussed.
Collapse
|
19
|
Berdiel-Acer M, Maia A, Hristova Z, Borgoni S, Vetter M, Burmester S, Becki C, Michels B, Abnaof K, Binenbaum I, Bethmann D, Chatziioannou A, Hasmann M, Thomssen C, Espinet E, Wiemann S. Stromal NRG1 in luminal breast cancer defines pro-fibrotic and migratory cancer-associated fibroblasts. Oncogene 2021; 40:2651-2666. [PMID: 33692466 PMCID: PMC8049869 DOI: 10.1038/s41388-021-01719-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 01/31/2023]
Abstract
HER3 is highly expressed in luminal breast cancer subtypes. Its activation by NRG1 promotes activation of AKT and ERK1/2, contributing to tumour progression and therapy resistance. HER3-targeting agents that block this activation, are currently under phase 1/2 clinical studies, and although they have shown favorable tolerability, their activity as a single agent has proven to be limited. Here we show that phosphorylation and activation of HER3 in luminal breast cancer cells occurs in a paracrine manner and is mediated by NRG1 expressed by cancer-associated fibroblasts (CAFs). Moreover, we uncover a HER3-independent NRG1 signaling in CAFs that results in the induction of a strong migratory and pro-fibrotic phenotype, describing a subtype of CAFs with elevated expression of NRG1 and an associated transcriptomic profile that determines their functional properties. Finally, we identified Hyaluronan Synthase 2 (HAS2), a targetable molecule strongly correlated with NRG1, as an attractive player supporting NRG1 signaling in CAFs.
Collapse
Affiliation(s)
- Mireia Berdiel-Acer
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ana Maia
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Faculty of Biosciences, Ruprecht-Karls-University, Heidelberg, Germany
| | - Zhivka Hristova
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Faculty of Biosciences, Ruprecht-Karls-University, Heidelberg, Germany
| | - Simone Borgoni
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Faculty of Biosciences, Ruprecht-Karls-University, Heidelberg, Germany
| | - Martina Vetter
- grid.9018.00000 0001 0679 2801Department of Gynecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Sara Burmester
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Corinna Becki
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Birgitta Michels
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Khalid Abnaof
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ilona Binenbaum
- grid.7497.d0000 0004 0492 0584Division of Medical Informatics for Translational Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.11047.330000 0004 0576 5395Department of Biology, University of Patras, Patras, Greece ,grid.22459.380000 0001 2232 6894Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | - Daniel Bethmann
- grid.9018.00000 0001 0679 2801Institute of Pathology Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Aristotelis Chatziioannou
- grid.22459.380000 0001 2232 6894Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece ,e-NIOS PC, Kallithea-Athens, Greece
| | - Max Hasmann
- grid.424277.0Roche Diagnostics, Penzberg, Germany
| | - Christoph Thomssen
- grid.9018.00000 0001 0679 2801Department of Gynecology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Elisa Espinet
- grid.7497.d0000 0004 0492 0584Divison of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.482664.aHeidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Stefan Wiemann
- grid.7497.d0000 0004 0492 0584Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
20
|
Szymańska K, Makowska K, Całka J, Gonkowski S. The Endocrine Disruptor Bisphenol A (BPA) Affects the Enteric Neurons Immunoreactive to Neuregulin 1 (NRG1) in the Enteric Nervous System of the Porcine Large Intestine. Int J Mol Sci 2020; 21:E8743. [PMID: 33228092 PMCID: PMC7699376 DOI: 10.3390/ijms21228743] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/08/2020] [Accepted: 11/16/2020] [Indexed: 12/19/2022] Open
Abstract
The enteric nervous system (ENS), located in the wall of the gastrointestinal (GI) tract, is characterized by complex organization and a high degree of neurochemical diversity of neurons. One of the less known active neuronal substances found in the enteric neurons is neuregulin 1 (NRG1), a factor known to be involved in the assurance of normal development of the nervous system. During the study, made up using the double immunofluorescence technique, the presence of NRG1 in the ENS of the selected segment of porcine large intestine (caecum, ascending and descending colon) was observed in physiological conditions, as well as under the impact of low and high doses of bisphenol A (BPA) which is commonly used in the production of plastics. In control animals in all types of the enteric plexuses, the percentage of NRG1-positive neurons oscillated around 20% of all neurons. The administration of BPA caused an increase in the number of NRG1-positive neurons in all types of the enteric plexuses and in all segments of the large intestine studied. The most visible changes were noted in the inner submucous plexus of the ascending colon, where in animals treated with high doses of BPA, the percentage of NRG1-positive neurons amounted to above 45% of all neuronal cells. The mechanisms of observed changes are not entirely clear, but probably result from neurotoxic, neurodegenerative and/or proinflammatory activity of BPA and are protective and adaptive in nature.
Collapse
Affiliation(s)
- Kamila Szymańska
- Department of Human Physiology and Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury in Olsztyn, Warszawska Str. 30, 10-082 Olsztyn, Poland
| | - Krystyna Makowska
- Department of Clinical Diagnostics, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Str. 13, 10-719 Olsztyn, Poland;
| | - Jarosław Całka
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Str. 13, 10-719 Olsztyn, Poland; (J.C.); (S.G.)
| | - Sławomir Gonkowski
- Department of Clinical Physiology, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego Str. 13, 10-719 Olsztyn, Poland; (J.C.); (S.G.)
| |
Collapse
|
21
|
Wei X, Wang W, Wang H, Wang Y, Wang Y, Li G, Ji C, Ren X, Song N, Qin C. Identification of an independent autophagy-gene prognostic index for papillary renal cell carcinoma. Transl Androl Urol 2020; 9:1945-1956. [PMID: 33209659 PMCID: PMC7658136 DOI: 10.21037/tau-20-906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background Autophagy was a significant catabolic process which played a critical role in the maintenance of cellular homeostasis and viability in a stressed state. The dysregulation of autophagy was correlated with various diseases. The aim of our study was to develop a prognostic signature for papillary renal cell carcinoma (RCC). Methods First, 40 differently expressed genes related with autophagy (ARGs) were examined via high-throughput sequencing and large-scale databases. Then, functional enrichment analysis was performed to explore the biological attributes of these ARGs. The Cox proportional hazard regression hinted that four ARGs (P4HB, BIRC5, NGR1 and PRKN) were significantly correlated with overall survival (OS). Thus, we got genes with prognostic value. Finally, a prognostic index (PI) was constructed. Results After identifying the 4 ARGs, we profiled our risk signature. Based on the PI we developed, papillary RCC patients were stratified into high-risk and low-risk groups. High-risk patients had significant shorter OS than low-risk patients (P<0.001) and the mortality of high scoring patients was higher than low scoring patients. Additionally, we explored the relationship between the 4 ARGs and clinical parameters and found that the expression of P4HB, BIRC5 and NGR1 was correlated with clinicopathological features. Conclusions Our study suggested that the four-gene signature was an independent prognostic factor which could act as a novel indicator for the prognosis of papillary RCC.
Collapse
Affiliation(s)
- Xiyi Wei
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Wang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongye Wang
- First Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Yamin Wang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yichun Wang
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guangyao Li
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chengjian Ji
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaohan Ren
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ninghong Song
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,The Affiliated Kezhou People's Hospital of Nanjing Medical University, Kezhou, China
| | - Chao Qin
- Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
22
|
Li X, Huang Q, Wang S, Huang Z, Yu F, Lin J. HER4 promotes the growth and metastasis of osteosarcoma via the PI3K/AKT pathway. Acta Biochim Biophys Sin (Shanghai) 2020; 52:345-362. [PMID: 32181480 DOI: 10.1093/abbs/gmaa004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 01/05/2020] [Accepted: 01/20/2020] [Indexed: 11/13/2022] Open
Abstract
Osteosarcoma is the most common primary malignant bone tumor, which occurs in adolescents. As reported by our previous studies, HER4 indicates a poor prognosis of primary osteosarcoma. However, its mechanisms in the pathogenesis of osteosarcoma have not yet been studied. The purpose of this study was to investigate the role of HER4 in osteosarcoma and whether the PI3K/AKT pathway is involved. In this study, western blot analysis was used to investigate the expression of HER4 protein in osteosarcoma tissues and cell lines. CCK8 and transwell assays were used to detect the effects of HER4 on the proliferation, migration, and invasion of osteosarcoma cells in vitro. The effects of HER4 on the growth and metastasis of osteosarcoma in vivo were detected by tumor formation and immunofluorescence in nude mice. The role of the PI3K/AKT pathway in HER4 regulation of the growth and metastasis of osteosarcoma was examined by western blot analysis and immunofluorescence assay. We found that HER4 protein was highly expressed in clinical osteosarcoma specimens and osteosarcoma cells. HER4 markedly promoted the proliferation, migration, and invasion of osteosarcoma cells in vitro as well as the growth and metastasis of osteosarcoma in vivo. HER4 overexpression upregulated the expression of phosphorylated protein kinase B (pAKT), proliferation marker antigen Ki67, and metastasis cell marker matrix metalloproteinase 9 (MMP9). Notably, PI3K/AKT inhibitor LY294002 significantly inhibited the effects of HER4 via the downregulation of pAKT, Ki67, and MMP9. Moreover, LY294002 markedly blocked the effects of HER4-induced upregulation of tumor malignancy. The present study suggests that HER4 may promote the growth and metastasis of osteosarcoma via the PI3K/AKT pathway. The HER4/PI3K/AKT pathway could serve as a potential target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Qingshan Huang
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Shenglin Wang
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Zhen Huang
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Fengqiang Yu
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| | - Jianhua Lin
- Department of Orthopedics, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China
| |
Collapse
|
23
|
Park Y, Koh J, Na HY, Kwak Y, Lee KW, Ahn SH, Park DJ, Kim HH, Lee HS. PD-L1 Testing in Gastric Cancer by the Combined Positive Score of the 22C3 PharmDx and SP263 Assay with Clinically Relevant Cut-offs. Cancer Res Treat 2020; 52:661-670. [PMID: 32019283 PMCID: PMC7373862 DOI: 10.4143/crt.2019.718] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/09/2020] [Indexed: 01/06/2023] Open
Abstract
Purpose We provide a comparison between 22C3 pharmDx and SP263 assay, for evaluating programmed death ligand 1 (PD-L1) expression in advanced gastric cancer (GC) patients. Materials and Methods The PD-L1 immunohistochemistry by 22C3 pharmDx and SP263 assays was performed in the center of the tumor (CT) and invasive margin (IM) in 379 GC tissues using tissue microarrays and interpreted as combined positive score (CPS) and tumor proportion score (TPS). Of the total samples, 55 samples were independently reviewed by five pathologists. Results The two assays showed a high correlation in both the CPS and TPS. At a CPS ≥ 1 cut-off, 219 (57.8%) and 231 (60.9%) GCs were positive for PD-L1 with the 22C3 and SP263 assays, and at ≥ 10 cut-off, 37 (9.8%) and 36 (9.5%) GCs were positive, respectively. The overall percent agreement (OPA) was greater than 90% with CPS ≥ 1 and ≥ 10 cut-offs, and TPS ≥ 1% and ≥ 10% cut-offs. There was higher OPA between the two assays with a CPS cut-off ≥ 10 (99.2%) than ≥ 1 (94.7%). The percent agreement between the CT and IM was higher with a CPS cut-off ≥ 10 (92.9%) than ≥ 1 (77.6%). Patient with positive expression at CPS ≥ 5 cut-off had a significantly better outcomes in both assays. Interobserver variability among five pathologists was higher than the assay variability. Conclusion Two assays for PD-L1 expression in GC showed high agreement. These results provide guidance for selecting eligible patients with GC for pembrolizumab treatment.
Collapse
Affiliation(s)
- Yujun Park
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jiwon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Young Na
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yoonjin Kwak
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Sang-Hoon Ahn
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Do Joong Park
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hyung-Ho Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
24
|
Hamzehlou S, Momeny M, Zandi Z, Kashani B, Yousefi H, Dehpour AR, Tavakkoly-Bazzaz J, Ghaffari SH. Anti-tumor activity of neratinib, a pan-HER inhibitor, in gastric adenocarcinoma cells. Eur J Pharmacol 2019; 863:172705. [DOI: 10.1016/j.ejphar.2019.172705] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/24/2022]
|
25
|
Clinicopathologic significance of human leukocyte antigen class I expression in patients with stage II and III gastric cancer. Cancer Immunol Immunother 2019; 68:1779-1790. [PMID: 31620857 DOI: 10.1007/s00262-019-02410-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 10/05/2019] [Indexed: 12/12/2022]
Abstract
Human leukocyte antigen class I (HLA I) molecules composed of alpha (heavy) chain, including HLA-A, -B, or -C encoded by HLA genes, and beta-2-microglobulin (β2M) are membrane proteins on all nucleated cells that display peptide antigens for recognition by CD8-positive cytotoxic T cells. Here, we examined the clinicopathologic signification of HLA I expression in patients with gastric cancer (GC). Immunohistochemistry was performed to detect HLA A/B/C, β2M, CD8, p53, and programmed death-ligand 1 (PD-L1) in the center and invasive margin of the tumor in 395 stage II and III GCs using tissue array method. Additionally, Epstein-Barr virus (EBV) infection and microsatellite instability (MSI) status were investigated. Negative expression of HLA A/B/C and β2M was observed in 258 (65.3%) and 235 (59.5%) of 395 stage II and III GCs, respectively. Negative HLA I expression was significantly associated with aggressive clinicopathologic features. Furthermore, negative expression of HLA A/B/C and β2M was inversely correlated with CD8-positive cytotoxic T cell infiltration, EBV-positivity, and PD-L1 expression (all p < 0.001). Patients with HLA A/B/C-negative GC had worse overall survival (OS) (p = 0.019) and combined analysis with both HLA A/B/C and β2M expression status significantly predicted OS in univariate (p = 0.004) and multivariate survival analysis (p = 0.016). Negative expression of HLA A/B/C and β2M was frequently observed in stage II and III GCs, particularly with the aggressive clinicopathologic features, and correlated with an unfavorable prognosis and host immune response status. These findings contribute to further development of immunotherapy.
Collapse
|
26
|
Integrated genomic profiling expands clinical options for patients with cancer. Nat Biotechnol 2019; 37:1351-1360. [PMID: 31570899 DOI: 10.1038/s41587-019-0259-z] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 08/05/2019] [Indexed: 01/14/2023]
Abstract
Genomic analysis of paired tumor-normal samples and clinical data can be used to match patients to cancer therapies or clinical trials. We analyzed 500 patient samples across diverse tumor types using the Tempus xT platform by DNA-seq, RNA-seq and immunological biomarkers. The use of a tumor and germline dataset led to substantial improvements in mutation identification and a reduction in false-positive rates. RNA-seq enhanced gene fusion detection and cancer type classifications. With DNA-seq alone, 29.6% of patients matched to precision therapies supported by high levels of evidence or by well-powered studies. This proportion increased to 43.4% with the addition of RNA-seq and immunotherapy biomarker results. Combining these data with clinical criteria, 76.8% of patients were matched to at least one relevant clinical trial on the basis of biomarkers measured by the xT assay. These results indicate that extensive molecular profiling combined with clinical data identifies personalized therapies and clinical trials for a large proportion of patients with cancer and that paired tumor-normal plus transcriptome sequencing outperforms tumor-only DNA panel testing.
Collapse
|
27
|
Pan Y, Yang Z, Xu Y, Bai Z, Pan D, Yang R, Wang L, Guan W, Yang M. Targeting HER2-positive gastric cancer with a novel 18F-labeled Z HER2:342 probe. RSC Adv 2019; 9:10990-10998. [PMID: 35515328 PMCID: PMC9062611 DOI: 10.1039/c8ra10271f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/17/2019] [Indexed: 11/21/2022] Open
Abstract
To realize the diagnosis of HER2-positive gastric cancer via PET imaging, herein, a new kind of 18F-labeled HER2 affibody probe was created; the bifunctional maleimide derivative 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA-MAL) was first coupled to a polypeptide, and the resulting compound was subsequently labeled with the 18FAl complex. The binding characteristics of the probe were assessed using both in vitro studies and in vivo microPET imaging and biodistribution experiments. Immunohistochemical staining was performed to confirm the expression level of HER2 in the studied cell lines and tumors. The probe was successfully produced with the radiochemical purity of more than 95%. The NCI N87 cell-associated radioactivity was 19.31 ± 1.01% AD, and it decreased to 0.83 ± 0.04% AD per 106 cells after blocking HER2 as early as 15 minutes post-incubation (p < 0.05). A competition binding assay between radiolabeled and non-radioactive affibody molecules with NCI N87 indicated that the IC50 was 8.10 nM. The microPET imaging and biodistribution of human gastric cancer xenografts demonstrated that the probe could specifically accumulate in tumors at early time points. Protein detection confirmed a strong HER2 expression in NCIN87 and a weak HER2 expression in SGC7901. In conclusion, 18FAl-NOTA-MAL-Cys-GGGRDN(M0)-ZHER2:342 was successfully prepared via a one-step method. The favorable preclinical data showed specific and effective tumor targeting capacity of the proposed probe; this revealed that the probe proposed herein might have potential application in gastric cancer imaging.
Collapse
Affiliation(s)
- Yunyun Pan
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing China 210008
| | - Zhengyang Yang
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing China 210008
| | - Yuping Xu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine Wuxi Jiangsu China 214063
| | - Zhicheng Bai
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine Wuxi Jiangsu China 214063
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine Wuxi Jiangsu China 214063
| | - Runlin Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine Wuxi Jiangsu China 214063
| | - Lizhen Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine Wuxi Jiangsu China 214063
| | - Wenxian Guan
- Department of General Surgery, Drum Tower Hospital, Medical School of Nanjing University 321 Zhongshan RD Nanjing China 210008
| | - Min Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine Wuxi Jiangsu China 214063
| |
Collapse
|
28
|
Khelwatty SA, Essapen S, Bagwan I, Green M, Seddon AM, Modjtahedi H. Co-expression and prognostic significance of putative CSC markers CD44, CD133, wild-type EGFR and EGFRvIII in metastatic colorectal cancer. Oncotarget 2019; 10:1704-1715. [PMID: 30899442 PMCID: PMC6422200 DOI: 10.18632/oncotarget.26722] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/15/2019] [Indexed: 12/24/2022] Open
Abstract
The presence of colorectal cancer stem cells (CSCs) have been associated with tumour initiation and resistance to therapy. This study investigated the co-expression and prognostic significance of the CSCs biomarkers CD44 and CD133 with wild-type EGFR (wtEGFR) and EGFRvIII in colorectal cancer (CRC). The expression of these biomarkers were determined in tumours from 70 patients with metastatic CRC by immunohistochemistry, and in a panel of human CRC cell lines, and their variants with acquired-resistance to EGFR inhibitors, by flow cytometry. The expression of CD44, CD133, wtEGFR and EGFRvIII were present in 17%, 23%, 26% and 13% of cases and the co-expression of CD44/CD133 with wtEGFR and EGFRvIII were present in 9% and 3% of the cases respectively. Only co-expression of CSCs/EGFRvIII (P = 0.037), and amphiregulin (P = 0.017) were associated with worse overall survival. Interestingly, disease-free survival was improved in BTC expressing patients (P = 0.025). In vitro CD133 expression and its co-expression with CD44 were associated with primary-resistance to irinotecan and acquired-resistance to anti-EGFR inhibitors respectively. Our results suggest co-expression of CSCs and EGFRvIII could be potential biomarkers of worse overall survival and resistance to therapy in patients with mCRC and warrants further validation in a larger cohort.
Collapse
Affiliation(s)
| | - Sharadah Essapen
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston, UK.,St. Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, Surrey, UK
| | - Izhar Bagwan
- Department of Histopathology, Royal Surrey County Hospital, Guildford, Surrey, UK
| | - Margaret Green
- Department of Histopathology, Royal Surrey County Hospital, Guildford, Surrey, UK
| | - Alan M Seddon
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston, UK
| | - Helmout Modjtahedi
- School of Life Sciences, Pharmacy and Chemistry, Kingston University London, Kingston, UK
| |
Collapse
|
29
|
Li X, Chen B, Chi D, Zhang Y, Jiang W. lncRNA CASC9 regulates cell migration and invasion in hemangioma endothelial cells by targeting miR-125a-3p/Nrg1. Onco Targets Ther 2019; 12:423-432. [PMID: 30662268 PMCID: PMC6327889 DOI: 10.2147/ott.s181914] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background Despite being one of the most common benign tumors, the prevalence and pathogenesis of hemangiomas (HAs) are poorly understood. We aimed to identify the biological role of the long non-coding RNA (lncRNA) CASC9 in the HA-derived endothelial cell (HDECs) phenotype as well as elucidate the mechanism involved. Methods The expression of CASC9 was identified by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). the effect of CASC9 on cell proliferation, migration and invasion of HDECs were examined by CCK8, wound healing, and transwell assay, respectively. Bioinformatics analysis and a luciferase reporter assay were utilized to investigated the mechanisms involved. The in vivo tumorigenesis capability of CASC9 on HA was also evaluated. Results The expression of CASC9 was significantly elevated in HA tissue compared to normal tissue. Down-regulation of CASC9 inhibited proliferation, migration, and invasion of HDECs. The translation of cyclinD1, N-cadherin, Twist, and MMP2 was also decreased by CASC9 knockdown treatment. Furthermore, CASC9 over-expression exerted the opposite effect of proliferation, migration, and invasion of HDECs. We also found that CASC9 interacts with miR-125a-3p/Nrg1 to regulate cellular functions. Interestingly, miR-125a-3p can reverse the effect of CASC9 on proliferation, migration, and invasion of HDECs. Together, the clinical data showed that CASC9 expression is negatively correlated with miR-125a-3p expression and positively correlated with Nrg1 expression. CASC9 also exerted anti-tumorigenesis capability in vivo. Conclusion Our study indicates that CASC9 accelerates cell growth and invasion of HDECs and provides new insights for the diagnosis and molecular therapy of HA.
Collapse
Affiliation(s)
- Xianwei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, People's Republic of China,
| | - Bo Chen
- Department of Vascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, People's Republic of China,
| | - Decai Chi
- Department of Vascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, People's Republic of China,
| | - Yingnan Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, People's Republic of China,
| | - Weiliang Jiang
- Department of Vascular Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, People's Republic of China,
| |
Collapse
|
30
|
Yun S, Kwak Y, Nam SK, Seo AN, Oh HK, Kim DW, Kang SB, Lee HS. Ligand-Independent Epidermal Growth Factor Receptor Overexpression Correlates with Poor Prognosis in Colorectal Cancer. Cancer Res Treat 2018; 50:1351-1361. [PMID: 29361822 PMCID: PMC6192927 DOI: 10.4143/crt.2017.487] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 01/15/2018] [Indexed: 12/14/2022] Open
Abstract
Purpose Molecular treatments targeting epidermal growth factor receptors (EGFRs) are important strategies for advanced colorectal cancer (CRC). However, clinicopathologic implications of EGFRs and EGFR ligand signaling have not been fully evaluated. We evaluated the expression of EGFR ligands and correlation with their receptors, clinicopathologic factors, and patients’ survival with CRC. Materials and Methods The expression of EGFR ligands, including heparin binding epidermal growth factor-like growth factor (HBEGF), transforming growth factor (TGF), betacellulin, and epidermal growth factor (EGF), were evaluated in 331 consecutive CRC samples using mRNA in situ hybridization (ISH). We also evaluated the expression status of EGFR, human epidermal growth factor receptor 2 (HER2), HER3, and HER4 using immunohistochemistry and/or silver ISH. Results Unlike low incidences of TGF (38.1%), betacellulin (7.9%), and EGF (2.1%), HBEGF expression was noted in 62.2% of CRC samples. However, the expression of each EGFR ligand did not reveal significant correlations with survival. The combined analyses of EGFR ligands and EGFR expression indicated that the ligands‒/EGFR+ group showed a significant association with the worst disease-free survival (DFS; p=0.018) and overall survival (OS; p=0.005). It was also an independent, unfavorable prognostic factor for DFS (p=0.026) and OS (p=0.007). Additionally, HER4 nuclear expression, regardless of ligand expression, was an independent, favorable prognostic factor for DFS (p=0.034) and OS (p=0.049), by multivariate analysis. Conclusion Ligand-independent EGFR overexpression was suggested to have a significant prognostic impact; thus, the expression status of EGFR ligands, in addition to EGFR, might be necessary for predicting patients' outcome in CRC.
Collapse
Affiliation(s)
- Sumi Yun
- Department of Diagnostic Pathology, Samkwang Medical Laboratories, Seoul, Korea.,Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Yoonjin Kwak
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Soo Kyung Nam
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - An Na Seo
- Department of Pathology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Heung-Kwon Oh
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Duck-Woo Kim
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sung-Bum Kang
- Department of Surgery, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea.,Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|