1
|
QUAN JINGDAN, WAN ZIXIN, WU WEI, CAO XINYUAN, QIU JIAYUAN, LIU XIAOYE, ZHANG ZHIWEI. Classical biomarkers and non-coding RNAs associated with diagnosis and treatment in gastric cancer. Oncol Res 2025; 33:1069-1089. [PMID: 40296904 PMCID: PMC12034007 DOI: 10.32604/or.2025.063005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/17/2025] [Indexed: 04/30/2025] Open
Abstract
One of the most prevalent malignant tumors worldwide, stomach cancer still has a high incidence and fatality rate in China, and the number of young people developing early-onset gastric cancer is steadily increasing. The 5-year survival rate of stomach cancer is typically 30%-35%, the prognosis is bad, the patients' quality of life is low, and the progression of advanced gastric cancer cannot be effectively managed despite the use of surgical surgery, chemotherapy, and other medicines. We urgently need molecular biomarkers with high specificity and sensitivity to increase the early gastric cancer detection rate, extend patient survival, and improve patient quality of life. The initial diagnosis of gastric cancer primarily depends on gastroscopy and biopsy, and invasive procedures cause significant discomfort to patients. Similar to this, treating advanced and metastatic stomach cancer is a pressing issue that requires attention. More and more immune checkpoint molecules have been discovered, and corresponding inhibitors are gradually being applied to clinical diagnosis and treatment. Recently, some non-coding RNAs have begun to be used as new targets for the treatment of gastric cancer. Some non-coding RNAs are highly present in the serum or urine of gastric cancer patients and can be used as diagnostic markers or prognostic indicators. Many clinical trials targeting non-coding RNAs have also shown good therapeutic effects. In general, targeting non-coding RNAs has shown good therapeutic effects. The biomarkers for gastric cancer detection and treatment are reviewed in this article, focusing on the new non-coding RNAs used in diagnosis, prognosis, and treatment. Patients with stomach cancer should have access to more precise and efficient diagnosis and treatment choices as a result of ongoing technological advancements and thorough research.
Collapse
Affiliation(s)
- JINGDAN QUAN
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - ZIXIN WAN
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - WEI WU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - XINYUAN CAO
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - JIAYUAN QIU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - XIAOYE LIU
- Hengyang Medical College, University of South China, Hengyang, 421001, China
| | - ZHIWEI ZHANG
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, 421001, China
| |
Collapse
|
2
|
Kim YA, Sung WJ. The immunohistochemical expression of c-MET and RON in lung adenocarcinoma with clinicopathologic correlation. INDIAN J PATHOL MICR 2025; 68:23-29. [PMID: 38847203 DOI: 10.4103/ijpm.ijpm_358_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 03/12/2024] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Mesenchymal epidermal transition (MET) and receptor originating from nantes (RON) are transmembrane tyrosine kinase receptors. Both are members of a proto-oncogene family and thus play a role in the pathogenesis of various cancers and acquired resistance to kinase inhibitors in lung cancer. AIMS The aim of this study was to investigate the immunohistochemical expression of c-MET and RON in lung adenocarcinoma and its clinicopathologic correlation. SETTINGS AND DESIGN Retrospective study. MATERIALS AND METHODS The immunohistochemical c-MET and RON expression in specimens obtained from lung adenocarcinoma ( n = 175) and associated clinicopathologic parameters were evaluated. STATISTICAL ANALYSIS USED The correlation between c-MET and RON expression was analyzed by Chi-square test. A Cox proportional hazards model and Kaplan-Meier curve analysis were used to evaluate the risk factors and prognosis. RESULTS High expression of the c-MET protein showed a strong correlation with that of RON ( P = 0.013, kappa = 0.183). Five-year survival and recurrence-free 5-year survival were not associated with high expression of c-MET or RON. High c-MET expression was significantly associated with age older than 60 years ( P = 0.000), tumor differentiation ( P = 0.009), lymphovascular invasion ( P = 0.016), and pleural invasion ( P = 0.005). High RON expression was associated with a solid growth pattern ( P = 0.001) and pleural invasion ( P = 0.002). CONCLUSIONS The results point to the potential of immunohistochemical expression of c-MET and RON as useful prognostic markers of unfavorable histopathologic features in lung adenocarcinoma.
Collapse
Affiliation(s)
- Young-Ah Kim
- Department of Pathology, Daegu Catholic University School of Medicine, Duryugongwon-ro 17-gil 33, Nam-gu, Daegu 42472, Republic of Korea
- Seoul Clinical Laboratories of Daegu, 95, Hwarang-ro, Dong-gu, Daegu, 41238, Republic of Korea
| | - Woo Jung Sung
- Department of Pathology, Daegu Catholic University School of Medicine, Duryugongwon-ro 17-gil 33, Nam-gu, Daegu 42472, Republic of Korea
| |
Collapse
|
3
|
Yu S, Chen C, Chen M, Liang J, Jiang K, Lou B, Lu J, Zhu X, Zhou D. MAGOH promotes gastric cancer progression via hnRNPA1 expression inhibition-mediated RONΔ160/PI3K/AKT signaling pathway activation. J Exp Clin Cancer Res 2024; 43:32. [PMID: 38268030 PMCID: PMC10809607 DOI: 10.1186/s13046-024-02946-8] [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: 10/05/2023] [Accepted: 01/05/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is associated with high mortality and heterogeneity and poses a great threat to humans. Gene therapies for the receptor tyrosine kinase RON and its spliceosomes are attracting increasing amounts of attention due to their unique characteristics. However, little is known about the mechanism involved in the formation of the RON mRNA alternative spliceosome RONΔ160. METHODS Fourteen human GC tissue samples and six normal gastric tissue samples were subjected to label-free relative quantitative proteomics analysis, and MAGOH was identified as a candidate protein for subsequent studies. The expression of MAGOH in clinical specimens was verified by quantitative real-time PCR and western blotting. We then determined the biological function of MAGOH in GC through in vitro and in vivo experiments. RNA pulldown, RNA sequencing and RNA immunoprecipitation (RIP) were subsequently conducted to uncover the underlying mechanism by which MAGOH regulated the formation of RONΔ160. RESULTS Proteomic analysis revealed that MAGOH, which is located at key nodes and participates in RNA processing and mRNA splicing, was upregulated in GC tissue and GC cell lines and was associated with poor prognosis. Functional analysis showed that MAGOH promoted the proliferation, migration and invasion of GC cells in vitro and in vivo. Mechanistically, MAGOH inhibited the expression of hnRNPA1 and reduced the binding of hnRNPA1 to RON mRNA, thereby promoting the formation of RONΔ160 to activate the PI3K/AKT signaling pathway and consequently facilitating GC progression. CONCLUSIONS Our study revealed that MAGOH could promote the formation of RONΔ160 and activate the PI3K/AKT signaling pathway through the inhibition of hnRNPA1 expression. We elucidate a novel mechanism and potential therapeutic targets for the growth and metastasis of GC based on the MAGOH-RONΔ160 axis, and these findings have important guiding significance and clinical value for the future development of effective therapeutic strategies for GC.
Collapse
Affiliation(s)
- Shanshan Yu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cheng Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ming Chen
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinxiao Liang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kecheng Jiang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bin Lou
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Lu
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohua Zhu
- Department of Oncology, Shaoxing People's Hospital, Shaoxing, China
| | - Donghui Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Diniz F, Lamas S, Osório H, Aguiar P, Freitas D, Gärtner F, Sarmento B, Reis CA, Gomes J. Nanoparticles targeting Sialyl-Tn for efficient tyrosine kinase inhibitor delivery in gastric cancer. Acta Biomater 2023; 170:142-154. [PMID: 37586448 DOI: 10.1016/j.actbio.2023.08.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 08/18/2023]
Abstract
Gastric cancer (GC) is the fourth leading cause of cancer-related deaths worldwide and, therefore, it is urgent to develop new and more efficient therapeutic approaches. Foretinib (FRT) is an oral multikinase inhibitor targeting MET (hepatocyte growth factor receptor) and RON (recepteur d'origine nantais) receptor tyrosine kinases (RTKs) that has been used in clinical trials for several solid tumors. Targeted uptake of therapeutic polymeric nanoparticles (NPs) represents a powerful approach in cancer cell drug delivery. Previously, a nanodelivery system composed of polymeric NPs functionalized with B72.3 antibody, which targets the tumor-associated antigen Sialyl-Tn (STn), has been developed. Herein, these NPs were loaded with FRT to evaluate its capacity in delivering the drug to multicellular tumors spheroids (MCTS) and mouse models. The data indicated that B72.3 functionalized FRT-loaded PLGA-PEG-COOH NPs (NFB72.3) specifically target gastric MCTS expressing the STn glycan (MKN45 SimpleCell (SC) cells), leading to a decrease in phospho-RTKs activation and reduced cell viability. In vivo evaluation using MKN45 SC xenograft mice revealed that NFB72.3 were able to decrease tumor growth, reduce cell proliferation and tumor necrosis. NFB72.3-treated tumors also showed inactivation of phospho-MET and phospho-RON. This study demonstrates the value of using NPs targeting STn for FRT delivery, highlighting its potential as a therapeutic application in GC. STATEMENT OF SIGNIFICANCE: Despite the advances in gastric cancer therapeutics, it remains one of the diseases with the highest incidence and mortality in the world. Combining targeted therapies with a controlled drug release is an attractive strategy to reduce drug cytotoxic effects and improve specific drug delivery efficiency to the cancer cells. Thus, we developed nanoparticles loaded with a tyrosine kinase inhibitor and targeting a specific tumor glycan exclusive of cancer cells. In in vivo gastric cancer xenograft mice models, these nanoparticles efficiently reduced tumor growth, cell proliferation and tumor necrosis area and inactivated phosphorylation of targeting receptors. This approach represents an innovative therapeutic strategy with high impact in gastric cancer.
Collapse
Affiliation(s)
- Francisca Diniz
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Sofia Lamas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, 4200-135, Portugal
| | - Hugo Osório
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Paulo Aguiar
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Daniela Freitas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Fátima Gärtner
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal; CESPU-IUCS, 4585-116 Gandra, Portugal
| | - Celso A Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal; FMUP - Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal.
| | - Joana Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal.
| |
Collapse
|
5
|
Epithelial-Mesenchymal Transition Gene Signature Is Associated with Neoadjuvant Chemoradiotherapy Resistance and Prognosis of Esophageal Squamous Cell Carcinoma. DISEASE MARKERS 2022; 2022:3534433. [PMID: 36072903 PMCID: PMC9442501 DOI: 10.1155/2022/3534433] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/02/2022] [Indexed: 11/28/2022]
Abstract
Background Neoadjuvant chemoradiotherapy (neo-CRT) in combination with surgery increases survival compared to surgery alone, as indicated by the esophageal squamous cell carcinoma (ESCC) treatment recommendations. However, the benefits of neo-CRT are diverse among patients. Consequently, the development of new biomarkers that correlate with neo-CRT might be important for the treatment of ESCC. Methods The differentially expressed genes (DEG) between responsive and resistant samples from the GSE45670 dataset were obtained. On the TCGA dataset, survival analysis was performed to identify prognosis-related-EMT-genes. For EMT score model construction, lasso regression analysis in the TCGA cohort was used to identify the genes. In the TCGA-ESCC cohort, age, stage, and EMT score were used to construct a nomogram. Results In total, 10 prognosis-related-EMT-genes were obtained. These 10 genes consisted of 6 risky genes and 4 protective genes. Based on the lasso analysis and univariate Cox regression, an EMT score model consisting of 7 genes (CLEC18A, PIR, KCNN4, MST1R, CAPG, ALDH5A1, and COX7B) was identified. ESCC patients with a high EMT score have a worse prognosis. These genes were differentially expressed between responsive and resistant patients and had a high accuracy for distinguishing resistant and responsive patients. Conclusions The identified genes have the potential to function as molecular biomarkers for predicting ESCC patients' resistance to neo-CRT. This research may aid in the elucidation of the molecular processes driving resistance and the identification of targets for improving the prognosis for ESCC.
Collapse
|
6
|
Xie XP, Yang W, Zhang L, Wang HQ. Identifying Biomarkers of Cisplatin Sensitivity in Non-Small Cell Lung
Cancer via Comprehensive Integrative Analysis. Curr Bioinform 2022; 17:498-509. [DOI: 10.2174/1574893617666220407105905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 11/22/2022]
Abstract
Background:
Only 30-40% of non-small cell lung cancer (NSCLC) patients are clinically
sensitive to cisplatin-based chemotherapy. Thus, it is necessary to identify biomarkers for personalized
cisplatin chemotherapy in NSCLC. However, data heterogeneity and low-value density make it challenging
to detect reliable cisplatin efficacy biomarkers using traditional analysis methods.
Objective:
This paper aims to find reliable cisplatin efficacy biomarkers for NSCLC patients using
comprehensive integrative analysis.
Method:
We searched online resources and collected six NSCLC transcriptomics data sets with responses
to cisplatin. The six data sets are divided into two groups: the learning group for biomarker
identification and the test group for independent validation. We performed comprehensive integrative
analysis under two kinds of frameworks, i.e., one-level and two-level, with three integrative models.
Pathway analysis was performed to estimate the biological significance of the resulting biomarkers. For
independent validation, logrank statistic was employed to test how significant the difference of Kaplan-
Meier (KM) curves between two patient groups is, and the Cox proportional-hazards model was used to
test how the expression of a gene is associated with patients’ survival time. Especially, a permutation
test was performed to verify the predictive power of a biomarker panel on cisplatin efficacy. For comparison,
we also analyzed each learning data set individually, in which three popular differential expression
models, Limma, SAM, and RankSum, were used.
Results:
A total of 318 genes were identified as a core panel of cisplatin efficacy markers for NSCLC
patients, exhibiting consistent differential expression between cisplatin-sensitive and –resistant groups
across studies. A total of 129 of 344 KEGG pathways were found to be enriched in the core panel, reflecting
a picture of the molecular mechanism of cisplatin resistance in NSCLC. By mapping onto the
KEGG pathway tree, we found that a KEGG pathway-level I module, genetic information processing, is
most active in the core panel with the highest activity ratio in response to cisplatin in NSCLC as expected.
Related pathways include mismatch repair, nucleotide excision repair, aminoacyl-tRNA biosynthesis,
and basal transcription factors, most of which respond to DNA double-strand damage in patients.
Evaluation on two independent data sets demonstrated the predictive power of the core marker panel for
cisplatin sensitivity in NSCLC. Also, some single markers, e.g., MST1R, were observed to be remarkably
predictive of cisplatin resistance in NSCLC.
Conclusion:
Integrative analysis is more powerful in detecting biomarkers for cisplatin efficacy by
overcoming data heterogeneity and low-value density in data sets, and the identified core panel (318
genes) can help develop personalized medicine of cisplatin chemotherapy for NSCLC patients.
Collapse
Affiliation(s)
- Xin-Ping Xie
- School of Mathematics and Physics, Anhui Jianzhu University, Hefei, China
| | - Wulin Yang
- Cancer hospital, Hefei Institutes of Physical
Science, Chinese Academy of Science, Hefei, China
| | - Lei Zhang
- Department of Pharmacy, The First Affiliated Hospital of
USTC, Division of Life Sciences and Medicine, University of Science & Technology of China, Hefei, China
| | - Hong-Qiang Wang
- Cancer hospital, Hefei Institutes of Physical
Science, Chinese Academy of Science, Hefei, China
| |
Collapse
|
7
|
Chen SL, Wang GP, Shi DR, Yao SH, Chen KD, Yao HP. RON in hepatobiliary and pancreatic cancers: Pathogenesis and potential therapeutic targets. World J Gastroenterol 2021; 27:2507-2520. [PMID: 34092972 PMCID: PMC8160627 DOI: 10.3748/wjg.v27.i20.2507] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/04/2021] [Accepted: 04/09/2021] [Indexed: 02/06/2023] Open
Abstract
The receptor protein tyrosine kinase RON belongs to the c-MET proto-oncogene family. Research has shown that RON has a role in cancer pathogenesis, which places RON on the frontline of the development of novel cancer therapeutic strategies. Hepatobiliary and pancreatic (HBP) cancers have a poor prognosis, being reported as having higher rates of cancer-related death. Therefore, to combat these malignant diseases, the mechanism underlying the aberrant expression and signaling of RON in HBP cancer pathogenesis, and the development of RON as a drug target for therapeutic intervention should be investigated. Abnormal RON expression and signaling have been identified in HBP cancers, and also act as tumorigenic determinants for HBP cancer malignant behaviors. In addition, RON is emerging as an important mediator of the clinical prognosis of HBP cancers. Thus, not only is RON significant in HBP cancers, but also RON-targeted therapeutics could be developed to treat these cancers, for example, therapeutic monoclonal antibodies and small-molecule inhibitors. Among them, antibody-drug conjugates have become increasingly popular in current research and their potential as novel anti-cancer biotherapeutics will be determined in future clinical trials.
Collapse
Affiliation(s)
- Shao-Long Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310000, Zhejiang Province, China
| | - Guo-Ping Wang
- Department of Surgical Oncology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, Zhejiang Province, China
| | - Dan-Rong Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Shu-Hao Yao
- Department of Stomatology, Wenzhou Medical University Renji College, Wenzhou 325035, Zhejiang Province, China
| | - Ke-Da Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310000, Zhejiang Province, China
| | - Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
8
|
Grojean M, Schwarz MA, Schwarz JR, Hassan S, von Holzen U, Zhang C, Schwarz RE, Awasthi N. Targeted dual inhibition of c-Met/VEGFR2 signalling by foretinib improves antitumour effects of nanoparticle paclitaxel in gastric cancer models. J Cell Mol Med 2021; 25:4950-4961. [PMID: 33939252 PMCID: PMC8178268 DOI: 10.1111/jcmm.16362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/16/2020] [Accepted: 01/19/2021] [Indexed: 12/24/2022] Open
Abstract
Elevated expression of multiple growth factors and receptors including c‐Met and VEGFR has been reported in gastric adenocarcinoma (GAC) and thus provides a potentially useful therapeutic target. The therapeutic efficacy of foretinib, a c‐Met/VEGFR2 inhibitor, was determined in combination with nanoparticle paclitaxel (NPT) in GAC. Animal studies were conducted in NOD/SCID mice in subcutaneous and peritoneal dissemination xenografts. The mechanism of action was assessed by Immunohistochemical and Immunoblot analyses. In c‐Met overexpressing MKN‐45 cell‐derived xenografts, NPT and foretinib demonstrated inhibition in tumour growth, while NPT plus foretinib showed additive effects. In c‐Met low‐expressing SNU‐1 or patient‐derived xenografts, the foretinib effect was smaller, while NPT had a similar effect compared with MKN‐45, as NPT plus foretinib still exhibited an additive response. Median mice survival was markedly improved by NPT (83%), foretinib (100%) and NPT plus foretinib (230%) in peritoneal dissemination xenografts. Subcutaneous tumour analyses exhibited that foretinib increased cancer cell death and decreased cancer cell proliferation and tumour vasculature. NPT and foretinib suppressed the proliferation of GAC cells in vitro and had additive effects in combination. Further, foretinib caused a dramatic decrease in phosphorylated forms of c‐Met, ERK, AKT and p38. Foretinib led to a decrease in Bcl‐2, and an increase in p27, Bax, Bim, cleaved PARP‐1 and cleaved caspase‐3. Thus, these findings highlight the antitumour impact of simultaneous suppression of c‐Met and VEGFR2 signalling in GAC and its potential to enhance nanoparticle paclitaxel response. This therapeutic approach might lead to a clinically beneficial combination to increase GAC patients’ survival.
Collapse
Affiliation(s)
- Meghan Grojean
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA
| | - Margaret A Schwarz
- Department of Pediatrics, Indiana University School of Medicine, South Bend, IN, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Johann R Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA
| | - Sazzad Hassan
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Urs von Holzen
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Goshen Center for Cancer Care, Goshen, IN, USA.,University of Basel, Basel, Switzerland
| | - Changhua Zhang
- Department of Gastrointestinal Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Roderich E Schwarz
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA.,Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,University of Buffalo, Buffalo, NY, USA
| | - Niranjan Awasthi
- Department of Surgery, Indiana University School of Medicine, South Bend, IN, USA.,Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| |
Collapse
|
9
|
Kim S, Ahn JM, Bae WJ, Han JH, Lee D. Quantitation of ligand is critical for ligand-dependent MET signalling activation and determines MET-targeted therapeutic response in gastric cancer. Gastric Cancer 2021; 24:577-588. [PMID: 33164142 DOI: 10.1007/s10120-020-01139-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 10/26/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Despite the promising preclinical antitumor activity of MET-targeting therapies, most clinical trials have failed. We introduced a new concept of quantitation of stroma-induced hepatocyte growth factor (HGF) to assess the actual MET signalling activity in gastric cancer (GC). METHODS We treated serially diluted HGF and conditioned media (CM) from cancer-associated fibroblasts (CAFs) on low MET-expressing cancer cells and investigated the phenotypical and signalling changes. Stromal proportion and MET expression in GC samples were assessed, and gene set enrichment analysis (GSEA) from the public database was performed. The antitumor effect of anti-MET treatment was examined, especially when cancer cells were activated in a ligand-dependent manner. RESULTS Relatively high doses of HGF or high-concentrated CM fully activated MET signalling cascades and promoted cell proliferation/invasion. High stromal proportion denoted worse patient survival in MET-positive GCs than in MET-negative ones. GSEA showed that the gene sets regarding proliferation, migration, and CAF as well as MET pathway signature were enriched in simultaneously MET- and HGF-positive samples. Sufficient ligand-dependent MET signalling activation increased the sensitivity to crizotinib. CONCLUSIONS We conclude that patients whose tumours have a high stromal proportion and at least low MET expression may benefit more from MET-targeted therapies.
Collapse
Affiliation(s)
- Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea
| | - Ji Mi Ahn
- Department of Pathology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea
| | - Won Jung Bae
- Department of Pathology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea
| | - Jae Ho Han
- Department of Pathology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea
| | - Dakeun Lee
- Department of Pathology, Ajou University School of Medicine, 164, Worldcup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, Republic of Korea.
| |
Collapse
|
10
|
Yang Y, Wang C, Dai C, Liu X, Li W, Huang M, Zhao X, Ji D, Li J, Guo W. Amplification and expression of c-MET correlate with poor prognosis of patients with gastric cancer and upregulate the expression of PDL1. Acta Biochim Biophys Sin (Shanghai) 2021; 53:547-557. [PMID: 33693450 DOI: 10.1093/abbs/gmab026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 12/22/2022] Open
Abstract
The prognostic significance of c-MET in gastric cancer (GC) remains uncertain. In the present study, we examined the amplification, expression, and the prognostic value of c-MET, human epidermal growth factor receptor 2 (HER2), and programmed cell death 1 ligand 1 (PDL1), together with the correlations among them in a large cohort of Chinese samples. A total of 444 patients were included. The immunohistochemistry (IHC) and the dual-color silver in situ hybridization (SISH) were performed to examine their expression and amplification. Univariate and multivariate analyses were performed by the Cox proportional hazard regression model, and survival curves were estimated by the Kaplan-Meier method. The positivity determined by IHC of c-MET was 24.8%, and the MET amplification rate was 2.3%. The positivity rates of HER2 and PDL1 were 8% and 34.7%, respectively. PDL1 expression had a significantly positive association with c-MET expression. c-MET positivity played a significant prognostic role in disease-free survival (DFS) (P = 0.032). Patients with mesenchymal-epithelial transition (MET) amplification had significantly poorer prognosis on both DFS and overall survival (OS). Subgroup analysis showed that in HER2-negative patients, but not in HER2-positive patients, MET-positive patients had significantly worse DFS (P = 0.000) and OS (P = 0.006). c-MET regulated the expression of PDL1 through an AKT-dependent pathway. c-MET inhibitor enhanced the T-cell killing ability and increased the efficacy of PD1 antibody. c-MET was found to be an independent prognostic factor for DFS of GC patients. A combination of c-MET inhibitors and PD1 antibodies could enhance the killing capacity of T cells, providing a preliminary basis for the clinical research on the same combination in GC treatment.
Collapse
Affiliation(s)
- Ya’nan Yang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chenchen Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Congqi Dai
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Xinyang Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Wenhua Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Mingzhu Huang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoying Zhao
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Dongmei Ji
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Medical Oncology, Tongji University Shanghai East Hospital, Shanghai 200120, China
| | - Weijian Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
11
|
Tabibzadeh A, Tameshkel FS, Moradi Y, Soltani S, Moradi-Lakeh M, Ashrafi GH, Motamed N, Zamani F, Motevalian SA, Panahi M, Esghaei M, Ajdarkosh H, Mousavi-Jarrahi A, Niya MHK. Signal transduction pathway mutations in gastrointestinal (GI) cancers: a systematic review and meta-analysis. Sci Rep 2020; 10:18713. [PMID: 33127962 PMCID: PMC7599243 DOI: 10.1038/s41598-020-73770-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
The present study was conducted to evaluate the prevalence of the signaling pathways mutation rate in the Gastrointestinal (GI) tract cancers in a systematic review and meta-analysis study. The study was performed based on the PRISMA criteria. Random models by confidence interval (CI: 95%) were used to calculate the pooled estimate of prevalence via Metaprop command. The pooled prevalence indices of signal transduction pathway mutations in gastric cancer, liver cancer, colorectal cancer, and pancreatic cancer were 5% (95% CI: 3-8%), 12% (95% CI: 8-18%), 17% (95% CI: 14-20%), and 20% (95% CI: 5-41%), respectively. Also, the mutation rates for Wnt pathway and MAPK pathway were calculated to be 23% (95% CI, 14-33%) and 20% (95% CI, 17-24%), respectively. Moreover, the most popular genes were APC (in Wnt pathway), KRAS (in MAPK pathway) and PIK3CA (in PI3K pathway) in the colorectal cancer, pancreatic cancer, and gastric cancer while they were beta-catenin and CTNNB1 in liver cancer. The most altered pathway was Wnt pathway followed by the MAPK pathway. In addition, pancreatic cancer was found to be higher under the pressure of mutation compared with others based on pooled prevalence analysis. Finally, APC mutations in colorectal cancer, KRAS in gastric cancer, and pancreatic cancer were mostly associated gene alterations.
Collapse
Affiliation(s)
- Alireza Tabibzadeh
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Safarnezhad Tameshkel
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Yousef Moradi
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Saber Soltani
- Department of Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Maziar Moradi-Lakeh
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
- Preventive Medicine and Public Health Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - G Hossein Ashrafi
- Cancer Theme SEC Faculty, Kingston University, Penrhyn Road, London, KT1 2EE, UK
| | - Nima Motamed
- Department of Social Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farhad Zamani
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Abbas Motevalian
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mahshid Panahi
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Esghaei
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Ajdarkosh
- Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | | |
Collapse
|
12
|
A combined FAK, c-MET, and MST1R three-protein panel risk-stratifies colorectal cancer patients. Transl Oncol 2020; 13:100836. [PMID: 32739842 PMCID: PMC7399195 DOI: 10.1016/j.tranon.2020.100836] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022] Open
Abstract
Focal adhesion kinase (FAK) is a key tyrosine kinase downstream of c-MET (or hepatocyte growth factor receptor, HGFR) and MST1R (macrophage-stimulating protein receptor or recepteur d'origine Nantais, RON) membrane receptors. The pathway plays an important role in cancer survival and invasion. In this study, we examined the protein expression of FAK, c-MET, and MST1R levels in a well-annotated cohort of 330 colorectal cancer patients. We found FAK to be overexpressed in colorectal adenocarcinomas (p = 0.0002), and FAK levels correlated positively with phospho-FAK levels (R2 = 0.81). In comparison, MST1R levels were not significantly different, and c-MET levels were slightly higher in the normal samples. We then developed a combined 3-protein panel of FAK, c-MET, and MST1R expression signatures that can robustly risk-stratify colorectal cancer across all stages into three clusters that differ in progression-free survival. The colorectal cancer subgroup with high FAK, low c-MET, and low MST1R protein levels showed the worst progression-free survival with particularly early progression of disease (p = 0.0053). Combined FAK, c-MET, and MST1R were independently prognostic for progression-free survival in stage II colorectal cancers in a multivariate model. The 3-protein panel provides a potentially clinically attractive method for risk-stratification and adjuvant therapy guidance, especially in stage II disease.
Collapse
|
13
|
Zhou D, Huang L, Zhou Y, Wei T, Yang L, Li C. RON and RONΔ160 promote gastric cancer cell proliferation, migration, and adaption to hypoxia via interaction with β-catenin. Aging (Albany NY) 2020; 11:2735-2748. [PMID: 31085796 PMCID: PMC6535062 DOI: 10.18632/aging.101945] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 04/27/2019] [Indexed: 01/08/2023]
Abstract
Aberrant accumulation of the receptor tyrosine kinase recepteur d’origine nantais (RON) has been verified in gastric adenocarcinoma. Upregulation of RON and its splice variant RONΔ160 contribute to the growth and migration in gastric cancer cells in vitro. However, the mechanisms of RON/RONΔ160-mediated gastric cancer growth and metastasis remain vague. We therefore examined the actions of RON, RONΔ160, and β-catenin in gastric cancer cells and tissue samples, and their effects on cell growth in vitro and in vivo. We found that in gastric cancer samples and cell lines, there was positive correlation between RON/RONΔ160 and β-catenin levels, and that they formed a RON/RONΔ160-β-catenin complex which was translocated to the nucleus. Hypoxia led the binding of hypoxia-inducible factor-1α to the RON/RONΔ160-β-catenin complex, which increased nuclear translocation and expression of downstream oncogenic signaling molecules. Overexpression of RON/RONΔ160 promoted the proliferation and migration of gastric cancer cells, which were also enhanced by hypoxia. Suppression of RON using siRNA or anti‑RON monoclonal antibody diminished gastric cancer cell and tumor growth in vitro and in vivo. These findings establish a link between the receptor tyrosine kinase RON and β-catenin and provide insight into the mechanism by which they contribute to gastric cancer progression.
Collapse
Affiliation(s)
- Donghui Zhou
- Department of Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Ling Huang
- Department of Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yong Zhou
- Department of Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Tao Wei
- Department of Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Lina Yang
- Department of Oncology, the Affiliated Dongnan Hospital of Xiamen University, Zhangzhou, Fujian 363000, China
| | - Chao Li
- Department of Medical Oncology, Affiliated Hospital of Inner Mongolia Medical University, Huhhot, Inner Mongolia 010030, China
| |
Collapse
|
14
|
Yao HP, Suthe SR, Tong XM, Wang MH. Targeting RON receptor tyrosine kinase for treatment of advanced solid cancers: antibody-drug conjugates as lead drug candidates for clinical trials. Ther Adv Med Oncol 2020; 12:1758835920920069. [PMID: 32426050 PMCID: PMC7222236 DOI: 10.1177/1758835920920069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/27/2020] [Indexed: 02/06/2023] Open
Abstract
The recepteur d'origine nantais (RON) receptor tyrosine kinase, belonging to the mesenchymal-to-epithelial transition proto-oncogene family, has been implicated in the pathogenesis of cancers derived from the colon, lung, breast, and pancreas. These findings lay the foundation for targeting RON for cancer treatment. However, development of RON-targeted therapeutics has not gained sufficient attention for the last decade. Although therapeutic monoclonal antibodies (TMABs) targeting RON have been validated in preclinical studies, results from clinical trials have met with limited success. This outcome diminishes pharmaceutical enthusiasm for further development of RON-targeted therapeutics. Recently, antibody-drug conjugates (ADCs) targeting RON have drawn special attention owing to their increased therapeutic activity. The rationale for developing anti-RON ADCs is based on the observation that cancer cells are not sufficiently addicted to RON signaling for survival. Thus, TMAB-mediated inhibition of RON signaling is ineffective for clinical application. In contrast, anti-RON ADCs combine a target-specific antibody with potent cytotoxins for cancer cell killing. This approach not only overcomes the shortcomings in TMAB-targeted therapies but also holds the promise for advancing anti-RON ADCs into clinical trials. In this review, we discuss the latest advancements in the development of anti-RON ADCs for targeted cancer therapy including drug conjugation profile, pharmacokinetic properties, cytotoxic effect in vitro, efficacy in tumor models, and toxicological activities in primates.
Collapse
Affiliation(s)
- Hang-Ping Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- National Clinical Research Center for Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sreedhar Reddy Suthe
- Cancer Biology Research Center, Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Xiang-Min Tong
- Department of Hematology, Zhejiang Provincial People’s Hospital and People’s Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ming-Hai Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Biology Research Center, Texas Tech University Health Sciences Jerry H. Hodge School of Pharmacy, 1406 Coulter Street, Amarillo, TX 79106, USA
| |
Collapse
|
15
|
Tran LKH, Tran PMH, Mysona DP, Purohit SB, Myers E, Lee WS, Dun B, Xu D, Liu H, Hopkins D, Nechtman J, Scelsi CL, Mittal PK, Kleven D, Wallbillich JJ, Rungruang B, Ghamande S, She JX. A 73-gene proliferative transcriptomic signature predicts uterine serous carcinoma patient survival and response to primary therapy. Gynecol Oncol 2020; 157:340-347. [PMID: 32067813 DOI: 10.1016/j.ygyno.2020.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/03/2020] [Accepted: 02/08/2020] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To develop a transcriptomic signature capable of predicting overall survival (OS) for uterine serous carcinoma (USC). METHODS RNAseq data for 58 USC patients were obtained from TCGA. Expression of 73 candidate genes was measured for 67 Augusta University (AU) samples using NanoString technology. RESULTS Analysis of the TCGA RNAseq data identified 73 genes that individually predict prognosis for USC patients and an elastic net model with all 73 genes (USC73) distinguishes a good OS group with low USC73 score from a poor OS group with high USC73 score (5-year OS = 83.3% and 13.3% respectively, HR = 40.1; p = 3 × 10-8). This finding was validated in the independent AU cohort (HR = 4.3; p = 0.0004). The poor prognosis group with high USC73 score consists of 37.9% and 32.8% of patients in the TCGA and AU cohort respectively. USC73 score and pathologic stage independently contribute to OS and together provide the best prognostic value. Early stage, low USC73 patients have the best prognosis (5-year OS = 85.1% in the combined dataset), while advanced stage, high USC73 patients have the worst prognosis (5-year OS = 6.4%, HR = 30.5, p = 1.2 × 10-12). Consistent with the observed poor survival, primary cell cultures from high USC73 patients had higher proliferation rate and cell cycle progression; and high USC73 patients had lower rates of complete response to standard therapy. CONCLUSIONS The USC73 transcriptomic signature and stage independently predict OS of USC patients and the best prediction is achieved using USC73 and stage. USC73 may also serve as a therapeutic biomarker to guide patient care.
Collapse
Affiliation(s)
- Lynn K H Tran
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Paul M H Tran
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - David P Mysona
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Sharad B Purohit
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA; Department of Obstetrics and Gynecology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA; Department of Undergraduate Health Professionals, College of Allied Health Sciences, Augusta University, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Emily Myers
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Won Sok Lee
- Department of Pathology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Boying Dun
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA; Department of Obstetrics and Gynecology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Duo Xu
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Haitao Liu
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Diane Hopkins
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - John Nechtman
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Chris L Scelsi
- Department of Radiology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Pardeep K Mittal
- Department of Radiology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Daniel Kleven
- Department of Pathology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - John J Wallbillich
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA; Department of Obstetrics and Gynecology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Bunja Rungruang
- Department of Obstetrics and Gynecology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Sharad Ghamande
- Department of Obstetrics and Gynecology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, 1120 15(th) Street, Augusta, GA 30912, USA; Department of Obstetrics and Gynecology, Medical College of Georgia, 1120 15(th) Street, Augusta, GA 30912, USA.
| |
Collapse
|
16
|
Therapeutic anti-cancer activity of antibodies targeting sulfhydryl bond constrained epitopes on unglycosylated RON receptor tyrosine kinase. Oncogene 2019; 38:7342-7356. [PMID: 31417186 DOI: 10.1038/s41388-019-0946-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/13/2019] [Accepted: 06/25/2019] [Indexed: 01/18/2023]
Abstract
Recepteur d'origine nantais (RON) receptor tyrosine kinase (RTK) and its ligand, serum macrophage-stimulating protein (MSP), are well-established oncogenic drivers for tumorigenesis and metastasis. RON is often found to be alternatively spliced resulting in various isoforms that are constitutively active. RON is therefore an attractive target for cancer therapeutics, including small molecular inhibitors and monoclonal antibodies. While small molecule inhibitors of RON may inhibit other protein kinases including the highly similar MET kinase, monoclonal antibodies targeting RON are more specific, potentially inducing fewer side effects. Although anti-RON monoclonal antibody therapies have been developed and tested in clinical trials, they were met with limited success. Cancer cells have been associated with aberrant glycosylation mechanisms. Notably for RON, the loss of N-bisected glycosylation is a direct cause for tumorigenesis and poorer prognosis in cancer patients. Particularly in gastric cancer, aberrant RON glycosylation augments RON activation. Here, we present a novel panel of monoclonal antibodies which potentially widens the specific targeting of not only the glycosylated RON, but also unglycosylated and aberrantly glycosylated RON. Our antibodies can bind strongly to deglycosylated RON from tunicamycin treated cells, recognise RON in IHC/IF and possess superior therapeutic efficacy in RON expressing xenograft tumours. Our most potent antibody in xenograft assays, is directed to the RON alpha chain and targets a sulfhydryl bond constrained epitope that appears to be cryptic in the crystal structure. This establishes the paradigm that such constrained and cryptic epitopes represent good targets for therapeutic antibodies.
Collapse
|
17
|
Kim SA, Lee KH, Lee DH, Lee JK, Lim SC, Joo YE, Chung IJ, Noh MG, Yoon TM. Receptor tyrosine kinase, RON, promotes tumor progression by regulating EMT and the MAPK signaling pathway in human oral squamous cell carcinoma. Int J Oncol 2019; 55:513-526. [PMID: 31268163 DOI: 10.3892/ijo.2019.4836] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 06/18/2019] [Indexed: 11/05/2022] Open
Abstract
The receptor tyrosine kinase, recepteur d'origine nantais (RON), is known to be associated with the progression, metastasis, and prognosis of various types of cancers. Nevertheless, the role of RON in human oral squamous cell carcinoma (OSCC) is unclear. This study evaluated whether RON affects oncogenic behavior, oncogenic signaling pathways, and clinical outcomes, including survival, in human OSCC. Reverse transcription‑PCR, quantitative PCR, western blotting and immunohistochemical staining were used to determine mRNA and protein expression levels of RON. Cell invasion, migration and apoptosis assays were used to assess the functional effects of small interfering RNA‑mediated knockdown of RON or snail family transcriptional repressor 2 (SLUG). RON knockdown suppressed tumor cell invasion and migration and enhanced apoptosis in human OSCC cells. RON knockdown also decreased the phosphorylation of MAPK signaling proteins, such as ERK1/2, JNK and p38. In addition, RON knockdown suppressed the expression of the epithelial mesenchymal transition (EMT)‑related transcription factor, SLUG. SLUG knockdown blocked the enhancement of cell invasion and migration induced by macrophage‑stimulation protein (MSP)‑mediated RON activation in OSCC cells. The cell morphology was changed to spindle‑like shape under MSP‑mediated RON activation in OSCC cells. RON was overexpressed in both fresh and paraffin‑embedded human OSCC tissues. Taken together, these results indicate that RON contributed to tumor progression by regulating the EMT‑related factor, SLUG, and the MAPK pathway in OSCC. This study may provide a theoretical basis for the application of RON‑targeting agents, currently being studied in various cancer fields, for the treatment of OSCC.
Collapse
Affiliation(s)
- Sun-Ae Kim
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Kyung-Hwa Lee
- Department of Pathology, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Dong Hoon Lee
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Joon Kyoo Lee
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Sang Chul Lim
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Ik-Joo Chung
- Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| | - Myung-Giun Noh
- Department of Genomic Medicine, Gwangju Institute of Science and Technology, Gwangju, Gwangju 61005, Republic of Korea
| | - Tae Mi Yoon
- Department of Otorhinolaryngology‑Head and Neck Surgery, Chonnam National University Medical School and Hwasun Hospital, Hwasun, Chonnam 58128, Republic of Korea
| |
Collapse
|
18
|
Mereiter S, Martins ÁM, Gomes C, Balmaña M, Macedo JA, Polom K, Roviello F, Magalhães A, Reis CA. O‐glycan truncation enhances cancer‐related functions of
CD
44 in gastric cancer. FEBS Lett 2019; 593:1675-1689. [DOI: 10.1002/1873-3468.13432] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 05/04/2019] [Accepted: 05/06/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Stefan Mereiter
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Álvaro M. Martins
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Catarina Gomes
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Meritxell Balmaña
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Joana A. Macedo
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Karol Polom
- Department of Surgical Oncology Medical University of Gdansk Poland
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Franco Roviello
- General Surgery and Surgical Oncology Department University of Siena Italy
| | - Ana Magalhães
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
| | - Celso A. Reis
- I3S – Instituto de Investigação e Inovação em Saúde Universidade do Porto Portugal
- IPATIMUP – Institute of Molecular Pathology and Immunology University of Porto Portugal
- Faculty of Medicine University of Porto Portugal
- Instituto de Ciências Biomédicas Abel Salazar University of Porto Portugal
| |
Collapse
|
19
|
Guo Q, Huang F, Goncalves C, Del Rincón SV, Miller WH. Translation of cancer immunotherapy from the bench to the bedside. Adv Cancer Res 2019; 143:1-62. [PMID: 31202357 DOI: 10.1016/bs.acr.2019.03.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The tremendous success of immune checkpoint blockades has revolutionized cancer management. Our increased understanding of the cell types that compose the tumor microenvironment (TME), including those of the innate and adaptive immune system, has helped to shape additional immune modulatory strategies in cancer care. Pre-clinical and clinical investigations targeting novel checkpoint interactions and key pathways that regulate cancer immunity continue to increase rapidly. Various combinatorial drug regimens are being tested in attempt to achieve durable response and survival rates of patients with cancer. This review provides an overview of specific components of the TME, an introduction to novel immune checkpoints, followed by a survey of present day and future combination immune modulatory therapies. The idea that the immune system can recognize and destroy tumor cells was first described in the cancer immunosurveillance hypothesis of Burnet and Thomas. However, early experimental evidence failed to support the concept. It was not until the late 1990s when seminal papers clearly showed the existence of cancer immunosurveillance, leading to the cancer immunoediting hypothesis. In this century, progress in the understanding of negative regulators of the immune response led to the discovery that inhibition of these regulators in patients with cancer could lead to dramatic and durable remissions. Drs. Tasuku Honjo and James P. Allison were awarded the Nobel Prize in 2018 for their pioneering work in this field. We now see rapid advances in cancer immunology and emerging effective therapies revolutionizing cancer care across tumor types in the clinic, while pre-clinical research is moving from a focus on the malignant cells themselves to dissect the highly heterogenic and complex multi-cellular tumor microenvironment (TME).
Collapse
Affiliation(s)
- Qianyu Guo
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Fan Huang
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Christophe Goncalves
- Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Wilson H Miller
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada; Rossy Cancer Network, Montreal, QC, Canada.
| |
Collapse
|
20
|
Baird AM, Easty D, Jarzabek M, Shiels L, Soltermann A, Klebe S, Raeppel S, MacDonagh L, Wu C, Griggs K, Kirschner MB, Stanfill B, Nonaka D, Goparaju CM, Murer B, Fennell DA, O'Donnell DM, Barr MP, Mutti L, Reid G, Finn S, Cuffe S, Pass HI, Opitz I, Byrne AT, O'Byrne KJ, Gray SG. When RON MET TAM in Mesothelioma: All Druggable for One, and One Drug for All? Front Endocrinol (Lausanne) 2019; 10:89. [PMID: 30863365 PMCID: PMC6399142 DOI: 10.3389/fendo.2019.00089] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/31/2019] [Indexed: 12/12/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive inflammatory cancer with a poor survival rate. Treatment options are limited at best and drug resistance is common. Thus, there is an urgent need to identify novel therapeutic targets in this disease in order to improve patient outcomes and survival times. MST1R (RON) is a trans-membrane receptor tyrosine kinase (RTK), which is part of the c-MET proto-oncogene family. The only ligand recognized to bind MST1R (RON) is Macrophage Stimulating 1 (MST1), also known as Macrophage Stimulating Protein (MSP) or Hepatocyte Growth Factor-Like Protein (HGFL). In this study, we demonstrate that the MST1-MST1R (RON) signaling axis is active in MPM. Targeting this pathway with a small molecule inhibitor, LCRF-0004, resulted in decreased proliferation with a concomitant increase in apoptosis. Cell cycle progression was also affected. Recombinant MST1 treatment was unable to overcome the effect of LCRF-0004 in terms of either proliferation or apoptosis. Subsequently, the effect of an additional small molecular inhibitor, BMS-777607 (which targets MST1R (RON), MET, Tyro3, and Axl) also resulted in a decreased proliferative capacity of MPM cells. In a cohort of MPM patient samples, high positivity for total MST1R by IHC was an independent predictor of favorable prognosis. Additionally, elevated expression levels of MST1 also correlated with better survival. This study also determined the efficacy of LCRF-0004 and BMS-777607 in xenograft MPM models. Both LCRF-0004 and BMS-777607 demonstrated significant anti-tumor efficacy in vitro, however BMS-777607 was far superior to LCRF-0004. The in vivo and in vitro data generated by this study indicates that a multi-TKI, targeting the MST1R/MET/TAM signaling pathways, may provide a more effective therapeutic strategy for the treatment of MPM as opposed to targeting MST1R alone.
Collapse
Affiliation(s)
- Anne-Marie Baird
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
- Cancer and Ageing Research Program, Queensland University of Technology, Brisbane, QLD, Australia
| | - David Easty
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Monika Jarzabek
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Liam Shiels
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alex Soltermann
- Department of Clinical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Sonja Klebe
- Department of Anatomical Pathology, Flinders University of South Australia, Bedford Park, SA, Australia
| | | | - Lauren MacDonagh
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Chengguang Wu
- Department of Clinical Pathology, University Hospital Zurich, Zurich, Switzerland
| | - Kim Griggs
- Department of Anatomical Pathology, Flinders University of South Australia, Bedford Park, SA, Australia
| | - Michaela B. Kirschner
- Asbestos Diseases Research Institute, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, NSW, Australia
| | - Bryan Stanfill
- The Commonwealth Scientific and Industrial Research Organization, Brisbane, QLD, Australia
| | - Daisuke Nonaka
- Department of Histopathology, The Christie NHS Foundation Trust, Manchester, United Kingdom
- Department of Cardiothoracic Surgery, New York University (NYU) Langone Medical Center, New York, NY, United States
| | - Chandra M. Goparaju
- Department of Cardiothoracic Surgery, New York University (NYU) Langone Medical Center, New York, NY, United States
| | - Bruno Murer
- Department of Clinical Pathology, Ospedale dell'Angelo, Venice, Italy
| | - Dean A. Fennell
- MRC Toxicology Unit, University of Leicester and Leicester University Hospitals, Leicester, United Kingdom
| | | | - Martin P. Barr
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
| | - Luciano Mutti
- Center for Biotechnology, Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Glen Reid
- Asbestos Diseases Research Institute, Sydney, NSW, Australia
- Sydney Medical School, University of Sydney, NSW, Australia
| | - Stephen Finn
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin, Ireland
| | - Sinead Cuffe
- HOPE Directorate, St James's Hospital, Dublin, Ireland
| | - Harvey I. Pass
- Department of Cardiothoracic Surgery, New York University (NYU) Langone Medical Center, New York, NY, United States
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Annette T. Byrne
- Department of Physiology and Medical Physics and Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Kenneth J. O'Byrne
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
- Cancer and Ageing Research Program, Queensland University of Technology, Brisbane, QLD, Australia
- HOPE Directorate, St James's Hospital, Dublin, Ireland
- Division of Cancer Services, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Steven G. Gray
- Thoracic Oncology Research Group, Labmed Directorate, St. James's Hospital, Dublin, Ireland
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- *Correspondence: Steven G. Gray
| |
Collapse
|
21
|
Faham N, Zhao L, Welm AL. mTORC1 is a key mediator of RON-dependent breast cancer metastasis with therapeutic potential. NPJ Breast Cancer 2018; 4:36. [PMID: 30456298 PMCID: PMC6226524 DOI: 10.1038/s41523-018-0091-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 10/11/2018] [Indexed: 02/02/2023] Open
Abstract
Metastasis is the biggest challenge in treating breast cancer, and it kills >40,000 breast cancer patients annually in the US. Aberrant expression of the RON receptor tyrosine kinase in breast tumors correlates with poor prognosis and has been shown to promote metastasis. However, the molecular mechanisms that govern how RON promotes metastasis, and how to block it, are still largely unknown. We sought to determine critical effectors of RON using a combination of mutational and pharmacologic strategies. High-throughput proteomic analysis of breast cancer cells upon activation of RON showed robust phosphorylation of ribosomal protein S6. Further analysis revealed that RON strongly signals through mTORC1/p70S6K, which is mediated predominantly by the PI3K pathway. A targeted mutation approach to modulate RON signaling validated the importance of PI3K/mTORC1 pathway for spontaneous metastasis in vivo. Finally, inhibition of mTORC1 with an FDA-approved drug, everolimus, resulted in transient shrinkage of established RON-dependent metastases, and combined blockade of mTORC1 and RON delayed progression. These studies have identified a key downstream mediator of RON-dependent metastasis in breast cancer cells and revealed that inhibition of mTORC1, or combined inhibition of mTORC1 and RON, may be effective for treatment of metastatic breast cancers with elevated expression of RON.
Collapse
Affiliation(s)
- Najme Faham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| | - Ling Zhao
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT USA
| |
Collapse
|
22
|
Maron SB, Alpert L, Kwak HA, Lomnicki S, Chase L, Xu D, O'Day E, Nagy RJ, Lanman RB, Cecchi F, Hembrough T, Schrock A, Hart J, Xiao SY, Setia N, Catenacci DVT. Targeted Therapies for Targeted Populations: Anti-EGFR Treatment for EGFR-Amplified Gastroesophageal Adenocarcinoma. Cancer Discov 2018; 8:696-713. [PMID: 29449271 PMCID: PMC5984701 DOI: 10.1158/2159-8290.cd-17-1260] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/11/2018] [Accepted: 02/09/2018] [Indexed: 02/07/2023]
Abstract
Previous anti-EGFR trials in unselected patients with gastroesophageal adenocarcinoma (GEA) were resoundingly negative. We identified EGFR amplification in 5% (19/363) of patients at the University of Chicago, including 6% (8/140) who were prospectively screened with intention-to-treat using anti-EGFR therapy. Seven patients received ≥1 dose of treatment: three first-line FOLFOX plus ABT-806, one second-line FOLFIRI plus cetuximab, and three third/fourth-line cetuximab alone. Treatment achieved objective response in 58% (4/7) and disease control in 100% (7/7) with a median progression-free survival of 10 months. Pretreatment and posttreatment tumor next-generation sequencing (NGS), serial plasma circulating tumor DNA (ctDNA) NGS, and tumor IHC/FISH for EGFR revealed preexisting and/or acquired genomic events, including EGFR-negative clones, PTEN deletion, KRAS amplification/mutation, NRAS, MYC, and HER2 amplification, and GNAS mutations serving as mechanisms of resistance. Two evaluable patients demonstrated interval increase of CD3+ infiltrate, including one who demonstrated increased NKp46+, and PD-L1 IHC expression from baseline, suggesting an immune therapeutic mechanism of action. EGFR amplification predicted benefit from anti-EGFR therapy, albeit until various resistance mechanisms emerged.Significance: This paper highlights the role of EGFR inhibitors in EGFR-amplified GEA-despite negative results in prior unselected phase III trials. Using serial ctDNA and tissue NGS, we identified mechanisms of primary and acquired resistance in all patients, as well as potential contribution of antibody-dependent cell-mediated cytotoxicity to their clinical benefit. Cancer Discov; 8(6); 696-713. ©2018 AACR.See related commentary by Strickler, p. 679This article is highlighted in the In This Issue feature, p. 663.
Collapse
Affiliation(s)
- Steven B Maron
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Lindsay Alpert
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Heewon A Kwak
- Department of Pathology, University of Chicago, Chicago, Illinois
| | | | - Leah Chase
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - David Xu
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Emily O'Day
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | | | | | | | | | - John Hart
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Shu-Yuan Xiao
- Department of Pathology, University of Chicago, Chicago, Illinois
| | - Namrata Setia
- Department of Pathology, University of Chicago, Chicago, Illinois
| | | |
Collapse
|
23
|
Abstract
Gastroesophageal cancer (GEC) remains a major cause of cancer-related mortality worldwide. Although the incidence of distal gastric adenocarcinoma (GC) is declining in the United States, proximal esophagogastric junction adenocarcinoma (EGJ) is increasing in incidence. GEC, including GC and EGJ, is treated uniformly in the metastatic setting. Overall survival in the metastatic setting remains poor. Molecular characterization of GEC has identified mutations and copy number variations, along with other oncogenes, biomarkers, and immuno-oncologic checkpoints that may serve as actionable therapeutic targets. This article reviews these key aberrations, their impact on protein expression, therapeutic implications, and clinical directions within each pathway.
Collapse
Affiliation(s)
- Steven B Maron
- Section of Hematology/Oncology, University of Chicago Comprehensive Cancer Center, 900 E 57th St, Suite 7128, Chicago, IL 60637, USA
| | - Daniel V T Catenacci
- Section of Hematology/Oncology, University of Chicago Comprehensive Cancer Center, 900 E 57th St, Suite 7128, Chicago, IL 60637, USA.
| |
Collapse
|
24
|
Milan M, Benvenuti S, Balderacchi AM, Virzì AR, Gentile A, Senetta R, Cassoni P, Comoglio PM, Stella GM. RON tyrosine kinase mutations in brain metastases from lung cancer. ERJ Open Res 2018. [PMID: 29531956 PMCID: PMC5838354 DOI: 10.1183/23120541.00083-2017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Brain metastases originate from cancer cells that have spread through the bloodstream and reached the brain from primary tumours in other organs. Metastatic masses are among the most common intracranial neoplasms, occurring in ∼25% of cancer patients. Most brain metastases derive from lung cancers (40–50% of cases), breast cancers (10–16%), melanoma (5–20%), kidney and ovarian cancers (5–10%) and more rarely colorectal cancers. Sometimes (5–10%) no primary site is detectable [1]. The onset of brain lesions from primary solid cancers is associated with poor prognosis with a median survival of 4–5 months [2]. Molecular mechanisms regulating metastatic spreading to the brain are largely unknown. RON (Recepteur d'Origine Nantais), also known as macrophage-stimulating receptor-1 (MSTR1) or stem cell derived tyrosine kinase (STK) in mice, belongs to the family of tyrosine kinase receptors of which MET is the prototype. It has been shown that RON regulates cellular proliferation, adhesion, motility and protection from apoptosis, all events resulting in the invasive growth genetic programme [3], which occurs in specific physiological conditions (i.e. embryonic development) and, when aberrantly regulated, contributes to tumour onset, progression and, above all, metastatic dissemination. RON mutations might identify actionable targets in highly aggressive lung tumourshttp://ow.ly/RTUp30hSBX6
Collapse
Affiliation(s)
- Melissa Milan
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Catholic University of Sacred Heart, Rome, Italy
| | | | | | - Anna Rita Virzì
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,University of Turin, Turin, Italy
| | | | | | - Paola Cassoni
- Dept of Medical Sciences, University of Turin, Turin, Italy
| | | | - Giulia Maria Stella
- Cardiothoracic Dept, Section of Respiratory System Diseases, IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
25
|
Catenacci DVT, Tebbutt NC, Davidenko I, Murad AM, Al-Batran SE, Ilson DH, Tjulandin S, Gotovkin E, Karaszewska B, Bondarenko I, Tejani MA, Udrea AA, Tehfe M, De Vita F, Turkington C, Tang R, Ang A, Zhang Y, Hoang T, Sidhu R, Cunningham D. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 18:1467-1482. [PMID: 28958504 DOI: 10.1016/s1470-2045(17)30566-1] [Citation(s) in RCA: 281] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Rilotumumab is a fully human monoclonal antibody that selectively targets the ligand of the MET receptor, hepatocyte growth factor (HGF). We aimed to assess the efficacy, safety, and pharmacokinetics of rilotumumab combined with epirubicin, cisplatin, and capecitabine, and to assess potential biomarkers, in patients with advanced MET-positive gastric or gastro-oesophageal junction adenocarcinoma. METHODS This multicentre, randomised, double-blind, placebo-controlled, phase 3 study was done at 152 centres in 27 countries. We recruited adults (aged ≥18 years) with unresectable locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, MET-positive tumours (≥25% of tumour cells with membrane staining of ≥1+ staining intensity), and evaluable disease, who had not received previous systemic therapy. Eligible patients were randomly assigned (1:1) via a computerised voice response system to receive rilotumumab 15 mg/kg intravenously or placebo in combination with open-label chemotherapy (epirubicin 50 mg/m2 intravenously; cisplatin 60 mg/m2 intravenously; capecitabine 625 mg/m2 orally twice daily) in 21-day cycles for up to ten cycles. After completion of chemotherapy, patients continued to receive rilotumumab or placebo monotherapy until disease progression, intolerability, withdrawal of consent, or study termination. Randomisation was stratified by disease extent and ECOG performance status. Both patients and physicians were masked to study treatment assignment. The primary endpoint was overall survival, analysed by intention to treat. We report the final analysis. This study is registered with ClinicalTrials.gov, number NCT01697072. FINDINGS Between Nov 7, 2012, and Nov 21, 2014, 609 patients were randomly assigned to rilotumumab plus epirubicin, cisplatin, and capecitabine (rilotumumab group; n=304) or placebo plus epirubicin, cisplatin, and capecitabine (placebo group; n=305). Study treatment was stopped early after an independent data monitoring committee found a higher number of deaths in the rilotumumab group than in the placebo group; all patients in the rilotumumab group subsequently discontinued all study treatment. Median follow-up was 7·7 months (IQR 3·6-12·0) for patients in the rilotumumab group and 9·4 months (5·3-13·1) for patients in the placebo group. Median overall survival was 8·8 months (95% CI 7·7-10·2) in the rilotumumab group compared with 10·7 months (9·6-12·4) in the placebo group (stratified hazard ratio 1·34, 95% CI 1·10-1·63; p=0·003). The most common grade 3 or worse adverse events in the rilotumumab and placebo groups were neutropenia (86 [29%] of 298 patients vs 97 [32%] of 299 patients), anaemia (37 [12%] vs 43 [14%]), and fatigue (30 [10%] vs 35 [12%]). The frequency of serious adverse events was similar in the rilotumumab and placebo groups (142 [48%] vs 149 [50%]). More deaths due to adverse events occurred in the rilotumumab group than the placebo group (42 [14%] vs 31 [10%]). In the rilotumumab group, 33 (11%) of 298 patients had fatal adverse events due to disease progression, and nine (3%) had fatal events not due to disease progression. In the placebo group, 23 (8%) of 299 patients had fatal adverse events due to disease progression, and eight (3%) had fatal events not due to disease progression. INTERPRETATION Ligand-blocking inhibition of the MET pathway with rilotumumab is not effective in improving clinical outcomes in patients with MET-positive gastric or gastro-oesophageal adenocarcinoma. FUNDING Amgen.
Collapse
Affiliation(s)
| | | | - Irina Davidenko
- State Budgetary Healthcare Institution, Clinical Oncology Dispensary #1, Krasnodar Region Ministry of Healthcare, Krasnodar, Russia
| | - André M Murad
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Horizonte, Brazil
| | - Salah-Eddin Al-Batran
- Institute of Clinical Cancer Research, Krankenhaus Nordwest, University Cancer Center, Frankfurt, Germany
| | - David H Ilson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Evengy Gotovkin
- Regional Budgetary Institution of Public Health Ivanovo Regional Oncology Dispensary, Ivanovo, Russia
| | | | - Igor Bondarenko
- Dnipropetrovsk Medical Academy, City Multifield Clinical Hospital 4, Dnipropetrovsk, Ukraine
| | - Mohamedtaki A Tejani
- University of Rochester Medical Center, James P Wilmot Cancer Center, Rochester, NY, USA
| | | | - Mustapha Tehfe
- Centre Hospitalier de L'Universite de Montreal Notre-Dame, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yang SY, Nguyen TT, Ung TT, Jung YD. Role of Recepteur D'origine Nantais on Gastric Cancer Development and Progression. Chonnam Med J 2017; 53:178-186. [PMID: 29026705 PMCID: PMC5636756 DOI: 10.4068/cmj.2017.53.3.178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 01/12/2023] Open
Abstract
Recepteur d'origine nantais (RON) is a receptor tyrosine kinase belonging to the subfamily of which c-MET is the prototype. Large epidemiologic studies have confirmed the strong association between RON and gastric cancer development. Constitutive activation of RON signaling directly correlates with tumorigenic phenotypes of gastric cancer and a poor survival rate in advanced gastric cancer patients. In this review, we focus on recent evidence of the aberrant expression and activation of RON in gastric cancer tumors and provide insights into the mechanism of RON signaling associated with gastric cancer progression and metastasis. Current therapeutics against RON in gastric cancer are summarized.
Collapse
Affiliation(s)
- Sung Yeul Yang
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Thi Thinh Nguyen
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Trong Thuan Ung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
27
|
Technological advances for interrogating the human kinome. Biochem Soc Trans 2017; 45:65-77. [PMID: 28202660 DOI: 10.1042/bst20160163] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/20/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022]
Abstract
There is increasing appreciation among researchers and clinicians of the value of investigating biology and pathobiology at the level of cellular kinase (kinome) activity. Kinome analysis provides valuable opportunity to gain insights into complex biology (including disease pathology), identify biomarkers of critical phenotypes (including disease prognosis and evaluation of therapeutic efficacy), and identify targets for therapeutic intervention through kinase inhibitors. The growing interest in kinome analysis has fueled efforts to develop and optimize technologies that enable characterization of phosphorylation-mediated signaling events in a cost-effective, high-throughput manner. In this review, we highlight recent advances to the central technologies currently available for kinome profiling and offer our perspectives on the key challenges remaining to be addressed.
Collapse
|
28
|
Abstract
Gastroesophageal cancer (GEC) remains a major cause of cancer-related mortality worldwide. Although the incidence of distal gastric adenocarcinoma (GC) is declining in the United States, proximal esophagogastric junction adenocarcinoma (EGJ) incidence is rising. GC and EGJ, together, are treated uniformly in the metastatic setting as GEC. Overall survival in the metastatic setting remains poor, with few molecular targeted approaches having been successfully incorporated into routine care to date-only first-line anti-HER2 therapy for ERBB2 amplification and second-line anti-VEGFR2 therapy. This article reviews aberrations in epidermal growth factor receptor, MET, and ERBB2, their therapeutic implications, and future directions in targeting these pathways.
Collapse
Affiliation(s)
- Steven B Maron
- Section of Hematology/Oncology, University of Chicago Comprehensive Cancer Center, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | - Daniel V T Catenacci
- The University of Chicago Medical Center & Biological Sciences, 900 East 57th Street, KCBD Building, Office 7128, Chicago, IL 60637, USA.
| |
Collapse
|
29
|
Zhang X, Maity T, Kashyap MK, Bansal M, Venugopalan A, Singh S, Awasthi S, Marimuthu A, Charles Jacob HK, Belkina N, Pitts S, Cultraro CM, Gao S, Kirkali G, Biswas R, Chaerkady R, Califano A, Pandey A, Guha U. Quantitative Tyrosine Phosphoproteomics of Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitor-treated Lung Adenocarcinoma Cells Reveals Potential Novel Biomarkers of Therapeutic Response. Mol Cell Proteomics 2017; 16:891-910. [PMID: 28331001 PMCID: PMC5417828 DOI: 10.1074/mcp.m117.067439] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/24/2017] [Indexed: 02/05/2023] Open
Abstract
Mutations in the Epidermal growth factor receptor (EGFR) kinase domain, such as the L858R missense mutation and deletions spanning the conserved sequence 747LREA750, are sensitive to tyrosine kinase inhibitors (TKIs). The gatekeeper site residue mutation, T790M accounts for around 60% of acquired resistance to EGFR TKIs. The first generation EGFR TKIs, erlotinib and gefitinib, and the second generation inhibitor, afatinib are FDA approved for initial treatment of EGFR mutated lung adenocarcinoma. The predominant biomarker of EGFR TKI responsiveness is the presence of EGFR TKI-sensitizing mutations. However, 30-40% of patients with EGFR mutations exhibit primary resistance to these TKIs, underscoring the unmet need of identifying additional biomarkers of treatment response. Here, we sought to characterize the dynamics of tyrosine phosphorylation upon EGFR TKI treatment of mutant EGFR-driven human lung adenocarcinoma cell lines with varying sensitivity to EGFR TKIs, erlotinib and afatinib. We employed stable isotope labeling with amino acids in cell culture (SILAC)-based quantitative mass spectrometry to identify and quantify tyrosine phosphorylated peptides. The proportion of tyrosine phosphorylated sites that had reduced phosphorylation upon erlotinib or afatinib treatment correlated with the degree of TKI-sensitivity. Afatinib, an irreversible EGFR TKI, more effectively inhibited tyrosine phosphorylation of a majority of the substrates. The phosphosites with phosphorylation SILAC ratios that correlated with the TKI-sensitivity of the cell lines include sites on kinases, such as EGFR-Y1197 and MAPK7-Y221, and adaptor proteins, such as SHC1-Y349/350, ERRFI1-Y394, GAB1-Y689, STAT5A-Y694, DLG3-Y705, and DAPP1-Y139, suggesting these are potential biomarkers of TKI sensitivity. DAPP1, is a novel target of mutant EGFR signaling and Y-139 is the major site of DAPP1 tyrosine phosphorylation. We also uncovered several off-target effects of these TKIs, such as MST1R-Y1238/Y1239 and MET-Y1252/1253. This study provides unique insight into the TKI-mediated modulation of mutant EGFR signaling, which can be applied to the development of biomarkers of EGFR TKI response.
Collapse
Affiliation(s)
- Xu Zhang
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Tapan Maity
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Manoj K Kashyap
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Mukesh Bansal
- ¶Department of System Biology, Columbia University, New York, New York, 10032
- ‖PsychoGenics Inc., Tarrytown, New York, 10591
| | - Abhilash Venugopalan
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Sahib Singh
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shivangi Awasthi
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | | | | | - Natalya Belkina
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Stephanie Pitts
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Constance M Cultraro
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Shaojian Gao
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Guldal Kirkali
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Romi Biswas
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892
| | - Raghothama Chaerkady
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
- **Medimmune LLC, Gaithersburg, Maryland, 20878
| | - Andrea Califano
- ¶Department of System Biology, Columbia University, New York, New York, 10032
| | - Akhilesh Pandey
- §Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205
| | - Udayan Guha
- From the ‡Thoracic and Gastrointestinal Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, 20892;
| |
Collapse
|
30
|
Chen JF, Yu BX, Yu R, Ma L, Lv XY, Cheng Y, Ma Q. Monoclonal antibody Zt/g4 targeting RON receptor tyrosine kinase enhances chemosensitivity of bladder cancer cells to Epirubicin by promoting G1/S arrest and apoptosis. Oncol Rep 2017; 37:721-728. [PMID: 28075465 PMCID: PMC5355669 DOI: 10.3892/or.2017.5356] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 11/21/2016] [Indexed: 02/05/2023] Open
Abstract
Epirubicin (EPI) is one of the most used intravesical chemotherapy agents after transurethral resection to non-muscle invasive bladder tumors (NMIBC) to prevent cancer recurrence and progression. However, even after resection of bladder tumors and intravesical chemotherapy, half of them will recur and progress. RON is a membrane tyrosine kinase receptor usually overexpressed in bladder cancer cells and associated with poor pathological features. This study aims to investigate the effects of anti-RON monoclonal antibody Zt/g4 on the chemosensitivity of bladder cells to EPI. After Zt/g4 treatment, cell cytotoxicity was significantly increased and cell invasion was markedly suppressed in EPI-treated bladder cancer cells. Further investigation indicated that combing Zt/g4 with EPI promoted cell G1/S-phase arrest and apoptosis, which are the potential mechanisms that RON signaling inhibition enhances chemosensitivity of EPI. Thus, combing antibody-based RON targeted therapy enhances the therapeutic effects of intravesical chemotherapy, which provides new strategy for further improvement of NMIBC patient outcomes.
Collapse
Affiliation(s)
- Jun-Feng Chen
- Translational Research Laboratory for Urology, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Bi-Xia Yu
- Translational Research Laboratory for Urology, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Rui Yu
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, Zhejiang 315212, P.R. China
| | - Liang Ma
- Translational Research Laboratory for Urology, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Xiu-Yi Lv
- Translational Research Laboratory for Urology, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Yue Cheng
- Translational Research Laboratory for Urology, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Qi Ma
- Translational Research Laboratory for Urology, Ningbo First Hospital, The Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
31
|
Faham N, Welm AL. RON Signaling Is a Key Mediator of Tumor Progression in Many Human Cancers. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2017; 81:177-188. [PMID: 28057847 DOI: 10.1101/sqb.2016.81.031377] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
With an increasing body of literature covering RON receptor tyrosine kinase function in different types of human cancers, it is becoming clear that RON has prominent roles in both cancer cells and in the tumor-associated microenvironment. RON not only activates several oncogenic signaling pathways in cancer cells, leading to more aggressive behavior, but also promotes an immunosuppressive, alternatively activated phenotype in macrophages and limits the antitumor immune response. These two unique functions of this oncogene, the strong correlation between RON expression and poor outcomes in cancer, and the high tolerability of a new RON inhibitor make it an exciting therapeutic target, the blocking of which offers an advantage toward improving the survival of cancer patients. Here, we discuss recent findings on the role of RON signaling in cancer progression and its potential in cancer therapy.
Collapse
Affiliation(s)
- Najme Faham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
32
|
Catenacci DVT, Ang A, Liao WL, Shen J, O'Day E, Loberg RD, Cecchi F, Hembrough T, Ruzzo A, Graziano F. MET tyrosine kinase receptor expression and amplification as prognostic biomarkers of survival in gastroesophageal adenocarcinoma. Cancer 2016; 123:1061-1070. [PMID: 27926778 PMCID: PMC5339041 DOI: 10.1002/cncr.30437] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/14/2016] [Accepted: 10/05/2016] [Indexed: 01/29/2023]
Abstract
BACKGROUND MET gene amplification and Met protein overexpression may be associated with a poor prognosis. The MET/Met status is typically determined with fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC), respectively. Targeted proteomics uses mass spectrometry–based selected reaction monitoring (SRM) to accurately quantitate Met expression. FISH, IHC, and SRM analyses were compared to characterize the prognostic value of MET/Met in gastroesophageal adenocarcinoma (GEC). METHODS Samples from 447 GEC patients were analyzed for MET gene amplification (FISH) and Met protein expression (IHC and SRM). Cox proportional hazards models and Kaplan‐Meier estimates were applied to explore relations between Met, overall survival (OS), and clinical/pathological characteristics. Spearman's rank coefficient was used to assess the correlation between parameters. RESULTS Patients with MET‐amplified tumors had worse OS when: the MET/centromere enumeration probe for chromosome 7 FISH ratio was ≥ 2 (hazard ratio [HR], 3.13; 95% confidence interval [CI], 1.84‐5.33), the MET gene copy number was ≥5 (HR, 2.51; 95% CI, 1.45‐4.34), or ≥ 10% of the cells had ≥15 copies (HR, 4.28; 95% CI, 2.18‐8.39). Similar observations were made with Met protein overexpression by IHC (≥1 + intensity in ≥ 25% of the tumor cell membrane: HR, 1.39; 95% CI, 1.04‐1.86) or SRM (≥400 amol/μg: HR, 1.76; 95% CI, 1.06‐2.90). A significant correlation was observed between MET FISH/Met IHC, MET FISH/Met SRM, and Met IHC/Met SRM; only MET FISH and Met SRM were independent negative prognostic biomarkers in multivariate analyses. CONCLUSIONS MET amplification and overexpression, assessed by multiple methods, were associated with a worse prognosis in univariate analyses. However, only MET amplification by FISH and Met expression by SRM were independent prognostic biomarkers. Compared with IHC, SRM may provide an added benefit for informed decisions about Met‐targeted therapy. Cancer 2017;123:1061–70. © 2016 American Cancer Society. In a large study, MET gene amplification, Met protein overexpression, or both, as assessed by various assays, are associated with a poor prognosis in univariate analyses. However, only MET amplification by fluorescence in situ hybridization and Met expression by selected reaction monitoring mass spectrometry are independent prognostic biomarkers; compared with immunohistochemistry, selected reaction monitoring may provide an added benefit for informed decisions about Met‐targeted therapy.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Agnes Ang
- Amgen, Inc, Thousand Oaks, California
| | | | - Jing Shen
- Amgen, Inc, Thousand Oaks, California
| | - Emily O'Day
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | | | | | | | - Annamaria Ruzzo
- Department of Biomolecular Science, University of Urbino, Urbino, Italy
| | - Francesco Graziano
- Department of Onco-Hematology, Azienda Ospedali Riuniti Marche Nord, Pesaro, Italy
| |
Collapse
|
33
|
Zarei O, Benvenuti S, Ustun-Alkan F, Hamzeh-Mivehroud M, Dastmalchi S. Strategies of targeting the extracellular domain of RON tyrosine kinase receptor for cancer therapy and drug delivery. J Cancer Res Clin Oncol 2016; 142:2429-2446. [PMID: 27503093 DOI: 10.1007/s00432-016-2214-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/01/2016] [Indexed: 01/22/2023]
Abstract
PURPOSE Cancer is one of the most important life-threatening diseases in the world. The current efforts to combat cancer are being focused on molecular-targeted therapies. The main purpose of such approaches is based on targeting cancer cell-specific molecules to minimize toxicity for the normal cells. RON (Recepteur d'Origine Nantais) tyrosine kinase receptor is one of the promising targets in cancer-targeted therapy and drug delivery. METHODS In this review, we will summarize the available agents against extracellular domain of RON with potential antitumor activities. RESULTS The presented antibodies and antibody drug conjugates against RON in this review showed wide spectrum of in vitro and in vivo antitumor activities promising the hope for them entering the clinical trials. CONCLUSION Due to critical role of extracellular domain of RON in receptor activation, the development of therapeutic agents against this region could lead to fruitful outcome in cancer therapy.
Collapse
Affiliation(s)
- Omid Zarei
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Students Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Silvia Benvenuti
- Molecular Therapeutics and Exploratory Research Laboratory, Candiolo Cancer Institute-FPO-IRCCS, Candiolo, Turin, Italy
| | - Fulya Ustun-Alkan
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Istanbul University, Istanbul, Turkey
| | - Maryam Hamzeh-Mivehroud
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Siavoush Dastmalchi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
34
|
Wu Z, Zhang Z, Ge X, Lin Y, Dai C, Chang J, Liu X, Geng R, Wang C, Chen H, Sun M, Guo W, Li J. Identification of short-form RON as a novel intrinsic resistance mechanism for anti-MET therapy in MET-positive gastric cancer. Oncotarget 2016; 6:40519-34. [PMID: 26528757 PMCID: PMC4747350 DOI: 10.18632/oncotarget.5816] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 09/22/2015] [Indexed: 12/30/2022] Open
Abstract
Despite the promising results from initial studies, there are significant limitations in the application of MET-targeted therapy in gastric cancer. Intrinsic resistance is one of the major obstacles. The aim of this study is to identify the responsible receptor tyrosine kinases (RTKs) that determine the unresponsiveness of MET inhibitor in MET-positive gastric cancer. through an RNA-interference-based functional screen targeting most human RTKs, we identified that activation of the fibroblast growth factor receptor 2 (FGFR2) and recepteur d'origine nantais (RON) pathways attenuated MET inhibitor-induced suppression of cell proliferation and migration. Notably, in the two forms of RON pathway activation, only upregulation of short-form RON (sf-RON), but not stimulation of full length RON with macrophage stimulating protein, conferred MET inhibitor resistance in vitro and in vivo. Furthermore, the profile of the gastric cancer samples observed that sf-RON was frequently upregulated in MET-positive gastric cancer. Our findings indicate that activation of the sf-RON signaling pathway represents a novel mechanism underlying MET inhibitor unresponsiveness. A combination strategy with drugs targeting both RON and MET pathways is believed to improve the efficacy of MET-targeted therapy.
Collapse
Affiliation(s)
- Zheng Wu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Zhe Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiaoxiao Ge
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ying Lin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Congqi Dai
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jinjia Chang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xinyang Liu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ruixuan Geng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chenchen Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Huan Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Menghong Sun
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Weijian Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jin Li
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Catenacci DVT, Liao WL, Zhao L, Whitcomb E, Henderson L, O'Day E, Xu P, Thyparambil S, Krizman D, Bengali K, Uzzell J, Darfler M, Cecchi F, Blackler A, Bang YJ, Hart J, Xiao SY, Lee SM, Burrows J, Hembrough T. Mass-spectrometry-based quantitation of Her2 in gastroesophageal tumor tissue: comparison to IHC and FISH. Gastric Cancer 2016; 19:1066-1079. [PMID: 26581548 PMCID: PMC4871781 DOI: 10.1007/s10120-015-0566-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/31/2015] [Indexed: 02/07/2023]
Abstract
BACKGROUND Trastuzumab has shown a survival benefit in cases of Her2-positive gastroesophageal cancer (GEC). Immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) currently determine eligibility for trastuzumab-based therapy. However, these low-throughput assays often produce discordant or equivocal results. METHODS We developed a targeted proteomic assay based on selected reaction monitoring mass spectrometry (SRM-MS) and quantified levels (amol/μg) of Her2-SRM protein in cell lines (n = 27) and GEC tissues (n = 139). We compared Her2-SRM protein expression with IHC/FISH, seeking to determine optimal SRM protein expression cutoffs in order to identify HER2 gene amplification. RESULTS After demonstrating assay development, precision, and stability, Her2-SRM protein measurement was observed to be highly concordant with the HER2/CEP17 ratio, particularly in a multivariate regression model adjusted for SRM expression of the covariates Met, Egfr, Her3, and HER2 heterogeneity, as well as their interactions (cell lines r (2) = 0.9842; FFPE r (2) = 0.7643). In GEC tissues, Her2-SRM protein was detected at any level in 71.2 % of cases. ROC curves demonstrated that Her2-SRM protein levels have a high specificity (100 %) at an upper-level cutoff of >750 amol/µg and sensitivity of 75 % at a lower-level cutoff of <450 amol/μg for identifying HER2 FISH-amplified tumors. An "equivocal zone" of 450-750 amol/µg of Her2-SRM protein was analogous to IHC2+ but represented fewer cases (9-16 % of cases versus 36-41 %). CONCLUSIONS Compared to IHC, targeted SRM-Her2 proteomics provided more objective and quantitative Her2 expression with excellent HER2/CEP17 FISH correlation and fewer equivocal cases. Along with its multiplex capability for other relevant oncoproteins, these results demonstrate a refined HER2 protein expression assay for clinical application.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA.
| | - Wei-Li Liao
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Lei Zhao
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Emma Whitcomb
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Les Henderson
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Emily O'Day
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Peng Xu
- Department of Medicine, Section of Hematology/Oncology, University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Sheeno Thyparambil
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - David Krizman
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Kathleen Bengali
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | | | - Marlene Darfler
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Fabiola Cecchi
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Adele Blackler
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, Korea
| | - John Hart
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Shu-Yuan Xiao
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Sang Mee Lee
- Department of Public Health Studies, University of Chicago, Chicago, IL, USA
| | - Jon Burrows
- OncoPlex Diagnostics Inc., Rockville, MD, USA
| | - Todd Hembrough
- OncoPlex Diagnostics Inc., Rockville, MD, USA
- NantOmics, LLC, Culver City, CA, USA
| |
Collapse
|
36
|
Pyo JS, Kang G, Cho H. Clinicopathological Significance and Diagnostic Accuracy of c-MET Expression by Immunohistochemistry in Gastric Cancer: A Meta-Analysis. J Gastric Cancer 2016; 16:141-151. [PMID: 27752391 PMCID: PMC5065943 DOI: 10.5230/jgc.2016.16.3.141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/09/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022] Open
Abstract
PURPOSE The aim of the present study was to elucidate the clinicopathological significance and diagnostic accuracy of immunohistochemistry (IHC) for determining the mesenchymal epidermal transition (c-MET) expression in patients with gastric cancer (GC). MATERIALS AND METHODS The present meta-analysis investigated the correlation between c-MET expression as determined by IHC and the clinicopathological parameters in 8,395 GC patients from 37 studies that satisfied the eligibility criteria. In addition, a concordance analysis was performed between c-MET expression as determined by IHC and c-MET amplification, and the diagnostic test accuracy was reviewed. RESULTS The estimated rate of c-MET overexpression was 0.403 (95% confidence interval [CI], 0.327~0.484) and it was significantly correlated with male patients, poor differentiation, lymph node metastasis, higher TNM stage, and human epidermal growth factor receptor 2 (HER2) positivity in IHC analysis. There was a significant correlation between c-MET expression and worse overall survival rate (hazard ratio, 1.588; 95% CI, 1.266~1.992). The concordance rates between c-MET expression and c-MET amplification were 0.967 (95% CI, 0.916~0.987) and 0.270 (95% CI, 0.173~0.395) for cases with non-overexpressed and overexpressed c-MET, respectively. In the diagnostic test accuracy review, the pooled sensitivity and specificity were 0.56 (95% CI, 0.50~0.63) and 0.79 (95% CI, 0.77~0.81), respectively. CONCLUSIONS The c-MET overexpression as determined by IHC was significantly correlated with aggressive tumor behavior and positive IHC status for HER2 in patients with GC. In addition, the c-MET expression status could be useful in the screening of c-MET amplification in patients with GC.
Collapse
Affiliation(s)
- Jung-Soo Pyo
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Guhyun Kang
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Hyunjin Cho
- Department of Surgery, Inje University Sanggye Paik Hospital, Seoul, Korea
| |
Collapse
|
37
|
Riquelme I, Saavedra K, Espinoza JA, Weber H, García P, Nervi B, Garrido M, Corvalán AH, Roa JC, Bizama C. Molecular classification of gastric cancer: Towards a pathway-driven targeted therapy. Oncotarget 2016; 6:24750-79. [PMID: 26267324 PMCID: PMC4694793 DOI: 10.18632/oncotarget.4990] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 07/17/2015] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer mortality worldwide. Although surgical resection is a potentially curative approach for localized cases of GC, most cases of GC are diagnosed in an advanced, non-curable stage and the response to traditional chemotherapy is limited. Fortunately, recent advances in our understanding of the molecular mechanisms that mediate GC hold great promise for the development of more effective treatment strategies. In this review, an overview of the morphological classification, current treatment approaches, and molecular alterations that have been characterized for GC are provided. In particular, the most recent molecular classification of GC and alterations identified in relevant signaling pathways, including ErbB, VEGF, PI3K/AKT/mTOR, and HGF/MET signaling pathways, are described, as well as inhibitors of these pathways. An overview of the completed and active clinical trials related to these signaling pathways are also summarized. Finally, insights regarding emerging stem cell pathways are described, and may provide additional novel markers for the development of therapeutic agents against GC. The development of more effective agents and the identification of biomarkers that can be used for the diagnosis, prognosis, and individualized therapy for GC patients, have the potential to improve the efficacy, safety, and cost-effectiveness for GC treatments.
Collapse
Affiliation(s)
- Ismael Riquelme
- Department of Pathology, School of Medicine, Universidad de La Frontera, CEGIN-BIOREN, Temuco, Chile
| | - Kathleen Saavedra
- Department of Pathology, School of Medicine, Universidad de La Frontera, CEGIN-BIOREN, Temuco, Chile
| | - Jaime A Espinoza
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Helga Weber
- Department of Pathology, School of Medicine, Universidad de La Frontera, CEGIN-BIOREN, Temuco, Chile
| | - Patricia García
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bruno Nervi
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcelo Garrido
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H Corvalán
- UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology Oncology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan Carlos Roa
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDIS), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Bizama
- Department of Pathology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.,UC-Center for Investigational Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
38
|
Chakedis J, French R, Babicky M, Jaquish D, Howard H, Mose E, Lam R, Holman P, Miyamoto J, Walterscheid Z, Lowy AM. A novel protein isoform of the RON tyrosine kinase receptor transforms human pancreatic duct epithelial cells. Oncogene 2016; 35:3249-59. [PMID: 26477314 PMCID: PMC4837108 DOI: 10.1038/onc.2015.384] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 07/27/2015] [Accepted: 08/28/2015] [Indexed: 02/04/2023]
Abstract
The MST1R gene is overexpressed in pancreatic cancer producing elevated levels of the RON tyrosine kinase receptor protein. While mutations in MST1R are rare, alternative splice variants have been previously reported in epithelial cancers. We report the discovery of a novel RON isoform discovered in human pancreatic cancer. Partial splicing of exons 5 and 6 (P5P6) produces a RON isoform that lacks the first extracellular immunoglobulin-plexin-transcription domain. The splice variant is detected in 73% of xenografts derived from pancreatic adenocarcinoma patients and 71% of pancreatic cancer cell lines. Peptides specific to RON P5P6 detected in human pancreatic cancer specimens by mass spectrometry confirm translation of the protein isoform. The P5P6 isoform is found to be constitutively phosphorylated, present in the cytoplasm, and it traffics to the plasma membrane. Expression of P5P6 in immortalized human pancreatic duct epithelial (HPDE) cells activates downstream AKT, and in human pancreatic epithelial nestin-expressing cells, activates both the AKT and MAPK pathways. Inhibiting RON P5P6 in HPDE cells using a small molecule inhibitor BMS-777607 blocked constitutive activation and decreased AKT signaling. P5P6 transforms NIH3T3 cells and induces tumorigenicity in HPDE cells. Resultant HPDE-P5P6 tumors develop a dense stromal compartment similar to that seen in pancreatic cancer. In summary, we have identified a novel and constitutively active isoform of the RON tyrosine kinase receptor that has transforming activity and is expressed in human pancreatic cancer. These findings provide additional insight into the biology of the RON receptor in pancreatic cancer and are clinically relevant to the study of RON as a potential therapeutic target.
Collapse
Affiliation(s)
- J Chakedis
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - R French
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - M Babicky
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - D Jaquish
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - H Howard
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - E Mose
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - R Lam
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - P Holman
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - J Miyamoto
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Z Walterscheid
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - A M Lowy
- Department of Surgery, Division of Surgical Oncology, Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
39
|
Mereiter S, Balmaña M, Gomes J, Magalhães A, Reis CA. Glycomic Approaches for the Discovery of Targets in Gastrointestinal Cancer. Front Oncol 2016; 6:55. [PMID: 27014630 PMCID: PMC4783390 DOI: 10.3389/fonc.2016.00055] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 02/24/2016] [Indexed: 12/22/2022] Open
Abstract
Gastrointestinal (GI) cancer is the most common group of malignancies and many of its types are among the most deadly. Various glycoconjugates have been used in clinical practice as serum biomarker for several GI tumors, however, with limited diagnose application. Despite the good accessibility by endoscopy of many GI organs, the lack of reliable serum biomarkers often leads to late diagnosis of malignancy and consequently low 5-year survival rates. Recent advances in analytical techniques have provided novel glycoproteomic and glycomic data and generated functional information and putative biomarker targets in oncology. Glycosylation alterations have been demonstrated in a series of glycoconjugates (glycoproteins, proteoglycans, and glycosphingolipids) that are involved in cancer cell adhesion, signaling, invasion, and metastasis formation. In this review, we present an overview on the major glycosylation alterations in GI cancer and the current serological biomarkers used in the clinical oncology setting. We further describe recent glycomic studies in GI cancer, namely gastric, colorectal, and pancreatic cancer. Moreover, we discuss the role of glycosylation as a modulator of the function of several key players in cancer cell biology. Finally, we address several state-of-the-art techniques currently applied in this field, such as glycomic and glycoproteomic analyses, the application of glycoengineered cell line models, microarray and proximity ligation assay, and imaging mass spectrometry, and provide an outlook to future perspectives and clinical applications.
Collapse
Affiliation(s)
- Stefan Mereiter
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal
| | - Meritxell Balmaña
- Biochemistry and Molecular Biology Unit, Department of Biology, University of Girona , Girona , Spain
| | - Joana Gomes
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (I3S), University of Porto, Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Porto, Portugal; Medical Faculty, University of Porto, Porto, Portugal
| |
Collapse
|
40
|
Mereiter S, Magalhães A, Adamczyk B, Jin C, Almeida A, Drici L, Ibáñez-Vea M, Gomes C, Ferreira JA, Afonso LP, Santos LL, Larsen MR, Kolarich D, Karlsson NG, Reis CA. Glycomic analysis of gastric carcinoma cells discloses glycans as modulators of RON receptor tyrosine kinase activation in cancer. Biochim Biophys Acta Gen Subj 2015; 1860:1795-808. [PMID: 26721331 DOI: 10.1016/j.bbagen.2015.12.016] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 01/16/2023]
Abstract
BACKGROUND Terminal α2-3 and α2-6 sialylation of glycans precludes further chain elongation, leading to the biosynthesis of cancer relevant epitopes such as sialyl-Lewis X (SLe(X)). SLe(X) overexpression is associated with tumor aggressive phenotype and patients' poor prognosis. METHODS MKN45 gastric carcinoma cells transfected with the sialyltransferase ST3GAL4 were established as a model overexpressing sialylated terminal glycans. We have evaluated at the structural level the glycome and the sialoproteome of this gastric cancer cell line applying liquid chromatography and mass spectrometry. We further validated an identified target expression by proximity ligation assay in gastric tumors. RESULTS Our results showed that ST3GAL4 overexpression leads to several glycosylation alterations, including reduced O-glycan extension and decreased bisected and increased branched N-glycans. A shift from α2-6 towards α2-3 linked sialylated N-glycans was also observed. Sialoproteomic analysis further identified 47 proteins with significantly increased sialylated N-glycans. These included integrins, insulin receptor, carcinoembryonic antigens and RON receptor tyrosine kinase, which are proteins known to be key players in malignancy. Further analysis of RON confirmed its modification with SLe(X) and the concomitant activation. SLe(X) and RON co-expression was validated in gastric tumors. CONCLUSION The overexpression of ST3GAL4 interferes with the overall glycophenotype of cancer cells affecting a multitude of key proteins involved in malignancy. Aberrant glycosylation of the RON receptor was shown as an alternative mechanism of oncogenic activation. GENERAL SIGNIFICANCE This study provides novel targets and points to an integrative tumor glycomic/proteomic-profiling for gastric cancer patients' stratification. This article is part of a Special Issue entitled "Glycans in personalised medicine" Guest Editor: Professor Gordan Lauc.
Collapse
Affiliation(s)
- Stefan Mereiter
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Portugal
| | - Ana Magalhães
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
| | - Barbara Adamczyk
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Chunsheng Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Andreia Almeida
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany; Free University Berlin, Berlin, Germany
| | - Lylia Drici
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Maria Ibáñez-Vea
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Catarina Gomes
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal
| | - José A Ferreira
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology of Porto, Portugal
| | - Luis P Afonso
- Department of Pathology, Portuguese Institute of Oncology of Porto, Portugal
| | - Lúcio L Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology of Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology of Porto, Portugal
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Niclas G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Celso A Reis
- I3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto - IPATIMUP, Porto, Portugal; Institute of Biomedical Sciences of Abel Salazar - ICBAS, University of Porto, Portugal; Medical Faculty, University of Porto, Portugal.
| |
Collapse
|
41
|
Kanteti R, Dhanasingh I, El-Hashani E, Riehm JJ, Stricker T, Nagy S, Zaborin A, Zaborina O, Biron D, Alverdy JC, Im HK, Siddiqui S, Padilla PA, Salgia R. C. elegans and mutants with chronic nicotine exposure as a novel model of cancer phenotype. Cancer Biol Ther 2015; 17:91-103. [PMID: 26574927 PMCID: PMC6093410 DOI: 10.1080/15384047.2015.1108495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
We previously investigated MET and its oncogenic mutants relevant to lung cancer
in C. elegans. The inactive orthlogues of the receptor tyrosine
kinase Eph and MET, namely vab-1 and RB2088 respectively, the
temperature sensitive constitutively active form of KRAS, SD551
(let-60; GA89) and the inactive c-CBL equivalent mutants in
sli-1 (PS2728, PS1258, and MT13032) when subjected to
chronic exposure of nicotine resulted in a significant loss in egg-laying
capacity and fertility. While the vab-1 mutant revealed
increased circular motion in response to nicotine, the other mutant strains
failed to show any effect. Overall locomotion speed increased with increasing
nicotine concentration in all tested mutant strains except in the
vab-1 mutants. Moreover, chronic nicotine exposure, in
general, upregulated kinases and phosphatases. Taken together, these studies
provide evidence in support of C. elegans as initial in
vivo model to study nicotine and its effects on oncogenic mutations
identified in humans.
Collapse
Affiliation(s)
- Rajani Kanteti
- a Department of Medicine , Section of Hematology/Oncology, University of Chicago , Chicago , IL , USA
| | - Immanuel Dhanasingh
- a Department of Medicine , Section of Hematology/Oncology, University of Chicago , Chicago , IL , USA
| | | | - Jacob J Riehm
- a Department of Medicine , Section of Hematology/Oncology, University of Chicago , Chicago , IL , USA
| | - Thomas Stricker
- c Department of Pathology , Microbiology and Immunology, Vanderbilt University School of Medicine , Nashville , TN , USA
| | - Stanislav Nagy
- d Department of Physics , James Franck Institute, and the College, University of Chicago , Chicago , IL , USA
| | - Alexander Zaborin
- e Department of Surgery , Pritzker School of Medicine, University of Chicago , Chicago , IL , USA
| | - Olga Zaborina
- e Department of Surgery , Pritzker School of Medicine, University of Chicago , Chicago , IL , USA
| | - David Biron
- d Department of Physics , James Franck Institute, and the College, University of Chicago , Chicago , IL , USA
| | - John C Alverdy
- e Department of Surgery , Pritzker School of Medicine, University of Chicago , Chicago , IL , USA
| | - Hae Kyung Im
- f Department of Medicine , Section of Genetic Medicine, University of Chicago , Chicago , IL , USA
| | - Shahid Siddiqui
- g Department of Medicine , University of Chicago, Chicago, IL and Department of Basic and Oral Biology, UQUDENT, U. Q. University , Makkah , KSA
| | - Pamela A Padilla
- h Department of Biological Sciences , University of North- Texas , Denton , TX , USA
| | - Ravi Salgia
- a Department of Medicine , Section of Hematology/Oncology, University of Chicago , Chicago , IL , USA
| |
Collapse
|
42
|
Inokuchi M, Otsuki S, Fujimori Y, Sato Y, Nakagawa M, Kojima K. Clinical significance of MET in gastric cancer. World J Gastrointest Oncol 2015; 7:317-327. [PMID: 26600931 PMCID: PMC4644854 DOI: 10.4251/wjgo.v7.i11.317] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 04/06/2015] [Accepted: 08/28/2015] [Indexed: 02/05/2023] Open
Abstract
Chemotherapy has become the global standard treatment for patients with metastatic or unresectable gastric cancer (GC), although outcomes remain unfavorable. Many molecular-targeted therapies inhibiting signaling pathways of various tyrosine kinase receptors have been developed, and monoclonal antibodies targeting human epidermal growth factor receptor 2 or vascular endothelial growth factor receptor 2 have become standard therapy for GC. Hepatocyte growth factor and its receptor, c-MET (MET), play key roles in tumor growth through activated signaling pathways from receptor in GC cells. Genomic amplification of MET leads to the aberrant activation found in GC tumors and is related to survival in patients with GC. This review discusses the clinical significance of MET in GC and examines MET as a potential therapeutic target in patients with GC. Preclinical studies in animal models have shown that MET antibodies or small-molecule MET inhibitors suppress tumor-cell proliferation and tumor progression in MET-amplified GC cells. These drugs are now being evaluated in clinical trials as treatments for metastatic or unresectable GC.
Collapse
|
43
|
Lin XL, Xiao XY. Histological and molecular classification of gastric cancer and personalized therapy. Shijie Huaren Xiaohua Zazhi 2015; 23:4141-4149. [DOI: 10.11569/wcjd.v23.i26.4141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is one of the most common malignancies of the digestive system, and the majority of gastric cancer patients have already had advanced disease at diagnosis. Although rapid progress has been made in medical diagnostic equipment, surgical techniques, chemotherapy drugs and targeted therapy, the prognosis of advanced gastric cancer is still poor. Gastric cancer still has the second highest mortality rate among all cancers and is highly heterogeneous in terms of biology and genetics. The existing pathological typing system for gastric cancer cannot well evaluate prognosis and guide treatment. This paper aims to analyze and discuss the relationship between different histological and molecular classification methods and susceptibility to drugs in order to seek a suitable pathological and molecular typing method to guide the personalized therapy of this malignancy.
Collapse
|
44
|
Metzger ML, Behrens HM, Böger C, Haag J, Krüger S, Röcken C. MET in gastric cancer--discarding a 10% cutoff rule. Histopathology 2015; 68:241-53. [PMID: 26033401 PMCID: PMC4744765 DOI: 10.1111/his.12745] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/26/2015] [Indexed: 12/20/2022]
Abstract
Aims We aimed to develop a putative predictive biomarker score for future hepatocyte growth factor receptor (MET)‐targeted therapy of gastric cancer (GC). Methods and results MET expression and MET amplification were analysed by immunohistochemistry (IHC) and chromogenic in‐situ hybridization (CISH) in 470 GC patients. Immunostaining was documented with the HistoScore. The percentage area of MET‐amplified tumour cell clones was assessed by virtual microscopy. The expression of MET was heterogeneous in primary and metastatic GC. Immunostaining intensity (MET‐IHC 2+/3+) correlated with MET amplification and a positive MET status was defined by a combination of MET‐IHC 2+ or 3+ with MET amplification, or MET‐IHC 3+ without MET amplification. The prognostic significance of the MET status was independent from the percentage area of positive tumour cells (e.g. <10 versus ≥10%). MET‐positive GCs were microsatellite stable and of KRAS/PIK3CA wild‐type. MET‐positive GCs had a very poor prognosis, with a median survival of 5.4 months and a hazard ratio of 2.126. Conclusions A combination of immunohistochemistry and CISH is suitable to assess MET status. If MET status is used as a predictive biomarker, prospective studies should pay specific attention to adequate tissue sampling, should ignore cutoff values for tumour areas, may consider the KRAS and PIK3CA genotype as negative predictive markers and should carry out the analysis expeditiously.
Collapse
Affiliation(s)
| | | | - Christine Böger
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Jochen Haag
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Sandra Krüger
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|
45
|
Batth IS, Yun H, Kumar AP. Recepteur d'origine nantais (RON), more than a kinase: Role in castrate-resistant prostate cancer. Mol Carcinog 2015; 54:937-46. [PMID: 26152593 DOI: 10.1002/mc.22354] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/20/2015] [Accepted: 05/28/2015] [Indexed: 12/30/2022]
Abstract
Prostate cancer (PCA) is the second leading cause of cancer-related deaths in men in the United States. It is natural for a hormone-driven malignancy such as prostate cancer that androgen deprivation therapy (ADT) would be the preferred treatment for clinical disease management. However, after initial treatment response a vast majority of patients develop metastatic castrate-resistant prostate cancer (CRPC), which is fatal. While great headway has been made to understand the possible mechanisms that drive castrate-resistant disease, a bonafide cure remains elusive. Reactivation of androgen receptor (AR) signaling partly contributes to the emergence of CRPC. Here we briefly examine some of the known mechanisms of AR reactivation including intratumoral synthesis of androgens, modulation of AR coregulators, and AR variants with constitutive activity as well as activation of receptor tyrosine kinases. We primarily focus on the emerging dual function of the receptor tyrosine kinase (recepteur d'origine nantais; RON) as a traditional tyrosine kinase and transcription factor. We further discuss activation of RON as an alternate mechanism in the development of CRPC and available therapeutic approaches for clinical management of CRPC by combined inhibition of RON and AR.
Collapse
Affiliation(s)
- Izhar Singh Batth
- Department of Urology, University of Texas Health Science Center, San Antonio, Texas
| | - Huiyoung Yun
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas
| | - Addankl P Kumar
- Department of Urology, University of Texas Health Science Center, San Antonio, Texas.,Department of Pharmacology, University of Texas Health Science Center, San Antonio, Texas.,Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas.,Cancer Therapy and Research Center, University of Texas Health Science Center, San Antonio, Texas.,South Texas Veterans Health Care System, San Antonio, Texas
| |
Collapse
|
46
|
Catenacci DVT. Next-generation clinical trials: Novel strategies to address the challenge of tumor molecular heterogeneity. Mol Oncol 2015; 9:967-96. [PMID: 25557400 PMCID: PMC4402102 DOI: 10.1016/j.molonc.2014.09.011] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Revised: 09/23/2014] [Accepted: 09/26/2014] [Indexed: 02/09/2023] Open
Abstract
The promise of 'personalized cancer care' with therapies toward specific molecular aberrations has potential to improve outcomes. However, there is recognized heterogeneity within any given tumor-type from patient to patient (inter-patient heterogeneity), and within an individual (intra-patient heterogeneity) as demonstrated by molecular evolution through space (primary tumor to metastasis) and time (after therapy). These issues have become hurdles to advancing cancer treatment outcomes with novel molecularly targeted agents. Classic trial design paradigms are challenged by heterogeneity, as they are unable to test targeted therapeutics against low frequency genomic 'oncogenic driver' aberrations with adequate power. Usual accrual difficulties to clinical trials are exacerbated by low frequencies of any given molecular driver. To address these challenges, there is need for innovative clinical trial designs and strategies implementing novel diagnostic biomarker technologies to account for inter-patient molecular diversity and scarce tissue for analysis. Importantly, there is also need for pre-defined treatment priority algorithms given numerous aberrations commonly observed within any one individual sample. Access to multiple available therapeutic agents simultaneously is crucial. Finally intra-patient heterogeneity through time may be addressed by serial biomarker assessment at the time of tumor progression. This report discusses various 'next-generation' biomarker-driven trial designs and their potentials and limitations to tackle these recognized molecular heterogeneity challenges. Regulatory hurdles, with respect to drug and companion diagnostic development and approval, are considered. Focus is on the 'Expansion Platform Design Types I and II', the latter demonstrated with a first example, 'PANGEA: Personalized Anti-Neoplastics for Gastro-Esophageal Adenocarcinoma'. Applying integral medium-throughput genomic and proteomic assays along with a practical biomarker assessment and treatment algorithm, 'PANGEA' attempts to address the problem of heterogeneity towards successful implementation of molecularly targeted therapies.
Collapse
Affiliation(s)
- Daniel V T Catenacci
- University of Chicago Medical Center, Department of Medicine, Section of Hematology & Oncology, 5841 S. Maryland Avenue, MC2115, Chicago, IL 60637, USA.
| |
Collapse
|
47
|
MET is a predictive factor for late recurrence but not for overall survival of early stage hepatocellular carcinoma. Tumour Biol 2015; 36:4993-5000. [PMID: 25874493 DOI: 10.1007/s13277-015-3150-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/26/2015] [Indexed: 01/09/2023] Open
Abstract
The MET and RON receptors are tyrosine kinases that form a non-covalent complex on the cell surface that functions in several steps of tumor progression. The purpose of this study was to determine the clinical significance of MET and RON expression on long-term survival and recurrence after curative resection in a large cohort of hepatocellular carcinoma (HCC) patients. We performed immunohistochemical analyses on microarrays of the tumors using antibodies against MET and RON. We evaluated the prognostic value of biomarker expression using Cox regression and the Kaplan-Meier method in 490 HCC patients. MET-positive patients had higher overall recurrence rates than MET-negative patients (P = 0.041); however, MET positivity was not associated with overall survival (OS) (P = .249). RON was not associated with overall recurrence rates and OS. MET was independently associated with late but not early phase recurrence. Particularly, the prognostic significance of MET is limited in early stage disease. MET+/RON+ patients had higher overall recurrence rates than those with the other expression patterns (P = 0.071), although the result did not reach statistical significance. Immunohistological activation of MET expression has no prognostic significance for OS in patients with HCC. However, MET positivity was correlated with late recurrence after HCC resection in early stage disease.
Collapse
|
48
|
Zhou DH, Li C, Yang LN. Variant RONΔ160 of the RON receptor tyrosine kinase promotes the growth and invasion in vitro and in vivo in gastric cancer cell lines. Cancer Cell Int 2015; 15:9. [PMID: 25685065 PMCID: PMC4326440 DOI: 10.1186/s12935-015-0157-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/05/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recepteur d'origine nantais (RON) is a receptor tyrosine kinase whose overexpression has been observed in human gastric cancers. This study aimed to determine whether overexpression of the variant RONΔ160 could induce tumorigenicity of gastric cancer cells in vitro or in vivo, and whether its specific small molecule inhibitor (Compound I) could inhibit the effect of RONΔ160. METHODS We constructed human gastric cancer cell line MGC-803 that was stably transfected with a recombinant plasmid expressing RONΔ160, and the effect of RONΔ160 overexpression and macrophage-stimulating protein (MSP) activation on proliferation, migration and invasion abilities of MGC-803 cells were evaluated. Tumor-bearing mice with gastric cancer cells were used to analyze the effects of RONΔ160 overexpression and Compound I on implanted tumor growth. RESULTS In vitro, overexpression of RONΔ160 in MGC-803 cells resulted changes to their cell morphology, and promoted cell proliferation, migration and invasion. In addition, overexpression of RONΔ160 increased the proportion of cells in the S phase. The effect of RONΔ160 was significantly enhanced by induction of MSP inducing (p < 0.05). In vivo, RONΔ160 promoted the growth of MGC-803 cells in nude mice, including increased tumor size and weight, and lower tumor incubation period. The Compound I inhibited the tumorigenic abilities of RONΔ160 (p <0.05). CONCLUSIONS The results indicate that overexpression of the variant RONΔ160 altered the phenotype and tumorigenicity of MGC-803 cells. Its specific small molecule inhibitor could inhibit the effect of RONΔ160. Therefore, the variant RONΔ160 may become a potential therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Dong-Hui Zhou
- Department of Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China
| | - Chao Li
- Department of Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003 China
| | - Li-Na Yang
- Department of Oncology, Dongnan Affiliated Hospital of Xiamen University, Zhangzhou, China
| |
Collapse
|
49
|
Hack SP, Bruey JM, Koeppen H. HGF/MET-directed therapeutics in gastroesophageal cancer: a review of clinical and biomarker development. Oncotarget 2015; 5:2866-80. [PMID: 24930887 PMCID: PMC4102777 DOI: 10.18632/oncotarget.2003] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aberrant activation of the HGF/MET signaling axis has been strongly implicated in the malignant transformation and progression of gastroesophageal cancer (GEC). MET receptor overexpression in tumor samples from GEC patients has been consistently correlated with an aggressive metastatic phenotype and poor prognosis. In preclinical GEC models, abrogation of HGF/MET signaling has been shown to induce tumor regression as well as inhibition of metastatic dissemination. Promising clinical results in patient subsets in which MET is overexpressed have spurned several randomized studies of HGF/MET-directed agents, including two pivotal global Phase III trials. Available data highlight the need for predictive biomarkers in order to select patients most likely to benefit from HGF/MET inhibition. In this review, we discuss the current knowledge of mechanisms of MET activation in GEC, the current status of the clinical evaluation of MET-targeted therapies in GEC, characteristics of ongoing randomized GEC trials and the associated efforts to identify and validate biomarkers. We also discuss the considerations and challenges for HGF/MET inhibitor drug development in the GEC setting.
Collapse
Affiliation(s)
- Stephen P Hack
- Product Development, Genentech Inc., South San Francisco, CA, USA
| | | | | |
Collapse
|
50
|
Noguchi E, Saito N, Kobayashi M, Kameoka S. Clinical significance of hepatocyte growth factor/c-Met expression in the assessment of gastric cancer progression. Mol Med Rep 2015; 11:3423-31. [PMID: 25592281 PMCID: PMC4368069 DOI: 10.3892/mmr.2015.3205] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 12/12/2014] [Indexed: 12/12/2022] Open
Abstract
Among the mechanisms that control cancer progression, cell mobility is a significant factor required for cellular liberation from the primary focus and infiltration. Hepatocyte growth factor (HGF) has been shown to facilitate cell mobility. In the present study, the clinical significance of the HGF/c-Met pathway in the assessment of gastric cancer progression was evaluated. From a cohort of patients with gastric cancer who underwent surgical resection between April 1999 and March 2003, 110 subjects were randomly selected. Preoperative serum HGF levels were measured and various pathological factors were analyzed. Furthermore, 50 subjects were randomly selected from within this group and immunohistochemical staining of tissue preparations for HGF and its receptor c-Met were performed. In the infiltrative growth pattern [(INF)α,β vs. INFγ], advanced progression was associated with elevated preoperative serum HGF levels (P<0.001). No correlation was identified between serum HGF levels and immunostaining for HGF or c-Met in the tissue preparations. Immunostaining revealed a significant correlation between c-Met expression and lymphatic vessel invasion (ly0.1 vs. 2.3; P=0.0416), lymph node metastasis (n0.1 vs. 2; P=0.0184) and maximum tumor diameter (≤50 mm vs. >50 mm; P=0.0469). Furthermore, c-Met-positivity was associated with a significant difference in overall survival (P=0.0342), despite stage I and II cases accounting for 82% of the total cohort (41 of 50 cases). These results suggested that the expression of the HGF/c-Met pathway in gastric cancer may be a potential predictive factor for disease progression.
Collapse
Affiliation(s)
- Eiichiro Noguchi
- Department of Surgery II, Tokyo Women's Medical University, Tokyo 162‑8666, Japan
| | - Noboru Saito
- Department of Surgery II, Tokyo Women's Medical University, Tokyo 162‑8666, Japan
| | - Makio Kobayashi
- Department of Pathology I, Tokyo Women's Medical University, Tokyo 162‑8666, Japan
| | - Shingo Kameoka
- Department of Surgery II, Tokyo Women's Medical University, Tokyo 162‑8666, Japan
| |
Collapse
|