1
|
Hanada K, Kawada K, Obama K. Targeting Asparagine Metabolism in Solid Tumors. Nutrients 2025; 17:179. [PMID: 39796613 PMCID: PMC11722615 DOI: 10.3390/nu17010179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025] Open
Abstract
Reprogramming of energy metabolism to support cellular growth is a "hallmark" of cancer, allowing cancer cells to balance the catabolic demands with the anabolic needs of producing the nucleotides, amino acids, and lipids necessary for tumor growth. Metabolic alterations, or "addiction", are promising therapeutic targets and the focus of many drug discovery programs. Asparagine metabolism has gained much attention in recent years as a novel target for cancer therapy. Asparagine is widely used in the production of other nutrients and plays an important role in cancer development. Nutritional inhibition therapy targeting asparagine has been used as an anticancer strategy and has shown success in the treatment of leukemia. However, in solid tumors, asparagine restriction alone does not provide ideal therapeutic efficacy. Tumor cells initiate reprogramming processes in response to asparagine deprivation. This review provides a comprehensive overview of asparagine metabolism in cancers. We highlight the physiological role of asparagine and current advances in improving survival and overcoming therapeutic resistance.
Collapse
Affiliation(s)
- Keita Hanada
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (K.H.); (K.O.)
- Department of Surgery, Rakuwakai Otowa Hospital, Kyoto 607-8062, Japan
| | - Kenji Kawada
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (K.H.); (K.O.)
- Department of General Surgery, Kurashiki Central Hospital, Kurashiki 710-8602, Japan
| | - Kazutaka Obama
- Department of Gastrointestinal Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; (K.H.); (K.O.)
| |
Collapse
|
2
|
Jiao Y, Peng X, Wang Y, Hao Z, Chen L, Wu M, Zhang Y, Li J, Li W, Zhan X. Malignant ascites supernatant enhances the proliferation of gastric cancer cells partially via the upregulation of asparagine synthetase. Oncol Lett 2023; 26:418. [PMID: 37664666 PMCID: PMC10472050 DOI: 10.3892/ol.2023.14005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/09/2023] [Indexed: 09/05/2023] Open
Abstract
Malignant ascites (MA) is a common manifestation of advanced gastric cancer (GC) with peritoneal metastasis (PM), which usually indicates a poor prognosis. The present study aimed to explore the effects of MA, a unique microenvironment of PM, on the proliferation of cancer cells and investigate the underlying mechanisms. Ex vivo experiments demonstrated that GC cells treated with MA exhibited enhanced proliferation. RNA sequencing indicated that asparagine synthetase (ASNS) was one of the differentially expressed genes in GC cells following incubation with MAs. Furthermore, the present study suggested that MA induced an upregulation of ASNS expression and the stimulatory effect of MA on cancer cell proliferation was alleviated upon ASNS downregulation. Activating transcription factor 4 (ATF4), a pivotal transcription factor regulating ASNS, was upregulated when cells were treated with MA supernatant. After ATF4 knockdown, the proliferation of MA-treated GC cells and the expression of ASNS decreased. In addition, the decline in the proliferation of the ATF4-downregulated AGS GC cell line was rescued by ASNS upregulation. The findings indicated that MA could promote the proliferation of GC cells via activation of the ATF4-ASNS axis. Hence, it may be a potential target for treating GC with PM and MA.
Collapse
Affiliation(s)
- Yuan Jiao
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yujie Wang
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Zhibin Hao
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Ling Chen
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Meihong Wu
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yingyi Zhang
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Jie Li
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Wenlin Li
- Department of Cell Biology, Naval Medical University, Shanghai 200433, P.R. China
| | - Xianbao Zhan
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
3
|
Tuna M, Ju Z, Yoshihara K, Amos CI, Tanyi JL, Mills GB. Clinical relevance of TP53 hotspot mutations in high-grade serous ovarian cancers. Br J Cancer 2020; 122:405-412. [PMID: 31780779 PMCID: PMC7000721 DOI: 10.1038/s41416-019-0654-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/28/2019] [Accepted: 11/06/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Mutation of TP53 is the most frequent genetic alteration in high-grade serous ovarian cancer (HGSOC). The impact of hotspot mutations of TP53 and protein levels on patient outcomes in HGSOC has not been fully elucidated. METHODS The study population (n = 791) comprised of HGSOC samples with TP53 mutation from TCGA and other publicly available data. Univariate and multivariate cox proportional hazards regression analyses were used to select variables that were correlated with patient survival. RESULTS We assessed the effects of TP53 mutations based on type and individual hotspot mutations on patient outcomes in HGSOC. Only hotspot mutations were associated with outcomes. Three hotspot mutations: G266, Y163C, and R282, in aggregate were associated with a worsened overall and recurrence-free survival compared with other hotspot mutations (p < 0.0001 and p = 0.001), other non-hotspot missense mutations (p < 0.0001 and p = 0.008), truncated mutations (p < 0.0001 and p = 0.001), and all other mutations (p < 0.0001 and p = 0.001). Specific hotspot mutations were associated with different protein expression patterns consistent with different functions. CONCLUSIONS This study provides evidence that individual TP53 hotspot mutations have different impact on HGSOC patient outcomes and potentially TP53 function. Thus the status of particular TP53 aberrations could influence response to therapy and selection of therapeutic agents.
Collapse
Affiliation(s)
- Musaffe Tuna
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
- Precision Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kosuke Yoshihara
- Department of Obstetrics and Gynecology, Niigata University, Niigata, Japan
| | | | - Janos L Tanyi
- Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Gordon B Mills
- Precision Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
4
|
Chiu M, Taurino G, Bianchi MG, Kilberg MS, Bussolati O. Asparagine Synthetase in Cancer: Beyond Acute Lymphoblastic Leukemia. Front Oncol 2020; 9:1480. [PMID: 31998641 PMCID: PMC6962308 DOI: 10.3389/fonc.2019.01480] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/10/2019] [Indexed: 12/12/2022] Open
Abstract
Asparagine Synthetase (ASNS) catalyzes the synthesis of the non-essential amino acid asparagine (Asn) from aspartate (Asp) and glutamine (Gln). ASNS expression is highly regulated at the transcriptional level, being induced by both the Amino Acid Response (AAR) and the Unfolded Protein Response (UPR) pathways. Lack of ASNS protein expression is a hallmark of Acute Lymphoblastic Leukemia (ALL) blasts, which, therefore, are auxotrophic for Asn. This peculiarity is the rationale for the use of bacterial L-Asparaginase (ASNase) for ALL therapy, the first example of anti-cancer treatment targeting a tumor-specific metabolic feature. Other hematological and solid cancers express low levels of ASNS and, therefore, should also be Asn auxotrophs and ASNase sensitive. Conversely, in the last few years, several reports indicate that in some cancer types ASNS is overexpressed, promoting cell proliferation, chemoresistance, and a metastatic behavior. However, enhanced ASNS activity may constitute a metabolic vulnerability in selected cancer models, suggesting a variable and tumor-specific role of the enzyme in cancer. Recent evidence indicates that, beyond its canonical role in protein synthesis, Asn may have additional regulatory functions. These observations prompt a re-appreciation of ASNS activity in the biology of normal and cancer tissues, with particular attention to the fueling of Asn exchange between cancer cells and the tumor microenvironment.
Collapse
Affiliation(s)
- Martina Chiu
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Taurino
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Massimiliano G. Bianchi
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Michael S. Kilberg
- Department of Biochemistry and Molecular Biology, University of Florida College of Medicine, Gainesville, FL, United States
| | - Ovidio Bussolati
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
5
|
Kenawy N, Kalirai H, Sacco JJ, Lake SL, Heegaard S, Larsen AC, Finger PT, Milman T, Chin K, Mosci C, Lanza F, Moulin A, Schmitt CA, Caujolle JP, Maschi C, Marinkovic M, Taktak AF, Heimann H, Damato BE, Coupland SE. Conjunctival melanoma copy number alterations and correlation with mutation status, tumor features, and clinical outcome. Pigment Cell Melanoma Res 2019; 32:564-575. [PMID: 30672666 PMCID: PMC6849808 DOI: 10.1111/pcmr.12767] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/09/2019] [Accepted: 01/17/2019] [Indexed: 01/03/2023]
Abstract
Relatively little is known about the genetic aberrations of conjunctival melanomas (CoM) and their correlation with clinical and histomorphological features as well as prognosis. The aim of this large collaborative multicenter study was to determine potential key biomarkers for metastatic risk and any druggable targets for high metastatic risk CoM. Using Affymetrix single nucleotide polymorphism genotyping arrays on 59 CoM, we detected frequent amplifications on chromosome (chr) 6p and deletions on 7q, and characterized mutation‐specific copy number alterations. Deletions on chr 10q11.21‐26.2, a region harboring the tumor suppressor genes, PDCD4, SUFU, NEURL1, PTEN, RASSF4, DMBT1, and C10orf90 and C10orf99, significantly correlated with metastasis (Fisher's exact, p ≤ 0.04), lymphatic invasion (Fisher's exact, p ≤ 0.02), increasing tumor thickness (Mann–Whitney, p ≤ 0.02), and BRAF mutation (Fisher's exact, p ≤ 0.05). This enhanced insight into CoM biology is a step toward identifying patients at risk of metastasis and potential therapeutic targets for systemic disease.
Collapse
Affiliation(s)
- Nihal Kenawy
- Liverpool Ocular Oncology Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.,Aintree University Hospital, Liverpool, UK
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Joseph J Sacco
- Liverpool Ocular Oncology Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.,Clatterbridge Cancer Centre, Wirral, UK
| | - Sarah L Lake
- Liverpool Ocular Oncology Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Steffen Heegaard
- Eye Pathology Section, Department of Pathology and Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Cathrine Larsen
- Eye Pathology Section, Department of Pathology and Department of Ophthalmology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | - Carlo Mosci
- Ocular Oncology Service, Galliera Hospital, Genoa, Italy
| | | | - Alexandre Moulin
- Ophthalmic Pathology Laboratory and Department of Ophthalmology, Jules Gonin Eye Hospital, Lausanne, Switzerland
| | | | | | - Célia Maschi
- Ophthalmology Department, University Hospital of Nice, Nice, France
| | - Marina Marinkovic
- Ophthalmology Department, Leiden University Medical Centre, Leiden, The Netherlands
| | - Azzam F Taktak
- Liverpool Ocular Oncology Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.,Department of Medical Physics and Clinical Engineering, Royal Liverpool University Hospital, Liverpool, UK
| | - Heinrich Heimann
- Liverpool Ocular Oncology Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.,Liverpool Ocular Oncology Centre, Royal Liverpool University Hospital, Liverpool, UK
| | - Bertil E Damato
- Oxford Eye Hospital and Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Sarah E Coupland
- Liverpool Ocular Oncology Research Group, Institute of Translational Medicine, University of Liverpool, Liverpool, UK.,Cellular Pathology, Liverpool Clinical Laboratories, Royal Liverpool University Hospital, Liverpool, UK
| |
Collapse
|
6
|
Poynter L, Galea D, Veselkov K, Mirnezami A, Kinross J, Nicholson J, Takáts Z, Darzi A, Mirnezami R. Network Mapping of Molecular Biomarkers Influencing Radiation Response in Rectal Cancer. Clin Colorectal Cancer 2019; 18:e210-e222. [PMID: 30928329 DOI: 10.1016/j.clcc.2019.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/12/2018] [Accepted: 01/23/2019] [Indexed: 02/07/2023]
Abstract
Preoperative radiotherapy (RT) plays an important role in the management of locally advanced rectal cancer (RC). Tumor regression after RT shows marked variability, and robust molecular methods are needed to help predict likely response. The aim of this study was to review the current published literature and use Gene Ontology (GO) analysis to define key molecular biomarkers governing radiation response in RC. A systematic review of electronic bibliographic databases (Medline, Embase) was performed for original articles published between 2000 and 2015. Biomarkers were then classified according to biological function and incorporated into a hierarchical GO tree. Both significant and nonsignificant results were included in the analysis. Significance was binarized on the basis of univariate and multivariate statistics. Significance scores were calculated for each biological domain (or node), and a direct acyclic graph was generated for intuitive mapping of biological pathways and markers involved in RC radiation response. Seventy-two individual biomarkers across 74 studies were identified. On highest-order classification, molecular biomarkers falling within the domains of response to stress, cellular metabolism, and pathways inhibiting apoptosis were found to be the most influential in predicting radiosensitivity. Homogenizing biomarker data from original articles using controlled GO terminology demonstrated that cellular mechanisms of response to RT in RC-in particular the metabolic response to RT-may hold promise in developing radiotherapeutic biomarkers to help predict, and in the future modulate, radiation response.
Collapse
Affiliation(s)
- Liam Poynter
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Dieter Galea
- Computational & Systems Medicine, Imperial College London, London, UK
| | - Kirill Veselkov
- Computational & Systems Medicine, Imperial College London, London, UK
| | | | - James Kinross
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Jeremy Nicholson
- Computational & Systems Medicine, Imperial College London, London, UK
| | - Zoltán Takáts
- Computational & Systems Medicine, Imperial College London, London, UK
| | - Ara Darzi
- Department of Surgery & Cancer, Imperial College London, London, UK
| | - Reza Mirnezami
- Department of Surgery & Cancer, Imperial College London, London, UK; St Mark's Hospital and Academic Institute, Harrow, London, UK.
| |
Collapse
|
7
|
Zhong Q, Fang J, Huang Z, Yang Y, Lian M, Liu H, Zhang Y, Ye J, Hui X, Wang Y, Ying Y, Zhang Q, Cheng Y. A response prediction model for taxane, cisplatin, and 5-fluorouracil chemotherapy in hypopharyngeal carcinoma. Sci Rep 2018; 8:12675. [PMID: 30139993 PMCID: PMC6107664 DOI: 10.1038/s41598-018-31027-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/31/2018] [Indexed: 01/03/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. The five-year survival rate of HNSCC has not improved even with major technological advancements in surgery and chemotherapy. Currently, docetaxel, cisplatin, and 5-fluoruracil (TPF) treatment has been the most popular chemotherapy method for HNSCC; but only a small percentage of HNSCC patients exhibit a good response to TPF treatment. Unfortunately, at present, no reasonably effective prediction model exists to assist clinicians with patient treatment. For this reason, patients have no other alternative but to risk neoadjuvant chemotherapy in order to determine their response to TPF. In this study, we analyzed the gene expression profile in TPF-sensitive and non-sensitive patient samples. We identified a gene expression signature between these two groups. We further chose 10 genes and trained a support vector machine (SVM) model. This model has 88.3% sensitivity and 88.9% specificity to predict the response to TPF treatment in our patients. In addition, four more TPF responsive and four more TPF non-sensitive patient samples were used for further validation. This SVM model has been proven to achieve approximately 75.0% sensitivity and 100% specificity to predict TPF response in new patients. This suggests that our 10-genes SVM prediction model has the potential to assist clinicians to personalize treatment for HNSCC patients.
Collapse
Affiliation(s)
- Qi Zhong
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Otolaryngology, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China
| | - Jugao Fang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China. .,Beijing Institute of Otolaryngology, Beijing, China. .,Key Laboratory of Otolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China.
| | - Zhigang Huang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Otolaryngology, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China
| | - Yifan Yang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Otolaryngology, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China
| | - Meng Lian
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Otolaryngology, Beijing, China.,Key Laboratory of Otolaryngology Head and Neck Surgery, Capital Medical University, Ministry of Education, Beijing, China
| | - Honggang Liu
- Beijing Key Laboratory of Head and Neck Molecular Diagnostic Pathology, Beijing, 100730, China
| | - Yixiang Zhang
- Department of Urology, The Second Affiliated Hospital of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, China
| | - Junhui Ye
- Neurontechnology, Shenzhen, Guangdong, China
| | - Xinjie Hui
- Department of Cell Biology and Medical Genetics, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Yejun Wang
- Department of Cell Biology and Medical Genetics, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Ying Ying
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Health Sciences Center, Shenzhen, 518060, China
| | - Qing Zhang
- Neurontechnology, Shenzhen, Guangdong, China.
| | - Yingduan Cheng
- Department of Urology, The Second Affiliated Hospital of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, China.
| |
Collapse
|
8
|
Yu Q, Wang X, Wang L, Zheng J, Wang J, Wang B. Knockdown of asparagine synthetase (ASNS) suppresses cell proliferation and inhibits tumor growth in gastric cancer cells. Scand J Gastroenterol 2016; 51:1220-6. [PMID: 27251594 DOI: 10.1080/00365521.2016.1190399] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Asparagine synthetase (ASNS) gene encodes an enzyme that catalyzes the glutamine- and ATP-dependent conversion of aspartic acid to asparagine. ASNS is deemed as a promising therapeutic target and its expression is associated with the chemotherapy resistance in several human cancers. However, its role in gastric cancer tumorigenesis has not been investigated. METHODS In this study, we employed small interfering RNA (siRNA) to transiently knockdown ASNS in two gastric cancer cell lines, AGS and MKN-45, followed by growth rate assay and colony formation assay. Dose response curve analysis was performed in AGS and MKN-45 cells with stable ASNS knockdown to assess sensitivity to cisplatin. Xenograft experiment was performed to examine in vivo synergistic effects of ASNS depletion and cisplatin on tumor growth. Expression level of ASNS was evaluated in human patient samples using quantitative PCR. Kaplan-Meier curve analysis was performed to evaluate association between ASNS expression and patient survival. RESULTS Transient knockdown of ASNS inhibited cell proliferation and colony formation in AGS and MKN-45 cells. Stable knockdown of ASNS conferred sensitivity to cisplatin in these cells. Depletion of ASNS and cisplatin treatment exerted synergistic effects on tumor growth in AGS xenografts. Moreover, ASNS was found to be up-regulated in human gastric cancer tissues compared with matched normal colon tissues. Low expression of ASNS was significantly associated with better survival in gastric cancer patients. CONCLUSION ASNS may contribute to gastric cancer tumorigenesis and may represent a novel therapeutic target for prevention or intervention of gastric cancer.
Collapse
Affiliation(s)
- Qingxiang Yu
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Xiaoyu Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Li Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Jia Zheng
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Jiang Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| | - Bangmao Wang
- a Department of Gastroenterology and Hepatology , General Hospital, Tianjin Medical University , Tianjin , PR China
| |
Collapse
|
9
|
Chang IW, Wu WJ, Wang YH, Wu TF, Liang PI, He HL, Yeh BW, Li CF. BCAT1 overexpression is an indicator of poor prognosis in patients with urothelial carcinomas of the upper urinary tract and urinary bladder. Histopathology 2016; 68:520-532. [PMID: 26173071 DOI: 10.1111/his.12778] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/06/2015] [Indexed: 02/06/2023]
Abstract
AIMS Amino acid biosynthesis is one of the cardinal events of carcinogenesis that has not been investigated in urothelial carcinoma (UC). By data-mining a published transcriptomic database of UCs of urinary bladder (UBUCs) (GSE31684), we identified branched-chain amino acid transaminase 1 (BCAT1) as the most significantly stepwise up-regulated gene during tumour progression among those associated with the amino acid biosynthetic process (GO:0008652). Accordingly, we analysed BCAT1 transcript and protein expression with their clinicopathological significance. METHODS AND RESULTS We used real-time reverse transcription-polymerase chain reaction (RT-PCR) to detect BCAT1 transcript levels in 20 UCs of upper tract (UTUCs) and 20 UBUCs, respectively. Immunohistochemical study was performed to determine BCAT1 protein expression in 340 UTUCs and 295 UBUCs. Higher BCAT1 transcript levels were associated with higher pT status in both groups (P < 0.05). BCAT1 protein overexpression was also associated significantly with adverse clinicopathological features, e.g. advanced pT stage, nodal metastasis, high pathological grade, etc. (P < 0.05). BCAT1 overexpression predicted worse disease-specific survival and metastasis-free survival in both univariate and multivariate analyses (P ≤ 0.001). CONCLUSION BCAT1 overexpression is associated with advanced tumour status, and implies adverse clinical outcomes of UCs, suggesting that its role in tumour progression could serve as a prognostic biomarker and a novel therapeutic target in UC.
Collapse
Affiliation(s)
- I-Wei Chang
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Wen-Jen Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Hui Wang
- Institute of Bioinformatics and Biosignal Transduction, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Feng Wu
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Peir-In Liang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hong-Lin He
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Bi-Wen Yeh
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Feng Li
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan
- National Cancer Research Institute, National Health Research Institutes, Tainan, Taiwan
- Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine and Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
10
|
Lee YE, He HL, Shiue YL, Lee SW, Lin LC, Wu TF, Chang IW, Lee HH, Li CF. The prognostic impact of lipid biosynthesis-associated markers, HSD17B2 and HMGCS2, in rectal cancer treated with neoadjuvant concurrent chemoradiotherapy. Tumour Biol 2015; 36:7675-7683. [PMID: 25929810 DOI: 10.1007/s13277-015-3503-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 04/23/2015] [Indexed: 11/27/2022] Open
Abstract
Neoadjuvant concurrent chemoradiotherapy has been widely used for rectal cancer to improve local tumor control. The varied response of individual tumors encouraged us to search for useful biomarkers to predict the therapeutic response. The study was aimed to evaluate the prognostic impact of lipid biosynthesis-associated biomarkers in rectal cancer patients treated with preoperative chemoradiotherapy. Through analysis of the previously published gene expression profiling database focusing on genes associated with lipid biosynthesis, we found that HSD17B2 and HMGCS2 were the top two significantly upregulated genes in the non-responders. We further evaluated their expression by immunohistochemistry in the pre-treatment tumor specimens from 172 patients with rectal cancer and statistically analyzed the associations between their expression and various clinicopathological factors, as well as survival. High expression of HMGCS2 or HSD17B2 was significantly associated with advanced pre- and post-treatment tumor or nodal status (P < 0.001) and lower tumor regression grade (P < 0.001). More importantly, high expression of either HMGCS2 or HSD17B2 was of prognostic significance, with HMGCS2 overexpression indicating poor prognosis for disease-free survival (P = 0.0003), local recurrence-free survival (P = 0.0115), and metastasis-free survival (P = 0.0119), while HSD17B2 overexpression was associated with poor prognosis for disease-free survival (P <0.0001), local recurrence-free survival (P = 0.0009), and metastasis-free survival (P < 0.0001). In multivariate analysis, only HSD17B2 overexpression remained as an independent prognosticator for shorter disease-free survival (P < 0.001) and metastasis-free survival (P = 0.008). In conclusion, high expression of either HSD17B2 or HMGCS2 predicted poor susceptibility of rectal cancer to preoperative chemoradiotherapy. Both acted as promising prognostic factors, particularly HSD17B2.
Collapse
Affiliation(s)
- Ying-En Lee
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hong-Lin He
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Sung-Wei Lee
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Ting-Feng Wu
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - I-Wei Chang
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hao-Hsien Lee
- Department of Surgery, Chi Mei Medical Center, Liouying, Tainan, Taiwan
| | - Chien-Feng Li
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan.
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan.
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.
| |
Collapse
|
11
|
Cecchi F, Lih CJ, Lee YH, Walsh W, Rabe DC, Williams PM, Bottaro DP. Expression array analysis of the hepatocyte growth factor invasive program. Clin Exp Metastasis 2015; 32:659-76. [PMID: 26231668 DOI: 10.1007/s10585-015-9735-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 07/13/2015] [Indexed: 02/17/2023]
Abstract
Signaling by human hepatocyte growth factor (hHGF) via its cell surface receptor (MET) drives mitogenesis, motogenesis and morphogenesis in a wide spectrum of target cell types and embryologic, developmental and homeostatic contexts. Oncogenic pathway activation also contributes to tumorigenesis and cancer progression, including tumor angiogenesis and metastasis, in several prevalent malignancies. The HGF gene encodes full-length hHGF and two truncated isoforms known as NK1 and NK2. NK1 induces all three HGF activities at modestly reduced potency, whereas NK2 stimulates only motogenesis and enhances HGF-driven tumor metastasis in transgenic mice. Prior studies have shown that mouse HGF (mHGF) also binds with high affinity to human MET. Here we show that, like NK2, mHGF stimulates cell motility, invasion and spontaneous metastasis of PC3M human prostate adenocarcinoma cells in mice through human MET. To identify target genes and signaling pathways associated with motogenic and metastatic HGF signaling, i.e., the HGF invasive program, gene expression profiling was performed using PC3M cells treated with hHGF, NK2 or mHGF. Results obtained using Ingenuity Pathway Analysis software showed significant overlap with networks and pathways involved in cell movement and metastasis. Interrogating The Cancer Genome Atlas project also identified a subset of 23 gene expression changes in PC3M with a strong tendency for co-occurrence in prostate cancer patients that were associated with significantly decreased disease-free survival.
Collapse
Affiliation(s)
- Fabiola Cecchi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA
| | - Chih-Jian Lih
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research, Inc. and Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - Young H Lee
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA
| | - William Walsh
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research, Inc. and Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - Daniel C Rabe
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA
| | - Paul M Williams
- Molecular Characterization and Clinical Assay Development Laboratory, Leidos Biomedical Research, Inc. and Frederick National Laboratory for Cancer Research, Frederick, MD, 21702-1201, USA
| | - Donald P Bottaro
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892-1501, USA. .,Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, Bldg 10 CRC Rm 2-3952, 10 Center Drive MSC 1107, Bethesda, MD, 20892-1107, USA.
| |
Collapse
|
12
|
He HL, Lee YE, Shiue YL, Lee SW, Lin LC, Chen TJ, Wu TF, Li CF. PLA2G2A overexpression is associated with poor therapeutic response and inferior outcome in rectal cancer patients receiving neoadjuvant concurrent chemoradiotherapy. Histopathology 2015; 66:991-1002. [PMID: 25393083 DOI: 10.1111/his.12613] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 11/09/2014] [Indexed: 11/30/2022]
Abstract
AIMS The aim of this study was to investigate the prognostic impact of group IIA phospholipase A2 (PLA2G2A) expression and its role in predicting the response to neoadjuvant concurrent cheomoradiotherapy (CCRT) in rectal cancer. METHODS AND RESULTS Through analysing a public transcriptome of rectal cancers, the PLA2G2A gene was identified as a significant predictor for CCRT response. We validated the expression of PLA2G2A using immunohistochemistry in the pretreatment tumour specimens from 172 patients with rectal cancer. The results were correlated with clinicopathological features, tumour regression grade, overall survival (OS), disease-free survival (DFS) and local recurrence-free survival (LRFS). High expression of PLA2G2A was associated with advanced pretreatment tumour status (P = 0.001), advanced pretreatment nodal status (P = 0.010), advanced post-treatment tumour status (P = 0.002) and lower tumour regression grade (P = 0.006). Furthermore, PLA2G2A expression was correlated negatively with gamma H2A histone family, member X (γ-H2AX) expression (P < 0.001, r = -0.580). More importantly, high expression of PLA2G2A emerged as an adverse prognostic factor for OS (P = 0.0190), DFS (P < 0.0001) and LRFS (P < 0.0001). In multivariate analysis, it remained independently prognostic for shorter DFS (P = 0.014) and LRFS (P = 0.012). CONCLUSIONS High expression of PLA2G2A was associated with poor therapeutic response and worse survival in patients with rectal cancer receiving neoadjuvant CCRT, justifying PLA2G2A as an important marker to predict CCRT response and outcome.
Collapse
Affiliation(s)
- Hong-Lin He
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ying-En Lee
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Sung-Wei Lee
- Department of Radiation Oncology, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Tzu-Ju Chen
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan
| | - Ting-Feng Wu
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chien-Feng Li
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| |
Collapse
|
13
|
He HL, Lee YE, Chen HP, Hsing CH, Chang IW, Shiue YL, Lee SW, Hsu CT, Lin LC, Wu TF, Li CF. Overexpression of DNAJC12 predicts poor response to neoadjuvant concurrent chemoradiotherapy in patients with rectal cancer. Exp Mol Pathol 2015; 98:338-345. [PMID: 25805104 DOI: 10.1016/j.yexmp.2015.03.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/20/2015] [Indexed: 12/16/2022]
Abstract
Genes associated with protein folding have been found to have certain prognostic significance in a subset of cancers. The aim of this study is to evaluate the clinical impact of DNAJC12 expression in patients with rectal cancers receiving neoadjuvant concurrent chemoradiotherapy (CCRT) followed by surgery. Through data mining from a public transcriptomic dataset of rectal cancer focusing on genes associated with protein folding, we found that DNAJC12, a member of the HSP40/DNAJ family, was the most significant such gene correlated with the CCRT response. We further evaluated the expression of DNAJC12 by immunohistochemistry in the pre-treatment tumor specimens from 172 patients with rectal cancers. From this set, we statistically analyzed the association of DNAJC12 expression with various clinicopathological factors, tumor regression grade, overall survival (OS), disease-free survival (DFS) and local recurrence-free survival (LRFS). High expression of DNAJC12 was significantly associated with advanced pre- and post-treatment tumor status (P<0.001), advanced pre- and post-treatment nodal status (P<0.001), increased vascular invasion (P=0.015), increased perineural invasion (P=0.023) and lower tumor regression grade (P=0.009). More importantly, high expression of DNAJC12 was found to be correlated with poor prognosis for OS (P=0.0012), DFS (P<0.0001) and LRFS (P=0.0001). In multivariate analysis, DNAJC12 overexpression still emerged as an independent prognosticator for shorter OS (P=0.040), DFS (P<0.001) and LRFS (P=0.016). The data indicate that DNAJC12 overexpression acts as a negative predictive factor for the response to neoadjuvant CCRT and was significantly associated with shorter survival in patients with rectal cancers receiving neoadjuvant CCRT followed by surgery.
Collapse
Affiliation(s)
- Hong-Lin He
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan; Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ying-En Lee
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hsin-Pao Chen
- Department of Colorectal Surgery, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan
| | - I-Wei Chang
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan; Institute of Biotechnology and Chemical Engineering, I-Shou University, Kaohsiung, Taiwan
| | - Yow-Ling Shiue
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Sung-Wei Lee
- Department of Radiation Oncology, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Chao-Tien Hsu
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Li-Ching Lin
- Department of Radiation Oncology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Ting-Feng Wu
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chien-Feng Li
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan.
| |
Collapse
|
14
|
Li CF, He HL, Wang JY, Huang HY, Wu TF, Hsing CH, Lee SW, Lee HH, Fang JL, Huang WT, Chen SH. Fibroblast growth factor receptor 2 overexpression is predictive of poor prognosis in rectal cancer patients receiving neoadjuvant chemoradiotherapy. J Clin Pathol 2014; 67:1056-1061. [PMID: 25271212 DOI: 10.1136/jclinpath-2014-202551] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
AIMS Neoadjuvant concurrent chemoradiotherapy (CCRT) followed by surgery is an increasingly used therapeutic strategy for advanced rectal cancer, but risk stratification and final outcomes remain suboptimal. Recently, the oncogenic role of the fibroblast growth factor/fibroblast growth factor receptor (FGFR) signalling pathway has been recognised; however, its clinical significance in rectal cancer has not been elucidated. In this study, we identify and validate targetable drivers associated with the FGFR signalling pathway in rectal cancer patients treated with CCRT. METHODS Using a published transcriptome of rectal cancers, we found FGFR2 gene significantly predicted response to CCRT. The expression levels of FGFR2, using immunohistochemistry assays, were further evaluated in 172 rectal cancer specimens that had not received any treatment. Expression levels of FGFR2 were statistically correlated with major clinicopathological features and clinical survival in this valid cohort. RESULTS High expression of FGFR2 was significantly related to advanced pretreatment tumour (p=0.022) and nodal status (p=0.026), post-treatment tumour (p<0.001) and nodal status (p=0.004), and inferior tumour regression grade (p<0.001). In survival analyses, high expression of FGFR2 was significantly associated with shorter local recurrence-free survival (p=0.0001), metastasis-free survival (MeFS; p=0.0003) and disease-specific survival (DSS; p<0.0001). Notably, high expression of FGFR2 was independently predictive of worse outcomes for MeFS (p=0.002, HR=5.387) and DSS (p=0.004, HR=4.997). CONCLUSIONS High expression of FGFR2 is correlated with advanced tumour stage, poor therapeutic response and worse survival in rectal cancer patients receiving neoadjuvant CCRT. These findings indicate that FGFR2 is a prognostic factor for treating rectal cancer.
Collapse
Affiliation(s)
- Chien-Feng Li
- Department of Pathology, Chi-Mei Medical Center, Tainan, Taiwan National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hong-Lin He
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Jaw-Yuan Wang
- Graduate Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan Division of Gastrointestinal and General Surgery, Department of Surgery Cancer, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Hsuan-Ying Huang
- Department of Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ting-Fe Wu
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi-Mei Medical Center, Tainan, Taiwan
| | - Sung-Wei Lee
- Department of Radiation Oncology, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Hao-Hsien Lee
- Department of Surgery, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Jui-Lung Fang
- Department of Radiology, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Wen-Tsung Huang
- Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| | - Shang-Hung Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan Division of Hematology and Oncology, Department of Internal Medicine, Chi-Mei Medical Center, Liouying, Tainan, Taiwan
| |
Collapse
|
15
|
He HL, Lee YE, Shiue YL, Lee SW, Lin LC, Chen TJ, Wu TF, Hsing CH, Huang HY, Wang JY, Li CF. Overexpression of REG4 confers an independent negative prognosticator in rectal cancers receiving concurrent chemoradiotherapy. J Surg Oncol 2014; 110:1002-1010. [PMID: 25155043 DOI: 10.1002/jso.23764] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/19/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND OBJECTIVES Neoadjuvant concurrent chemoradiotherapy (CCRT) followed by surgery is the standard treatment for locally advanced rectal cancer. Through data mining from published transcriptomic database, we identified Regenerating Gene Type IV (REG4) as the most significantly associated gene with resistance to CCRT. This study examined the prognostic impact of REG4 expression in patients with rectal cancer receiving neoadjuvant CCRT. METHODS REG4 immunohistochemistry was retrospectively assessed for pre-treatment biopsy specimens from 172 rectal cancer patients who received neoadjuvant CCRT followed by surgery without initial distant metastasis. The results were correlated with the clinicopathological variables, disease-specific survival (DSS), local recurrence-free survival (LRFS), and distant metastasis-free survival (DMFS), as well as γ-H2AX expression in post-treatment tumor samples. RESULTS High expression of REG4 was associated with advanced pre-treatment nodal status (P = 0.026), advanced post-treatment tumor status (P = 0.006), advanced post-treatment nodal status (P = 0.001), advanced post-treatment tumor stage (P < 0.001), and inferior tumor regression grade (P = 0.001). Of note, high expression of REG4 emerged as an adverse prognosticator for DSS (P = 0.0004), LRFS (P = 0.0009), and MeFS (P = 0.0254). After multivariate comparisons, it remained independently prognostic for worse DSS (hazard ratio [HR] = 2.731; P = 0.025) and LRFS (HR = 2.676; P = 0.029). High expression of REG4 was also negatively associated with γ-H2AX expression (P < 0.0001, r = -0.708). CONCLUSIONS High expression of REG4 is associated with poor therapeutic response, adverse outcome and an aggressive phenotype in rectal cancer patients treated with neoadjuvant CCRT, justifying REG4 is a surrogate marker to predict CCRT resistance.
Collapse
Affiliation(s)
- Hong-Lin He
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan; Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Lee YY, Li CF, Lin CY, Lee SW, Sheu MJ, Lin LC, Chen TJ, Wu TF, Hsing CH. Overexpression of CPS1 is an independent negative prognosticator in rectal cancers receiving concurrent chemoradiotherapy. Tumour Biol 2014; 35:11097-11105. [PMID: 25099619 DOI: 10.1007/s13277-014-2425-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 07/29/2014] [Indexed: 12/13/2022] Open
Abstract
Locally advanced rectal cancers are currently treated with neoadjuvant concurrent chemoradiotherapy (CCRT) followed by surgery, but stratification of risk and final outcomes remain suboptimal. In view of the fact that glutamine metabolism is usually altered in cancer, we profiled and validated the significance of genes involved in this pathway in rectal cancers treated with CCRT. From a published transcriptome of rectal cancers (GSE35452), we focused on glutamine metabolic process-related genes (GO:0006541) and found upregulation of carbamoyl phosphate synthetase 1 (CPS1) gene most significantly predicted poor response to CCRT. We evaluated the expression levels of CPS1 using immunohistochemistry to analyze tumor specimens obtained during colonoscopy from 172 rectal cancer patients. Expression levels of CPS1 were further correlated with major clinicopathological features and survivals in this validation cohort. To further confirm CPS1 expression levels, Western blotting was performed for human colon epithelial primary cell (HCoEpiC) and four human colon cancer cells, including HT29, SW480, LoVo, and SW620. CPS1 overexpression was significantly related to advanced posttreatment tumor (T3, T4; P = 0.006) and nodal status (N1, N2; P < 0.001), and inferior tumor regression grade (P = 0.004). In survival analyses, CPS1 overexpression was significantly associated with shorter disease-specific survival (DSS) and metastasis-free survival (MeFS). Furthermore, using multivariate analysis, it was also independently predictive of worse DSS (P = 0.021, hazard ratio = 2.762) and MeFS (P = 0.004, hazard ratio = 3.897). CPS1 protein expression, as detected by Western blotting, is more abundant in colon cancer cells than nonneoplastic HCoEpiC. Overexpression of CPS1 is associated with poor therapeutic response and adverse outcomes among rectal cancer patients receiving CCRT, justifying the potential theranostic value of CPS1 for such patients.
Collapse
Affiliation(s)
- Yi-Ying Lee
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Sheu MJ, Li CF, Lin CY, Lee SW, Lin LC, Chen TJ, Ma LJ. Overexpression of ANXA1 confers independent negative prognostic impact in rectal cancers receiving concurrent chemoradiotherapy. Tumour Biol 2014; 35:7755-7763. [PMID: 24810927 DOI: 10.1007/s13277-014-2032-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 04/28/2014] [Indexed: 01/01/2023] Open
Abstract
Neoadjuvant concurrent chemoradiation therapy (CCRT) is an increasingly common therapeutic strategy for rectal cancer. Clinically, it remains a major challenge to predict therapeutic response and patient outcomes after CCRT. Annexin I (ANXA1), encoded by ANXA1, is a Ca(2+)/phospholipid-binding protein that mediates actin dynamics and cellular proliferation, as well as suggesting tumor aggressiveness and predicting therapeutic response in certain malignancies. However, expression of ANXA1 has never been reported in rectal cancer receiving CCRT. This study examined the predictive and prognostic impact of ANXA1 expression in patients with rectal cancer following neoadjuvant CCRT. We identified ANXA1 as associated with resistance to CCRT through data mining from a published transcriptomic dataset. Its immunoexpression was retrospectively assessed using H scores on pre-treatment biopsies from 172 rectal cancer patients treated with neoadjuvant CCRT followed by curative surgery. Results were correlated with clinicopathological features, therapeutic response, tumor regression grade (TRG), and metastasis-free survival (MeFS), as well as local recurrent-free survival (LRFS) and disease-specific survival (DSS). High expression of ANXA1 was associated with advanced pre-treatment tumor status (T3, T4, p = 0.022), advanced pre-treatment nodal status (N1, N2, p = 0.004), advanced post-treatment tumor status (T3, T4, p < 0.001), advanced post-treatment nodal status (N1, N2, p = 0.001) and inferior TRG (p = 0.009). In addition, high expression of ANXA1 emerged as an adverse prognosticator for DSS (p < 0.0001), LRFS (p = 0.0001) and MeFS (p = 0.0004). Moreover, high expression of ANXA1 also remained independently prognostic of worse DSS (hazard ratio [HR] = 3.998; p = 0.007), LRFS (HR = 3.206; p = 0.028) and MeFS (HR = 3.075; p = 0.017). This study concludes that high expression of ANXA1 is associated with poor therapeutic response and adverse outcomes in rectal cancer patients treated with neoadjuvant CCRT.
Collapse
Affiliation(s)
- Ming-Jen Sheu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Chi Mei Foundation Medical Center, Tainan, Taiwan
| | | | | | | | | | | | | |
Collapse
|