1
|
Wang X, Feng B, Guo HY, Yao FF, Song HN, Wang XY, Sun XC, Wang K, Ge YC, Cui R. Roles of cathepsin S expression levels on the prognosis and tumour microenvironment in clear cell renal cell carcinoma. Discov Oncol 2024; 15:690. [PMID: 39570472 PMCID: PMC11582264 DOI: 10.1007/s12672-024-01547-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Increasing evidence suggests a link between the enzyme cathepsin S (CTSS) and tumour development. However, the potential involvement and molecular functions of CTSS in clear cell renal cell carcinoma (ccRCC) remain unclear. METHODS We downloaded original data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases and integrated them using R. Kaplan-Meier plots of integrated expression scores were used to analyse survival outcomes. Additionally, we investigated mRNA expression, clinicopathological features, immune infiltrates, and single-cell sequencing analysis of CTSS in ccRCC. In vitro experiments were conducted with qRT-PCR and IHC staining. RESULTS CTSS transcriptomic and proteomic levels were higher in ccRCC than in para-cancerous tissues. Low CTSS expression was correlated with poor prognosis in patients with ccRCC. Our data demonstrated that the expression of CTSS was strongly correlated with immune cell infiltration levels and gene markers of immune cells, chemokines, and receptors. Single-cell sequencing analysis demonstrated that CTSS expression was detectable in monocytes/macrophages. Finally, certain chemicals were confirmed to affect CTSS expression. CONCLUSION Our findings indicate that CTSS offers promise as a prognostic biomarker and novel immune-related therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Nephrology, The First People's Hospital in Jinzhou, Dalian, China
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Bei Feng
- Department of Nephrology, Jingzhou Central Hospital, Hubei, China
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Hai-Ying Guo
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Fei-Fei Yao
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hui-Nan Song
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi-Yue Wang
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiao-Chen Sun
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Kai Wang
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Yu-Chen Ge
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Rui Cui
- Department of Nephrology, Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
2
|
Kim S, Lee KW, Yoo Y, Park SH, Lee JW, Jeon S, Illia S, Joshi P, Park HW, Lo HE, Seo J, Kim Y, Chang M, Lee TJ, Seo JB, Kim SH, Croce CM, Kim I, Suh SS, Jeon YJ. MiR-29 and MiR-140 regulate TRAIL-induced drug tolerance in lung cancer. Anim Cells Syst (Seoul) 2024; 28:184-197. [PMID: 38693921 PMCID: PMC11062278 DOI: 10.1080/19768354.2024.2345644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/11/2024] [Indexed: 05/03/2024] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has chemotherapeutic potential as a regulator of an extrinsic apoptotic ligand, but its effect as a drug is limited by innate and acquired resistance. Recent findings suggest that an intermediate drug tolerance could mediate acquired resistance, which has made the main obstacle for limited utility of TRAIL as an anti-cancer therapeutics. We propose miRNA-dependent epigenetic modification drives the drug tolerant state in TRAIL-induced drug tolerant (TDT). Transcriptomic analysis revealed miR-29 target gene activation in TDT cells, showing oncogenic signature in lung cancer. Also, the restored TRAIL-sensitivity was associated with miR-29ac and 140-5p expressions, which is known as tumor suppressor by suppressing oncogenic protein RSK2 (p90 ribosomal S6 kinase), further confirmed in patient samples. Moreover, we extended this finding into 119 lung cancer cell lines from public data set, suggesting a significant correlation between TRAIL-sensitivity and RSK2 mRNA expression. Finally, we found that increased RSK2 mRNA is responsible for NF-κB activation, which we previously showed as a key determinant in both innate and acquired TRAIL-resistance. Our findings support further investigation of miR-29ac and -140-5p inhibition to maintain TRAIL-sensitivity and improve the durability of response to TRAIL in lung cancer.
Collapse
Affiliation(s)
- Suyeon Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ki Wook Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yongjin Yoo
- Department of Stem Cell Biology and Regenerative Medicine Institute, Stanford University, Stanford, CA, USA
| | - Sang Hee Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ji Won Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Suhyun Jeon
- Department of Biosciences, Mokpo National University, Muan, Republic of Korea
| | - Shaginyan Illia
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Pooja Joshi
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hyun Woo Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Han-En Lo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jimin Seo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeonwoo Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Min Chang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tae Jin Lee
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jong Bae Seo
- Department of Biosciences, Mokpo National University, Muan, Republic of Korea
| | - Sung-Hak Kim
- Department of Animal Science, Chonnam National University, Gwangju, Republic of Korea
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Inki Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
| | - Sung-Suk Suh
- Department of Biosciences, Mokpo National University, Muan, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
3
|
Strzelczyk J, Wójcik-Giertuga M, Strzelczyk JK, Seńkowska AP, Biernacki K, Kos-Kudła B. Selected Parameters of Bone Turnover in Neuroendocrine Tumors-A Potential Clinical Use? J Clin Med 2023; 12:4608. [PMID: 37510722 PMCID: PMC10380215 DOI: 10.3390/jcm12144608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/04/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Currently, there are no effective markers to diagnose and monitor patients with neuroendocrine tumors (NETs). The aim of this study was to assess bone metabolism based on selected markers of bone turnover: OST, OPG, and IGFBP-3, in both the group of patients with NETs and the control group. Associations with selected sociodemographic, biochemical, and clinicopathological characteristics were examined. We also evaluated any potential associations between these markers and selected biochemical markers of NETs commonly used in clinical practice. METHODS The study group included 60 patients with GEP-NETs and BP-NETs, while the control group comprised 62 healthy individuals. The serum concentrations of OST, OPG and IGFBP-3 were assessed using ELISA. RESULTS OST and OPG levels were significantly higher in the study group compared to the control group. In the study group, we observed a significant correlation between OPG and the clinical stage and chromogranin A. Additionally, an association was found between OPG and histological grade, Ki-67, and metastasis in GEP-NET cases. CONCLUSIONS Markers of bone turnover cannot be used in the routine diagnostics of neuroendocrine tumors. Nonetheless, these markers may help evaluate the skeletal system in patients with NETs. Further research is needed to determine the utility of osteocalcin (OST) and osteoprotegerin (OPG) as potential biomarkers for neuroendocrine tumors.
Collapse
Affiliation(s)
- Janusz Strzelczyk
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-514 Katowice, Poland
| | - Monika Wójcik-Giertuga
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-514 Katowice, Poland
| | - Joanna Katarzyna Strzelczyk
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana St., 41-808 Zabrze, Poland
| | - Alicja Prawdzic Seńkowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana St., 41-808 Zabrze, Poland
| | - Krzysztof Biernacki
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana St., 41-808 Zabrze, Poland
| | - Beata Kos-Kudła
- Department of Endocrinology and Neuroendocrine Tumors, Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 35 Ceglana St., 40-514 Katowice, Poland
| |
Collapse
|
4
|
Groysman L, Carlsen L, Huntington KE, Shen WH, Zhou L, El-Deiry WS. Chemotherapy-induced cytokines and prognostic gene signatures vary across breast and colorectal cancer. Am J Cancer Res 2021; 11:6086-6106. [PMID: 35018244 PMCID: PMC8727797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/12/2021] [Indexed: 06/14/2023] Open
Abstract
The mechanisms by which chemotherapeutic drugs mediate efficacy and toxicity in patients across cancers are not fully understood. A poorly understood aspect of the tumor cell response to chemotherapy is cytokine regulation. Some drug-induced cytokines promote the anti-cancer activity of the drugs, but others may promote proliferation, metastasis, and drug resistance. We evaluated effects of clinical chemotherapeutics oxaliplatin, cisplatin, 5-fluorouracil (5-FU), doxorubicin, paclitaxel, docetaxel, and carboplatin on a panel of 52 cytokines in MCF7 breast cancer (BC) cells. We observed pan-drug effects, such as the upregulation of TRAIL-R2 and Chitinase 3-like 1 and drug-specific effects on interleukin and CXCL cytokines. We compared cytokine regulation in MCF7 BC and HCT116 colorectal cancer (CRC) cells, revealing tissue-specific drug effects such as enhanced upregulation of TRAIL-R2 and downregulation of IFN-β and TRAIL in MCF7 by cisplatin, oxaliplatin, and 5-FU. We found that chemotherapy-inducible transcripts have varying potential for prognostic significance in CRC versus BC. Among the non-prognostic CRC genes that were prognostic in BC were NFKBIA and GADD45A, both of which support anti-cancer drug mechanisms. Thus, we establish a novel 7-drug, 52-cytokine signature in MCF7 BC cells and a 3-drug, 40-cytokine signature in HCT116 CRC cells that suggest drug-specific and tissue-specific cytokine regulation. Distinct differences across prognostic gene signatures in BC and CRC further support tissue specificity in the relative impact of drug-regulated genes on patient survival.
Collapse
Affiliation(s)
- Leya Groysman
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Macaulay Honors College at Hunter College, CUNYManhattan, NY 10065, USA
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell UniversityNY 10065, USA
| | - Lindsey Carlsen
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Graduate Program in Pathobiology, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Kelsey E Huntington
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Graduate Program in Pathobiology, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Wen H Shen
- Department of Radiation Oncology, Weill Cornell Medicine, Cornell UniversityNY 10065, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Graduate Program in Pathobiology, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University and The Lifespan Health SystemProvidence, RI 02903, USA
- Hematology-Oncology Division, Brown University and The Lifespan Cancer InstituteProvidence, RI 02903, USA
- Cancer Center at Brown University, The Warren Alpert Medical School, Brown UniversityProvidence, RI 02903, USA
| |
Collapse
|
5
|
Giorello MB, Borzone FR, Labovsky V, Piccioni FV, Chasseing NA. Cancer-Associated Fibroblasts in the Breast Tumor Microenvironment. J Mammary Gland Biol Neoplasia 2021; 26:135-155. [PMID: 33398516 DOI: 10.1007/s10911-020-09475-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/30/2020] [Indexed: 12/11/2022] Open
Abstract
Years of investigation have shed light on a theory in which breast tumor epithelial cells are under the effect of the stromal microenvironment. This review aims to discuss recent findings concerning the phenotypic and functional characteristics of cancer associated fibroblasts (CAFs) and their involvement in tumor evolution, as well as their potential implications for anti-cancer therapy. In this manuscript, we reviewed that CAFs play a fundamental role in initiation, growth, invasion, and metastasis of breast cancer, and also serve as biomarkers in the clinical diagnosis, therapy, and prognosis of this disease.
Collapse
Affiliation(s)
- María Belén Giorello
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| | - Francisco Raúl Borzone
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Vivian Labovsky
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Flavia Valeria Piccioni
- Laboratorio de Oncología Molecular y Nuevos Blancos Terapéuticos (IBYME) y Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Norma Alejandra Chasseing
- Laboratorio de Inmunohematología (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Biología y Medicina Experimental, Vuelta de Obligado 2490, CP, 1428, Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
6
|
Fein MR, He XY, Almeida AS, Bružas E, Pommier A, Yan R, Eberhardt A, Fearon DT, Van Aelst L, Wilkinson JE, Dos Santos CO, Egeblad M. Cancer cell CCR2 orchestrates suppression of the adaptive immune response. J Exp Med 2021; 217:151949. [PMID: 32667673 PMCID: PMC7537399 DOI: 10.1084/jem.20181551] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 12/27/2019] [Accepted: 05/26/2020] [Indexed: 01/22/2023] Open
Abstract
C-C chemokine receptor type 2 (CCR2) is expressed on monocytes and facilitates their recruitment to tumors. Though breast cancer cells also express CCR2, its functions in these cells are unclear. We found that Ccr2 deletion in cancer cells led to reduced tumor growth and approximately twofold longer survival in an orthotopic, isograft breast cancer mouse model. Deletion of Ccr2 in cancer cells resulted in multiple alterations associated with better immune control: increased infiltration and activation of cytotoxic T lymphocytes (CTLs) and CD103+ cross-presenting dendritic cells (DCs), as well as up-regulation of MHC class I and down-regulation of checkpoint regulator PD-L1 on the cancer cells. Pharmacological or genetic targeting of CCR2 increased cancer cell sensitivity to CTLs and enabled the cancer cells to induce DC maturation toward the CD103+ subtype. Consistently, Ccr2−/− cancer cells did not induce immune suppression in Batf3−/− mice lacking CD103+ DCs. Our results establish that CCR2 signaling in cancer cells can orchestrate suppression of the immune response.
Collapse
Affiliation(s)
- Miriam R Fein
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Graduate Program in Genetics, Stony Brook University, Stony Brook, NY
| | - Xue-Yan He
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Ana S Almeida
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Emilis Bružas
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Watson School of Biological Sciences, Cold Spring Harbor, NY
| | | | - Ran Yan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Watson School of Biological Sciences, Cold Spring Harbor, NY
| | - Anaïs Eberhardt
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Département de Biologie, École Normale Supérieure de Lyon, Lyon, France
| | - Douglas T Fearon
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY.,Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK.,Weill Cornell Medical College, New York, NY
| | | | - John Erby Wilkinson
- Departments of Molecular and Integrative Physiology and Internal Medicine, University of Michigan, Ann Arbor, MI
| | | | | |
Collapse
|
7
|
Liu F, Wu H. CC Chemokine Receptors in Lung Adenocarcinoma: The Inflammation-Related Prognostic Biomarkers and Immunotherapeutic Targets. J Inflamm Res 2021; 14:267-285. [PMID: 33574689 PMCID: PMC7872903 DOI: 10.2147/jir.s278395] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common type of lung cancer with a high incidence and increased mortality. CC chemokine receptors were participating in the modulation of the tumor microenvironment and involved in carcinogenesis and tumor development. However, the potential mechanistic values of CC chemokine receptors as clinical biomarkers and therapeutic targets in LUAD have not been fully clarified. Methodology ONCOMINE, UALCAN, GEPIA, Kaplan-Meier Plotter, SurvExpress, MethSurv, SurvivalMeth, cBioPortal, String, GeneMANIA, DAVID, Metascape, TRRUST, LinkedOmics, and Timer were applied in this work. Results The transcriptional levels of CCR1/10 in LUAD tissues were significantly reduced while the transcriptional levels of CCR3/6/7/8 were significantly elevated, and the expression of CCR1 was the highest in LUAD among these CC chemokine receptors. A significant correlation was found between the expression of CCR2/4/6/7 and the pathological stage of LUAD patients. There were significant associations between CCR2/3/4/5/6/10 expression levels and OS in LUAD, and LUAD patients with high transcriptional levels of CCR3/4 had inferior first-progression survival. In addition, the prognostic values of CC chemokine receptors signature in LUAD were explored in three independent cohorts, the high-risk group displayed unfavorable OS compared with the low-risk group, and the LUAD cases in the high-risk group also suffered inferior RFS than that in the low-risk group. And for the prognostic value of the DNA methylation of CC chemokine receptors, we found 1 CpG of CCR2, 2 CpGs of CCR3, 1 CpG of CCR4, 3 CpGs of CCR6, 3 CpGs of CCR7, 1 CpG of CCR8, and 3 CpGs of CCR9 were significantly associated with prognosis in LUAD patients. However, the DNA methylation signature analysis showed there was no statistically significant association between the high- and low-risk group. For potential mechanism, the neighbor gene networks, interaction analyses, functional enrichment analyses of CC chemokine receptors in LUAD were performed, the transcription factor targets, kinase targets, and miRNA targets of CC chemokine receptors were also identified in LUAD. We also found significant correlations among CC chemokine receptors expression and the infiltration of immune cells, the tumor infiltration levels among LUAD with different somatic copy number alterations of these chemokine receptors were also assessed. Moreover, the Cox proportional hazard model showed that CCR1/2/10, B_cell, CD4_Tcell were significantly related to the clinical outcome of LUAD patients. Conclusion CC chemokine receptors might serve as immunotherapeutic targets and prognostic biomarkers in LUAD.
Collapse
Affiliation(s)
- Fangteng Liu
- Department of Breast Surgery, The Third Hospital of Nanchang, Nanchang, Jiangxi, 330009, People's Republic of China.,Faculty of Medicine, University of Munich, Munich, 80336, Germany
| | - Hengyu Wu
- Department of Breast Surgery, The Third Hospital of Nanchang, Nanchang, Jiangxi, 330009, People's Republic of China
| |
Collapse
|
8
|
Evangelista AF, Oliveira RJ, O Silva VA, D C Vieira RA, Reis RM, C Marques MM. Integrated analysis of mRNA and miRNA profiles revealed the role of miR-193 and miR-210 as potential regulatory biomarkers in different molecular subtypes of breast cancer. BMC Cancer 2021; 21:76. [PMID: 33461524 PMCID: PMC7814437 DOI: 10.1186/s12885-020-07731-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/13/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Breast cancer is the most frequently diagnosed malignancy among women. However, the role of microRNA (miRNA) expression in breast cancer progression is not fully understood. In this study we examined predictive interactions between differentially expressed miRNAs and mRNAs in breast cancer cell lines representative of the common molecular subtypes. Integrative bioinformatics analysis identified miR-193 and miR-210 as potential regulatory biomarkers of mRNA in breast cancer. Several recent studies have investigated these miRNAs in a broad range of tumors, but the mechanism of their involvement in cancer progression has not previously been investigated. METHODS The miRNA-mRNA interactions in breast cancer cell lines were identified by parallel expression analysis and miRNA target prediction programs. The expression profiles of mRNA and miRNAs from luminal (MCF-7, MCF-7/AZ and T47D), HER2 (BT20 and SK-BR3) and triple negative subtypes (Hs578T e MDA-MB-231) could be clearly separated by unsupervised analysis using HB4A cell line as a control. Breast cancer miRNA data from TCGA patients were grouped according to molecular subtypes and then used to validate these findings. Expression of miR-193 and miR-210 was investigated by miRNA transient silencing assays using the MCF7, BT20 and MDA-MB-231 cell lines. Functional studies included, xCELLigence system, ApoTox-Glo triplex assay, flow cytometry and transwell inserts were performed to determine cell proliferation, cytotoxicity, apoptosis, migration and invasion, respectively. RESULTS The most evident effects were associated with cell proliferation after miR-210 silencing in triple negative subtype cell line MDA-MB-231. Using in silico prediction algorithms, TNFRSF10 was identified as one of the potential regulated downstream targets for both miRNAs. The TNFRSF10C and TNFRSF10D mRNA expression inversely correlated with the expression levels of miR-193 and miR210 in breast cell lines and breast cancer patients, respectively. Other potential regulated genes whose expression also inversely correlated with both miRNAs were CCND1, a known mediator on invasion and metastasis, and the tumor suppressor gene RUNX3. CONCLUSIONS In summary, our findings identify miR-193 and miR-210 as potential regulatory miRNA in different molecular subtypes of breast cancer and suggest that miR-210 may have a specific role in MDA-MB-231 proliferation. Our results highlight important new downstream regulated targets that may serve as promising therapeutic pathways for aggressive breast cancers.
Collapse
Affiliation(s)
- Adriane F Evangelista
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, 14784-400, Brazil
| | - Renato J Oliveira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, 14784-400, Brazil.
| | - Viviane A O Silva
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, 14784-400, Brazil
| | - Rene A D C Vieira
- Department of Mastology and Breast Reconstruction, Barretos Cancer Hospital, Barretos, São Paulo, 14784-400, Brazil
| | - Rui M Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, 14784-400, Brazil.,Life and Health Sciences Research Institute (ICVS), Health Sciences School, University of Minho, Braga, 4710-057, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, 4710-057, Portugal
| | - Marcia M C Marques
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, 14784-400, Brazil.,Tumor Biobank, Barretos Cancer Hospital, Barretos, São Paulo, 14784-400, Brazil.,Barretos School of Health Sciences, FACISB, Barretos, São Paulo, 14784-400, Brazil
| |
Collapse
|
9
|
Singh D, Tewari M, Singh S, Narayan G. Revisiting the role of TRAIL/TRAIL-R in cancer biology and therapy. Future Oncol 2021; 17:581-596. [PMID: 33401962 DOI: 10.2217/fon-2020-0727] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily, can induce apoptosis in cancer cells, sparing normal cells when bound to its associated death receptors (DR4/DR5). This unique mechanism makes TRAIL a potential anticancer therapeutic agent. However, clinical trials of recombinant TRAIL protein and TRAIL receptor agonist monoclonal antibodies have shown disappointing results due to its short half-life, poor pharmacokinetics and the resistance of the cancer cells. This review summarizes TRAIL-induced apoptotic and survival pathways as well as mechanisms leading to apoptotic resistance. Recent development of methods to overcome cancer cell resistance to TRAIL-induced apoptosis, such as protein modification, combination therapy and TRAIL-based gene therapy, appear promising. We also discuss the challenges and opportunities in the development of TRAIL-based therapies for the treatment of human cancers.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, 221005, India
| | - Gopeshwar Narayan
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
10
|
Geerts D, Chopra C, Connelly L. Osteoprotegerin: Relationship to Breast Cancer Risk and Prognosis. Front Oncol 2020; 10:462. [PMID: 32318347 PMCID: PMC7154067 DOI: 10.3389/fonc.2020.00462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 03/16/2020] [Indexed: 01/14/2023] Open
Abstract
Osteoprotegerin (OPG) is a secreted member of the Tumor Necrosis Factor (TNF) receptor superfamily (TNFRSF11B), that was first characterized and named for its protective role in bone remodeling. In this context, OPG binds to another TNF superfamily member Receptor Activator of NF-kappaB Ligand (RANKL; TNFSF11) and blocks interaction with RANK (TNFRSF11A), preventing RANKL/RANK stimulation of osteoclast maturation, and bone breakdown. Further studies revealed that OPG protein is also expressed by tumor cells and led to investigation of the role of OPG in tumor biology. An increasing body of data has demonstrated that OPG modulates breast tumor behavior. Initially, research was focused on OPG in the bone microenvironment as a potential inhibitor of RANKL-driven osteolysis. More recently, attention has shifted to include OPG expression and interactions in the primary breast tumor independent of RANKL. In the primary tumor, OPG may interact with another TNF superfamily member, TNF-Related Apoptosis Inducing Ligand (TRAIL; TNFSF10) to prevent apoptosis induction. Additional interest in OPG in breast cancer has been stimulated by the tumor-promoting role of its binding partner RANKL in association with BRCA1 gene mutations. We and others have previously summarized the functional studies on OPG and breast cancer (1, 2). After basic research studies on the in vitro role for OPG (and RANKL) in breast cancer, the field now expands to assess the in vivo role for OPG by examining the correlation between OPG expression and breast cancer risk or patient prognosis. However, the data reported so far is conflicting, since OPG expression appears linked to both good and poor patient survival. In the current review we will summarize these studies. Our goal is to provide stimulus for further research to bridge the basic research findings and clinical data regarding OPG in breast cancer.
Collapse
Affiliation(s)
- Dirk Geerts
- Department of Medical Biology, Academic Medical Center Amsterdam, Amsterdam, Netherlands
| | - Christina Chopra
- School of Medicine, California University of Science and Medicine, San Bernardino, CA, United States
| | - Linda Connelly
- School of Medicine, California University of Science and Medicine, San Bernardino, CA, United States
| |
Collapse
|
11
|
Singh D, Prasad CB, Biswas D, Tewari M, Kar AG, Ansari MA, Singh S, Narayan G. TRAIL receptors are differentially regulated and clinically significant in gallbladder cancer. Pathology 2020; 52:348-358. [PMID: 32111400 DOI: 10.1016/j.pathol.2019.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/26/2019] [Accepted: 12/01/2019] [Indexed: 12/15/2022]
Abstract
Deregulation of the receptors of TNF-related apoptosis inducing ligand (TRAIL) has been reported in various cancers. In an effort to define the role of these receptors we profiled their expression in gallbladder cancer (GBC) and explored their clinical significance. Expression of TRAIL receptors' mRNA in GBC was analysed through reverse transcriptase polymerase chain reaction (RT-PCR), and protein through western blotting, immunohistochemistry and enzyme-linked immunosorbent assay (ELISA). mRNA data show frequent higher expression of TRAIL receptors in GBC samples. Death receptors DR4 and DR5 showed significant negative correlation with tumour stage, T stage and tumour grade; DcR1 transcript showed positive correlation with tumour stage, N stage, M stage and tumour grade. Similarly, IHC showed frequent positive staining for DR4, DR5 and DcR1in GBC samples. Cytoplasmic and nuclear DR4 protein showed negative correlation with T stage and tumour grade, whereas cytoplasmic DcR1 protein showed positive correlation with tumour stage and N stage. Nuclear DcR1 showed positive correlation with N stage. ELISA results showed significantly higher expression of secretory DcR1 in GBC patients. Kaplan-Meier analysis demonstrated significantly decreased mean survival of patients with positive staining of cytoplasmic DcR1. High level of death receptors identified the patients with early gallbladder cancer, whereas high DcR1 expression served as a prognostic factor for poor outcome.
Collapse
Affiliation(s)
- Deepika Singh
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Chandra Bhushan Prasad
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Dipanjan Biswas
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Amrita Ghosh Kar
- Department of Pathology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mumtaz Ahmed Ansari
- Department of General Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, India
| | - Gopeshwar Narayan
- Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
12
|
TRAIL Mediated Signaling in Breast Cancer: Awakening Guardian Angel to Induce Apoptosis and Overcome Drug Resistance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:243-252. [PMID: 31456187 DOI: 10.1007/978-3-030-20301-6_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
Sequencing technologies have allowed us to characterize highly heterogeneous molecular landscape of breast cancer with unprecedented details. Tremendous breakthroughs have been made in unraveling contributory role of signaling pathways in breast cancer development and progression. It is becoming progressively more understandable that deregulation of spatio-temporally controlled pathways underlie development of resistance against different drugs. TRAIL mediated signaling has attracted considerable appreciation because of its characteristically unique ability to target cancer cells while leaving normal cells intact. Discovery of TRAIL was considered as a paradigm shift in molecular oncology because of its conspicuous ability to selectively target cancer cells. There was an exponential growth in the number of high-quality reports which highlighted cancer targeting ability of TRAIL and scientists worked on the development of TRAIL-based therapeutics and death receptor targeting agonistic antibodies to treat cancer. However, later studies challenged simplistic view related to tumor targeting ability of TRAIL. Detailed mechanistic insights revealed that overexpression of anti-apoptotic proteins, inactivation of pro-apoptotic proteins and downregulation of death receptors were instrumental in impairing apoptosis in cancer cells. Therefore researchers started to give attention to identification of methodologies and strategies to overcome the stumbling blocks associated with TRAIL-based therapeutics. Subsequent studies gave us a clear picture of signaling cascade of TRAIL and how deregulation of different proteins abrogated apoptosis. In this chapter we have attempted to provide an overview of the TRAIL induced signaling, list of proteins frequently deregulated and modern approaches to strategically restore apoptosis in TRAIL-resistant breast cancers.
Collapse
|
13
|
Heterogeneous intracellular TRAIL-receptor distribution predicts poor outcome in breast cancer patients. J Mol Med (Berl) 2019; 97:1155-1167. [PMID: 31183506 DOI: 10.1007/s00109-019-01805-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 05/22/2019] [Accepted: 05/24/2019] [Indexed: 12/20/2022]
Abstract
Upon ligand binding, plasma membrane-located TNF-related apoptosis-inducing ligand (TRAIL)-receptors 1 and 2 induce apoptosis as well as cancer-promoting signaling in cancer cells. TRAIL-R3 and TRAIL-R4 are believed to negatively regulate TRAIL-mediated apoptosis. Intracellular localization of TRAIL-receptors, as observed in many tumor cells, has been associated with oncogenic features, which are distinct from membrane-associated TRAIL-R signaling. Here, analyzing a panel of 354 breast cancer specimens, we found that an unfavorable outcome correlating with cancer-promoting properties of TRAIL-R1, TRAIL-R2, and TRAIL-R4 was most significantly defined by their intracellular distribution and mutual co-expression. A nuclear or cytoplasmic heterogeneous expression pattern correlated with markedly decreased overall survival and discriminated high-risk breast cancer patients from low-risk patients with a homogeneous distribution of expression, i.e., nuclear and cytoplasmic expression. The homogeneous TRAIL-R expression was associated with favorable breast cancer surrogate markers corresponding with excellent survival prognoses at 5 years after diagnosis (hazard ratio, 0.043) and over the complete course of follow-up (hazard ratio, 0.098; both p < 0.001). No associations with specific intrinsic breast cancer subtypes were found. Our data suggest that the determination of intracellular co-expression patterns of TRAIL-R1, TRAIL-R2, and TRAIL-R4 provides an innovative and robust method for risk stratification in breast cancer patients beyond conventional prognostic markers. KEY MESSAGES: A total of 70% of breast cancer specimens show comparably high levels of intracellular TRAIL-Rs. Nuclear or cytoplasmic TRAIL-R co-expression occurs in the majority of tumors. A total of 25% of tumors show a heterogeneous expression of cytoplasmic or nuclear TRAIL-Rs. Patients with a heterogeneous TRAIL-R expression present with poor prognoses. Additive TRAIL-R-based risk stratification comprises different breast cancer subtypes.
Collapse
|
14
|
Toll-Like Receptor 4 Promotes Th17 Lymphocyte Infiltration Via CCL25/CCR9 in Pathogenesis of Experimental Autoimmune Encephalomyelitis. J Neuroimmune Pharmacol 2019; 14:493-502. [PMID: 31065973 DOI: 10.1007/s11481-019-09854-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 04/22/2019] [Indexed: 12/11/2022]
Abstract
Toll-like receptor 4 (TLR4) is a key component in innate immunity and has been linked to central nervous system (CNS) inflammation diseases, such as multiple sclerosis (MS), an inflammatory disorder induced by autoreactive Th17 cells. In our study, we found that TLR4 deficient (TLR4-/-) mice were inadequate to induce experimental autoimmune encephalomyelitis (EAE), characterized by low clinic score and weight loss, alleviative demyelinating, as well as decreased inflammatory cell infiltration in the spinal cord. In the lesion area of EAE mice, loss of TLR4 down-regulated the secretion of inflammatory cytokines and chemokine CCL25. Furthermore, the expression of CCR9 was decreased and chemotactic migration was attenuated in TLR4-/- Th17 cells. Our results demonstrate that TLR4 may mediate Th17 infiltration through CCL25/CCR9 signal during pathogenesis of EAE. Graphical Abstract Immunofluorescent staining of RORγt (green) and CCR9 (red) in spinal cords. TLR4 deficiency down-regulates CCR9 expression in infiltrating lymphocytes.
Collapse
|
15
|
Chrétien S, Zerdes I, Bergh J, Matikas A, Foukakis T. Beyond PD-1/PD-L1 Inhibition: What the Future Holds for Breast Cancer Immunotherapy. Cancers (Basel) 2019; 11:E628. [PMID: 31060337 PMCID: PMC6562626 DOI: 10.3390/cancers11050628] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/01/2019] [Accepted: 05/02/2019] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapy has altered the management of human malignancies, improving outcomes in an expanding list of diseases. Breast cancer - presumably due to its perceived low immunogenicity - is a late addition to this list. Furthermore, most of the focus has been on the triple negative subtype because of its higher tumor mutational load and lymphocyte-enriched stroma, although emerging data show promise on the other breast cancer subtypes as well. To this point the clinical use of immunotherapy is limited to the inhibition of two immune checkpoints, Programmed Cell Death Protein 1 (PD-1) and Cytotoxic T-lymphocyte-associated Protein 4 (CTLA-4). Consistent with the complexity of the regulation of the tumor - host interactions and their lack of reliance on a single regulatory pathway, combinatory approaches have shown improved efficacy albeit at the cost of increased toxicity. Beyond those two checkpoints though, a large number of co-stimulatory or co-inhibitory molecules play major roles on tumor evasion from immunosurveillance. These molecules likely represent future targets of immunotherapy provided that the promise shown in early data is translated into improved patient survival in randomized trials. The biological role, prognostic and predictive implications regarding breast cancer and early clinical efforts on exploiting these immune-related therapeutic targets are herein reviewed.
Collapse
Affiliation(s)
- Sebastian Chrétien
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Ioannis Zerdes
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Jonas Bergh
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Alexios Matikas
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| | - Theodoros Foukakis
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, 171 76, Sweden.
| |
Collapse
|
16
|
Sarink D, Schock H, Johnson T, Chang-Claude J, Overvad K, Olsen A, Tjønneland A, Arveux P, Fournier A, Kvaskoff M, Boeing H, Karakatsani A, Trichopoulou A, La Vecchia C, Masala G, Agnoli C, Panico S, Tumino R, Sacerdote C, van Gils CH, Peeters PHM, Weiderpass E, Agudo A, Rodríguez-Barranco M, Huerta JM, Ardanaz E, Gil L, Kaw KT, Schmidt JA, Dossus L, His M, Aune D, Riboli E, Kaaks R, Fortner RT. Receptor activator of nuclear factor kB ligand, osteoprotegerin, and risk of death following a breast cancer diagnosis: results from the EPIC cohort. BMC Cancer 2018; 18:1010. [PMID: 30348163 PMCID: PMC6196438 DOI: 10.1186/s12885-018-4887-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 10/02/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Receptor activator of nuclear factor kappa-B (RANK)-signaling is involved in tumor growth and spread in experimental models. Binding of RANK ligand (RANKL) to RANK activates signaling, which is inhibited by osteoprotegerin (OPG). We have previously shown that circulating soluble RANKL (sRANKL) and OPG are associated with breast cancer risk. Here we extend these findings to provide the first data on pre-diagnosis concentrations of sRANKL and OPG and risk of breast cancer-specific and overall mortality after a breast cancer diagnosis. METHODS Two thousand six pre- and postmenopausal women with incident invasive breast cancer (1620 (81%) with ER+ disease) participating in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort were followed-up for mortality. Pre-diagnosis concentrations of sRANKL and OPG were quantified in baseline serum samples using an enzyme-linked immunosorbent assay and electrochemiluminescent assay, respectively. Hazard ratios (HRs) and 95% confidence intervals (CIs) for breast cancer-specific and overall mortality were calculated using Cox proportional hazards regression models. RESULTS Especially in women with ER+ disease, higher circulating OPG concentrations were associated with higher risk of breast cancer-specific (quintile 5 vs 1 HR 1.77 [CI 1.03, 3.04]; ptrend 0.10) and overall mortality (q5 vs 1 HR 1.39 [CI 0.94, 2.05]; ptrend 0.02). sRANKL and the sRANKL/OPG ratio were not associated with mortality following a breast cancer diagnosis. CONCLUSIONS High pre-diagnosis endogenous concentrations of OPG, the decoy receptor for RANKL, were associated with increased risk of death after a breast cancer diagnosis, especially in those with ER+ disease. These results need to be confirmed in well-characterized patient cohorts.
Collapse
Affiliation(s)
- Danja Sarink
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Helena Schock
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Theron Johnson
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Kim Overvad
- Section for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Anja Olsen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | | | - Patrick Arveux
- Breast and Gynaecologic Cancer Registry of Côte d’Or, Georges-François Leclerc Comprehensive Cancer Care Centre, Dijon, France
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Agnès Fournier
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Marina Kvaskoff
- Université Paris-Saclay, Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
- Gustave Roussy, Villejuif, France
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Anna Karakatsani
- Hellenic Health Foundation, Athens, Greece
- 2nd Pulmonary Medicine Department, School of Medicine, National and Kapodistrian University of Athens, “ATTIKON” University Hospital, Haidari, Athens, Greece
| | - Antonia Trichopoulou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Department of Hygiene, Epidemiology and Medical Statistics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Carlo La Vecchia
- Hellenic Health Foundation, Athens, Greece
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giovanna Masala
- Cancer Risk Factors and Life-Style Epidemiology Unit, Cancer Research and Prevention Institute – ISP, Florence, Italy
| | - Claudia Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Salvatore Panico
- Dipartimento di Medicine Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Rosario Tumino
- Cancer Registry and Histopathology Department, “Civic M.P.Arezzo” Hospital, Azienda Sanitaria Provinciale, Ragusa, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, Città della Salute e della Scienza University Hospital and Center for Cancer Prevention (CPO), Turin, Italy
| | - Carla H. van Gils
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Petra H. M. Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
- MRC-PHE Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, UK
| | - Elisabete Weiderpass
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø, The Arctic University of Norway, Tromsø, Norway
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
| | - Antonio Agudo
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology-IDIBELL, L’Hospitalet de Llobregat, Barcelona, Spain
| | - Miguel Rodríguez-Barranco
- Escuela Andaluza de Salud Pública. Instituto de Investigación Biosanitaria ibs.GRANADA, Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - José María Huerta
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council, IMIB-Arrixaca, Murcia, Spain
| | - Eva Ardanaz
- CIBER de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Navarra Public Health Institute, Pamplona, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Leire Gil
- Public Health Division of Gipuzkoa, Biodonostia Health Research Institute, San Sebastian, Spain
| | - Kay Tee Kaw
- Cancer Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Julie A. Schmidt
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Laure Dossus
- International Agency for Research on Cancer, Lyon, France
| | - Mathilde His
- International Agency for Research on Cancer, Lyon, France
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, The School of Public Health, Imperial College London, London, UK
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Renée T. Fortner
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|