701
|
Prediction of recurrence-free survival using a protein expression-based risk classifier for head and neck cancer. Oncogenesis 2015; 4:e147. [PMID: 25893634 PMCID: PMC4491610 DOI: 10.1038/oncsis.2015.7] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/28/2015] [Accepted: 02/09/2015] [Indexed: 12/21/2022] Open
Abstract
Loco-regional recurrence in 50% of oral squamous cell carcinoma (OSCC) patients poses major challenge for oncologists. Lack of biomarkers that can predict disease aggressiveness and recurrence risk makes the scenario more dismal. On the basis of our earlier global proteomic analyses we identified five differentially expressed proteins in OSCC. This study aimed to develop protein biomarkers-based prognostic risk prediction model for OSCC. Sub-cellular expression of five proteins, S100A7, heterogeneous nuclear ribonucleoproteinK (hnRNPK), prothymosin α (PTMA), 14-3-3ζ and 14-3-3σ was analyzed by immunohistochemistry in test set (282 Indian OSCCs and 209 normal tissues), correlated with clinic-pathological parameters and clinical outcome over 12 years to develop a risk model for prediction of recurrence-free survival. This risk classifier was externally validated in 135 Canadian OSCC and 96 normal tissues. Biomarker signature score based on PTMA, S100A7 and hnRNPK was associated with recurrence free survival of OSCC patients (hazard ratio=1.11; 95% confidence interval 1.08, 1.13, P<0.001, optimism-corrected c-statistic=0.69) independent of clinical parameters. Biomarker signature score stratified OSCC patients into high- and low-risk groups with significant difference for disease recurrence. The high-risk group had median survival 14 months, and 3-year survival rate of 30%, whereas low-risk group survival probability did not reach 50%, and had 3-year survival rate of 71%. As a powerful predictor of 3-year recurrence-free survival in OSCC patients, the newly developed biomarkers panel risk classifier will facilitate patient counseling for personalized treatment.
Collapse
|
702
|
Fuller T, Pearson M, Peters J, Anderson R. What affects authors' and editors' use of reporting guidelines? Findings from an online survey and qualitative interviews. PLoS One 2015; 10:e0121585. [PMID: 25875918 PMCID: PMC4398362 DOI: 10.1371/journal.pone.0121585] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 02/13/2015] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES To identify and understand, through data from multiple sources, some of the factors that affect authors' and editors' decisions to use reporting guidelines in the publication of health research. DESIGN Mixed methods study comprising an online survey and semi-structured interviews with a sample of authors (online survey: n = 56; response rate = 32%; semi-structured interviews: n = 5) and journal editors (online survey: n = 43; response rate = 27%; semi-structured interviews: n = 6) involved in publishing health and medical research. Participants were recruited from an earlier study examining the effectiveness of the TREND reporting guideline. RESULTS Four types of factors interacted to affect authors' and editors' likelihood of reporting guideline use; individual (e.g., having multiple reasons for use of reporting guidelines); the professional culture in which people work; environmental (e.g., policies of journals); and, practical (e.g., having time to use reporting guidelines). Having multiple reasons for using reporting guidelines was a particularly salient factor in facilitating reporting guidelines use for both groups of participants. CONCLUSIONS Improving the completeness and consistency of reporting of research studies is critical to the integrity and synthesis of health research. The use of reporting guidelines offers one potentially efficient and effective means for achieving this, but decisions to use (or not use) reporting guidelines take many factors into account. These findings could be used to inform future studies that might, for example, test the factors that we have identified within a wider theoretical framework for understanding changes in professional practices. The use of reporting guidelines by senior professionals appears to shape the expectations of what constitutes best practice and can be assimilated into the culture of a field or discipline. Without evidence of effectiveness of reporting guidelines, and sustained, multifaceted efforts to improve reporting, little progress seems likely to be made.
Collapse
Affiliation(s)
- Thomas Fuller
- Collaboration for Leadership in Applied Health Research and Care (CLAHRC) for the South West Peninsula, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Mark Pearson
- Collaboration for Leadership in Applied Health Research and Care (CLAHRC) for the South West Peninsula, University of Exeter Medical School, Exeter, Devon, United Kingdom
- Evidence Synthesis & Modelling for Health Improvement (ESMI), University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Jaime Peters
- Evidence Synthesis & Modelling for Health Improvement (ESMI), University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Rob Anderson
- Collaboration for Leadership in Applied Health Research and Care (CLAHRC) for the South West Peninsula, University of Exeter Medical School, Exeter, Devon, United Kingdom
- Evidence Synthesis & Modelling for Health Improvement (ESMI), University of Exeter Medical School, Exeter, Devon, United Kingdom
| |
Collapse
|
703
|
Starlinger P, Haegele S, Wanek D, Zikeli S, Schauer D, Alidzanovic L, Fleischmann E, Gruenberger B, Gruenberger T, Brostjan C. Plasma thrombospondin 1 as a predictor of postoperative liver dysfunction. Br J Surg 2015; 102:826-36. [PMID: 25871570 DOI: 10.1002/bjs.9814] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/15/2015] [Accepted: 02/26/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Liver regeneration following liver resection involves a complex interplay of growth factors and their antagonists. Thrombospondin 1 has recently been identified as a critical inhibitor of liver regeneration by the activation of transforming growth factor β1 in mice, and preliminary data seem to confirm its relevance in humans. This study aimed to confirm these observations in an independent validation cohort. METHODS Perioperative circulating levels of thrombospondin 1 were measured in patients undergoing liver resection between January 2012 and September 2013. Postoperative liver dysfunction was defined according to the International Study Group of Liver Surgery and classification of morbidity was based on the criteria by Dindo et al. RESULTS In 85 patients (44 major and 41 minor liver resections), plasma levels of thrombospondin 1 increased 1 day after liver resection (mean 51·6 ng/ml before surgery and 68·3 ng/ml on postoperative day 1; P = 0·001). Circulating thrombospondin 1 concentration on the first postoperative day specifically predicted liver dysfunction (area under the receiver operating characteristic (ROC) curve 0·818, P = 0·003) and was confirmed as a significant predictor in multivariable analysis (Exp(B) 1·020, 95 per cent c.i. 1·005 to 1·035; P = 0·009). Patients with a high thrombospondin 1 concentration (over 80 ng/ml) on postoperative day 1 more frequently had postoperative liver dysfunction than those with a lower level (28 versus 2 per cent) and severe morbidity (44 versus 15 per cent), and their length of hospital stay was more than doubled (19·7 versus 9·9 days). CONCLUSION Thrombospondin 1 may prove a helpful clinical marker to predict postoperative liver dysfunction as early as postoperative day 1.
Collapse
Affiliation(s)
- P Starlinger
- Departments of Surgery, General Hospital, Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
704
|
Hu ZD, Huang YL, Qin BD, Tang QQ, Yang M, Ma N, Fu HT, Wei TT, Zhong RQ. Prognostic value of neutrophil to lymphocyte ratio for gastric cancer. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:50. [PMID: 25861605 DOI: 10.3978/j.issn.2305-5839.2015.03.26] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 03/02/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND Although the prognostic value of the neutrophil to lymphocyte ratio (NLR) in gastric cancer (GC) patients has been investigated by many studies, the results are heterogeneous. The objective of this systematic review is to ascertain the prognostic value of NLR in GC patients. METHODS PubMed and Embase were retrieved to identify potential studies published before 8 June, 2014. Newcastle-Ottawa Scale (NOS) for cohort study was used to assess the quality of all eligible studies. RESULTS Of the 20 studies included in this systematic review, 17 studies investigated the effect of NLR on overall survival (OS), 11 studies reported that NLR negatively affected OS in their multivariante analysis, and 16 studies reported that NLR negatively affected OS in univariate analysis. Three studies investigated the effect of NLR on progression-free survival (PFS), reporting that increased NLR was associated with worse PFS. Four studies investigated the effect of NLR on disease-free survival (DFS), two of which reported that increased NLR was associated with worse DFS. Two studies investigated the effect of NLR on disease special survival (DSS), but neither observed any significant association between NLR and DSS. The major design deficiencies of the studies available were retrospective data collection, inadequacy of follow-up cohorts, and unavailability of the method used for outcome assessment. CONCLUSIONS Based on the above findings, we conclude that NLR may be a useful prognostic index (PI) for GC. In addition, future studies with prospective design, long-term follow-up and fully adjusted confounding factors are needed to rigorously assess the prognostic value of NLR for GC.
Collapse
Affiliation(s)
- Zhi-De Hu
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Yuan-Lan Huang
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Bao-Dong Qin
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Qing-Qin Tang
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Min Yang
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Ning Ma
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Hai-Tao Fu
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Ting-Ting Wei
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| | - Ren-Qian Zhong
- 1 Department of Laboratory Diagnosis, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China ; 2 Department of Laboratory Medicine, General Hospital of Ji'nan Military Command Region, Ji'nan 250031, China ; 3 Department of Laboratory Medicine, No. 455 Hospital of Chinese People's Liberation Army, Shanghai 200052, China
| |
Collapse
|
705
|
Sun S, Liang X, Zhang X, Liu T, Shi Q, Song Y, Jiang Y, Wu H, Jiang Y, Lu X, Pang D. Phosphoglycerate kinase-1 is a predictor of poor survival and a novel prognostic biomarker of chemoresistance to paclitaxel treatment in breast cancer. Br J Cancer 2015; 112:1332-9. [PMID: 25867275 PMCID: PMC4402453 DOI: 10.1038/bjc.2015.114] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 02/16/2015] [Accepted: 03/03/2015] [Indexed: 02/07/2023] Open
Abstract
Background: Phosphoglycerate kinase-1 (PGK1) has been recently documented in various malignancies; however, the molecular mechanisms of the variable PGK1 expression and its clinical significance in terms of survival status remain unclear. Methods: Real-time quantitative PCR (real-time qPCR) and western blotting were used to verify PGK1 expression in 46 fresh breast cancer tissues and matched normal tissues. A tissue microarray (TMA) comprising 401 breast cancer tissues and 123 matched normal tissues was investigated by immunohistochemistry for PGK1 expression. Then, the correlation between PGK1 expression and the clinicopathologic features was analysed. Results: PGK1 mRNA and protein expression were significantly increased in breast cancer tissues compared with that in normal breast tissues. High PGK1 expression was significantly associated with higher histologic grade (P=0.009) and positive status of ER (P=0.004), Her-2 (P=0.026) and P53 (P=0.012). High levels of PGK1 expression were associated with worse overall survival (OS, P=0.02). Furthermore, patients who underwent paclitaxel chemotherapy with high levels PGK1 expression had shorter OS than did those with low levels of PGK1 expression (P<0.001). Multivariate analysis indicated that PGK1 (P=0.001) was an independent predictor in the patients treated with paclitaxel. Conclusions: PGK1 is a prognostic biomarker of chemoresistance to paclitaxel treatment in breast cancer.
Collapse
Affiliation(s)
- S Sun
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - X Liang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - X Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - T Liu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Q Shi
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Y Song
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Y Jiang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - H Wu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - Y Jiang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - X Lu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China
| | - D Pang
- 1] Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150081, China [2] North China Translational Medicine Research and Cooperation Center (NTMRC), Harbin 150081, China
| |
Collapse
|
706
|
Insights into the application of let-7 family as promising biomarker in cancer screening. Tumour Biol 2015; 36:5233-9. [PMID: 25801240 DOI: 10.1007/s13277-015-3180-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 01/27/2015] [Indexed: 10/24/2022] Open
Abstract
Cancer is a leading cause of death worldwide with its low 5-year survival rate. Studies on the accuracy of let-7 family for human cancers have inconsistent conclusions, leading us to conduct this meta-analysis. This meta-analysis comprised of 11 studies from eight articles involving 805 cancer patients and 483 controls. The pooled parameters were as follows: sensitivity, 77 % (95 % confidence interval (CI) 73-81 %); specificity, 80 % (95 % CI 68-88 %); positive likelihood ratio (PLR), 3.8; negative likelihood ratio (NLR), 0.29; and diagnostic odds ratio (DOR) 13.0. In addition, we plotted the SROC and calculated the area under the curve (AUC) of 0.81 (95 % CI 0.78-0.84), which indicated a relatively high descriptive accuracy. In summary, our data suggested that let-7 family might be applied in noninvasive screening tests for human cancers, which needed to be validated in further large-scale studies.
Collapse
|
707
|
Sabatier R, Finetti P, Mamessier E, Adelaide J, Chaffanet M, Ali HR, Viens P, Caldas C, Birnbaum D, Bertucci F. Prognostic and predictive value of PDL1 expression in breast cancer. Oncotarget 2015; 6:5449-64. [PMID: 25669979 PMCID: PMC4467160 DOI: 10.18632/oncotarget.3216] [Citation(s) in RCA: 369] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 12/26/2014] [Indexed: 12/30/2022] Open
Abstract
Expression of programmed cell death receptor ligand 1 (PDL1) has been scarcely studied in breast cancer. Recently PD1/PDL1-inhibitors have shown promising results in different carcinomas with correlation between PDL1 tumor expression and responses. We retrospectively analyzed PDL1 mRNA expression in 45 breast cancer cell lines and 5,454 breast cancers profiled using DNA microarrays. Compared to normal breast samples, PDL1 expression was upregulated in 20% of clinical samples and 38% of basal tumors. High expression was associated with poor-prognosis features (large tumor size, high grade, ER-negative, PR-negative, ERBB2-positive status, high proliferation, basal and ERBB2-enriched subtypes). PDL1 upregulation was associated with biological signs of strong cytotoxic local immune response. PDL1 upregulation was not associated with survival in the whole population, but was associated with better metastasis-free and overall specific survivals in basal tumors, independently of clinicopathological features. Pathological complete response after neoadjuvant chemotherapy was higher in case of PDL1 upregulation (50% versus 21%). In conclusion, PDL1 upregulation, more frequent in basal breast cancers, was associated with increased T-cell cytotoxic immune response. In this aggressive subtype, upregulation was associated with better survival and response to chemotherapy. Reactivation of dormant tumor-infiltrating lymphocytes by PDL1-inhibitors could represent promising strategy in PDL1-upregulated basal breast cancer.
Collapse
MESH Headings
- B7-H1 Antigen/genetics
- Biomarkers, Tumor/genetics
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Carcinoma, Ductal, Breast/genetics
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/secondary
- Carcinoma, Lobular/genetics
- Carcinoma, Lobular/mortality
- Carcinoma, Lobular/secondary
- Carcinoma, Medullary/genetics
- Carcinoma, Medullary/mortality
- Carcinoma, Medullary/secondary
- Comparative Genomic Hybridization
- Female
- Follow-Up Studies
- Gene Expression Profiling
- Humans
- Lymphatic Metastasis
- Middle Aged
- Neoplasm Grading
- Neoplasm Invasiveness
- Neoplasm Staging
- Oligonucleotide Array Sequence Analysis
- Prognosis
- Retrospective Studies
- Survival Rate
Collapse
Affiliation(s)
- Renaud Sabatier
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
- Département d'Oncologie Médicale, CRCM, Institut Paoli-Calmettes, Marseille, France
- Faculté de Médecine, Aix-Marseille Université, Marseille, France
| | - Pascal Finetti
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Emilie Mamessier
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - José Adelaide
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Max Chaffanet
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - Hamid Raza Ali
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Patrice Viens
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
- Département d'Oncologie Médicale, CRCM, Institut Paoli-Calmettes, Marseille, France
- Faculté de Médecine, Aix-Marseille Université, Marseille, France
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Daniel Birnbaum
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
| | - François Bertucci
- Département d'Oncologie Moléculaire, “Equipe labellisée Ligue Contre le Cancer”, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes, INSERM UMR1068, CNRS UMR725, Marseille, France
- Département d'Oncologie Médicale, CRCM, Institut Paoli-Calmettes, Marseille, France
- Faculté de Médecine, Aix-Marseille Université, Marseille, France
| |
Collapse
|
708
|
Qi XW, Zhang F, Wu H, Liu JL, Zong BG, Xu C, Jiang J. Wilms' tumor 1 (WT1) expression and prognosis in solid cancer patients: a systematic review and meta-analysis. Sci Rep 2015; 5:8924. [PMID: 25748047 PMCID: PMC4352850 DOI: 10.1038/srep08924] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/10/2015] [Indexed: 01/08/2023] Open
Abstract
Though proposed as a promising target antigen for cancer immunotherapy, the prognostic value of Wilms' tumor 1 (WT1) in solid tumors remains inconclusive. Here, we report a systematic review and meta-analysis of the association between WT1 expression and prognosis in solid tumors. PubMed, Web of Science and Google Scholar were searched to identify studies exploring the impact of WT1 on clinical outcomes, including overall survival (OS), disease-specific survival (DSS), disease-free survival (DFS), relapse/recurrence-free survival (RFS) or progression-free survival (PFS), in solid cancer patients. Hazard ratio (HR) and 95% confidence interval (CI) were applied to assess the strength of these associations. Finally, a total of 29 eligible studies with 4090 patients were identified for qualitative analysis, and 22 studies with 3620 patients were enrolled for quantitative synthesis. Overall, positive expression of WT1 was significantly associated with worse OS (metaHR = 1.48, 95% CI = 1.11–1.97) and DFS/RFS/PFS (metaHR = 2.14, 95% CI = 1.42–3.21). Subgroup analyses showed that WT1 positive expression could independently predict unfavorable DFS/RFS/PFS (metaHR = 1.86, 95%CI = 1.04–3.35). In summary, our study suggests that WT1 may be a potential marker to predict DFS/RFS/PFS in solid tumor patients. Further studies are needed to confirm the role of WT1 expression in clinical practice.
Collapse
Affiliation(s)
- Xiao-wei Qi
- 1] Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China [2] Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Fan Zhang
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Hong Wu
- Department of Oncology, Luzhou Medical College, Luzhou, Sichuan, China
| | - Jun-lan Liu
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Bei-ge Zong
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Chuan Xu
- 1] Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China [2] Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Jun Jiang
- Breast Disease Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
709
|
Colbert LE, Fisher SB, Balci S, Saka B, Chen Z, Kim S, El-Rayes BF, Adsay NV, Maithel SK, Landry JC, Curran WJ. High nuclear hypoxia-inducible factor 1 alpha expression is a predictor of distant recurrence in patients with resected pancreatic adenocarcinoma. Int J Radiat Oncol Biol Phys 2015; 91:631-9. [PMID: 25596110 PMCID: PMC5746186 DOI: 10.1016/j.ijrobp.2014.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 10/29/2014] [Accepted: 11/04/2014] [Indexed: 01/19/2023]
Abstract
PURPOSE To evaluate nuclear hypoxia-inducible factor 1α (HIF-1α) expression as a prognostic factor for distant recurrence (DR) and local recurrence (LR) after pancreatic adenocarcinoma resection. METHODS AND MATERIALS Tissue specimens were collected from 98 patients with pancreatic adenocarcinoma who underwent resection without neoadjuvant therapy between January 2000 and December 2011. Local recurrence was defined as radiographic or pathologic evidence of progressive disease in the pancreas, pancreatic bed, or associated nodal regions. Distant recurrence was defined as radiographically or pathologically confirmed recurrent disease in other sites. Immunohistochemical staining was performed and scored by an independent pathologist blinded to patient outcomes. High HIF-1α overall expression score was defined as high percentage and intensity staining and thus score >1.33. Univariate analysis was performed for HIF-1α score with LR alone and with DR. Multivariate logistic regression was used to determine predictors of LR and DR. RESULTS Median follow-up time for all patients was 16.3 months. Eight patients (8%) demonstrated isolated LR, 26 patients (26.5%) had isolated DR, and 13 patients had both LR and DR. Fifty-three patients (54%) had high HIF-1α expression, and 45 patients (46%) had low HIF-1α expression. High HIF-1α expression was significantly associated with DR (P=.03), and low HIF-1α expression was significantly associated with isolated LR (P=.03). On multivariate logistic regression analysis, high HIF-1α was the only significant predictor of DR (odds ratio 2.46 [95% confidence interval 1.06-5.72]; P=.03). In patients with a known recurrence, an HIF-1α score ≥2.5 demonstrated a specificity of 100% for DR. CONCLUSIONS High HIF-1α expression is a significant predictor of distant failure versus isolated local failure in patients undergoing resection of pancreatic adenocarcinoma. Expression of HIF-1α may have utility in determining candidates for adjuvant local radiation therapy and systemic chemotherapy.
Collapse
Affiliation(s)
- Lauren E Colbert
- Department of Radiation Oncology, Emory University, Atlanta, Georgia; Winship Cancer Institute, Emory University, Atlanta, Georgia; Memorial Sloan-Kettering Cancer Center, New York, New York
| | - Sarah B Fisher
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Division of Surgical Oncology, Department of Surgery, Emory University, Atlanta, Georgia
| | - Serdar Balci
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Department of Pathology, Emory University, Atlanta, Georgia
| | - Burcu Saka
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Department of Pathology, Emory University, Atlanta, Georgia
| | - Zhengjia Chen
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Biostatistics and Bioinformatics Shared Resource, Emory University, Atlanta, Georgia
| | - Sungjin Kim
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Biostatistics and Bioinformatics Shared Resource, Emory University, Atlanta, Georgia
| | - Bassel F El-Rayes
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Department of Hematology and Medical Oncology, Emory University, Atlanta, Georgia
| | - N Volkan Adsay
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Department of Pathology, Emory University, Atlanta, Georgia; Biostatistics and Bioinformatics Shared Resource, Emory University, Atlanta, Georgia
| | - Shishir K Maithel
- Winship Cancer Institute, Emory University, Atlanta, Georgia; Division of Surgical Oncology, Department of Surgery, Emory University, Atlanta, Georgia; Biostatistics and Bioinformatics Shared Resource, Emory University, Atlanta, Georgia
| | - Jerome C Landry
- Department of Radiation Oncology, Emory University, Atlanta, Georgia; Winship Cancer Institute, Emory University, Atlanta, Georgia; Biostatistics and Bioinformatics Shared Resource, Emory University, Atlanta, Georgia
| | - Walter J Curran
- Department of Radiation Oncology, Emory University, Atlanta, Georgia; Winship Cancer Institute, Emory University, Atlanta, Georgia; Biostatistics and Bioinformatics Shared Resource, Emory University, Atlanta, Georgia.
| |
Collapse
|
710
|
Sun DW, Zhang YY, Qi Y, Zhou XT, Lv GY. Prognostic significance of MMP-7 expression in colorectal cancer: a meta-analysis. Cancer Epidemiol 2015; 39:135-42. [PMID: 25677090 DOI: 10.1016/j.canep.2015.01.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 01/07/2015] [Accepted: 01/11/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Matrix metalloproteinase-7 (MMP-7) is a member of the family of matrix metalloproteinases (MMPs); it is associated with invasive tumor growth and distant metastasis in colorectal cancer (CRC). However, the prognostic value of MMP-7 in CRC remains controversial. Thus, we conducted a meta-analysis to investigate the prognostic value of MMP-7 expression in CRC. METHODS We systematically searched for studies evaluating the relationship between MMP-7 expression and the outcome of CRC patients in PubMed, the Cochrane library, Embase, Google Scholar, and Wanfang databases updated to August 2014. Studies were assessed for quality using REMARK (REporting recommendations for tumor MARKer prognostic studies). Hazard ratios (HRs) with 95% confidence intervals (95%CIs) were pooled to estimate the hazard for overall survival (OS) and disease-free survival (DFS), and odds ratios (ORs) with 95%CIs were pooled to estimate the impact of MMP-7 on the 5-year survival rate. RESULTS In total, 2985 patients from 17 studies were included in this meta-analysis. Our results showed that MMP-7 over-expression predicted poor OS (HR=3.57, 95%CI 2.21-5.75, P=0.000, random-effect model), poor DFS (HR=2.49, 95%CI 1.73-3.57, P=0.000, fixed-effect model), and decreased 5-year survival rate (OR=0.26, 95%CI 0.19-0.37, P=0.000, fixed-effect model). Therefore, MMP-7 is an indicator of poor survival and high recurrence rate in CRC patients. CONCLUSION MMP-7 can serve as a prognostic indicator and a potential novel target for treatment in CRC patients. More well-designed prospective studies with better methodology for MMP-7 assessment are required to clarify the prognostic significance of MMP-7 expression in CRC patients.
Collapse
Affiliation(s)
- Da-wei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Bethune Hospital, Jilin University, Changchun 130021, China
| | - Ying-yi Zhang
- Department of Chemotherapy, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, China
| | - Yue Qi
- Department of Hepatobiliary and Pancreatic Medicine, The First Bethune Hospital, Jilin University, Changchun 130021, China
| | - Xing-tong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, China
| | - Guo-yue Lv
- Department of Hepatobiliary and Pancreatic Surgery, The First Bethune Hospital, Jilin University, Changchun 130021, China.
| |
Collapse
|
711
|
Kumar M, Srivastava G, Kaur J, Assi J, Alyass A, Leong I, MacMillan C, Witterick I, Shukla NK, Thakar A, Duggal R, Roychoudhury A, Sharma MC, Walfish PG, Chauhan SS, Ralhan R. Prognostic significance of cytoplasmic S100A2 overexpression in oral cancer patients. J Transl Med 2015; 13:8. [PMID: 25591983 PMCID: PMC4324434 DOI: 10.1186/s12967-014-0369-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 12/18/2014] [Indexed: 12/16/2022] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) patients are at high risk of loco-regional recurrence and 5-year survival rates are about 50%. Identification of patients at high risk of recurrence will enable rigorous personalized post-treatment management. Most novel biomarkers have failed translation for clinical use because of their limited successful validation in external patient cohorts. The aim of this study was to determine the prognostic significance of alterations in sub-cellular expression of S100A2, a pro-tumorigenic calcium binding protein, identified as a candidate biomarker in our proteomic analysis in OSCC and validation of its clinical utility in an external cohort. Methods In a retrospective study, immunohistochemical analysis of S100A2 was carried out in 235 Indian OSCC (Test set) and 129 normal oral tissues, correlated with clinicopathological parameters and disease outcome over 122 months for OSCC patients following the REMARK criteria. The findings were validated in an external cohort (Validation set 115 Canadian OSCC and 51 normal tissues) and data analyzed using the R package. Results Significant increase in cytoplasmic and decrease in nuclear S100A2 expression was observed in OSCC in comparison with normal tissues. Cox multivariable regression analysis internally and externally validated cytoplasmic S100A2 association with tumor recurrence. Kaplan Meier analysis of patients stratified to high and low risk groups showed significantly different recurrence free survival (Test set- log rank test, p = 0.005, median survival 16 and 69 months respectively and Validation set - p < 0.00001, median survival 9.4 and 59.9 months respectively); 86% and 81% of patients who had recurrence were correctly stratified into the high risk group. Seventy percent and 81% patients stratified into low risk group did not show cancer recurrence within 1 year in Test and Validation sets. Conclusions Our study provided clinical evidence for the potential of cytoplasmic S100A2 overexpression as a predictor of recurrence risk in OSCC patients. A unique translational aspect of our study is validation of S100A2 as prognostic marker in two independent cohorts (Canadian and Indian) suggesting this protein is likely to find widespread utility in clinical practice for identifying oral cancer patients at high risk of disease recurrence.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Gunjan Srivastava
- Alex and Simona Shnaider Laboratory of Molecular Oncology, Mount Sinai Hospital, Toronto, Ontario, Canada.
| | - Jatinder Kaur
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Jasmeet Assi
- Alex and Simona Shnaider Laboratory of Molecular Oncology, Mount Sinai Hospital, Toronto, Ontario, Canada.
| | - Akram Alyass
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Canada.
| | - Iona Leong
- Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada. .,Department of Oral Pathology and Oral Medicine, Faculty of Dentistry, University of Toronto, 124 Edward Street, Toronto, Ontario, M5G 1G6, Canada.
| | - Christina MacMillan
- Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada.
| | - Ian Witterick
- Department of Otolaryngology - Head and Neck Surgery, Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada. .,Department of Otolaryngology - Head and Neck Surgery, University of Toronto, 190 Elizabeth Street, Toronto, Ontario, M5G 2N2, Canada.
| | - Nootan Kumar Shukla
- Department of Surgery, Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India.
| | - Alok Thakar
- Department of Otorhinolaryngology, All India Institute of Medical Sciences, New Delhi, India.
| | - Ritu Duggal
- Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India.
| | - Ajoy Roychoudhury
- Centre for Dental Education and Research, All India Institute of Medical Sciences, New Delhi, India.
| | - Mehar Chand Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Paul G Walfish
- Alex and Simona Shnaider Laboratory of Molecular Oncology, Mount Sinai Hospital, Toronto, Ontario, Canada. .,Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada. .,Department of Otolaryngology - Head and Neck Surgery, Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada. .,Department of Otolaryngology - Head and Neck Surgery, University of Toronto, 190 Elizabeth Street, Toronto, Ontario, M5G 2N2, Canada. .,Department of Medicine, Endocrine Division, Mount Sinai Hospital and University of Toronto, Toronto, Ontario, M5G 1X5, Canada.
| | - Shyam Singh Chauhan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, 110029, India.
| | - Ranju Ralhan
- Alex and Simona Shnaider Laboratory of Molecular Oncology, Mount Sinai Hospital, Toronto, Ontario, Canada. .,Department of Pathology & Laboratory Medicine, Mount Sinai Hospital, Joseph & Wolf Lebovic Health Complex, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada. .,Department of Otolaryngology - Head and Neck Surgery, Joseph and Mildred Sonshine Family Centre for Head and Neck Diseases, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario, M5G 1X5, Canada. .,Department of Otolaryngology - Head and Neck Surgery, University of Toronto, 190 Elizabeth Street, Toronto, Ontario, M5G 2N2, Canada.
| |
Collapse
|
712
|
Zhang ZZ, Hua R, Zhang JF, Zhao WY, Zhao EH, Tu L, Wang CJ, Cao H, Zhang ZG. TEM7 (PLXDC1), a key prognostic predictor for resectable gastric cancer, promotes cancer cell migration and invasion. Am J Cancer Res 2015; 5:772-781. [PMID: 25973314 PMCID: PMC4396023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Accepted: 01/05/2015] [Indexed: 06/04/2023] Open
Abstract
Tumor endothelial marker 7 (TEM7) is a new candidate of molecular target for antiangiogenic therapy. This study aims to evaluate its expression in gastric cancer (GC) and to explore the correlation between its expression and the clinical outcome of patients. Expression of TEM7 was analyzed in both tumor tissues and cell lines of GC by real-time quantitative RT-PCR (qRT-PCR) and Western blot. RNA interference (RNAi) approaches were used to investigate the biological functions of TEM7. The effects of TEM7 on cell migration and invasion were evaluated by Transwell assays. In vitro experiments revealed that TEM7 was significantly overexpressed in GC cell lines (N87, AGS and SGC-7901) by 2-fold to 4-fold, and knockdown of TEM7 could significantly inhibit cancer cell migration and invasion. For GC patients, TEM7 gene expression was elevated in tumors in most cases (25/31), and its expression was closely correlated with tumor differentiation, depth of cancer invasion, lymphatic metastasis and TNM stage. The overall survival of TEM7 (-) group was significantly higher than that of TEM7 (+) group (P = 0.048) and TEM7 (++) group (P = 0.003). TEM7 is highly expressed in GC and is likely correlated with tumor invasion and migration, and thus its expression is closely related to the clinical outcome of patients.
Collapse
Affiliation(s)
- Zi-Zhen Zhang
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - Rong Hua
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - Jun-Feng Zhang
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - Wen-Yi Zhao
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - En-Hao Zhao
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - Lin Tu
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - Chao-Jie Wang
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - Hui Cao
- Department of General Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200127, PR China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong UniversityShanghai 200240, China
| |
Collapse
|
713
|
Hansen TF, Carlsen AL, Heegaard NHH, Sørensen FB, Jakobsen A. Changes in circulating microRNA-126 during treatment with chemotherapy and bevacizumab predicts treatment response in patients with metastatic colorectal cancer. Br J Cancer 2015; 112:624-9. [PMID: 25584492 PMCID: PMC4333496 DOI: 10.1038/bjc.2014.652] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 10/25/2014] [Accepted: 12/03/2014] [Indexed: 12/19/2022] Open
Abstract
Background: This study investigated the predictive value of circulating microRNA-126 (cir-miRNA-126) in patients with metastatic colorectal cancer (mCRC) treated with first-line chemotherapy combined with bevacizumab. Methods: The study included 68 patients. Blood samples (plasma) were collected before the treatment initiation, at the first clinical evaluation after 3 weeks and at progression. Levels of cir-miRNA-126 were determined by qRT–PCR after purification of total RNA from plasma. Primary clinical end points were response rates evaluated according to the Response Evaluation Criteria In Solid Tumours (RECIST) and progression-free survival (PFS). Results: Changes in circulating miRNA-126 during treatment were predictive of tumour response. Non-responding patients had a median increase in cir-miRNA-126 of 0.244 (95% confidence interval (CI), 0.050–0.565) compared with a median decrease of −0.374 (95% CI, −0.472 to −0.111) in the responding patients, P=0.002. A significant positive correlation was demonstrated by comparing the changes in tumour size with the changes in cir-miRNA-126, r=0.48, P=0.0001. Grouping the patients according to the changes in cir-miRNA-126 disclosed a borderline significant separation of the groups in the PFS analysis favouring patients with decreasing miRNA-126 levels, hazard ratio (HR) 0.60 (95% CI, 0.33–1.09), P=0.07. Conclusions: The present results indicate that changes in cir-miRNA-126 during treatment are related to the response to chemotherapy and bevacizumab in patients with mCRC, thus representing a possible biomarker for the resistance to anti-angiogenic containing treatments.
Collapse
Affiliation(s)
- T F Hansen
- 1] Department of Oncology, Vejle Hospital, Part of Lillebaelt Hospital, Vejle, Denmark [2] Institute of Regional Health Research, University of Southern Denmark, Denmark
| | - A L Carlsen
- Department of Clinical Biochemistry, Immunology & Genetics, Statens Serum Institute, Copenhagen, Denmark
| | - N H H Heegaard
- Department of Clinical Biochemistry, Immunology & Genetics, Statens Serum Institute, Copenhagen, Denmark
| | - F B Sørensen
- 1] Institute of Regional Health Research, University of Southern Denmark, Denmark [2] Department of Clinical Pathology, Vejle Hospital, Part of Lillebaelt Hospital, Vejle, Denmark
| | - A Jakobsen
- 1] Department of Oncology, Vejle Hospital, Part of Lillebaelt Hospital, Vejle, Denmark [2] Institute of Regional Health Research, University of Southern Denmark, Denmark
| |
Collapse
|
714
|
Hidalgo M, Cascinu S, Kleeff J, Labianca R, Löhr JM, Neoptolemos J, Real FX, Van Laethem JL, Heinemann V. Addressing the challenges of pancreatic cancer: future directions for improving outcomes. Pancreatology 2015; 15:8-18. [PMID: 25547205 DOI: 10.1016/j.pan.2014.10.001] [Citation(s) in RCA: 356] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 10/01/2014] [Accepted: 10/03/2014] [Indexed: 12/11/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), which accounts for more than 90% of all pancreatic tumours, is a devastating malignancy with an extremely poor prognosis, as shown by a 1-year survival rate of around 18% for all stages of the disease. The low survival rates associated with PDAC primarily reflect the fact that tumours progress rapidly with few specific symptoms and are thus at an advanced stage at diagnosis in most patients. As a result, there is an urgent need to develop accurate markers of pre-invasive pancreatic neoplasms in order to facilitate prediction of cancer risk and to help diagnose the disease at an earlier stage. However, screening for early diagnosis of prostate cancer remains challenging and identifying a highly accurate, low-cost screening test for early PDAC for use in clinical practice remains an important unmet need. More effective therapies are also crucial in PDAC, since progress in identifying novel therapies has been hampered by the genetic complexity of the disease and treatment remains a major challenge. Presently, the greatest step towards improved treatment efficacy has been made in the field of palliative chemotherapy by introducing FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan and oxaliplatin) and gemcitabine/nab-paclitaxel. Strategies designed to raise the profile of PDAC in research and clinical practice are a further requirement in order to ensure the best treatment for patients. This article proposes a number of approaches that may help to accelerate progress in treating patients with PDAC, which, in turn, may be expected to improve the quality of life and survival for those suffering from this devastating disease.
Collapse
Affiliation(s)
- Manuel Hidalgo
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain.
| | - Stefano Cascinu
- Department of Medical Oncology, University of Ancona, Ancona, Italy
| | - Jörg Kleeff
- Department of General Surgery, Technische Universität München, Munich, Germany
| | | | - J-Matthias Löhr
- Department of Clinical Science, Intervention and Technology, Karolinska Institute, Stockholm, Sweden
| | - John Neoptolemos
- National Institutes of Health Research Liverpool Pancreas Biomedical Research Unit and Cancer Research UK Liverpool Clinical Trials Unit Director, University of Liverpool and Royal Liverpool University Hospital, Liverpool, UK
| | - Francisco X Real
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid and Universitat Pompeu Fabra, Barcelona, Spain
| | - Jean-Luc Van Laethem
- Department of Gastroenterology-GI Cancer Unit, Erasme University Hospital, Brussels, Belgium
| | - Volker Heinemann
- Comprehensive Cancer Centre Munich, Klinikum der Universität München, Munich, Germany
| |
Collapse
|
715
|
Eriksson H, Frohm-Nilsson M, Järås J, Kanter-Lewensohn L, Kjellman P, Månsson-Brahme E, Vassilaki I, Hansson J. Prognostic factors in localized invasive primary cutaneous malignant melanoma: results of a large population-based study. Br J Dermatol 2015; 172:175-86. [PMID: 24910143 DOI: 10.1111/bjd.13171] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2014] [Indexed: 01/19/2023]
Abstract
BACKGROUND The prognostic impact of several histopathological prognostic features in cutaneous malignant melanoma (CMM) remains controversial. OBJECTIVES To assess the independent prognostic value of mitotic rate, regression, tumour-infiltrating lymphocytes (TILs) and growth phase in primary stage I and II CMMs. METHODS Clinicohistopathological data were obtained from the Stockholm-Gotland registry for 4237 patients diagnosed with an incident primary stage I or II CMM followed up to December 2011. The risk of CMM-specific death was evaluated by a Cox regression model. RESULTS A mitotic rate of 1-10 mitoses per mm(2) [hazard ratio (HR) 1·69, 95% confidence interval (CI) 1·16-2·45] and > 10 mitoses per mm(2) (HR 2·27, 95% CI 1·46-3·52) were significant; TILs and regression were not. A more detailed analysis of data assessed between 1989 and 1995 confirmed significantly increased HRs for the presence vs. absence of mitoses (HR1-5/mm² 2·25, 95% CI 1·36-3·76; HR6-10/mm² 2·34, 95% CI 1·23-4·44; HR> 10/mm² 2·64, 95% CI 1·39-4·99). Other prognosticators were increasing T-stage vs. T1, presence of ulceration and presence of vertical growth phase (VGP). In T1 CMMs, an increasing tumour thickness vs. < 0·7 mm (HR0·7-0·8 mm 2·24, 95% CI 1·24-4·04; HR>0·8 mm 2·92, 95% CI 1·57-5·43) and presence of ulceration were significantly associated with higher HRs; mitotic rate, TILs, regression and growth phase were not. CONCLUSIONS Determinants of increased risk of CMM death in stage I and II CMMs were increasing T-stage, presence of ulceration, presence of mitoses and VGP. This was not found for TILs or regression.
Collapse
Affiliation(s)
- H Eriksson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
716
|
Vollan HKM, Rueda OM, Chin SF, Curtis C, Turashvili G, Shah S, Lingjærde OC, Yuan Y, Ng CK, Dunning MJ, Dicks E, Provenzano E, Sammut S, McKinney S, Ellis IO, Pinder S, Purushotham A, Murphy LC, Kristensen VN, Brenton JD, Pharoah PDP, Børresen-Dale AL, Aparicio S, Caldas C. A tumor DNA complex aberration index is an independent predictor of survival in breast and ovarian cancer. Mol Oncol 2015; 9:115-27. [PMID: 25169931 PMCID: PMC4286124 DOI: 10.1016/j.molonc.2014.07.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 07/23/2014] [Accepted: 07/25/2014] [Indexed: 01/27/2023] Open
Abstract
Complex focal chromosomal rearrangements in cancer genomes, also called "firestorms", can be scored from DNA copy number data. The complex arm-wise aberration index (CAAI) is a score that captures DNA copy number alterations that appear as focal complex events in tumors, and has potential prognostic value in breast cancer. This study aimed to validate this DNA-based prognostic index in breast cancer and test for the first time its potential prognostic value in ovarian cancer. Copy number alteration (CNA) data from 1950 breast carcinomas (METABRIC cohort) and 508 high-grade serous ovarian carcinomas (TCGA dataset) were analyzed. Cases were classified as CAAI positive if at least one complex focal event was scored. Complex alterations were frequently localized on chromosome 8p (n = 159), 17q (n = 176) and 11q (n = 251). CAAI events on 11q were most frequent in estrogen receptor positive (ER+) cases and on 17q in estrogen receptor negative (ER-) cases. We found only a modest correlation between CAAI and the overall rate of genomic instability (GII) and number of breakpoints (r = 0.27 and r = 0.42, p < 0.001). Breast cancer specific survival (BCSS), overall survival (OS) and ovarian cancer progression free survival (PFS) were used as clinical end points in Cox proportional hazard model survival analyses. CAAI positive breast cancers (43%) had higher mortality: hazard ratio (HR) of 1.94 (95%CI, 1.62-2.32) for BCSS, and of 1.49 (95%CI, 1.30-1.71) for OS. Representations of the 70-gene and the 21-gene predictors were compared with CAAI in multivariable models and CAAI was independently significant with a Cox adjusted HR of 1.56 (95%CI, 1.23-1.99) for ER+ and 1.55 (95%CI, 1.11-2.18) for ER- disease. None of the expression-based predictors were prognostic in the ER- subset. We found that a model including CAAI and the two expression-based prognostic signatures outperformed a model including the 21-gene and 70-gene signatures but excluding CAAI. Inclusion of CAAI in the clinical prognostication tool PREDICT significantly improved its performance. CAAI positive ovarian cancers (52%) also had worse prognosis: HRs of 1.3 (95%CI, 1.1-1.7) for PFS and 1.3 (95%CI, 1.1-1.6) for OS. This study validates CAAI as an independent predictor of survival in both ER+ and ER- breast cancer and reveals a significant prognostic value for CAAI in high-grade serous ovarian cancer.
Collapse
Affiliation(s)
- Hans Kristian Moen Vollan
- Cancer Research UK, Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway; The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Department of Oncology, Division for Surgery, Cancer and Transplantation, Oslo University Hospital, The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway
| | - Oscar M Rueda
- Cancer Research UK, Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Suet-Feung Chin
- Cancer Research UK, Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Christina Curtis
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Gulisa Turashvili
- Department of Pathology and Laboratory Medicine, University of British Colombia, Vancouver, British Colombia V6T 2B5, Canada; Molecular Oncology, British Colombia Cancer Research Center, Vancouver, British Columbia V5Z 1L3, Canada
| | - Sohrab Shah
- Department of Pathology and Laboratory Medicine, University of British Colombia, Vancouver, British Colombia V6T 2B5, Canada; Molecular Oncology, British Colombia Cancer Research Center, Vancouver, British Columbia V5Z 1L3, Canada
| | - Ole Christian Lingjærde
- The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Biomedical Informatics Division, Department of Computer Science, University of Oslo, Oslo, Norway; Center for Cancer Biomedicine, University of Oslo, Norway
| | - Yinyin Yuan
- Division of Molecular Pathology, The Institute of Cancer Research, 237 Fulham Road, SW3 6JB, London, UK
| | - Charlotte K Ng
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Mark J Dunning
- Cancer Research UK, Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Ed Dicks
- Strangeways Research Laboratories, University of Cambridge, Cambridge CB1 9RN, UK
| | - Elena Provenzano
- Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK; Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
| | - Stephen Sammut
- Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
| | - Steven McKinney
- Molecular Oncology, British Colombia Cancer Research Center, Vancouver, British Columbia V5Z 1L3, Canada
| | - Ian O Ellis
- Department of Histopathology, School of Molecular Medical Sciences, University of Nottingham, Nottingham, NG5 1PB, UK
| | - Sarah Pinder
- King's College London, Breakthrough Breast Cancer Research Unit, London WC2R 2LS, UK; NIHR Comprehensive Biomedical Research Centre at Guy's and St. Thomas NHS Foundation Trust and King's College London, London WC2R 2LS, UK
| | - Arnie Purushotham
- King's College London, Breakthrough Breast Cancer Research Unit, London WC2R 2LS, UK; NIHR Comprehensive Biomedical Research Centre at Guy's and St. Thomas NHS Foundation Trust and King's College London, London WC2R 2LS, UK
| | - Leigh C Murphy
- Manitoba Institute of Cell Biology, CancerCare Manitoba, University of Manitoba, Manitoba R3E 0V9, Canada
| | - Vessela N Kristensen
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway; The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway; Department of Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital, Lørenskog, Norway
| | - James D Brenton
- Cancer Research UK, Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; Department of Oncology, University of Cambridge, Hills Road, Cambridge CB2 2XZ, UK; Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK; Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
| | - Paul D P Pharoah
- Department of Oncology, University of Cambridge, Hills Road, Cambridge CB2 2XZ, UK; Strangeways Research Laboratories, University of Cambridge, Cambridge CB1 9RN, UK; Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK; Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
| | - Anne-Lise Børresen-Dale
- Department of Genetics, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Montebello, 0310 Oslo, Norway; The K.G. Jebsen Center for Breast Cancer Research, Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Norway
| | - Samuel Aparicio
- Department of Pathology and Laboratory Medicine, University of British Colombia, Vancouver, British Colombia V6T 2B5, Canada; Molecular Oncology, British Colombia Cancer Research Center, Vancouver, British Columbia V5Z 1L3, Canada.
| | - Carlos Caldas
- Cancer Research UK, Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK; Department of Oncology, University of Cambridge, Hills Road, Cambridge CB2 2XZ, UK; Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK; Cambridge Breast Unit, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust and NIHR Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK.
| |
Collapse
|
717
|
Abstract
As highlighted in a recent editorial in the Journal (Am J Epidemiol. 2014;180(2):127-128), the research area of "-omics" includes genomics, proteomics, metabolomics, and nascent fields of scientific inquiry such as epigenomics and exposomics. These fields can be collectively referred to as "global -omics." Increasing efforts have been made over the past 2 decades to identify and modify environmental risk factors among persons who are susceptible to disease because of their genotype and to integrate genetic information and other biological variables with information about individual-level risk factors and group-level or societal factors related to the broader residential, behavioral, or cultural context. In genome-wide association studies, only a small proportion of heritability is explained by genetic variants identified to date, which has prompted researchers in bioinformatics and biostatistics to take into account nonlinear relationships due to gene-environment or gene-gene interactions. The exposome, which is dynamic and variable, consists of all of the internal and external exposures an individual incurs over a lifetime. Both the epigenome and exposome change with age. The prenatal and perinatal periods are thought to be important times for epigenetic marking. Once the human epigenome has been fully mapped, identification of the effects of all deleterious environmental exposures according to duration of exposure and time period will be a complex undertaking, requiring collaborative epidemiologic studies.
Collapse
Affiliation(s)
- Steven S Coughlin
- Department of Epidemiology, Rollins School of Public Health, Emory University, 1432 Central Avenue, No. 910, Memphis, TN.
| |
Collapse
|
718
|
Gioacchini FM, Alicandri-Ciufelli M, Kaleci S, Magliulo G, Presutti L, Re M. The prognostic value of cyclin D1 expression in head and neck squamous cell carcinoma. Eur Arch Otorhinolaryngol 2014; 273:801-9. [DOI: 10.1007/s00405-014-3426-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/01/2014] [Indexed: 10/24/2022]
|
719
|
Mao Y, Qu Q, Zhang Y, Liu J, Chen X, Shen K. The value of tumor infiltrating lymphocytes (TILs) for predicting response to neoadjuvant chemotherapy in breast cancer: a systematic review and meta-analysis. PLoS One 2014; 9:e115103. [PMID: 25501357 PMCID: PMC4264870 DOI: 10.1371/journal.pone.0115103] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 11/18/2014] [Indexed: 12/31/2022] Open
Abstract
Background We carried out a systematic review and meta-analysis to evaluate the predictive roles of tumor infiltrating lymphocytes (TILs) in response to neoadjuvant chemotherapy (NAC) in breast cancer. Method A PubMed and Web of Science literature search was designed. Random or fixed effect models were adopted to estimate the summary odds ratio (OR). Heterogeneity and sensitivity analyses were performed to explore heterogeneity among studies and to assess the effects of study quality. Publication bias was evaluated using a funnel plot, Egger's test and Begg's test. We included studies where the predictive significance of TILs, and/or TILs subset on the pathologic complete response (pCR) were determined in NAC of breast cancer. Results A total of 13 published studies (including 3251 patients) were eligible. In pooled analysis, the detection of higher TILs numbers in pre-treatment biopsy was correlated with better pCR to NAC (OR = 3.93, 95% CI, 3.26–4.73). Moreover, TILs predicted higher pCR rates in triple negative (OR = 2.49, 95% CI: 1.61–3.83), HER2 positive (OR = 5.05, 95% CI: 2.86–8.92) breast cancer, but not in estrogen receptor (ER) positive (OR = 6.21, 95%CI: 0.86–45.15) patients. In multivariate analysis, TILs were still an independent marker for high pCR rate (OR = 1.41, 95% CI: 1.19–1.66). For TILs subset, higher levels of CD8+ and FOXP3+ T-lymphocytes in pre-treatment biopsy respectively predicted better pathological response to NAC (OR = 6.44, 95% CI: 2.52–16.46; OR = 2.94, 95% CI: 1.05–8.26). Only FOXP3+ lymphocytes in post-NAC breast tissue were a predictive marker for low pCR rate in univariate (OR = 0.41, 95% CI: 0.21–0.80) and multivariate (OR = 0.36, 95% CI: 0.13–0.95) analysis. Conclusion Higher TILs levels in pre-treatment biopsy indicated higher pCR rates for NAC. TILs subset played different roles in predicting response to NAC.
Collapse
Affiliation(s)
- Yan Mao
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Qu
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuzi Zhang
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjun Liu
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaosong Chen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kunwei Shen
- Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail:
| |
Collapse
|
720
|
Köbel M, Madore J, Ramus SJ, Clarke BA, Pharoah PDP, Deen S, Bowtell DD, Odunsi K, Menon U, Morrison C, Lele S, Bshara W, Sucheston L, Beckmann MW, Hein A, Thiel FC, Hartmann A, Wachter DL, Anglesio MS, Høgdall E, Jensen A, Høgdall C, Kalli KR, Fridley BL, Keeney GL, Fogarty ZC, Vierkant RA, Liu S, Cho S, Nelson G, Ghatage P, Gentry-Maharaj A, Gayther SA, Benjamin E, Widschwendter M, Intermaggio MP, Rosen B, Bernardini MQ, Mackay H, Oza A, Shaw P, Jimenez-Linan M, Driver KE, Alsop J, Mack M, Koziak JM, Steed H, Ewanowich C, DeFazio A, Chenevix-Trench G, Fereday S, Gao B, Johnatty SE, George J, Galletta L, Goode EL, Kjær SK, Huntsman DG, Fasching PA, Moysich KB, Brenton JD, Kelemen LE. Evidence for a time-dependent association between FOLR1 expression and survival from ovarian carcinoma: implications for clinical testing. An Ovarian Tumour Tissue Analysis consortium study. Br J Cancer 2014; 111:2297-307. [PMID: 25349970 PMCID: PMC4264456 DOI: 10.1038/bjc.2014.567] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 09/03/2014] [Accepted: 10/02/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Folate receptor 1 (FOLR1) is expressed in the majority of ovarian carcinomas (OvCa), making it an attractive target for therapy. However, clinical trials testing anti-FOLR1 therapies in OvCa show mixed results and require better understanding of the prognostic relevance of FOLR1 expression. We conducted a large study evaluating FOLR1 expression with survival in different histological types of OvCa. METHODS Tissue microarrays composed of tumour samples from 2801 patients in the Ovarian Tumour Tissue Analysis (OTTA) consortium were assessed for FOLR1 expression by centralised immunohistochemistry. We estimated associations for overall (OS) and progression-free (PFS) survival using adjusted Cox regression models. High-grade serous ovarian carcinomas (HGSC) from The Cancer Genome Atlas (TCGA) were evaluated independently for association between FOLR1 mRNA upregulation and survival. RESULTS FOLR1 expression ranged from 76% in HGSC to 11% in mucinous carcinomas in OTTA. For HGSC, the association between FOLR1 expression and OS changed significantly during the years following diagnosis in OTTA (Pinteraction=0.01, N=1422) and TCGA (Pinteraction=0.01, N=485). In OTTA, particularly for FIGO stage I/II tumours, patients with FOLR1-positive HGSC showed increased OS during the first 2 years only (hazard ratio=0.44, 95% confidence interval=0.20-0.96) and patients with FOLR1-positive clear cell carcinomas (CCC) showed decreased PFS independent of follow-up time (HR=1.89, 95% CI=1.10-3.25, N=259). In TCGA, FOLR1 mRNA upregulation in HGSC was also associated with increased OS during the first 2 years following diagnosis irrespective of tumour stage (HR: 0.48, 95% CI: 0.25-0.94). CONCLUSIONS FOLR1-positive HGSC tumours were associated with an increased OS in the first 2 years following diagnosis. Patients with FOLR1-negative, poor prognosis HGSC would be unlikely to benefit from anti-FOLR1 therapies. In contrast, a decreased PFS interval was observed for FOLR1-positive CCC. The clinical efficacy of FOLR1-targeted interventions should therefore be evaluated according to histology, stage and time following diagnosis.
Collapse
Affiliation(s)
- M Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - J Madore
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5E 4E6, Canada
- Melanoma Institute Australia, University of Sydney, Royal Prince Alfred Hospital, Gloucester House–level 3, Missenden Road, Camperdown, NSW 2050, Australia
| | - S J Ramus
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Harlyne Norris Research Tower, 1450 Biggy Street, Office 2517G, Los Angeles, CA 90033, USA
| | - B A Clarke
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Centre, University of Toronto, 610 Univeristy Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - P D P Pharoah
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - S Deen
- Department of Histopathology, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK
| | - D D Bowtell
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, 30 Flemington Road, Melbourne, VIC 3010, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, 30 Flemington Road, Melbourne, VIC 3010, Australia
| | - K Odunsi
- Department of Gynecological Oncology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - U Menon
- Gynaecological Cancer Research Centre, Department of Women's Cancer, Institute for Women's Health, University College London, Maple House 1st Floor, 149 Tottenham Court Road, London W1T 7DN, UK
| | - C Morrison
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - S Lele
- Department of Gynecological Oncology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - W Bshara
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - L Sucheston
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - M W Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - A Hein
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - F C Thiel
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - A Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - D L Wachter
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - M S Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5E 4E6, Canada
| | - E Høgdall
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Ø, Denmark
- Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2370 Herlev, Denmark
| | - A Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Ø, Denmark
| | - C Høgdall
- The Juliane Marie Center, Department of Obstetrics and Gynecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Ø, Denmark
| | - K R Kalli
- Department of Medical Oncology, Mayo Clinic, 200 First Street SW, Charlton 6, Rochester, MN 55905, USA
| | - B L Fridley
- Department of Biostatistics, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - G L Keeney
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology, Mayo Clinic, 200 First Street SW, Stabile 13, Rochester, MN 55905, USA
| | - Z C Fogarty
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Charlton 6, Rochester, MN 55905, USA
| | - R A Vierkant
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Charlton 6, Rochester, MN 55905, USA
| | - S Liu
- Anatomic Pathology Research Laboratory, Calgary Laboratory Services, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - S Cho
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - G Nelson
- Department of Obstetrics and Gynecology, Division of Oncology, Tom Baker Cancer Centre, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - P Ghatage
- Department of Obstetrics and Gynecology, Division of Oncology, Tom Baker Cancer Centre, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - A Gentry-Maharaj
- Gynaecological Cancer Research Centre, Department of Women's Cancer, Institute for Women's Health, University College London, Maple House 1st Floor, 149 Tottenham Court Road, London W1T 7DN, UK
| | - S A Gayther
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Harlyne Norris Research Tower, 1450 Biggy Street, Office 2517G, Los Angeles, CA 90033, USA
| | - E Benjamin
- Department of Pathology, Cancer Institute, University College London, Maple House, 149 Tottenham Court Road, London WC1E 6JJ, UK
| | - M Widschwendter
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London WC1E 6AU, UK
| | - M P Intermaggio
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Harlyne Norris Research Tower, 1450 Biggy Street, Office 2517G, Los Angeles, CA 90033, USA
| | - B Rosen
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - M Q Bernardini
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - H Mackay
- Department of Medicine, Division of Medical Oncology, University of Toronto, Princess Margaret Hospital, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - A Oza
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - P Shaw
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - M Jimenez-Linan
- Department of Pathology, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
| | - K E Driver
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - J Alsop
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - M Mack
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - J M Koziak
- Department of Population Health Research, Alberta Health Services-Cancer Care, 2210 2nd Street SW, Calgary, AB, T2S 3C3, Canada
| | - H Steed
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Royal Alexandra Hospital, 10240 Kingsway Ave, Edmonton, AB T5H 3V9, Canada
| | - C Ewanowich
- Department of Laboratory Medicine and Pathology, Royal Alexandra Hospital, 10240 Kingsway Ave, Edmonton, AB T5H 3V9, Canada
| | - A DeFazio
- Department of Gynaecological Oncology and Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, NSW 2145, Australia
| | - G Chenevix-Trench
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD,4006, Australia
| | - S Fereday
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
| | - B Gao
- Department of Gynaecological Oncology and Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, NSW 2145, Australia
| | - S E Johnatty
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD,4006, Australia
| | - J George
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
| | - L Galletta
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
| | - AOCS Study Group
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
| | - E L Goode
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, 200 First Street SW Charlton 6, Rochester, MN 55905, USA
| | - S K Kjær
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Ø, Denmark
- The Juliane Marie Center, Department of Obstetrics and Gynecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Ø, Denmark
| | - D G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5E 4E6, Canada
- Centre For Translational and Applied Genomics, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | - P A Fasching
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - K B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - J D Brenton
- National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
- Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
- Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK
| | - L E Kelemen
- Department of Public Health Sciences, Medical University of South Carolina and Hollings Cancer Center, 135 Cannon Street, Charleston, SC 29425, USA
| |
Collapse
|
721
|
Köbel M, Madore J, Ramus SJ, Clarke BA, Pharoah PDP, Deen S, Bowtell DD, Odunsi K, Menon U, Morrison C, Lele S, Bshara W, Sucheston L, Beckmann MW, Hein A, Thiel FC, Hartmann A, Wachter DL, Anglesio MS, Høgdall E, Jensen A, Høgdall C, Kalli KR, Fridley BL, Keeney GL, Fogarty ZC, Vierkant RA, Liu S, Cho S, Nelson G, Ghatage P, Gentry-Maharaj A, Gayther SA, Benjamin E, Widschwendter M, Intermaggio MP, Rosen B, Bernardini MQ, Mackay H, Oza A, Shaw P, Jimenez-Linan M, Driver KE, Alsop J, Mack M, Koziak JM, Steed H, Ewanowich C, DeFazio A, Chenevix-Trench G, Fereday S, Gao B, Johnatty SE, George J, Galletta L, Goode EL, Kjær SK, Huntsman DG, Fasching PA, Moysich KB, Brenton JD, Kelemen LE. Evidence for a time-dependent association between FOLR1 expression and survival from ovarian carcinoma: implications for clinical testing. An Ovarian Tumour Tissue Analysis consortium study. Br J Cancer 2014. [PMID: 25349970 DOI: 10.1038/bjc.2014.567] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Folate receptor 1 (FOLR1) is expressed in the majority of ovarian carcinomas (OvCa), making it an attractive target for therapy. However, clinical trials testing anti-FOLR1 therapies in OvCa show mixed results and require better understanding of the prognostic relevance of FOLR1 expression. We conducted a large study evaluating FOLR1 expression with survival in different histological types of OvCa. METHODS Tissue microarrays composed of tumour samples from 2801 patients in the Ovarian Tumour Tissue Analysis (OTTA) consortium were assessed for FOLR1 expression by centralised immunohistochemistry. We estimated associations for overall (OS) and progression-free (PFS) survival using adjusted Cox regression models. High-grade serous ovarian carcinomas (HGSC) from The Cancer Genome Atlas (TCGA) were evaluated independently for association between FOLR1 mRNA upregulation and survival. RESULTS FOLR1 expression ranged from 76% in HGSC to 11% in mucinous carcinomas in OTTA. For HGSC, the association between FOLR1 expression and OS changed significantly during the years following diagnosis in OTTA (Pinteraction=0.01, N=1422) and TCGA (Pinteraction=0.01, N=485). In OTTA, particularly for FIGO stage I/II tumours, patients with FOLR1-positive HGSC showed increased OS during the first 2 years only (hazard ratio=0.44, 95% confidence interval=0.20-0.96) and patients with FOLR1-positive clear cell carcinomas (CCC) showed decreased PFS independent of follow-up time (HR=1.89, 95% CI=1.10-3.25, N=259). In TCGA, FOLR1 mRNA upregulation in HGSC was also associated with increased OS during the first 2 years following diagnosis irrespective of tumour stage (HR: 0.48, 95% CI: 0.25-0.94). CONCLUSIONS FOLR1-positive HGSC tumours were associated with an increased OS in the first 2 years following diagnosis. Patients with FOLR1-negative, poor prognosis HGSC would be unlikely to benefit from anti-FOLR1 therapies. In contrast, a decreased PFS interval was observed for FOLR1-positive CCC. The clinical efficacy of FOLR1-targeted interventions should therefore be evaluated according to histology, stage and time following diagnosis.
Collapse
Affiliation(s)
- M Köbel
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - J Madore
- 1] Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5E 4E6, Canada [2] Melanoma Institute Australia, University of Sydney, Royal Prince Alfred Hospital, Gloucester House-level 3, Missenden Road, Camperdown, NSW 2050, Australia
| | - S J Ramus
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Harlyne Norris Research Tower, 1450 Biggy Street, Office 2517G, Los Angeles, CA 90033, USA
| | - B A Clarke
- Department of Laboratory Medicine and Pathobiology, Princess Margaret Cancer Centre, University of Toronto, 610 Univeristy Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - P D P Pharoah
- 1] Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK [2] Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - S Deen
- Department of Histopathology, Queen's Medical Centre, Nottingham University Hospitals NHS Trust, Nottingham NG7 2UH, UK
| | - D D Bowtell
- 1] Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia [2] Department of Biochemistry and Molecular Biology, University of Melbourne, 30 Flemington Road, Melbourne, VIC 3010, Australia [3] Sir Peter MacCallum Department of Oncology, University of Melbourne, 30 Flemington Road, Melbourne, VIC 3010, Australia
| | - K Odunsi
- Department of Gynecological Oncology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - U Menon
- Gynaecological Cancer Research Centre, Department of Women's Cancer, Institute for Women's Health, University College London, Maple House 1st Floor, 149 Tottenham Court Road, London W1T 7DN, UK
| | - C Morrison
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - S Lele
- 1] Department of Gynecological Oncology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA [2] Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - W Bshara
- Department of Pathology and Laboratory Medicine, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - L Sucheston
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - M W Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - A Hein
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - F C Thiel
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany
| | - A Hartmann
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - D L Wachter
- Institute of Pathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Krankenhausstrasse 8-10, 91054 Erlangen, Germany
| | - M S Anglesio
- Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5E 4E6, Canada
| | - E Høgdall
- 1] Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Ø, Denmark [2] Department of Pathology, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, DK-2370 Herlev, Denmark
| | - A Jensen
- Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Ø, Denmark
| | - C Høgdall
- The Juliane Marie Center, Department of Obstetrics and Gynecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Ø, Denmark
| | - K R Kalli
- Department of Medical Oncology, Mayo Clinic, 200 First Street SW, Charlton 6, Rochester, MN 55905, USA
| | - B L Fridley
- Department of Biostatistics, University of Kansas Medical Center, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| | - G L Keeney
- Department of Laboratory Medicine and Pathology, Division of Anatomic Pathology, Mayo Clinic, 200 First Street SW, Stabile 13, Rochester, MN 55905, USA
| | - Z C Fogarty
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Charlton 6, Rochester, MN 55905, USA
| | - R A Vierkant
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic, 200 First Street SW, Charlton 6, Rochester, MN 55905, USA
| | - S Liu
- Anatomic Pathology Research Laboratory, Calgary Laboratory Services, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - S Cho
- Department of Pathology and Laboratory Medicine, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - G Nelson
- Department of Obstetrics and Gynecology, Division of Oncology, Tom Baker Cancer Centre, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - P Ghatage
- Department of Obstetrics and Gynecology, Division of Oncology, Tom Baker Cancer Centre, University of Calgary, Foothills Medical Center, 1403 29 ST NW, Calgary, AB T2N 2T9, Canada
| | - A Gentry-Maharaj
- Gynaecological Cancer Research Centre, Department of Women's Cancer, Institute for Women's Health, University College London, Maple House 1st Floor, 149 Tottenham Court Road, London W1T 7DN, UK
| | - S A Gayther
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Harlyne Norris Research Tower, 1450 Biggy Street, Office 2517G, Los Angeles, CA 90033, USA
| | - E Benjamin
- Department of Pathology, Cancer Institute, University College London, Maple House, 149 Tottenham Court Road, London WC1E 6JJ, UK
| | - M Widschwendter
- Department of Women's Cancer, UCL EGA Institute for Women's Health, University College London, 74 Huntley Street, London WC1E 6AU, UK
| | - M P Intermaggio
- Department of Preventive Medicine, Keck School of Medicine, USC/Norris Comprehensive Cancer Center, University of Southern California, Harlyne Norris Research Tower, 1450 Biggy Street, Office 2517G, Los Angeles, CA 90033, USA
| | - B Rosen
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - M Q Bernardini
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - H Mackay
- Department of Medicine, Division of Medical Oncology, University of Toronto, Princess Margaret Hospital, 610 University Avenue, Toronto, ON M5G 2M9, Canada
| | - A Oza
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - P Shaw
- Department of Obstetrics and Gynecology, University of Toronto, Princess Margaret Cancer Centre, 610 University Avenue, M-700, Toronto, ON M5T 2M9, Canada
| | - M Jimenez-Linan
- 1] Department of Pathology, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, UK [2] National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK
| | - K E Driver
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - J Alsop
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - M Mack
- Department of Oncology, University of Cambridge, Strangeways Research Laboratory, Worts Causeway, Cambridge CB1 8RN, UK
| | - J M Koziak
- Department of Population Health Research, Alberta Health Services-Cancer Care, 2210 2nd Street SW, Calgary, AB, T2S 3C3, Canada
| | - H Steed
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Royal Alexandra Hospital, 10240 Kingsway Ave, Edmonton, AB T5H 3V9, Canada
| | - C Ewanowich
- Department of Laboratory Medicine and Pathology, Royal Alexandra Hospital, 10240 Kingsway Ave, Edmonton, AB T5H 3V9, Canada
| | - A DeFazio
- Department of Gynaecological Oncology and Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, NSW 2145, Australia
| | - G Chenevix-Trench
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD,4006, Australia
| | - S Fereday
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
| | - B Gao
- Department of Gynaecological Oncology and Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, NSW 2145, Australia
| | - S E Johnatty
- Genetics and Computational Biology Department, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston, QLD,4006, Australia
| | - J George
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
| | - L Galletta
- Department of Cancer Genomics and Genetics, Peter MacCallum Cancer Centre, Locked Bag I, A'Beckett Street, East Melbourne, VIC 8006, Australia
| | | | - E L Goode
- Department of Health Sciences Research, Division of Epidemiology, Mayo Clinic, 200 First Street SW Charlton 6, Rochester, MN 55905, USA
| | - S K Kjær
- 1] Department of Virus, Lifestyle and Genes, Danish Cancer Society Research Center, Strandboulevarden 49, DK-2100 Copenhagen, Ø, Denmark [2] The Juliane Marie Center, Department of Obstetrics and Gynecology, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, DK-2100 Copenhagen, Ø, Denmark
| | - D G Huntsman
- 1] Department of Pathology and Laboratory Medicine, University of British Columbia, BC Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5E 4E6, Canada [2] Centre For Translational and Applied Genomics, British Columbia Cancer Agency, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | - P A Fasching
- 1] Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Comprehensive Cancer Center Erlangen-EMN, Universitaetsstrasse 21-23, 91054 Erlangen, Germany [2] Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - K B Moysich
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - J D Brenton
- 1] National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge CB2 2QQ, UK [2] Department of Oncology, University of Cambridge, Hutchison/MRC Research Centre, Hills Road, Cambridge CB2 0XZ, UK [3] Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK [4] Cambridge Experimental Cancer Medicine Centre, Cambridge CB2 0RE, UK
| | - L E Kelemen
- Department of Public Health Sciences, Medical University of South Carolina and Hollings Cancer Center, 135 Cannon Street, Charleston, SC 29425, USA
| |
Collapse
|
722
|
Shen W, Xi H, Zhang K, Cui J, Li J, Wang N, Wei B, Chen L. Prognostic role of EphA2 in various human carcinomas: a meta-analysis of 23 related studies. Growth Factors 2014; 32:247-53. [PMID: 25418013 DOI: 10.3109/08977194.2014.984806] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The prognostic role of EphA2 in human carcinomas remains controversial. We systematically reviewed the evidence of assessment of EphA2 expression in cancers to help clarify this issue. PubMed, Embase and Web of Science databases were searched to identify eligible studies to evaluate the association of EphA2 expression and overall survival (OS) of cancers. Hazard ratios (HRs) were pooled to estimate the effect. EphA2 overexpression was significantly correlated with poor OS of patients with cancer (HR: 1.94, 95% confidence interval [CI]: 1.65-2.28). Subgroup analysis also indicated a significant relation between EphA2 overexpression and OS in gastric cancer (HR: 1.95, 95% CI: 1.48-2.59). However, there was no significant relation between EphA2 overexpression and OS in lung cancer (HR: 1.30, 95% CI: 0.93-1.83). Our analyses demonstrate that EphA2 overexpression was effectively predictive of worse prognosis in various human carcinomas. For certain cancers, EphA2 might be a marker of poor prognosis in patients with cancer, except for lung cancer.
Collapse
Affiliation(s)
- Weisong Shen
- Department of General Surgery, Chinese People's Liberation Army General Hospital , Beijing , China
| | | | | | | | | | | | | | | |
Collapse
|
723
|
Xu XL, Zheng WH, Fu ZX, Li ZP, Xie HX, Li XX, Jiang LH, Wang Y, Zhu SM, Mao WM. Topo2A as a prognostic biomarker for patients with resectable esophageal squamous cell carcinomas. Med Oncol 2014; 32:396. [PMID: 25432700 DOI: 10.1007/s12032-014-0396-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/20/2014] [Indexed: 10/24/2022]
Abstract
Topoisomerase 2α (Topo2A) is a key enzyme in replication. It functions as a cell proliferation and cell cycle-specific marker and it is identified mainly in the interphase nuclei of proliferating cells. Many studies have shown that Topo2A protein expression is up-regulated in various cancers including esophageal cancer. However, to date, no studies have adequately addressed the prognostic value of Topo2A in patients with resectable esophageal squamous cell carcinoma (ESCC). Therefore, we conducted a large-scale retrospective study investigating the expression of Topo2A and the clinicopathological characteristics or prognosis of ESCC patients. Eight hundred and twenty-nine specimens of ESCC from patients who underwent complete esophageal cancer resection were evaluated using an immunohistochemical assay. Among them, 404 (48.7 %) cases with a score >2 were determined to be positive for Topo2A expression. Topo2A overexpression was significantly associated with poorer differentiation (P = 0.007) and perineural invasion (P = 0.046). The median progression-free survival (PFS) of 319 patients with Topo2A-positive expression and 336 patients with Topo2A-negative expression was 19.5 and 26.5 months, respectively (P = 0.000). The overall survival (OS) in patients with and without Topo2A expression was 34.0 and 44.5 months, respectively (P = 0.002). In the multivariate analysis, Topo2A overexpression was identified as an independent prognostic factor for PFS (P = 0.001) and OS (P = 0.009). We determined that Topo2A overexpression was not only associated with poorer differentiation and perineural invasion, but it could also act as an independent risk factor for ESCC.
Collapse
Affiliation(s)
- Xiao-Ling Xu
- Department of Medical Oncology, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou City, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
724
|
Cheng H, Wang X, Li T, Chen L. Bcl-2 expression and patient survival in gastric cancer: a systematic review of the literature with meta-analysis. Med Oncol 2014; 32:389. [PMID: 25428392 DOI: 10.1007/s12032-014-0389-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 11/18/2014] [Indexed: 12/30/2022]
Abstract
The prognostic role of Bcl-2 in gastric cancer remains controversial. We systematically reviewed the evidence for the effect of Bcl-2 expression in gastric cancer to elucidate this issue. PubMed, EMBASE, and Web of Science were searched to identify eligible studies to evaluate the association between Bcl-2 expression and overall survival (OS) and clinicopathological features of gastric cancer. Hazard ratios (HRs) or odds ratios (ORs) with 95 % confidence interval (95 % CI) were pooled to estimate the effect. Bcl-2 expression was not significantly correlated with OS of gastric cancer patients (combined HR 0.88, 95 % CI 0.62-1.25). Subgroup analysis indicated that Bcl-2 expression had a favorable impact on OS in Asian countries (HR 0.60, 95 % CI 0.47-0.77) and cutoff value <15 % (HR 0.69, 95 % CI 0.48-0.97). Furthermore, Bcl-2 expression was significantly associated with TNM stage (OR 0.58, 95 % CI 0.41-0.80), the depth of invasion (OR 0.53, 95 % CI 0.37-0.76), and lymph node metastasis (OR 0.55, 95 % CI 0.42-0.71). Bcl-2 expression predicts less invasion and malignancy in clinicopathological features. Moreover, Bcl-2 could be a favorable prognostic marker for patients of Asian countries.
Collapse
Affiliation(s)
- Haidong Cheng
- Medical Center, Tsinghua University, Beijing, 100084, China
| | | | | | | |
Collapse
|
725
|
Prensner JR, Zhao S, Erho N, Schipper M, Iyer MK, Dhanasekaran SM, Magi-Galluzzi C, Mehra R, Sahu A, Siddiqui J, Davicioni E, Den RB, Dicker AP, Karnes RJ, Wei JT, Klein EA, Jenkins RB, Chinnaiyan AM, Feng FY. RNA biomarkers associated with metastatic progression in prostate cancer: a multi-institutional high-throughput analysis of SChLAP1. Lancet Oncol 2014; 15:1469-1480. [PMID: 25456366 DOI: 10.1016/s1470-2045(14)71113-1] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Improved clinical predictors for disease progression are needed for localised prostate cancer, since only a subset of patients develop recurrent or refractory disease after first-line treatment. Therefore, we undertook an unbiased analysis to identify RNA biomarkers associated with metastatic progression after prostatectomy. METHODS Prostate cancer samples from patients treated with radical prostatectomy at three academic institutions were analysed for gene expression by a high-density Affymetrix GeneChip platform, encompassing more than 1 million genomic loci. In a discovery cohort, all protein-coding genes and known long non-coding RNAs were ranked by fold change in expression between tumours that subsequently metastasised versus those that did not. The top ranked gene was then validated for its prognostic value for metastatic progression in three additional independent cohorts. 95% of the gene expression assays were done in a Clinical Laboratory Improvements Amendments certified laboratory facility. All genes were assessed for their ability to predict metastatic progression by receiver-operating-curve area-under-the-curve analyses. Multivariate analyses were done for the primary endpoint of metastatic progression, with variables including Gleason score, preoperative prostate-specific antigen concentration, seminal vesicle invasion, surgical margin status, extracapsular extension, lymph node invasion, and expression of the highest ranked gene. FINDINGS 1008 patients were included in the study: 545 in the discovery cohort and 463 in the validation cohorts. The long non-coding RNA SChLAP1 was identified as the highest-ranked overexpressed gene in cancers with metastatic progression. Validation in three independent cohorts confirmed the prognostic value of SChLAP1 for metastatic progression. On multivariate modelling, SChLAP1 expression (high vs low) independently predicted metastasis within 10 years (odds ratio [OR] 2·45, 95% CI 1·70-3·53; p<0·0001). The only other variable that independently predicted metastasis within 10 years was Gleason score (8-10 vs 5-7; OR 2·14, 95% CI 1·77-2·58; p<0·0001). INTERPRETATION We identified and validated high SChLAP1 expression as significantly prognostic for metastatic disease progression of prostate cancer. Our findings suggest that further development of SChLAP1 as a potential biomarker, for treatment intensification in aggressive prostate cancer, warrants future study. FUNDING Prostate Cancer Foundation, National Institutes of Health, Department of Defense, Early Detection Research Network, Doris Duke Charitable Foundation, and Howard Hughes Medical Institute.
Collapse
Affiliation(s)
- John R Prensner
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Shuang Zhao
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Matthew Schipper
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthew K Iyer
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Saravana M Dhanasekaran
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Cristina Magi-Galluzzi
- Anatomic Pathology, Robert J Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Rohit Mehra
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anirban Sahu
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Javed Siddiqui
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Robert B Den
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam P Dicker
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - John T Wei
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Urology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA; Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Felix Y Feng
- Michigan Center for Translational Pathology, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, USA; Comprehensive Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
726
|
Klein EA, Yousefi K, Haddad Z, Choeurng V, Buerki C, Stephenson AJ, Li J, Kattan MW, Magi-Galluzzi C, Davicioni E. A genomic classifier improves prediction of metastatic disease within 5 years after surgery in node-negative high-risk prostate cancer patients managed by radical prostatectomy without adjuvant therapy. Eur Urol 2014; 67:778-86. [PMID: 25466945 DOI: 10.1016/j.eururo.2014.10.036] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/22/2014] [Indexed: 11/18/2022]
Abstract
BACKGROUND Surgery is a standard first-line therapy for men with intermediate- or high-risk prostate cancer. Clinical factors such as tumor grade, stage, and prostate-specific antigen (PSA) are currently used to identify those who are at risk of recurrence and who may benefit from adjuvant therapy, but novel biomarkers that improve risk stratification and that distinguish local from systemic recurrence are needed. OBJECTIVE To determine whether adding the Decipher genomic classifier, a validated metastasis risk-prediction model, to standard risk-stratification tools (CAPRA-S and Stephenson nomogram) improves accuracy in predicting metastatic disease within 5 yr after surgery (rapid metastasis [RM]) in an independent cohort of men with adverse pathologic features after radical prostatectomy (RP). DESIGN, SETTING, AND PARTICIPANTS The study population consisted of 169 patients selected from 2641 men who underwent RP at the Cleveland Clinic between 1987 and 2008 who met the following criteria: (1) preoperative PSA>20 ng/ml, stage pT3 or margin positive, or Gleason score≥8; (2) pathologic node negative; (3) undetectable post-RP PSA; (4) no neoadjuvant or adjuvant therapy; and (5) minimum of 5-yr follow-up for controls. The final study cohort consisted of 15 RM patients and 154 patients as non-RM controls. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The performance of Decipher was evaluated individually and in combination with clinical risk factors using concordance index (c-index), decision curve analysis, and logistic regression for prediction of RM. RESULTS AND LIMITATIONS RM patients developed metastasis at a median of 2.3 yr (interquartile range: 1.7-3.3). In multivariable analysis, Decipher was a significant predictor of RM (odds ratio: 1.48; p=0.018) after adjusting for clinical risk factors. Decipher had the highest c-index, 0.77, compared with the Stephenson model (c-index: 0.75) and CAPRA-S (c-index: 0.72) as well as with a panel of previously reported prostate cancer biomarkers unrelated to Decipher. Integration of Decipher into the Stephenson nomogram increased the c-index from 0.75 (95% confidence interval [CI], 0.65-0.85) to 0.79 (95% CI, 0.68-0.89). CONCLUSIONS Decipher was independently validated as a genomic metastasis signature for predicting metastatic disease within 5 yr after surgery in a cohort of high-risk men treated with RP and managed conservatively without any adjuvant therapy. Integration of Decipher into clinical nomograms increased prediction of RM. Decipher may allow identification of men most at risk for metastatic progression who should be considered for multimodal therapy or inclusion in clinical trials. PATIENT SUMMARY Use of Decipher in addition to standard clinical information more accurately identified men who developed metastatic disease within 5 yr after surgery. The results suggest that Decipher allows improved identification of the men who should consider secondary therapy from among the majority that may be managed conservatively after surgery.
Collapse
Affiliation(s)
- Eric A Klein
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA.
| | - Kasra Yousefi
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | - Zaid Haddad
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| | | | | | - Andrew J Stephenson
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jianbo Li
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Michael W Kattan
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA
| | | | - Elai Davicioni
- GenomeDx Biosciences, Vancouver, British Columbia, Canada
| |
Collapse
|
727
|
Dasgupta A, Mahapatra M, Saxena R. A study for proposal of use of regulatory T cells as a prognostic marker and establishing an optimal threshold level for their expression in chronic lymphocytic leukemia. Leuk Lymphoma 2014; 56:1831-8. [PMID: 25263321 DOI: 10.3109/10428194.2014.966245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Although regulatory T cells (Tregs) have been extensively studied in chronic lymphocytic leukemia, there is no uniform guideline or consensus regarding their use as a prognostic marker. This study describes the methodology used to develop an optimal threshold level for Tregs in these patients. Treg levels were assessed in the peripheral blood of 130 patients and 150 controls. Treg frequencies were linked to established prognostic markers as well as overall survival and time to first treatment. The cut-offs for Treg positivity were assessed by receiver operating characteristic (ROC) analysis. A cut-off of 5.7% for Treg cell percentage and of 35 cells/μL for absolute Treg cell count were determined as optimal in patients with CLL along with a median Treg percentage of 15.5% used to separate patients with low- and high-risk disease. The experiments presented here will possibly aid in the use of Treg frequencies as a potential prognostic marker in CLL.
Collapse
Affiliation(s)
- Alakananda Dasgupta
- Department of Hematology, All India Institute of Medical Sciences , New Delhi , India
| | | | | |
Collapse
|
728
|
Georgoulias V, Apostolaki S, Bozionelou V, Politaki E, Perraki M, Georgoulia N, Kalbakis K, Kotsakis A, Xyrafas A, Agelaki S, Mavroudis D. Effect of front-line chemotherapy on circulating CK-19 mRNA-positive cells in patients with metastatic breast cancer. Cancer Chemother Pharmacol 2014; 74:1217-25. [PMID: 25344760 DOI: 10.1007/s00280-014-2598-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 09/22/2014] [Indexed: 11/29/2022]
Abstract
PURPOSE To evaluate the effect of front-line chemotherapy on CK-19mRNA+ circulating tumor cells (CTCs) and their relevance in patients with metastatic breast cancer (MBC). PATIENTS AND METHODS The presence of CK-19mRNA+ CTCs was assessed using a real-time RT-PCR assay in 298 previously untreated patients with MBC before and after the administration of front-line chemotherapy. RESULTS CK-19mRNA+ CTCs were detected in the blood of 199 (66.8 %) and 148 (49.7 %) patients before and after chemotherapy, respectively. There was no correlation between the detection of CK-19mRNA+ CTCs after chemotherapy and the various known clinicopathologic parameters except with HER2 status. The incidence of detection of CK-19mRNA+ CTCs was significantly decreased after the administration of 3 (47.8 %; p < 0.001) or 6 (44.3 %; p = 0.001) chemotherapy cycles. The persistent detection of >2.25 CK-19mRNA+ CTCs both before and after chemotherapy (persistently high group) was associated with a significantly (p = 0.003) decreased overall survival. In addition, chemotherapy-induced decrease of CK-19mRNA+ CTCs (≤2.25 CTCs) was associated with a better survival (47 vs 34 months; p < 0.001). Failure of chemotherapy to decrease the CK-19mRNA+ CTCs ≤2.25 was associated with decreased overall survival (HR 1.405, 95 % CI 1.044-1.891; p = 0.025) whereas in multivariate analysis the persistence of >2.25 CTCs both before and after chemotherapy was emerged as an independent prognostic factor (HR 1.661, 95 % CI 1.070-2.579; p = 0.024). CONCLUSION Detection of CK-19mRNA+ CTCs after the completion of front-line chemotherapy in patients with MBC is associated with poor survival and may be a useful tool for the evaluation of front-line chemotherapy.
Collapse
Affiliation(s)
- V Georgoulias
- Department of Medical Oncology, University General Hospital of Heraklion, P.O. Box 1352, Voutes, 71110, Heraklion, Crete, Greece,
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
729
|
Goossens-Beumer IJ, Derr RS, Buermans HPJ, Goeman JJ, Böhringer S, Morreau H, Nitsche U, Janssen KP, van de Velde CJH, Kuppen PJK. MicroRNA classifier and nomogram for metastasis prediction in colon cancer. Cancer Epidemiol Biomarkers Prev 2014; 24:187-97. [PMID: 25315964 DOI: 10.1158/1055-9965.epi-14-0544-t] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Colon cancer prognosis and treatment are currently based on a classification system still showing large heterogeneity in clinical outcome, especially in TNM stages II and III. Prognostic biomarkers for metastasis risk are warranted as development of distant recurrent disease mainly accounts for the high lethality rates of colon cancer. miRNAs have been proposed as potential biomarkers for cancer. Furthermore, a verified standard for normalization of the amount of input material in PCR-based relative quantification of miRNA expression is lacking. METHODS A selection of frozen tumor specimens from two independent patient cohorts with TNM stage II-III microsatellite stable primary adenocarcinomas was used for laser capture microdissection. Next-generation sequencing was performed on small RNAs isolated from colorectal tumors from the Dutch cohort (N = 50). Differential expression analysis, comparing in metastasized and nonmetastasized tumors, identified prognostic miRNAs. Validation was performed on colon tumors from the German cohort (N = 43) using quantitative PCR (qPCR). RESULTS miR25-3p and miR339-5p were identified and validated as independent prognostic markers and used to construct a multivariate nomogram for metastasis risk prediction. The nomogram showed good probability prediction in validation. In addition, we recommend combination of miR16-5p and miR26a-5p as standard for normalization in qPCR of colon cancer tissue-derived miRNA expression. CONCLUSIONS In this international study, we identified and validated a miRNA classifier in primary cancers, and propose a nomogram capable of predicting metastasis risk in microsatellite stable TNM stage II-III colon cancer. IMPACT In conjunction with TNM staging, by means of a nomogram, this miRNA classifier may allow for personalized treatment decisions based on individual tumor characteristics.
Collapse
Affiliation(s)
| | - Remco S Derr
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Henk P J Buermans
- Department of Human Genetics/Leiden Genome Technology Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Jelle J Goeman
- Biostatistics, Department for Health Evidence, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Stefan Böhringer
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans Morreau
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ulrich Nitsche
- Department of Surgery, Klinikum rechts der Isar, Technical University München, Munich, Germany
| | - Klaus-Peter Janssen
- Department of Surgery, Klinikum rechts der Isar, Technical University München, Munich, Germany
| | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
730
|
Markkula A, Simonsson M, Ingvar C, Rose C, Jernström H. IL6 genotype, tumour ER-status, and treatment predicted disease-free survival in a prospective breast cancer cohort. BMC Cancer 2014; 14:759. [PMID: 25305747 PMCID: PMC4198733 DOI: 10.1186/1471-2407-14-759] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 10/03/2014] [Indexed: 02/07/2023] Open
Abstract
Background In breast cancer, high levels of the inflammatory cytokine interleukin-6 (IL-6) have been associated with disease-free survival and treatment resistance. Increased serum levels of IL-6 have been correlated with increased levels of NF-κβ and aromatase expression in adipose tissue. Several IL6 single nucleotide polymorphisms have been associated with breast cancer prognosis, but the impact may differ depending on tumour oestrogen receptor (ER) status. This translational study investigated the association between IL6 genotypes, ER-status, and treatment on the risk of early events among breast cancer patients. Methods The study included 634 25- to 99-year-old primary breast cancer patients in Sweden from 2002–2008. Genotyped IL6 single nucleotide polymorphisms rs1800797, rs1800796, rs1800795, and rs2069849 were analysed separately and as diplotypes. Disease-free survival was assessed for 567 patients. Clinical data, patient-, and tumour-characteristics were obtained from questionnaires, patient charts, population registries, and pathology reports. Results The median follow-up time was 5.1 years. IL6 diplotype was not associated with early events for all 567 patients, but AGCC/AGCC diplotype-carriers with ER-negative tumours had an increased risk, (adjusted Hazard Ratio (HR) = 5.91, 95% CI: 1.28–27.42). Any C-carriers (rs1800795) with ER-negative tumours had a higher risk of early events than GG-carriers with ER-negative tumours, (adjusted HR = 3.76, 95% CI: 1.05–13.43), particularly after radiotherapy (adjusted HR = 7.17, 95% CI: 1.16–32.28). Irrespective of ER-status, chemotherapy-treated Any C-carriers had a higher risk of early events than GG-carriers (adjusted HR = 3.42, 95% CI: 1.01–11.54). Conclusions The main finding of the present study was that IL6 genotype was strongly associated with early events among patients with ER-negative tumours, particularly among radiotherapy-treated patients, and among chemotherapy-treated patients irrespective of ER-status. The high risk for early events observed in these subgroups of patients suggests that combined information on IL6 genotype, tumour ER-status, and breast cancer treatment may represent a tool for identifying patients who require more personalised treatment. Electronic supplementary material The online version of this article (doi:10.1186/1471-2407-14-759) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | - Helena Jernström
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund, Lund University, Barngatan 2B, Lund SE-22185, Sweden.
| |
Collapse
|
731
|
Visconti L, Nelissen K, Deckx L, van den Akker M, Adriaensen W, Daniels L, Matheï C, Linsen L, Hellings N, Stinissen P, Buntinx F. Prognostic value of circulating cytokines on overall survival and disease-free survival in cancer patients. Biomark Med 2014; 8:297-306. [PMID: 24521026 DOI: 10.2217/bmm.13.122] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Through their tumor-promoting and/or tumor-suppressive properties, cytokines can influence progression of cancer. We systematically reviewed the current literature on the prognostic value of the circulating cytokines IL-1α/β, IL-6, IL-8, IL-10, IL-12, TNF-α, TGF-β and IFN-γ to predict overall and disease-free survival in any type of cancer patients. PubMed was systematically searched and based on eligibility assessment using our five criteria of the Reporting Recommendations for Tumor Marker Prognostic Studies (REMARK) checklist, six unique studies were reviewed. Elevated IL-6 and IL-10 levels seem independently associated with worse prognosis in terms of overall and disease-free survival. The prognostic value of IL-1α/β, IL-8, IL-12, TNF-α, TGF-β and IFN-γ could not be demonstrated. The small number of selected studies underlines the need for large well-designed prospective studies, using the REMARK checklist as a guideline, to determine which cytokines have prognostic value on survival in cancer patients.
Collapse
Affiliation(s)
- Laura Visconti
- Faculty of Medicine & Life Sciences, Hasselt University, Campus Diepenbeek, Martelarenlaan 42, 3500 Hasselt, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
732
|
HER4 and its cytoplasmic isoforms are associated with progression-free survival of malignant melanoma. Melanoma Res 2014; 24:88-91. [PMID: 24366194 DOI: 10.1097/cmr.0000000000000040] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
HER4 belongs to the epidermal growth factor (EGF) family. Mutations in HER4 are associated with malignant melanoma. This points to HER4 as an important receptor in malignant melanoma and also raises the question of whether the other receptors in the EGF system could be involved. RT-qPCR mRNA quantification was carried out of all four EGF receptors (EGFR, HER2, HER3, and HER4) and the HER4 cytoplasmic isoforms in lymph node metastases from patients with malignant melanoma. We related their expression to progression of the disease. HER4 expression was found to be an indicator of short progression-free survival (P=0.0340). Interestingly, of the two cytoplasmic splice variants of HER4, the association of CYT1 (P=0.0176) with progression-free survival was more pronounced than that for CYT2 (P=0.0458). Also, HER3 was associated with progression-free survival (P=0.0169), whereas no association was found for EGFR or HER2 with time to progression. Our results further emphasize the role of HER4 as an important oncogene in malignant melanoma and point to HER4 as a possible drug target in this disease.
Collapse
|
733
|
Moons KGM, de Groot JAH, Bouwmeester W, Vergouwe Y, Mallett S, Altman DG, Reitsma JB, Collins GS. Critical appraisal and data extraction for systematic reviews of prediction modelling studies: the CHARMS checklist. PLoS Med 2014; 11:e1001744. [PMID: 25314315 PMCID: PMC4196729 DOI: 10.1371/journal.pmed.1001744] [Citation(s) in RCA: 1031] [Impact Index Per Article: 103.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Carl Moons and colleagues provide a checklist and background explanation for critically appraising and extracting data from systematic reviews of prognostic and diagnostic prediction modelling studies. Please see later in the article for the Editors' Summary.
Collapse
Affiliation(s)
- Karel G. M. Moons
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Joris A. H. de Groot
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Walter Bouwmeester
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Yvonne Vergouwe
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Susan Mallett
- Department of Primary Care Health Sciences, New Radcliffe House, University of Oxford, Oxford, United Kingdom
| | - Douglas G. Altman
- Centre for Statistics in Medicine, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, United Kingdom
| | - Johannes B. Reitsma
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht, The Netherlands
| | - Gary S. Collins
- Centre for Statistics in Medicine, University of Oxford, Botnar Research Centre, Windmill Road, Oxford, United Kingdom
| |
Collapse
|
734
|
Bernstein JM, Homer JJ, West CM. Dynamic contrast-enhanced magnetic resonance imaging biomarkers in head and neck cancer: Potential to guide treatment? A systematic review. Oral Oncol 2014; 50:963-70. [PMID: 25116700 DOI: 10.1016/j.oraloncology.2014.07.011] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/22/2014] [Indexed: 11/16/2022]
Affiliation(s)
- Jonathan M Bernstein
- University Department of Otolaryngology - Head & Neck Surgery, Manchester Royal Infirmary, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK; (b)Translational Radiobiology Group, Institute of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, UK.
| | - Jarrod J Homer
- University Department of Otolaryngology - Head & Neck Surgery, Manchester Royal Infirmary, Manchester Academic Health Science Centre, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK.
| | - Catharine M West
- Translational Radiobiology Group, Institute of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester, Wilmslow Road, Withington, Manchester M20 4BX, UK.
| |
Collapse
|
735
|
Xu XL, Zheng WH, Tao KY, Li XX, Xu WZ, Wang Y, Zhu SM, Mao WM. p53 is an independent prognostic factor in operable esophageal squamous cell carcinoma: a large-scale study with a long follow-up. Med Oncol 2014; 31:257. [PMID: 25270283 DOI: 10.1007/s12032-014-0257-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 09/18/2014] [Indexed: 02/07/2023]
Abstract
The p53 protein is involved in many biological functions in cancer, such as cell cycle arrest, DNA repair, apoptosis, senescence, DNA metabolism, angiogenesis, and cellular differentiation. However, the association between p53 expression and clinicopathological findings or prognosis in esophageal squamous cell carcinoma (ESCC) is controversial. We designed a large-scale study of 830 operable ESCC patients with a long follow-up to investigate the relationship between p53 expression and the clinicopathological characteristics and prognosis of patients. Immunohistochemistry was used to detect p53 protein expression. When the patients were divided into two groups, a positive expression group and a negative expression group, p53-positive expression positively correlated with a poorer differentiation level (P = 0.044). The overexpression of p53 was associated with a more advanced clinical stage (P = 0.015). A total of 775 patients were available for survival analysis. The median OS of 160 patients who had p53-positive expression and 486 patients who had p53-negative expression were 58.8 and 46.3 months, respectively (P = 0.021); the median PFS of the two groups were 39.6 and 27.5 months, respectively (P = 0.015). Lymph node metastasis, gender, differentiation, depth of invasion, and p53 protein expression were proven to have an influence on both OS and PFS in a univariate analysis. In the multivariate analysis, p53-positive expression maintained its independent prognostic impact on OS (P = 0.048) and PFS (P = 0.039), as did lymph node metastasis, differentiation, and depth of invasion. We identified that p53 protein-positive expression can serve as an independent, unfavorable prognosis biomarker in ESCC.
Collapse
Affiliation(s)
- Xiao-Ling Xu
- Department of Medical Oncology, Zhejiang Cancer Hospital, 38 Guangji Road, Hangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
736
|
Benard A, Goossens-Beumer IJ, van Hoesel AQ, Horati H, Putter H, Zeestraten ECM, van de Velde CJH, Kuppen PJK. Prognostic value of polycomb proteins EZH2, BMI1 and SUZ12 and histone modification H3K27me3 in colorectal cancer. PLoS One 2014; 9:e108265. [PMID: 25243792 PMCID: PMC4171510 DOI: 10.1371/journal.pone.0108265] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 08/20/2014] [Indexed: 02/07/2023] Open
Abstract
Numerous changes in epigenetic mechanisms have been described in various types of tumors. In search for new biomarkers, we investigated the expression of Polycomb-group (PcG) proteins EZH2, BMI1 and SUZ12 and associated histone modification H3K27me3 in colorectal cancer. Nuclear expression of PcG proteins and histone modification H3K27me3 were immunohistochemically (IHC) stained on a tissue microarray (TMA), including 247 tumor tissues and 47 normal tissues, and scored using the semi-automated Ariol system. Tumor tissues showed higher expression of EZH2 (p = 0.05) and H3K27me3 (p<0.001) as compared to their normal counterparts. Combined marker trend analyses indicated that an increase in the number of markers showing high expression was associated with better prognosis. High expression of all four markers in the combined marker analyses was correlated with the best patient survival and the longest recurrence-free survival, with overall survival (p = 0.01, HR 0.42(0.21-0.84)), disease-free survival (p = 0.007, HR 0.23(0.08-0.67) and local recurrence-free survival (p = 0.02, HR 0.30(0.11-0.84)). In conclusion, we found that expression of PcG proteins and H3K27me3 showed prognostic value in our study cohort. Better stratification of patients was obtained by combining the expression data of the investigated biomarkers as compared to the individual markers, underlining the importance of investigating multiple markers simultaneously.
Collapse
Affiliation(s)
- Anne Benard
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
- * E-mail:
| | | | - Anneke Q. van Hoesel
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Hamed Horati
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Hein Putter
- Department of Medical Statistics, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Peter J. K. Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
737
|
Ullenhag GJ, Al-Attar A, Mukherjee A, Green AR, Ellis IO, Durrant LG. The TRAIL system is over-expressed in breast cancer and FLIP a marker of good prognosis. J Cancer Res Clin Oncol 2014; 141:505-14. [PMID: 25230899 DOI: 10.1007/s00432-014-1822-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/01/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND Breast cancer is the most common cancer in women. The tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway transmits apoptotic signals. Novel anticancer agents that activate this system are in clinical development, including anti-breast cancer. METHODS The tissue microarray technique was applied. We used an array of breast cancer tissues from a large group of patients (>800) to assess the protein expression of TRAIL-R1, TRAIL-R2, the long isoform of FLICE-inhibitory protein and total FLICE-inhibitory protein (FLIP(L) and FLIP(T)). Disease-free survival was examined by Kaplan-Meier estimates and the log-rank test. The independence of prognostic factors was determined by Cox multivariate analysis. RESULTS High intra-tumoral expression of all these proteins of the TRAIL pathway was found. The TRAIL receptors and FLIP(L) were not associated with survival. On univariate analysis, strong FLIP(T) expression was associated with a significantly better survival (p = 0.001). On multivariate analysis using the Cox proportional hazards model, FLIP(T) phenotype was significantly associated with a good prognosis in this series (HR 0.52, 95 % CI 0.35-0.78, p = 0.039). Results indicate that this association is valid for all the biological subtypes of breast cancer. The expression of FLIP(T) was especially high in the luminal subtype, known for its good prognosis. CONCLUSIONS These findings support the use of agonistic TRAIL antibodies and drugs targeting FLIP in breast cancer patients. Over-expression of FLIP(T) but not TRAIL-R1, TRAIL-R2 or FLIP(L) provides stage-independent prognostic information in breast cancer patients. This indicates a clinically less aggressive phenotype.
Collapse
Affiliation(s)
- Gustav J Ullenhag
- Section of Oncology, Department of Radiology, Oncology and Radiation Science, Uppsala University, Uppsala, Sweden,
| | | | | | | | | | | |
Collapse
|
738
|
Bernstein JM, Andrews TD, Slevin NJ, West CML, Homer JJ. Prognostic value of hypoxia-associated markers in advanced larynx and hypopharynx squamous cell carcinoma. Laryngoscope 2014; 125:E8-15. [PMID: 25230150 DOI: 10.1002/lary.24933] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Revised: 07/13/2014] [Accepted: 08/25/2014] [Indexed: 11/07/2022]
Abstract
OBJECTIVES/HYPOTHESIS To determine the prognostic value of hypoxia-associated markers carbonic anhydrase-9 (CA-9) and hypoxia-inducible factor-1α (HIF-1α) in advanced larynx and hypopharynx squamous cell carcinoma (SCCa) treated by organ preservation strategies. STUDY DESIGN Retrospective cohort study. METHODS Pretreatment CA-9 and HIF-1α expression, clinicopathologic data, and tumor volume were analyzed in a series of 114 patients with T3-4 SCCa larynx or hypopharynx treated by (chemo)radiation. RESULTS Adverse prognostic factors for locoregional control were T4 classification (P = 0.008), and for disease-specific survival were CA-9 positivity (P = 0.039), T4 classification (P = 0.001), larger tumor volume (P = 0.004), N1-3 classification (P = 0.002), and pretreatment hemoglobin < 13.0 g/dl (P = 0.014). With increasing CA-9 expression, there was a trend to increasing tumor recurrence (P trend = 0.009) and decreasing survival (P trend = 0.002). On multivariate analysis, independent variables were T4 classification (hazard ratio [HR] 13.54, P = 0.01) for locoregional failure, and CA-9 positivity (HR = 8.02, P = 0.042) and higher tumor volume (HR = 3.33, P = 0.007) for disease-specific mortality. CONCLUSION This is the first study to look specifically at T3 and T4 SCCa larynx and hypopharynx for a relationship between hypoxia-associated marker expression and clinical outcome. Pretreatment immunohistochemical CA-9 expression is an adverse prognostic factor for disease-specific survival, indicating that CA-9 expression may confer a more aggressive tumor phenotype.
Collapse
Affiliation(s)
- Jonathan M Bernstein
- University Department of Otolaryngology-Head & Neck Surgery, Manchester Royal Infirmary and Manchester Academic Health Science Centre; Translational Radiobiology Group, Institute of Cancer Sciences, Manchester Academic Health Science Centre, University of Manchester
| | | | | | | | | |
Collapse
|
739
|
Birch SE, Kench JG, Takano E, Chan P, Chan AL, Chiam K, Veillard AS, Stricker P, Haupt S, Haupt Y, Horvath L, Fox SB. Expression of E6AP and PML predicts for prostate cancer progression and cancer-specific death. Ann Oncol 2014; 25:2392-2397. [PMID: 25231954 DOI: 10.1093/annonc/mdu454] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The promyelocytic leukemia (PML) tumor suppressor plays an important role in the response to a variety of cellular stressors and its expression is downregulated or lost in a range of human tumors. We have previously shown that the E3 ligase E6-associated protein (E6AP) is an important regulator of PML protein stability but the relationship and clinical impact of PML and E6AP expression in prostatic carcinoma is unknown. METHODS E6AP and PML expression was assessed in tissue microarrays from a phase I discovery cohort of 170 patients treated by radical prostatectomy for localized prostate cancer (PC). Correlation analysis was carried out between PML and E6AP expression and clinicopathological variates including PSA as a surrogate of disease recurrence. The results were confirmed in a phase II validation cohort of 318 patients with associated clinical recurrence and survival data. RESULTS Survival analysis of the phase I cohort revealed that patients whose tumors showed reduced PML and high E6AP expression had reduced time to PSA relapse (P = 0.012). This was confirmed in the phase II validation cohort where the expression profile of high E6AP/low PML was significantly associated with reduced time to PSA relapse (P < 0.001), clinical relapse (P = 0.016) and PC-specific death (P = 0.014). In multivariate analysis, this expression profile was an independent prognostic indicator of PSA relapse and clinical relapse independent of clinicopathologic factors predicting recurrence. CONCLUSION This study identifies E6AP and PML as potential prognostic markers in localized prostate carcinoma and supports a role for E6AP in driving the downregulation or loss of PML expression in prostate carcinomas.
Collapse
Affiliation(s)
- S E Birch
- Department of Pathology, Peter MacCallum Cancer, East Melbourne.
| | - J G Kench
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Camperdown, Sydney; Sydney Medical School, University of Sydney, Sydney; The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney
| | - E Takano
- Department of Pathology, Peter MacCallum Cancer, East Melbourne
| | - P Chan
- Department of Pathology, Peter MacCallum Cancer, East Melbourne
| | - A-L Chan
- Department of Pathology, University of Melbourne, Melbourne
| | - K Chiam
- The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney
| | - A-S Veillard
- NHMRC Clinical Trial Centre, University of Sydney, Sydney
| | - P Stricker
- The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney; Department of Urology, St Vincent's Clinic, Sydney
| | - S Haupt
- Department of Pathology, University of Melbourne, Melbourne
| | - Y Haupt
- Department of Pathology, Peter MacCallum Cancer, East Melbourne; Department of Pathology, University of Melbourne, Melbourne; Department of Biochemistry and Molecular Biology, Monash University, Melbourne
| | - L Horvath
- Sydney Medical School, University of Sydney, Sydney; The Kinghorn Cancer Centre, Garvan Institute for Medical Research, Sydney; Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, Australia
| | - S B Fox
- Department of Pathology, Peter MacCallum Cancer, East Melbourne; Department of Pathology, University of Melbourne, Melbourne; Department of Pathology, University of Melbourne, Melbourne
| |
Collapse
|
740
|
Hansen TF, Kjær-Frifeldt S, Morgenthaler S, Blondal T, Lindebjerg J, Jakobsen A, Sørensen FB. The prognostic value of microRNA-126 and microvessel density in patients with stage II colon cancer: results from a population cohort. J Transl Med 2014; 12:254. [PMID: 25199818 PMCID: PMC4174250 DOI: 10.1186/s12967-014-0254-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/03/2014] [Indexed: 12/21/2022] Open
Abstract
Background Angiogenesis plays a pivotal role in malignant tumour growth and the metastatic process. We analysed the prognostic value of two angiogenesis parameters, microRNA-126 (miRNA-126) and microvessel density (MVD), in a population based cohort of patients operated for stage II colon cancer. Methods A total of 560 patients were included. Analyses were performed on formalin fixed paraffin embedded tissue from the primary tumours. The analysis of miRNA-126 expression was performed by qPCR. Microvessels were visualised by CD105 and quantified in hot spots using a light microscope. The analyses were correlated with recurrence-free cancer specific survival (RF-CSS) and overall survival (OS). Results Low miRNA-126 expression was significantly correlated to T4, high malignancy grade, tumour perforation, fixation, and the presence of microsatellite instability. A prognostic impact on OS was detected in the simple analysis favouring patients with high miRNA-126 expression p = 0.03, and borderline significance as to RF-CSS, p = 0.08. The impact on OS demonstrated borderline significance in a following multiple Cox regression analysis, hazard ratio 0.76 (95% confidence interval, 0.58-1.00), p = 0.051. The MVD estimate was not associated with either RF-CSS, p = 0.49, or OS, p = 0.94. Conclusion The current population based study of patients operated for stage II colon cancer demonstrated correlations between several prognostic unfavourable characteristics and miRNA-126 and argues for a possible prognostic impact on overall survival. An influence on survival by the MVD estimate was not detected.
Collapse
|
741
|
Bertucci F, Finetti P, Viens P, Birnbaum D. EndoPredict predicts for the response to neoadjuvant chemotherapy in ER-positive, HER2-negative breast cancer. Cancer Lett 2014; 355:70-5. [PMID: 25218596 DOI: 10.1016/j.canlet.2014.09.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/04/2014] [Accepted: 09/04/2014] [Indexed: 12/30/2022]
Abstract
The EndoPredict (EP) signature is a prognostic 11-gene expression signature specifically developed in ER+/HER2- node-negative/positive breast cancer. It is associated with relapse-free survival in patients treated with adjuvant hormone therapy, suggesting that EP low-risk patients could be treated with adjuvant hormone therapy alone whereas high-risk patients would deserve addition of adjuvant chemotherapy. Thus, it is important to determine whether EP high-risk patients are or are not more sensitive to chemotherapy than low-risk patients. Here, we have assessed the EP predictive value for pathological complete response to neoadjuvant chemotherapy in ER+/HER2- breast cancer. We gathered gene expression and histoclinical data of 553 pre-treatment ER+/HER2- breast carcinomas treated with anthracycline-based neoadjuvant chemotherapy. We searched for correlation between the pathological complete response (pCR) and the EP score-based classification. The overall pCR rate was 12%. Fifty-one percent of samples were classified as low-risk according to the EP score and 49% as high-risk. EP classification was associated with a pCR rate of 7% in the low-risk group and 17% in the high-risk group (p < 0.001). In multivariate analysis, the EP score remained significantly associated with pCR. Many genes upregulated in the high-risk tumours were involved in cell proliferation, whereas many genes upregulated in the low-risk tumours were involved in ER-signalling and stroma. Despite higher chemosensitivity, the high-risk group was associated with worse disease-free survival. In conclusion, EP high-risk ER+/HER2- breast cancers are more likely to respond to anthracycline-based chemotherapy.
Collapse
Affiliation(s)
- François Bertucci
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes (IPC), UMR1068 Inserm, 13009 Marseille, France; Faculté de Médecine, Aix-Marseille Université, 13001 Marseille, France.
| | - Pascal Finetti
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes (IPC), UMR1068 Inserm, 13009 Marseille, France
| | - Patrice Viens
- Faculté de Médecine, Aix-Marseille Université, 13001 Marseille, France; Département d'Oncologie Médicale, CRCM, IPC; UMR1068 Inserm, France
| | - Daniel Birnbaum
- Département d'Oncologie Moléculaire, Centre de Recherche en Cancérologie de Marseille (CRCM), Institut Paoli-Calmettes (IPC), UMR1068 Inserm, 13009 Marseille, France
| |
Collapse
|
742
|
Lin YS, Liu YF, Chou YE, Yang SF, Chien MH, Wu CH, Chou CH, Cheng CW, Wang PH. Correlation of chitinase 3-like 1 single nucleotide polymorphisms and haplotypes with uterine cervical cancer in Taiwanese women. PLoS One 2014; 9:e104038. [PMID: 25203433 PMCID: PMC4159225 DOI: 10.1371/journal.pone.0104038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/04/2014] [Indexed: 02/04/2023] Open
Abstract
Background This study aimed to investigate the relationships of chitinase 3-like 1 (CHI3L1) single nucleotide polymorphisms (SNPs) and haplotypes with the development of uterine cervical cancer in Taiwanese women. The SNPs frequencies and haplotypes were also correlated with the clinicopathologic variables of cervical cancer, cancer recurrence, and patient survival. Methodology and Principal Findings Ninety-nine patients with invasive cancer and 61 with pre-cancerous lesions of the uterine cervix were compared to 310 healthy control subjects. Three SNPs rs6691378 (−1371, G/A), rs10399805 (−247, G/A) and rs4950928 (−131, C/G) in the promoter region, and one SNP rs880633 (+2950, T/C) in exon 5 were analyzed by real time polymerase chain reaction and genotyping. The results showed that the mutant homozygous genotype AA of CHI3L1 SNP rs6691378 and AA of rs10399805, and haplotypes AACC and AACT increased the risk of developing pre-cancerous lesions and invasive cancer. The patients with these risk haplotypes had higher than stage I tumors, larger tumors, and vaginal invasion. In logistic regression model, they also tended to have poor survival event [p = 0.078; odds ratio (OR): 2.99, 95% confidence interval (CI): 0.89–10.08] and a higher probability of recurrence event (p = 0.081; OR: 3.07, 95% CI: 0.87–10.81). There was a significant association between the CHI3L1 risk haplotypes and probability of recurrence (p = 0.002; hazard ratio: 6.21, 95% CI: 1.90–20.41), and a marginal association between the risk haplotypes and overall survival (p = 0.051; hazard ratio: 3.76, 95% CI: 0.99–14.29) in the patients with SCC, using Cox proportional hazard model. Conclusion The CHI3L1 SNPs rs6691378 and rs10399805 and CHI3L1 haplotypes all correlated with the development of cervical pre-cancerous lesions and invasive cancer. The cervical cancer patients with the CHI3L1 haplotypes AACC or AACT had poor clinicopathologic characteristics and poor recurrence and survival events. These risk haplotypes were associated with higher recurrence, especially in the patients with SCC.
Collapse
Affiliation(s)
- Yue-Shan Lin
- Department of Obstetrics and Gynecology, Chi-Mei Foundation Medical Center, Tainan, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Ying-Erh Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hsien Wu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chi-Hung Chou
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Division of Cardiology, Department of Internal Medicine, Yuan-Sheng Hospital and Changhua Christian Hospital, Yuanlin Branch, Yuanlin, Taiwan
| | - Chao-Wen Cheng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hui Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan
- * E-mail:
| |
Collapse
|
743
|
Tian C, Sargent DJ, Krivak TC, Powell MA, Gabrin MJ, Brower SL, Coleman RL. Evaluation of a chemoresponse assay as a predictive marker in the treatment of recurrent ovarian cancer: further analysis of a prospective study. Br J Cancer 2014; 111:843-50. [PMID: 25003664 PMCID: PMC4150278 DOI: 10.1038/bjc.2014.375] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/23/2014] [Accepted: 06/12/2014] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Recently, a prospective study reported improved clinical outcomes for recurrent ovarian cancer patients treated with chemotherapies indicated to be sensitive by a chemoresponse assay, compared with those patients treated with non-sensitive therapies, thereby demonstrating the assay's prognostic properties. Due to cross-drug response over different treatments and possible association of in vitro chemosensitivity of a tumour with its inherent biology, further analysis is required to ascertain whether the assay performs as a predictive marker as well. METHODS Women with persistent or recurrent epithelial ovarian cancer (n=262) were empirically treated with one of 15 therapies, blinded to assay results. Each patient's tumour was assayed for responsiveness to the 15 therapies. The assay's ability to predict progression-free survival (PFS) was assessed by comparing the association when the assayed therapy matches the administered therapy (match) with the association when the assayed therapy is randomly selected, not necessarily matching the administered therapy (mismatch). RESULTS Patients treated with assay-sensitive therapies had improved PFS vs patients treated with non-sensitive therapies, with the assay result for match significantly associated with PFS (hazard ratio (HR)=0.67, 95% confidence interval (CI)=0.50-0.91, P=0.009). On the basis of 3000 simulations, the mean HR for mismatch was 0.81 (95% range=0.66-0.99), with 3.4% of HRs less than 0.67, indicating that HR for match is lower than for mismatch. While 47% of tumours were non-sensitive to all assayed therapies and 9% were sensitive to all, 44% displayed heterogeneity in assay results. Improved outcome was associated with the administration of an assay-sensitive therapy, regardless of homogeneous or heterogeneous assay responses across all of the assayed therapies. CONCLUSIONS These analyses provide supportive evidence that this chemoresponse assay is a predictive marker, demonstrating its ability to discern specific therapies that are likely to be more effective among multiple alternatives.
Collapse
Affiliation(s)
- C Tian
- Precision Therapeutics, Inc., 2516 Jane Street, Pittsburgh, PA 15203, USA
| | - D J Sargent
- Health Sciences Research, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - T C Krivak
- The Western Pennsylvania Hospital, 4800 Friendship Avenue, Pittsburgh, PA 15224, USA
| | - M A Powell
- Washington University School of Medicine, 4911 Barnes-Jewish Hospital Plaza, St. Louis, MO 63110, USA
| | - M J Gabrin
- Precision Therapeutics, Inc., 2516 Jane Street, Pittsburgh, PA 15203, USA
| | - S L Brower
- Precision Therapeutics, Inc., 2516 Jane Street, Pittsburgh, PA 15203, USA
| | - R L Coleman
- University of Texas MD Anderson Cancer Center, 1155 Herman Pressler Drive, Houston, TX 77030, USA
| |
Collapse
|
744
|
Wirsing AM, Rikardsen OG, Steigen SE, Uhlin-Hansen L, Hadler-Olsen E. Characterisation and prognostic value of tertiary lymphoid structures in oral squamous cell carcinoma. BMC Clin Pathol 2014; 14:38. [PMID: 25177210 PMCID: PMC4148494 DOI: 10.1186/1472-6890-14-38] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/18/2014] [Indexed: 12/15/2022] Open
Abstract
Background Oral squamous cell carcinomas are often heavily infiltrated by immune cells. The organization of B-cells, follicular dendritic cells, T-cells and high-endothelial venules into structures termed tertiary lymphoid structures have been detected in various types of cancer, where their presence is found to predict favourable outcome. The purpose of the present study was to evaluate the incidence of tertiary lymphoid structures in oral squamous cell carcinomas, and if present, analyse whether they were associated with clinical outcome. Methods Tumour samples from 80 patients with oral squamous cell carcinoma were immunohistochemically stained for B-cells, follicular dendritic cells, T-cells, germinal centre B-cells and high-endothelial venules. Some samples were sectioned at multiple levels to assess whether the presence of tertiary lymphoid structures varied within the tumour. Results Tumour-associated tertiary lymphoid structures were detected in 21 % of the tumours and were associated with lower disease-specific death. The presence of tertiary lymphoid structures varied within different levels of a tissue block. Conclusions Tertiary lymphoid structure formation was found to be a positive prognostic factor for patients with oral squamous cell carcinoma. Increased knowledge about tertiary lymphoid structure formation in oral squamous cell carcinoma might help to develop and guide immune-modulatory cancer treatments.
Collapse
Affiliation(s)
- Anna M Wirsing
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø 9037, Norway
| | - Oddveig G Rikardsen
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø 9037, Norway ; Department of Otorhinolaryngology, University Hospital of North Norway, Tromsø 9038, Norway
| | - Sonja E Steigen
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø 9037, Norway ; Diagnostic Clinic - Clinical Pathology, University Hospital of North Norway, Tromsø 9038, Norway
| | - Lars Uhlin-Hansen
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø 9037, Norway ; Diagnostic Clinic - Clinical Pathology, University Hospital of North Norway, Tromsø 9038, Norway
| | - Elin Hadler-Olsen
- Department of Medical Biology, Faculty of Health Sciences, University of Tromsø, Tromsø 9037, Norway
| |
Collapse
|
745
|
Mahon KL, Qu W, Devaney J, Paul C, Castillo L, Wykes RJ, Chatfield MD, Boyer MJ, Stockler MR, Marx G, Gurney H, Mallesara G, Molloy PL, Horvath LG, Clark SJ. Methylated Glutathione S-transferase 1 (mGSTP1) is a potential plasma free DNA epigenetic marker of prognosis and response to chemotherapy in castrate-resistant prostate cancer. Br J Cancer 2014; 111:1802-9. [PMID: 25144624 PMCID: PMC4453725 DOI: 10.1038/bjc.2014.463] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/16/2014] [Accepted: 07/24/2014] [Indexed: 12/25/2022] Open
Abstract
Background: Glutathione S-transferase 1 (GSTP1) inactivation is associated with CpG island promoter hypermethylation in the majority of prostate cancers (PCs). This study assessed whether the level of circulating methylated GSTP1 (mGSTP1) in plasma DNA is associated with chemotherapy response and overall survival (OS). Methods: Plasma samples were collected prospectively from a Phase I exploratory cohort of 75 men with castrate-resistant PC (CRPC) and a Phase II independent validation cohort (n=51). mGSTP1 levels in free DNA were measured using a sensitive methylation-specific PCR assay. Results: The Phase I cohort identified that detectable baseline mGSTP1 DNA was associated with poorer OS (HR, 4.2 95% CI 2.1–8.2; P<0.0001). A decrease in mGSTP1 DNA levels after cycle 1 was associated with a PSA response (P=0.008). In the Phase II cohort, baseline mGSTP1 DNA was a stronger predictor of OS than PSA change after 3 months (P=0.02). Undetectable plasma mGSTP1 after one cycle of chemotherapy was associated with PSA response (P=0.007). Conclusions: We identified plasma mGSTP1 DNA as a potential prognostic marker in men with CRPC as well as a potential surrogate therapeutic efficacy marker for chemotherapy and corroborated these findings in an independent Phase II cohort. Prospective Phase III assessment of mGSTP1 levels in plasma DNA is now warranted.
Collapse
Affiliation(s)
- K L Mahon
- 1] Chris O'Brien Lifehouse, Missenden Rd, Camperdown, New South Wales, 2050, Australia [2] Cancer Research Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, New South Wales, 2010, Australia [3] Sydney Medical School, University of Sydney, Camperdown, New South Wales, 2050, Australia
| | - W Qu
- Cancer Research Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, New South Wales, 2010, Australia
| | - J Devaney
- Cancer Research Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, New South Wales, 2010, Australia
| | - C Paul
- Cancer Research Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, New South Wales, 2010, Australia
| | - L Castillo
- Cancer Research Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, New South Wales, 2010, Australia
| | - R J Wykes
- Royal Prince Alfred Hospital, Missenden Rd, Camperdown, New South Wales, 2050, Australia
| | - M D Chatfield
- Menzies School of Health Research, Darwin, Northern Territory, Australia
| | - M J Boyer
- 1] Chris O'Brien Lifehouse, Missenden Rd, Camperdown, New South Wales, 2050, Australia [2] Sydney Medical School, University of Sydney, Camperdown, New South Wales, 2050, Australia [3] Royal Prince Alfred Hospital, Missenden Rd, Camperdown, New South Wales, 2050, Australia
| | - M R Stockler
- 1] Chris O'Brien Lifehouse, Missenden Rd, Camperdown, New South Wales, 2050, Australia [2] Sydney Medical School, University of Sydney, Camperdown, New South Wales, 2050, Australia [3] NHMRC Clinical Trials Centre, University of Sydney, New South Wales, 2050, Australia
| | - G Marx
- 1] Sydney Medical School, University of Sydney, Camperdown, New South Wales, 2050, Australia [2] Northern Haematology and Oncology Group, SAN Clinic, Wahroonga, New South Wales, 2076, Australia
| | - H Gurney
- 1] Sydney Medical School, University of Sydney, Camperdown, New South Wales, 2050, Australia [2] Westmead Hospital, Sydney, New South Wales, Australia
| | - G Mallesara
- Calvary Mater Newcastle, New South Wales, Australia
| | - P L Molloy
- CSIRO Animal, Food and Health Sciences, North Ryde, New South Wales, 2113, Australia
| | - L G Horvath
- 1] Chris O'Brien Lifehouse, Missenden Rd, Camperdown, New South Wales, 2050, Australia [2] Cancer Research Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, New South Wales, 2010, Australia [3] Sydney Medical School, University of Sydney, Camperdown, New South Wales, 2050, Australia [4] Royal Prince Alfred Hospital, Missenden Rd, Camperdown, New South Wales, 2050, Australia
| | - S J Clark
- 1] Cancer Research Division, Garvan Institute of Medical Research/The Kinghorn Cancer Centre, Darlinghurst, New South Wales, 2010, Australia [2] St Vincent's Clinical School, University of NSW, Sydney, 2010, New South Wales, Australia
| | | |
Collapse
|
746
|
Derr RS, van Hoesel AQ, Benard A, Goossens-Beumer IJ, Sajet A, Dekker-Ensink NG, de Kruijf EM, Bastiaannet E, Smit VTHBM, van de Velde CJH, Kuppen PJK. High nuclear expression levels of histone-modifying enzymes LSD1, HDAC2 and SIRT1 in tumor cells correlate with decreased survival and increased relapse in breast cancer patients. BMC Cancer 2014; 14:604. [PMID: 25139823 PMCID: PMC4148956 DOI: 10.1186/1471-2407-14-604] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/08/2014] [Indexed: 11/21/2022] Open
Abstract
Background Breast cancer is a heterogeneous disease with a highly variable clinical outcome in which both genetic and epigenetic changes have critical roles. We investigated tumor expression levels of histone-modifying enzymes LSD1, HDAC2 and SIRT1 in relation with patient survival and tumor relapse in a retrospective cohort of 460 breast cancer patients. Additionally, we correlated expression levels with tumor differentiation and tumor cell proliferation. Methods Immunohistochemical staining for LSD1, HDAC2 and SIRT1 was performed on tissue microarrays of tumor and corresponding normal formalin-fixed paraffin-embedded tissues from breast cancer patients. Median nuclear expression levels in tumor tissues were used to divide the patients into low and high expression categories. In combined expression analyses, patients were divided into four subgroups: 1, all enzymes below-median; 2, one enzyme above-median; 3, two enzymes above-median; 4, all three enzymes above-median. The Cox proportional hazard model was used for univariate and multivariate survival analyses. The Pearson Chi-square method was used to assess correlation of combined expression levels with tumor cell proliferation and tumor differentiation. Results Expression of LSD1 and SIRT1, but not of HDAC2, was significantly increased in tumor tissues compared to their normal counterparts (both p < 0.001). Multivariate survival analyses identified SIRT1 as independent prognostic factor for relapse-free survival (RFS) with a hazard ratio (HR) of 1.34 (95% CI = 1.04-1.74, p = 0.02). For overall survival (OS), no significant differences were found when the individual enzymes were analyzed. Analyses of combined expression levels of the three histone-modifying enzymes correlated with OS (p = 0.03) and RFS (p = 0.006) with a HR of respectively 1.49 (95% CI = 1.07-2.08) and 1.68 (95% CI = 1.16-2.44) in multivariate analyses and were also related to tumor differentiation (p < 0.001) and tumor cell proliferation (p = 0.002). Conclusions When the combined expression levels were analyzed, high expression of LSD1, HDAC2 and SIRT1 showed shorter patient survival time and shorter time to tumor relapse and correlated with poor tumor differentiation and a high level of tumor cell proliferation. Expression of these histone-modifying enzymes might therefore be involved in breast cancer pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Peter J K Kuppen
- Department of Surgery, K6-R, Leiden University Medical Center (LUMC), P,O, Box 9600, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
747
|
Hansen TF, Nielsen BS, Sørensen FB, Johnsson A, Jakobsen A. Epidermal growth factor-like domain 7 predicts response to first-line chemotherapy and bevacizumab in patients with metastatic colorectal cancer. Mol Cancer Ther 2014; 13:2238-45. [PMID: 25140000 DOI: 10.1158/1535-7163.mct-14-0131] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The number of approved antiangiogenic drugs is constantly growing and emphasizes the need for predictive biomarkers. The aim of this study was to analyze the predictive value of epidermal growth factor-like domain 7 (EGFL7) and microRNA-126 (miR126) to first-line chemotherapy combined with bevacizumab, in patients with metastatic colorectal cancer (mCRC). A total of 158 patients from two different, but comparable, cohorts were included. Analyses were performed on tumor tissue from the primary tumor either based on a whole-tumor resection or an endoscopic biopsy. EGFL7 was analyzed by immunohistochemistry (IHC) and miR126 by in situ hybridization (ISH). Both biomarkers were quantified by image-guided analyses. Endpoints were response rate (RR) and progression-free survival (PFS). The EGFL7 vessel area (VA) in tumor resections was closely related to treatment response with a median EGFL7 VA in responding patients of 4 [95% confidence interval (CI), 4-6] compared with 8.5 (95% CI, 7-11) in nonresponders, P = 0.0008. This difference translated into a borderline significant difference in PFS (P = 0.06). Furthermore, a significant relationship between high EGFL7 VA and KRAS mutation was detected (P = 0.049). The results showed no significant relationship between the miR126 VA and the clinical endpoints. Our study suggests a predictive value of EGFL7 in regard to first-line chemotherapy and bevacizumab in patients with mCRC and supports the mechanism of a dual blocking of the vascular endothelial growth factor-A and EGFL7 axis in this setting.
Collapse
Affiliation(s)
- Torben Frøstrup Hansen
- Department of Oncology, Vejle Hospital, part of Lillebaelt Hospital, Vejle, Denmark. Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.
| | | | - Flemming Brandt Sørensen
- Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark. Department of Clinical Pathology, Vejle Hospital, Part of Lillebaelt Hospital, Vejle, Denmark
| | | | - Anders Jakobsen
- Department of Oncology, Vejle Hospital, part of Lillebaelt Hospital, Vejle, Denmark. Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
748
|
Pretherapeutic gamma-glutamyltransferase is an independent prognostic factor for patients with renal cell carcinoma. Br J Cancer 2014; 111:1526-31. [PMID: 25117808 PMCID: PMC4200090 DOI: 10.1038/bjc.2014.450] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/13/2014] [Accepted: 07/15/2014] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Gamma-glutamyltransferase (GGT) regulates apoptotic balance and promotes cancer progression and invasion. Higher pretherapeutic GGT serum levels have been associated with worse outcomes in various malignancies, but there are no data for renal cell carcinoma (RCC). METHODS Pretherapeutic GGT serum levels and clinicopathological parameters were retrospectively evaluated in 921 consecutive RCC patients treated with nephrectomy at a single institution between 1998 and 2013. Gamma-glutamyltransferase was analysed as continuous and categorical variable. Associations with RCC-specific survival were assessed with Cox proportional hazards models. Discrimination was measured with the C-index. Decision-curve analysis was used to evaluate the clinical net benefit. The median postoperative follow-up was 45 months. RESULTS Median pretherapeutic serum GGT level was 25 U l(-1). Gamma-glutamyltransferase levels increased with advancing T (P<0.001), N (P=0.006) and M stages (P<0.001), higher grades (P<0.001), and presence of tumour necrosis (P<0.001). An increase of GGT by 10 U l(-1) was associated with an increase in the risk of death from RCC by 4% (HR 1.04, P<0.001). Based on recursive partitioning-based survival tree analysis, we defined four prognostic categories of GGT: normal low (<17.5 U l(-1)), normal high (17.5 to <34.5 U l(-1)), elevated (34.5 to <181.5 U l(-1)), and highly elevated (⩾181.5 U l(-1)). In multivariable analyses that adjusted for the effect of standard features, both continuously and categorically coded GGT were independent prognostic factors. Adding GGT to a model that included standard features increased the discrimination by 0.9% to 1.8% and improved the clinical net benefit. CONCLUSIONS Pretherapeutic serum GGT is a novel and independent prognostic factor for patients with RCC. Stratifying patients into prognostic subgroups according to GGT may be used for patient counselling, tailoring surveillance, individualised treatment planning, and clinical trial design.
Collapse
|
749
|
Agulló-Ortuño MT, Díaz-García CV, Agudo-López A, Pérez C, Cortijo A, Paz-Ares L, López-Ríos F, Pozo F, de Castro J, Cortés-Funes H, López Martín JA. Relevance of insulin-like growth factor 1 receptor gene expression as a prognostic factor in non-small-cell lung cancer. J Cancer Res Clin Oncol 2014; 141:43-53. [PMID: 25081930 DOI: 10.1007/s00432-014-1787-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/22/2014] [Indexed: 01/05/2023]
Abstract
PURPOSE Signalling through the insulin-like growth factor 1 receptor (IGF-1R) is implicated in carcinogenesis, metastasis, and resistance to cytotoxic cancer therapies. The purpose of this study was to investigate the prognostic role of IGF-1R expression in surgically resected non-small-cell lung cancer (NSCLC), and responses to IGF-1R tyrosine kinase inhibitor NVP-ADW742 in a panel of lung cancer cell lines. METHODS Insulin-like growth factor 1 receptor (IGF-1R) expression was evaluated by quantitative RT-PCR in 115 NSCLC samples and in a panel of 6 NSCLC cell lines. Cytotoxicity experiments with IGF-1R inhibitor and conventional systemic drugs such as paclitaxel in cell lines were realised. RESULTS Insulin-like growth factor 1 receptor (IGF-1R) was differentially expressed across histologic subtypes, with the lowest levels observed in squamous cell tumours. Median survival was longer in patients with squamous tumour histology expressing low IGF-1R levels. In multivariable analysis, ageing and high tumour stage were significant predictors of worse overall survival. The hazard of death was lower in patients with squamous histology and low IGF-1R gene expression. There was no correlation between IGF-1R expression and response to tyrosine kinase inhibitor in cell lines tested. However, combination drug treatment resulted in synergistically enhanced antiproliferative effects on several cell lines. CONCLUSIONS These findings suggest that IGF-1R is a potential target for therapy in NSCLC patients. Combination therapies will have an important role in treatment.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/mortality
- Adenocarcinoma/pathology
- Adult
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Carcinoma, Large Cell/genetics
- Carcinoma, Large Cell/mortality
- Carcinoma, Large Cell/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/pathology
- Female
- Follow-Up Studies
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Neoplasm Grading
- Neoplasm Staging
- Prognosis
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, IGF Type 1/genetics
- Retrospective Studies
- Reverse Transcriptase Polymerase Chain Reaction
- Survival Rate
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- M Teresa Agulló-Ortuño
- Translational Oncology, Instituto de Investigación Hospital 12 de Octubre (i+12), Avda de Córdoba S/N, 28041, Madrid, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
750
|
Glover AR, Lee JC, Sidhu SB. Is there an accurate biomarker test for thyroid cancer recurrence on the horizon? INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2014. [DOI: 10.2217/ije.14.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Anthony R Glover
- Kolling Institute of Medical Research, Cancer Genetics Laboratory, Royal North Shore Hospital & University of Sydney, Sydney, Australia
- University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia
| | - James C Lee
- Kolling Institute of Medical Research, Cancer Genetics Laboratory, Royal North Shore Hospital & University of Sydney, Sydney, Australia
- Endocrine Surgical Unit, The Alfred Hospital, Melbourne, Australia
| | - Stan B Sidhu
- Kolling Institute of Medical Research, Cancer Genetics Laboratory, Royal North Shore Hospital & University of Sydney, Sydney, Australia
- University of Sydney Endocrine Surgical Unit, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|