1
|
Zeng D, Song Z, Liu Q, Huang J, Wang X, Tang Z. Radiomics analysis of dual-layer detector spectral CT-derived iodine maps for predicting Ki-67 PI in pancreatic ductal adenocarcinoma. BMC Med Imaging 2025; 25:124. [PMID: 40247246 PMCID: PMC12007212 DOI: 10.1186/s12880-025-01664-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE To evaluate the feasibility of radiomics analysis using dual-layer detector spectral CT (DLCT)-derived iodine maps for the preoperative prediction of the Ki-67 proliferation index (PI) in pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS A total of 168 PDAC patients who underwent DLCT examination were included and randomly allocated to the training (n = 118) and validation (n = 50) sets. A clinical model was constructed using independent clinicoradiological features identified through multivariate logistic regression analysis in the training set. The radiomics signature was generated based on the coefficients of selected features from iodine maps in the arterial and portal venous phases of DLCT. Finally, a radiomics-clinical model was developed by integrating the radiomics signature and significant clinicoradiological features. The predictive performance of three models was evaluated using the Receiver Operating Characteristic (ROC) curve and Decision Curve Analysis. The optimal model was then used to develop a nomogram, with goodness-of-fit evaluated through the calibration curve. RESULTS The radiomics-clinical model integrating radiomics signature, CA19-9, and CT-reported regional lymph node status demonstrated excellent performance in predicting Ki-67 PI in PDAC, which showed an area under the ROC curve of 0.979 and 0.956 in the training and validation sets, respectively. The radiomics-clinical nomogram demonstrated the improved net benefit and exhibited satisfactory consistency. CONCLUSIONS This exploratory study demonstrated the feasibility of using DLCT-derived iodine map-based radiomics to predict Ki-67 PI preoperatively in PDAC patients. While preliminary, our findings highlight the potential of functional imaging combined with radiomics for personalized treatment planning.
Collapse
Affiliation(s)
- Dan Zeng
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Zuhua Song
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Qian Liu
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Jie Huang
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Xinwei Wang
- Department of Radiology, Chongqing General Hospital, Chongqing, China
| | - Zhuoyue Tang
- Department of Radiology, Chongqing General Hospital, Chongqing, China.
| |
Collapse
|
2
|
Miti T, Desai B, Miroshnychenko D, Basanta D, Marusyk A. Dissecting the Spatially Restricted Effects of Microenvironment-Mediated Resistance on Targeted Therapy Responses. Cancers (Basel) 2024; 16:2405. [PMID: 39001467 PMCID: PMC11240540 DOI: 10.3390/cancers16132405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
The response of tumors to anti-cancer therapies is defined not only by cell-intrinsic therapy sensitivities but also by local interactions with the tumor microenvironment. Fibroblasts that make tumor stroma have been shown to produce paracrine factors that can strongly reduce the sensitivity of tumor cells to many types of targeted therapies. Moreover, a high stroma/tumor ratio is generally associated with poor survival and reduced therapy responses. However, in contrast to advanced knowledge of the molecular mechanisms responsible for stroma-mediated resistance, its effect on the ability of tumors to escape therapeutic eradication remains poorly understood. To a large extent, this gap of knowledge reflects the challenge of accounting for the spatial aspects of microenvironmental resistance, especially over longer time frames. To address this problem, we integrated spatial inferences of proliferation-death dynamics from an experimental animal model of targeted therapy responses with spatial mathematical modeling. With this approach, we dissected the impact of tumor/stroma distribution, magnitude and distance of stromal effects. While all of the tested parameters affected the ability of tumor cells to resist elimination, spatial patterns of stroma distribution within tumor tissue had a particularly strong impact.
Collapse
Affiliation(s)
- Tatiana Miti
- Department of Integrative Mathematical Oncology, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA;
| | - Bina Desai
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL 33620, USA
| | - Daria Miroshnychenko
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
| | - David Basanta
- Department of Integrative Mathematical Oncology, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA;
| | - Andriy Marusyk
- Department of Tumor Microenvironment and Metastasis, H. Lee Moffitt Cancer Centre and Research Institute, Tampa, FL 33612, USA (D.M.)
| |
Collapse
|
3
|
Stillger MN, Kurowski K, Bronsert P, Brombacher E, Kreutz C, Werner M, Tang L, Timme-Bronsert S, Schilling O. Neoadjuvant chemo- or chemo-radiation-therapy of pancreatic ductal adenocarcinoma differentially shift ECM composition, complement activation, energy metabolism and ribosomal proteins of the residual tumor mass. Int J Cancer 2024; 154:2162-2175. [PMID: 38353498 DOI: 10.1002/ijc.34867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/08/2023] [Accepted: 12/20/2023] [Indexed: 04/14/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer, often diagnosed at stages that dis-qualify for surgical resection. Neoadjuvant therapies offer potential tumor regression and improved resectability. Although features of the tumor biology (e.g., molecular markers) may guide adjuvant therapy, biological alterations after neoadjuvant therapy remain largely unexplored. We performed mass spectrometry to characterize the proteomes of 67 PDAC resection specimens of patients who received either neoadjuvant chemo (NCT) or chemo-radiation (NCRT) therapy. We employed data-independent acquisition (DIA), yielding a proteome coverage in excess of 3500 proteins. Moreover, we successfully integrated two publicly available proteome datasets of treatment-naïve PDAC to unravel proteome alterations in response to neoadjuvant therapy, highlighting the feasibility of this approach. We found highly distinguishable proteome profiles. Treatment-naïve PDAC was characterized by enrichment of immunoglobulins, complement and extracellular matrix (ECM) proteins. Post-NCT and post-NCRT PDAC presented high abundance of ribosomal and metabolic proteins as compared to treatment-naïve PDAC. Further analyses on patient survival and protein expression identified treatment-specific prognostic candidates. We present the first proteomic characterization of the residual PDAC mass after NCT and NCRT, and potential protein candidate markers associated with overall survival. We conclude that residual PDAC exhibits fundamentally different proteome profiles as compared to treatment-naïve PDAC, influenced by the type of neoadjuvant treatment. These findings may impact adjuvant or targeted therapy options.
Collapse
Affiliation(s)
- Maren N Stillger
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Konrad Kurowski
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Core Facility for Histopathology and Digital Pathology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Peter Bronsert
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Core Facility for Histopathology and Digital Pathology, Medical Center-University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Brombacher
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Faculty of Medicine and Medical Center, Institute of Medical Biometry and Statistics, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
- Centre for Integrative Biological Signaling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Clemens Kreutz
- Faculty of Medicine and Medical Center, Institute of Medical Biometry and Statistics, University of Freiburg, Freiburg, Germany
| | - Martin Werner
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Core Facility for Histopathology and Digital Pathology, Medical Center-University of Freiburg, Freiburg, Germany
| | - Laura Tang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sylvia Timme-Bronsert
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
- Faculty of Medicine, Core Facility for Histopathology and Digital Pathology, Medical Center-University of Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Li Q, Song Z, Li X, Zhang D, Yu J, Li Z, Huang J, Su K, Liu Q, Zhang X, Tang Z. Development of a CT radiomics nomogram for preoperative prediction of Ki-67 index in pancreatic ductal adenocarcinoma: a two-center retrospective study. Eur Radiol 2024; 34:2934-2943. [PMID: 37938382 DOI: 10.1007/s00330-023-10393-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/31/2023] [Accepted: 09/13/2023] [Indexed: 11/09/2023]
Abstract
OBJECTIVES To develop and validate a contrast-enhanced computed tomography (CECT)-based radiomics nomogram for the preoperative evaluation of Ki-67 proliferation status in pancreatic ductal adenocarcinoma (PDAC). METHODS In this two-center retrospective study, a total of 181 patients (95 in the training cohort; 42 in the testing cohort, and 44 in the external validation cohort) with PDAC who underwent CECT examination were included. Radiomic features were extracted from portal venous phase images. The radiomics signatures were built by using two feature-selecting methods (relief and recursive feature elimination) and four classifiers (support vector machine, naive Bayes, linear discriminant analysis (LDA), and logistic regression (LR)). Multivariate LR was used to build a clinical model and radiomics-clinical nomogram. The predictive performances of the models were evaluated using area under receiver operating characteristic curve (AUC) and decision curve analysis (DCA). RESULTS The relief selector and LDA classifier using twelve features built the optimal radiomics signature, with AUCs of 0.948, 0.927, and 0.824 in the training, testing, and external validation cohorts, respectively. The radiomics-clinical nomogram incorporating the optimal radiomics signature, CT-reported lymph node status, and CA19-9 showed better predictive performance with AUCs of 0.976, 0.955, and 0.882 in the training, testing, and external validation cohorts, respectively. The calibration curve and DCA demonstrated goodness-of-fit and improved benefits in clinical practice of the nomogram. CONCLUSIONS The radiomics-clinical nomogram is an effective and non-invasive computer-aided tool to predict the Ki-67 expression status in patients with PDAC. CLINICAL RELEVANCE STATEMENT The radiomics-clinical nomogram is an effective and non-invasive computer-aided tool to predict the Ki-67 expression status in patients with pancreatic ductal adenocarcinoma. KEY POINTS The radiomics analysis could be helpful to predict Ki-67 expression status in patients with pancreatic ductal adenocarcinoma (PDAC). The radiomics-clinical nomogram integrated with the radiomics signature, clinical data, and CT radiological features could significantly improve the differential diagnosis of Ki-67 expression status. The radiomics-clinical nomogram showed satisfactory calibration and net benefit for discriminating high and low Ki-67 expression status in PDAC.
Collapse
Affiliation(s)
- Qian Li
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Zuhua Song
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Xiaojiao Li
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Dan Zhang
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Jiayi Yu
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Zongwen Li
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Jie Huang
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Kai Su
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Qian Liu
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China
| | - Xiaodi Zhang
- Department of Clinical Science, Philips Healthcare, Chengdu, China
| | - Zhuoyue Tang
- Department of Radiology, Chongqing General Hospital, Yuzhong District, No. 104, Pipashan Main Street, Chongqing, 400014, China.
| |
Collapse
|
5
|
Wen Y, Song Z, Li Q, Zhang D, Li X, Yu J, Li Z, Ren X, Zhang J, Liu Q, Huang J, Zeng D, Tang Z. Development and validation of a model for predicting the expression of Ki-67 in pancreatic ductal adenocarcinoma with radiological features and dual-energy computed tomography quantitative parameters. Insights Imaging 2024; 15:41. [PMID: 38353857 PMCID: PMC10866831 DOI: 10.1186/s13244-024-01617-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 12/21/2023] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVE To construct and validate a model based on the dual-energy computed tomography (DECT) quantitative parameters and radiological features to predict Ki-67 expression levels in pancreatic ductal adenocarcinoma (PDAC). MATERIALS AND METHODS Data from 143 PDAC patients were analysed. The variables of clinic, radiology and DECT were evaluated. In the arterial phase and portal venous phase (PVP), the normalized iodine concentration (NIC), normalized effective atomic number and slope of the spectral attenuation curves were measured. The extracellular volume fraction (ECVf) was measured in the equilibrium phase. Univariate analysis was used to screen independent risk factors to predict Ki-67 expression. The Radiology, DECT and DECT-Radiology models were constructed, and their diagnostic effectiveness and clinical applicability were obtained through area under the curve (AUC) and decision curve analysis, respectively. The nomogram was established based on the optimal model, and its goodness-of-fit was assessed by a calibration curve. RESULTS Computed tomography reported regional lymph node status, NIC of PVP, and ECVf were independent predictors for Ki-67 expression prediction. The AUCs of the Radiology, DECT, and DECT-Radiology models were 0.705, 0.884, and 0.905, respectively, in the training cohort, and 0.669, 0.835, and 0.865, respectively, in the validation cohort. The DECT-Radiology nomogram was established based on the DECT-Radiology model, which showed the highest net benefit and satisfactory consistency. CONCLUSIONS The DECT-Radiology model shows favourable predictive efficacy for Ki-67 expression, which may be of value for clinical decision-making in PDAC patients. CRITICAL RELEVANCE STATEMENT The DECT-Radiology model could contribute to the preoperative and non-invasive assessment of Ki-67 expression of PDAC, which may help clinicians to screen out PDAC patients with high Ki-67 expression. KEY POINTS • Dual-energy computed tomography (DECT) can predict Ki-67 in pancreatic ductal adenocarcinoma (PDAC). • The DECT-Radiology model facilitates preoperative and non-invasive assessment of PDAC Ki-67 expression. • The nomogram may help screen out PDAC patients with high Ki-67 expression.
Collapse
Affiliation(s)
- Youjia Wen
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Zuhua Song
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Qian Li
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Dan Zhang
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Xiaojiao Li
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Jiayi Yu
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Zongwen Li
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Xiaofang Ren
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Jiayan Zhang
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Qian Liu
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Jie Huang
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Dan Zeng
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China
| | - Zhuoyue Tang
- Chongqing General Hospital, No.118, Xingguang Avenue, Liangjiang New Area, Chongqing, China.
| |
Collapse
|
6
|
Li B, Yin X, Ding X, Zhang G, Jiang H, Chen C, Guo S, Jin G. Combined utility of Ki-67 index and tumor grade to stratify patients with pancreatic ductal adenocarcinoma who underwent upfront surgery. BMC Surg 2023; 23:370. [PMID: 38066512 PMCID: PMC10704770 DOI: 10.1186/s12893-023-02256-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
OBJECTIVE To investigate the prognostic prediction of a new indicator, combined by tumor grade and Ki-67, in patients with resected pancreatic ductal adenocarcinoma (PDAC). METHODS Data were retrospectively collected from consecutive patients who underwent primary resection of pancreas from December 2012 to December 2017. Tumor grade and Ki-67 were reviewed from routine pathological reports. G-Ki67 was classified as three categories as I (G1/2 and Ki-67 < 40%), II (G1/2 and Ki-67 ≥ 40%), and III(G3/4 and all Ki-67). RESULTS Cox regression analyses revealed that tumor stage (II vs. I: hazard ratio (HR), 3.781; 95% confidence index (CI), 2.844-5.025; P < 0.001; III vs. I: HR, 7.476; 95% CI, 5.481-10.20; P < 0.001) and G-Ki67 (II vs. I: HR, 1.299; 95% CI, 1.038-1.624; P = 0.022; III vs. I: HR, 1.942; 95% CI, 1.477-2.554; P < 0.001) were independent prognostic factors in the developing cohort. The result was rectified in the validation cohort. In subgroups analysis, G-Ki67 (II vs. I: HR, 1.866 ; 95% CI, 1.045-3.334; P = 0.035; III vs. I: HR, 2.333 ; 95% CI, 1.156-4.705; P = 0.018) also had a high differentiation for survival prediction. CONCLUSION Our findings indicate that three-categories of G-Ki67 in resectable PDAC according to the routine pathological descriptions provided additional prognostic information complementary to the TNM staging system.
Collapse
Affiliation(s)
- Bo Li
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China
- Department of Hepatobiliary Pancreatic Surgery, Naval Medical Center of People's Liberation Army, Naval Medical University (Second Military Medical University), 338 West Huaihai Road, Shanghai, 200052, China
| | - Xiaoyi Yin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China
| | - Xiuwen Ding
- Clinical Research Center, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China
| | - Guoxiao Zhang
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China
| | - Hui Jiang
- Department of Pathology, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China
| | - Cuimin Chen
- Clinical Research Center, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China.
| | - Shiwei Guo
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China.
| | - Gang Jin
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Naval Medical University (Second Military Medical University), 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
7
|
Miroshnychenko D, Miti T, Kumar P, Miller A, Laurie M, Giraldo N, Bui MM, Altrock PM, Basanta D, Marusyk A. Stroma-Mediated Breast Cancer Cell Proliferation Indirectly Drives Chemoresistance by Accelerating Tumor Recovery between Chemotherapy Cycles. Cancer Res 2023; 83:3681-3692. [PMID: 37791818 PMCID: PMC10646478 DOI: 10.1158/0008-5472.can-23-0398] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/28/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
The ability of tumors to survive therapy reflects both cell-intrinsic and microenvironmental mechanisms. Across many cancers, including triple-negative breast cancer (TNBC), a high stroma/tumor ratio correlates with poor survival. In many contexts, this correlation can be explained by the direct reduction of therapy sensitivity induced by stroma-produced paracrine factors. We sought to explore whether this direct effect contributes to the link between stroma and poor responses to chemotherapies. In vitro studies with panels of TNBC cell line models and stromal isolates failed to detect a direct modulation of chemoresistance. At the same time, consistent with prior studies, fibroblast-produced secreted factors stimulated treatment-independent enhancement of tumor cell proliferation. Spatial analyses indicated that proximity to stroma is often associated with enhanced tumor cell proliferation in vivo. These observations suggested an indirect link between stroma and chemoresistance, where stroma-augmented proliferation potentiates the recovery of residual tumors between chemotherapy cycles. To evaluate this hypothesis, a spatial agent-based model of stroma impact on proliferation/death dynamics was developed that was quantitatively parameterized using inferences from histologic analyses and experimental studies. The model demonstrated that the observed enhancement of tumor cell proliferation within stroma-proximal niches could enable tumors to avoid elimination over multiple chemotherapy cycles. Therefore, this study supports the existence of an indirect mechanism of environment-mediated chemoresistance that might contribute to the negative correlation between stromal content and poor therapy outcomes. SIGNIFICANCE Integration of experimental research with mathematical modeling reveals an indirect microenvironmental chemoresistance mechanism by which stromal cells stimulate breast cancer cell proliferation and highlights the importance of consideration of proliferation/death dynamics. See related commentary by Wall and Echeverria, p. 3667.
Collapse
Affiliation(s)
- Daria Miroshnychenko
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Tatiana Miti
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Pragya Kumar
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Cancer Biology PhD Program, University of South Florida, Tampa, Florida
| | - Anna Miller
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mark Laurie
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Nathalia Giraldo
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Molecular Medicine, University of South Florida, Tampa, Florida
| | - Marilyn M. Bui
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Philipp M. Altrock
- Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, Ploen, Schleswig-Holstein, Germany
| | - David Basanta
- Department of Integrated Mathematical Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Andriy Marusyk
- Department of Metabolism and Physiology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Molecular Medicine, University of South Florida, Tampa, Florida
| |
Collapse
|
8
|
Miroshnychenko D, Miti T, Miller A, Kumar P, Laurie M, Bui MM, Altrock PM, Basanta D, Marusyk A. Paracrine enhancement of tumor cell proliferation provides indirect stroma-mediated chemoresistance via acceleration of tumor recovery between chemotherapy cycles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.07.527543. [PMID: 36798328 PMCID: PMC9934626 DOI: 10.1101/2023.02.07.527543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The ability of tumors to survive therapy is mediated not only by cell-intrinsic but also cell-extrinsic, microenvironmental mechanisms. Across many cancers, including triple-negative breast cancer (TNBC), a high stroma/tumor ratio correlates with poor survival. In many contexts, this correlation can be explained by the direct reduction of therapy sensitivity by stroma-produced paracrine factors through activating pro-survival signaling and stemness. We sought to explore whether this direct effect contributes to the link between stroma and poor responses to chemotherapies in TNBC. Our in vitro studies with panels of TNBC cell line models and stromal isolates failed to detect a direct modulation of chemoresistance. However, we found that fibroblasts often enhance baseline tumor cell proliferation. Consistent with this in vitro observation, we found evidence of stroma-enhanced TNBC cell proliferation in vivo , in xenograft models and patient samples. Based on these observations, we hypothesized an indirect link between stroma and chemoresistance, where stroma-augmented proliferation potentiates the recovery of residual tumors between chemotherapy cycles. To test this hypothesis, we developed a spatial agent-based model of tumor response to repeated dosing of chemotherapy. The model was quantitatively parameterized from histological analyses and experimental studies. We found that even a slight enhancement of tumor cell proliferation within stroma-proximal niches can strongly enhance the ability of tumors to survive multiple cycles of chemotherapy under biologically and clinically feasible parameters. In summary, our study uncovered a novel, indirect mechanism of chemoresistance. Further, our study highlights the limitations of short-term cytotoxicity assays in understanding chemotherapy responses and supports the integration of experimental and in silico modeling.
Collapse
|
9
|
Organic cation transporter 2 activation enhances sensitivity to oxaliplatin in human pancreatic ductal adenocarcinoma. Biomed Pharmacother 2022; 153:113520. [DOI: 10.1016/j.biopha.2022.113520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/31/2022] [Accepted: 08/08/2022] [Indexed: 11/18/2022] Open
|
10
|
Polymorphism of Baculoviral Inhibitor of Apoptosis Repeat-Containing 5 (BIRC5) Can Be Associated with Clinical Outcome of Non-Small Cell Lung Cancer. Cells 2022; 11:cells11060956. [PMID: 35326407 PMCID: PMC8946487 DOI: 10.3390/cells11060956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 01/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) comprises about 85% of all lung cancers. Currently, NSCLC therapy is based on the analysis of specific predictors, whose presence qualifies patients for appropriate treatment. Baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5), also known as “survivin”, is a protein whose expression is characteristic for most malignant tumors and fetal tissue, while absent in mature cells. The biological role of BIRC5 is to counteract apoptosis by inhibiting the initiating and effector activities of caspases and binding to microtubules of the mitotic spindle. In our study, we looked for a relationship between BIRC5 gene polymorphism and the effectiveness of platinum-based chemotherapy. The study group consisted of 104 patients with newly diagnosed locally advanced or metastatic NSCLC. DNA was isolated from pretreatment blood samples, and SNPs of BIRC5 gene were analyzed. All patients received first-line platinum-based chemotherapy. Univariate analysis showed that a specific BIRC5 genotype was significantly associated with a higher risk of early progression (homozygous GG vs. heterozygous CG or CC: 28.9% vs. 11.9%). The presence of a homozygous GG genotype of the BIRC5 gene was insignificantly related to PFS shortening and TTP shortening. Moreover, significantly higher risk of overall survival shortening was associated with the BIRC5 homozygous GG genotype. Thus, studies on polymorphisms of selected genes affecting apoptosis may have a practical benefit for clinicians who monitor and treat NSCLC.
Collapse
|
11
|
Mitsuda J, Tsujikawa T, Yoshimura K, Saburi S, Suetsugu M, Kitamoto K, Takenaka M, Ohmura G, Arai A, Ogi H, Itoh K, Hirano S. A 14-Marker Multiplexed Imaging Panel for Prognostic Biomarkers and Tumor Heterogeneity in Head and Neck Squamous Cell Carcinoma. Front Oncol 2021; 11:713561. [PMID: 34490110 PMCID: PMC8417535 DOI: 10.3389/fonc.2021.713561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/15/2021] [Indexed: 01/10/2023] Open
Abstract
Recent advances made in treatment for head and neck squamous cell carcinoma (HNSCC) highlight the need for new prediction tools to guide therapeutic strategies. In this study, we aimed to develop a HNSCC-targeting multiplex immunohistochemical (IHC) panel that can evaluate prognostic factors and the intratumor heterogeneity of HNSCC. To identify IHC-based tissue biomarkers that constitute new multiplex IHC panel, a systematic review and meta-analysis were performed to analyze reported IHC biomarkers in laryngeal and pharyngeal SCC in the period of 2008–2018. The Cancer Genome Atlas (TCGA) and Reactome pathway databases were used to validate the prognostic and functional significance of the identified biomarkers. A 14-marker chromogenic multiplex IHC panel including identified biomarkers was used to analyze untreated HNSCC tissue. Forty-five high-quality studies and thirty-one candidate tissue biomarkers were identified (N = 7062). Prognostic validation in TCGA laryngeal and pharyngeal SCC cohort (N = 205) showed that β-catenin, DKK1, PINCH1, ADAM10, and TIMP1 were significantly associated with poor prognosis, which were related to functional categories such as immune system, cellular response, cell cycle, and developmental systems. Selected biomarkers were assembled to build a 14-marker panel, evaluating heterogeneity and polarized expression of tumor biomarkers in the tissue structures, which was particularly related to activation of Wnt/β-catenin pathway. Integrated IHC analysis based on a systemic review and meta-analysis provides an in situ proteomics tool to assess the aggressiveness and intratumor heterogeneity of HNSCC.
Collapse
Affiliation(s)
- Junichi Mitsuda
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiro Tsujikawa
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Cell, Developmental & Cancer Biology, Oregon Health and Science University, Portland, OR, United States
| | - Kanako Yoshimura
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Sumiyo Saburi
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masaho Suetsugu
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kayo Kitamoto
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Mari Takenaka
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Gaku Ohmura
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akihito Arai
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroshi Ogi
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.,SCREEN Holdings Co., Ltd., Kyoto, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Hirano
- Department of Otolaryngology-Head and Neck Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
12
|
McGuigan AJ, Coleman HG, McCain RS, Kelly PJ, Johnston DI, Taylor MA, Turkington RC. Immune cell infiltrates as prognostic biomarkers in pancreatic ductal adenocarcinoma: a systematic review and meta-analysis. J Pathol Clin Res 2021; 7:99-112. [PMID: 33481339 PMCID: PMC7869931 DOI: 10.1002/cjp2.192] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/15/2020] [Accepted: 11/16/2020] [Indexed: 12/24/2022]
Abstract
Immune cell infiltration has been identified as a prognostic biomarker in several cancers. However, no immune based biomarker has yet been validated for use in pancreatic ductal adenocarcinoma (PDAC). We undertook a systematic review and meta-analysis of immune cell infiltration, measured by immunohistochemistry (IHC), as a prognostic biomarker in PDAC. All other IHC prognostic biomarkers in PDAC were also summarised. MEDLINE, EMBASE and Web of Science were searched between 1998 and 2018. Studies investigating IHC biomarkers and prognosis in PDAC were included. REMARK score and Newcastle-Ottawa scale were used for qualitative analysis. Random-effects meta-analyses were used to pool results, where possible. Twenty-six articles studied immune cell infiltration IHC biomarkers and PDAC prognosis. Meta-analysis found high infiltration with CD4 (hazard ratio [HR] = 0.65, 95% confidence interval [CI] = 0.51-0.83.) and CD8 (HR = 0.68, 95% CI = 0.55-0.84.) T-lymphocytes associated with better disease-free survival. Reduced overall survival was associated with high CD163 (HR = 1.62, 95% CI = 1.03-2.56). Infiltration of CD3, CD20, FoxP3 and CD68 cells, and PD-L1 expression was not prognostic. In total, 708 prognostic biomarkers were identified in 1101 studies. In summary, high CD4 and CD8 infiltration are associated with better disease-free survival in PDAC. Increased CD163 is adversely prognostic. Despite the publication of 708 IHC prognostic biomarkers in PDAC, none has been validated for clinical use. Further research should focus on reproducibility of prognostic biomarkers in PDAC in order to achieve this.
Collapse
MESH Headings
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- B7-H1 Antigen/genetics
- B7-H1 Antigen/metabolism
- Biomarkers/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/pathology
- Disease-Free Survival
- Humans
- Immunohistochemistry
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/pathology
- Prognosis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Reproducibility of Results
- CD163 Antigen
Collapse
Affiliation(s)
- Andrew J McGuigan
- The Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastBelfastUK
| | - Helen G Coleman
- The Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastBelfastUK
- Centre for Public HealthQueen's University BelfastBelfastUK
| | - R Stephen McCain
- Centre for Public HealthQueen's University BelfastBelfastUK
- Department of Hepatobiliary SurgeryMater Hospital, Belfast Health and Social Care TrustBelfastUK
| | - Paul J Kelly
- Department of Tissue PathologyRoyal Victoria Hospital, Belfast Health and Social Care TrustBelfastUK
| | - David I Johnston
- Northern Ireland Cancer CentreBelfast Health and Social Care TrustBelfastUK
| | - Mark A Taylor
- Department of Hepatobiliary SurgeryMater Hospital, Belfast Health and Social Care TrustBelfastUK
| | - Richard C Turkington
- The Patrick G Johnston Centre for Cancer ResearchQueen's University BelfastBelfastUK
| |
Collapse
|
13
|
Son M, Kim H, Han D, Kim Y, Huh I, Han Y, Hong SM, Kwon W, Kim H, Jang JY, Kim Y. A Clinically Applicable 24-Protein Model for Classifying Risk Subgroups in Pancreatic Ductal Adenocarcinomas using Multiple Reaction Monitoring-Mass Spectrometry. Clin Cancer Res 2021; 27:3370-3382. [PMID: 33593883 DOI: 10.1158/1078-0432.ccr-20-3513] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 01/12/2021] [Accepted: 02/12/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) subtypes have been identified using various methodologies. However, it is a challenge to develop classification system applicable to routine clinical evaluation. We aimed to identify risk subgroups based on molecular features and develop a classification model that was more suited for clinical applications. EXPERIMENTAL DESIGN We collected whole dissected specimens from 225 patients who underwent surgery at Seoul National University Hospital [Seoul, Republic of Korea (South)], between October 2009 and February 2018. Target proteins with potential relevance to tumor progression or prognosis were quantified with robust quality controls. We used hierarchical clustering analysis to identify risk subgroups. A random forest classification model was developed to predict the identified risk subgroups, and the model was validated using transcriptomic datasets from external cohorts (N = 700), with survival analysis. RESULTS We identified 24 protein features that could classify the four risk subgroups associated with patient outcomes: stable, exocrine-like; activated, and extracellular matrix (ECM) remodeling. The "stable" risk subgroup was characterized by proteins that were associated with differentiation and tumor suppressors. "Exocrine-like" tumors highly expressed pancreatic enzymes. Two high-risk subgroups, "activated" and "ECM remodeling," were enriched in terms such as cell cycle, angiogenesis, immunocompetence, tumor invasion metastasis, and metabolic reprogramming. The classification model that included these features made prognoses with relative accuracy and precision in multiple cohorts. CONCLUSIONS We proposed PDAC risk subgroups and developed a classification model that may potentially be useful for routine clinical implementations, at the individual level. This clinical system may improve the accuracy of risk prediction and treatment guidelines.See related commentary by Thakur and Singh, p. 3272.
Collapse
Affiliation(s)
- Minsoo Son
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Hongbeom Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Dohyun Han
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea (South)
| | - Yoseop Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Iksoo Huh
- College of Nursing and Research Institute of Nursing Science, Seoul National University, Seoul, Republic of Korea (South)
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Seung-Mo Hong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea (South)
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Haeryoung Kim
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea (South)
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea (South).
| | - Youngsoo Kim
- Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, Republic of Korea (South).
| |
Collapse
|
14
|
Yusufu A, Tuerdi R, Redati D, Rehemutula A, Zhao ZL, Wang HJ. Expression and clinical correlation of Survivin and PTEN in gastric cancer patients. Oncol Lett 2020; 20:297. [PMID: 33101491 PMCID: PMC7576991 DOI: 10.3892/ol.2020.12160] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 02/04/2020] [Indexed: 12/24/2022] Open
Abstract
Reports on the correlation between the expression of Survivin/phosphatase and tensin homolog (PTEN) proteins and clinical factors in gastric cancer (GC) are varied, and the sample sizes were also not sufficient. The present study aimed to detect the expression of Survivin and PTEN proteins in GC patients on the basis of a greater number of specimens and to analyze the correlation with clinical features and survival. The results revealed that the Survivin expression rates in GC, normal tissues and metastatic lymph nodes were 72% (232/322), 5% (6/120) and 80% (36/45), respectively, while the PTEN expression rates were 34% (109/322), 92.5% (111/120) and 24.4% (11/45), respectively, and the differences between cancer and normal tissue or metastatic lymph nodes were significant for both proteins (P<0.05). The expression of Survivin was significantly associated with gross type, depth of invasion, distant metastasis, tumor, necrosis and metastasis (TNM) stage and vascular invasion, while PTEN expression was predominantly associated with age, tumor size, invasion depth, TNM stage and lymphatic invasion in GC patients (P<0.05). The expression of both was associated with postoperative metastasis and metastatic site (P=0.007 and P=0.011 for Survivin, and P=0.002 and P=0.005 for PTEN). There was a negative association between the expression levels of Survivin and PTEN (P=0.001, r=-0.524). The expression levels of both were also associated with prognosis. The expression of Survivin and PTEN protein exhibit opposing trends in GC, which may indicate adverse biological effects in the occurrence of GC. The Survivin and PTEN expression levels are likely to be an important molecular event in gastric tumorigenesis and may be considered as molecular markers of GC progression and reliable prognostic indicators of GC.
Collapse
Affiliation(s)
- Aikeremu Yusufu
- Department of Gastrointestinal Surgery, Tumor Hospital Affiliated to Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Rousidan Tuerdi
- Department of Gastrointestinal Surgery, Tumor Hospital Affiliated to Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Darebai Redati
- Department of Gastrointestinal Surgery, Tumor Hospital Affiliated to Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Aizimaiti Rehemutula
- Department of Gastrointestinal Surgery, Tumor Hospital Affiliated to Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Ze-Liang Zhao
- Department of Gastrointestinal Surgery, Tumor Hospital Affiliated to Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| | - Hai-Jiang Wang
- Department of Gastrointestinal Surgery, Tumor Hospital Affiliated to Xin Jiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830011, P.R. China
| |
Collapse
|
15
|
Wang Y, Zhong X, Zhou L, Lu J, Jiang B, Liu C, Guo J. Prognostic Biomarkers for Pancreatic Ductal Adenocarcinoma: An Umbrella Review. Front Oncol 2020; 10:1466. [PMID: 33042793 PMCID: PMC7527774 DOI: 10.3389/fonc.2020.01466] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 07/09/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) leads to the majority of cancer-related deaths due to its morbidity with similar mortality. Lack of effective prognostic biomarkers are the main reason for belated post-operative intervention of recurrence which causes high mortality. Numerous systematic reviews and meta-analyses have explored the prognostic value of biomarkers in PDAC so far. In this article, we performed an umbrella review analyzing these studies to provide an overview of associations between prognostic biomarkers and PDAC survival outcome and synthesized these results to guide better clinical practice. Methods: Systematic reviews and meta-analyses investigating the associations between PDAC survival outcomes and prognostic biomarkers were acquired via the PubMed and Embase databases from inception till February 1, 2020. Associations supported by nominally statistically significant results were classified into strong, highly suggestive, suggestive, and weak based on several critical factors such as the statistical significance of summary estimates, the number of events, the estimate of the largest study included, interstudy heterogeneity, small-study effects, 95% predictive interval (PI), excess significance bias, and the results of credibility ceiling sensitivity analyses. Results: We included 41 meta-analyses containing 63 associations between PDAC survival outcomes and prognostic biomarkers. Although, none was supported by strong evidence among these associations, an association between C-reactive protein to albumin ratio (CAR) and PDAC overall survival (OS) and an association between neutrophil-lymphocyte ratio (NLR) and PDAC OS were supported by highly suggestive evidence. Otherwise, the association between lactate dehydrogenase (LDH) and PDAC OS was supported by suggestive evidence. The remaining 60 associations were supported by weak or not suggestive evidence. Conclusion: Associations between CAR or NLR and PDAC OS were supported by highly suggestive evidence. And the association between LDH and PDAC OS was supported by suggestive evidence. Although the methodological quality of the included systematic reviews and meta-analyses which were evaluated by AMSTAR2.0 is generally poor, the identification of the relatively robust prognostic biomarkers of PDAC may guide better post-operative intervention and follow-up to prolong patients' survival.
Collapse
Affiliation(s)
- Yizhi Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Xi Zhong
- Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Department of Surgical Oncology and General Surgery, Ministry of Education, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jun Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Bolun Jiang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Chengxi Liu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Abstract
Objective: We aimed to define preoperative clinical and molecular characteristics that would allow better patient selection for operative resection. Background: Although we use molecular selection methods for systemic targeted therapies, these principles are not applied to surgical oncology. Improving patient selection is of vital importance for the operative treatment of pancreatic cancer (pancreatic ductal adenocarcinoma). Although surgery is the only chance of long-term survival, 80% still succumb to the disease and approximately 30% die within 1 year, often sooner than those that have unresected local disease. Method: In 3 independent pancreatic ductal adenocarcinoma cohorts (total participants = 1184) the relationship between aberrant expression of prometastatic proteins S100A2 and S100A4 and survival was assessed. A preoperative nomogram based on clinical variables available before surgery and expression of these proteins was constructed and compared to traditional measures, and a postoperative nomogram. Results: High expression of either S100A2 or S100A4 was independent poor prognostic factors in a training cohort of 518 participants. These results were validated in 2 independent patient cohorts (Glasgow, n = 198; Germany, n = 468). Aberrant biomarker expression stratified the cohorts into 3 distinct prognostic groups. A preoperative nomogram incorporating S100A2 and S100A4 expression predicted survival and nomograms derived using postoperative clinicopathological variables. Conclusions: Of those patients with a poor preoperative nomogram score, approximately 50% of patients died within a year of resection. Nomograms have the potential to improve selection for surgery and neoadjuvant therapy, avoiding surgery in aggressive disease, and justifying more extensive resections in biologically favorable disease.
Collapse
|
17
|
Jones MC, Zha J, Humphries MJ. Connections between the cell cycle, cell adhesion and the cytoskeleton. Philos Trans R Soc Lond B Biol Sci 2019; 374:20180227. [PMID: 31431178 PMCID: PMC6627016 DOI: 10.1098/rstb.2018.0227] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2018] [Indexed: 12/18/2022] Open
Abstract
Cell division, the purpose of which is to enable cell replication, and in particular to distribute complete, accurate copies of genetic material to daughter cells, is essential for the propagation of life. At a morphological level, division not only necessitates duplication of cellular structures, but it also relies on polar segregation of this material followed by physical scission of the parent cell. For these fundamental changes in cell shape and positioning to be achieved, mechanisms are required to link the cell cycle to the modulation of cytoarchitecture. Outside of mitosis, the three main cytoskeletal networks not only endow cells with a physical cytoplasmic skeleton, but they also provide a mechanism for spatio-temporal sensing via integrin-associated adhesion complexes and site-directed delivery of cargoes. During mitosis, some interphase functions are retained, but the architecture of the cytoskeleton changes dramatically, and there is a need to generate a mitotic spindle for chromosome segregation. An economical solution is to re-use existing cytoskeletal molecules: transcellular actin stress fibres remodel to create a rigid cortex and a cytokinetic furrow, while unipolar radial microtubules become the primary components of the bipolar spindle. This remodelling implies the existence of specific mechanisms that link the cell-cycle machinery to the control of adhesion and the cytoskeleton. In this article, we review the intimate three-way connection between microenvironmental sensing, adhesion signalling and cell proliferation, particularly in the contexts of normal growth control and aberrant tumour progression. As the morphological changes that occur during mitosis are ancient, the mechanisms linking the cell cycle to the cytoskeleton/adhesion signalling network are likely to be primordial in nature and we discuss recent advances that have elucidated elements of this link. A particular focus is the connection between CDK1 and cell adhesion. This article is part of a discussion meeting issue 'Forces in cancer: interdisciplinary approaches in tumour mechanobiology'.
Collapse
Affiliation(s)
| | | | - Martin J. Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
18
|
Gao X, Zhang L, Wei Y, Yang Y, Li J, Wu H, Yin Y. Prognostic Value of XIAP Level in Patients with Various Cancers: A Systematic Review and Meta-Analysis. J Cancer 2019; 10:1528-1537. [PMID: 31031863 PMCID: PMC6485232 DOI: 10.7150/jca.28229] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 12/15/2018] [Indexed: 02/06/2023] Open
Abstract
Background: X-linked inhibitor of apoptosis protein (XIAP) plays an important role in cancer pathogenesis, which has been found to be overexpressed in multiple human cancers and associated with survival rates. Herein, we performed a meta-analysis to explore the predictive value of XIAP level in patients with various solid tumors. Methods: Relevant articles exploring the relationship between XIAP expression and survival of cancer patients were retrieved in PubMed, PMC, EMBASE and Web of Science published from 2001 to 2018. The combined hazard ratios (HRs) and 95% confidence intervals (CIs) were calculated to evaluate the significance. Results: A total of 6554 patients from 40 articles were included in this meta-analysis. It was shown in 37 studies with 4864 cases that the over-expression of XIAP was associated with poorer overall survival (OS) (combined HR=1.61, 95% CI: 1.33-1.96). Meanwhile, 8 studies with 1862 cases revealed that elevated XIAP level predicted shorter disease-free survival (DFS) (HR=2.17, 95% CI: 1.03-4.59). Subgroup analyses showed that higher XIAP detection was related to worse OS in gastric cancer (HR=1.42, 95% CI: 1.18-1.72) and head and neck cancer (HNC) (HR=2.97, 95% CI: 1.97-4.47). Conclusion: Our results suggested that elevated XIAP level seemed to represent an unfavorable prognostic factor for clinical outcomes in cancer patients. However, there were limited studies describing the association between XIAP expression and clinical prognosis in each different type of tumors. Therefore, concrete roles of XIAP in various cancers need to be further explored.
Collapse
Affiliation(s)
- Xian Gao
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lei Zhang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Yong Wei
- Department of Urology, Nanjing Gaochun People's Hospital, Nanjing, 211300, China
| | - Yiqi Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Hao Wu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
19
|
Ntellas P, Dadouli K, Perivoliotis K, Sogka E, Pentheroudakis G, Ioannou M, Hadjichristodoulou C, Tepetes K, Mauri D. Microvessel Density and Impact of Angiogenesis on Survival of Resected Pancreatic Cancer Patients: A Systematic Review and Meta-analysis. Pancreas 2019; 48:233-241. [PMID: 30629030 DOI: 10.1097/mpa.0000000000001237] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVES Angiogenesis plays a major role in tumor progression and metastasis; however, its role in pancreatic cancer (PC) remains unclear. The aim of the study was to explore the cumulative evidence concerning the impact of microvessel density (MVD), an estimator of angiogenesis, on resected PC patients. METHODS A systematic review of literature and a meta-analysis of relevant reports were performed. Overall survival and disease-free survival were scrutinized. RESULTS One thousand five hundred patients were analyzed. Overall survival (hazard ratio, 2.0; 95% confidence interval, 1.57-2.54; P < 0.001) and disease-free survival (hazard ratio, 1.99; 95% confidence interval, 1.24-3.2; P = 0.004) were in favor of the low-MVD group. Use of CD105 antibody and of a computerized image analysis system was found to significantly reduce the heterogeneity. Disease staging, tumor location, and grading showed significant effect on survival. CONCLUSIONS High-MVD expression was strongly associated with poorer survival and recurrence among resected PC patients, demonstrating a negative prognostic value. Use of CD105 antibody and of a computerized image analysis system is recommended in future studies because they reduce heterogeneity of results. The potential role of MVD as a marker to select PC patients who would benefit from antiangiogenetic treatment should be further explored in clinical trials.
Collapse
Affiliation(s)
| | - Katerina Dadouli
- Department of Hygiene and Epidemiology, Faculty of Medicine, University of Thessaly, Larissa
| | | | - Eleni Sogka
- Medical Oncology, University Hospital of Larissa, Larissa
| | | | - Maria Ioannou
- Department of Pathology, Faculty of Medicine, University of Thessaly, Larissa, Greece
| | | | | | | |
Collapse
|
20
|
Zhou L, Lu J, Liang ZY, Zhou WX, Yuan D, Li BQ, You L, Guo JC, Zhao YP. High nuclear Survivin expression as a poor prognostic marker in pancreatic ductal adenocarcinoma. J Surg Oncol 2018; 118:1115-1121. [PMID: 30261114 DOI: 10.1002/jso.25253] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 09/04/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Survivin, one of the key regulators of mitosis and apoptosis, has long been well recognized to play important biological roles in many neoplasms, including pancreatic ductal adenocarcinoma (PDAC). However, its prognostic value in PDAC remains controversial. PATIENTS AND METHODS Nuclear expression of Survivin was detected, using tissue microarray-based immunohistochemistry, in paired-tumor and nontumor samples from 306 patients with radically resected PDAC. The staining H scores were further correlated with clinicopathologic features and disease-specific survival (DSS). RESULTS Nuclear Survivin expression was much higher in tumor than in nontumor tissues (P < 0.001). No significant association between tumoral Survivin expression and clinicopathologic variables was found. For prognosis, high Survivin expression was associated with shortened DSS in all eligible patients and four subgroups, that is, male and nondiabetic patients as well as those with head-located and G1-2 tumors, shown by univariate analyses. In addition, a statistically marginal significance was revealed in eight subgroups. For the entire cohort and two subgroups, nuclear Survivin expression was also multivariate identified as an independent predictor for DSS. For patients with G1-2 tumors, it was the single prognostic marker. CONCLUSION Our data suggest an association between high nuclear Survivin expression and poor prognosis in PDAC. However, further confirmation might be necessary.
Collapse
Affiliation(s)
- Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jun Lu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Zhi-Yong Liang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Wei-Xun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Da Yuan
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Bing-Qi Li
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Jun-Chao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| | - Yu-Pei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China
| |
Collapse
|
21
|
Saukkonen K, Hagström J, Mustonen H, Lehtinen L, Carpen O, Andersson LC, Seppänen H, Haglund C. Prognostic and diagnostic value of REG4 serum and tissue expression in pancreatic ductal adenocarcinoma. Tumour Biol 2018. [PMID: 29542402 DOI: 10.1177/1010428318761494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Expression of regenerating islet-derived protein 4 (REG4), a secretory protein involved in cell differentiation and proliferation, is upregulated in inflammatory bowel diseases and in many gastrointestinal malignancies. The prognostic significance of its expression in pancreatic ductal adenocarcinoma is unknown. Our aim was to investigate tumor tissue and serum REG4 expression in pancreatic ductal adenocarcinoma patients. We also evaluated as a control the diagnostic value of serum REG4 level in patients with chronic pancreatitis. Immunohistochemical expression of REG4 was evaluated in 154 surgical specimens and serum REG4 level in 130 samples from pancreatic ductal adenocarcinoma patients treated at Helsinki University Hospital, Finland, in 2000-2011. REG4 tissue and serum expression was assessed in relation to clinicopathological parameters and patient survival. A chronic pancreatitis control group comprised 34 patients who underwent pancreatic resection because of suspicion of malignancy. Significant survival differences were detectable in subgroups: in tumor stages IA-IIA, high serum REG4 level predicted worse survival (p=0.046). In patients with grade I tumor, positive tissue REG4 expression predicted better survival (p=0.006). In multivariate analysis, neither tissue nor serum REG4 expression was independent prognostic factors. Serum REG4 levels were higher in pancreatic ductal adenocarcinoma than in chronic pancreatitis (p=0.002), with diagnostic sensitivity of 45% and specificity of 91%. In logistic regression analysis, a multivariate model with REG4, CA19-9, and age provided sensitivity of 82% and specificity of 79%. REG4 tissue expression is a prognostic marker in subgroups of pancreatic ductal adenocarcinoma patients. Serum REG4 level might be useful in differential diagnosis between pancreatic ductal adenocarcinoma and chronic pancreatitis.
Collapse
Affiliation(s)
- Kapo Saukkonen
- 1 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,2 Translational Cancer Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Jaana Hagström
- 2 Translational Cancer Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland.,3 Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harri Mustonen
- 1 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Laura Lehtinen
- 4 Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Olli Carpen
- 3 Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,4 Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland.,5 Genome Scale Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Leif C Andersson
- 3 Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hanna Seppänen
- 1 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- 1 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,2 Translational Cancer Biology Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| |
Collapse
|
22
|
Tang Y, Shao A, Cao J, Li H, Li Q, Zeng M, Liu M, Cheng Y, Zhu W. cNGR-based synergistic-targeted NIR fluorescent probe for tracing and bioimaging of pancreatic ductal adenocarcinoma. Sci China Chem 2018; 61:184-191. [DOI: 10.1007/s11426-017-9092-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
23
|
Arpalahti L, Saukkonen K, Hagström J, Mustonen H, Seppänen H, Haglund C, Holmberg CI. Nuclear ubiquitin C-terminal hydrolase L5 expression associates with increased patient survival in pancreatic ductal adenocarcinoma. Tumour Biol 2017; 39:1010428317710411. [PMID: 28653876 DOI: 10.1177/1010428317710411] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a lethal disease with an overall 5-year survival of less than 5%. Prognosis among surgically treated patients is difficult and identification of new biomarkers is essential for accurate prediction of patient outcome. As part of one of the major cellular protein degradation systems, the proteasome plays a fundamental role in both physiological and pathophysiological conditions including cancer. The proteasome-associated deubiquitinating enzyme ubiquitin C-terminal hydrolase L5 (UCHL5)/Uch37 is a modulator of proteasome activity with cancer prognostic marker potential. Cytoplasmic and nuclear immunoexpression of UCHL5 was evaluated in 154 surgical specimens from pancreatic ductal adenocarcinoma patients treated at Helsinki University Hospital, Finland, in 2000-2011. UCHL5 expression in relation to clinicopathological parameters and the association between UCHL5 In this study, positive expression and patient survival were assessed. Positive nuclear UCHL5 expression was associated with increased patient survival ( p = 0.005). A survival benefit was also detectable in these subgroups of patients: over 65 years ( p < 0.001), at tumor stages IIB to III ( p = 0.007), or with lymph-node positivity ( p = 0.006). In stages IIB to III disease, patients with positive nuclear UCHL5 expression showed a twofold increase in 5-year cancer-specific survival compared to those with negative expression. Multivariate analysis identified positive nuclear UCHL5 expression as an independent prognostic factor ( p = 0.012). In conclusion, UCHL5 expression could function as a prognostic marker in pancreatic ductal adenocarcinoma, particularly at disease stages IIB to III. As UCHL5 is one of the few markers predicting increased survival, our results may be of clinical relevance.
Collapse
Affiliation(s)
- Leena Arpalahti
- 1 Research Programs Unit, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - Kapo Saukkonen
- 1 Research Programs Unit, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland.,2 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Jaana Hagström
- 1 Research Programs Unit, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland.,3 Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Harri Mustonen
- 2 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Hanna Seppänen
- 2 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Caj Haglund
- 1 Research Programs Unit, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland.,2 Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Carina I Holmberg
- 1 Research Programs Unit, Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| |
Collapse
|
24
|
Berlin A, Castro-Mesta JF, Rodriguez-Romo L, Hernandez-Barajas D, González-Guerrero JF, Rodríguez-Fernández IA, González-Conchas G, Verdines-Perez A, Vera-Badillo FE. Prognostic role of Ki-67 score in localized prostate cancer: A systematic review and meta-analysis. Urol Oncol 2017. [PMID: 28648414 DOI: 10.1016/j.urolonc.2017.05.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Ki-67 for quantifying tumor proliferation is widely used. In localized prostate cancer (PCa), despite a suggested predictive role of Ki-67 for outcomes after therapies, it has not been incorporated into clinical practice. Herein, we conduct a systematic review and meta-analysis of the literature reporting the association of Ki-67 and disease outcomes in PCa treated radically. METHODS Medline and EMBASE databases were searched without date or language restrictions, using "KI67" and "prostate cancer" MeSH terms. Studies reporting Ki-67 association with clinical outcomes (disease-free survival [DFS], biochemical failure-free survival, rate of distant metastases [DM], disease-specific survival [DSS], or overall survival [OS], or all of these) in patients with PCa managed actively were included, and relevant data extracted by 2 independent reviewers. Odds ratios (OR) were weighted and pooled in a meta-analysis using Mantel-Haenszel random-effect modeling. RESULTS Twenty-one studies comprising 5,419 patients met eligibility for analysis, and 67.6% of patients had low Ki-67. Mean Ki-67 was 6.14%. High Ki-67 was strongly associated with worse clinical outcomes. DFS was better in those patients with low Ki-67 at 5 and 10 years (OR = 0.32, 95% CI: 0.23-0.44, P<0.00001; OR = 0.31, 95% CI: 0.20-0.48, P<0.00001). Similarly, low Ki-67 was related to improved DSS at 5 and 10 years (OR = 0.15, 95% CI: 0.10-0.21, P<0.00001; OR = 0.16, 95% CI: 0.06-0.40, P<0.00001). Association between low Ki-67 scores with improved OS (OR = 0.47; 95% CI: 0.37-0.61; P<0.00001) and high Ki-67 scores with DM at 5 years (OR = 4.07; 95% CI: 2.52-6.58; P<0.00001) was consistently observed. CONCLUSIONS High Ki-67 expression in localized PCa is a factor of poor prognosis for DSS, biochemical failure-free survival, DFS, DM, and OS after curative-intent treatments. Incorporation into clinical routine of this widely available and standardized biomarker should be strongly considered.
Collapse
Affiliation(s)
- Alejandro Berlin
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Julio F Castro-Mesta
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Laura Rodriguez-Romo
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - David Hernandez-Barajas
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Juan F González-Guerrero
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Iván A Rodríguez-Fernández
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Galileo González-Conchas
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Adrian Verdines-Perez
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México
| | - Francisco E Vera-Badillo
- Centro Universitario Contra el Cáncer, Hospital Universitario, Universidad Autónoma de Nuevo León, Monterrey, México; Department of Medical Oncology, Faculty of Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
25
|
Massihnia D, Avan A, Funel N, Maftouh M, van Krieken A, Granchi C, Raktoe R, Boggi U, Aicher B, Minutolo F, Russo A, Leon LG, Peters GJ, Giovannetti E. Phospho-Akt overexpression is prognostic and can be used to tailor the synergistic interaction of Akt inhibitors with gemcitabine in pancreatic cancer. J Hematol Oncol 2017; 10:9. [PMID: 28061880 PMCID: PMC5219723 DOI: 10.1186/s13045-016-0371-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Accepted: 12/08/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND There is increasing evidence of a constitutive activation of Akt in pancreatic ductal adenocarcinoma (PDAC), associated with poor prognosis and chemoresistance. Therefore, we evaluated the expression of phospho-Akt in PDAC tissues and cells, and investigated molecular mechanisms influencing the therapeutic potential of Akt inhibition in combination with gemcitabine. METHODS Phospho-Akt expression was evaluated by immunohistochemistry in tissue microarrays (TMAs) with specimens tissue from radically-resected patients (n = 100). Data were analyzed by Fisher and log-rank test. In vitro studies were performed in 14 PDAC cells, including seven primary cultures, characterized for their Akt1 mRNA and phospho-Akt/Akt levels by quantitative-RT-PCR and immunocytochemistry. Growth inhibitory effects of Akt inhibitors and gemcitabine were evaluated by SRB assay, whereas modulation of Akt and phospho-Akt was investigated by Western blotting and ELISA. Cell cycle perturbation, apoptosis-induction, and anti-migratory behaviors were studied by flow cytometry, AnnexinV, membrane potential, and migration assay, while pharmacological interaction with gemcitabine was determined with combination index (CI) method. RESULTS Immunohistochemistry of TMAs revealed a correlation between phospho-Akt expression and worse outcome, particularly in patients with the highest phospho-Akt levels, who had significantly shorter overall and progression-free-survival. Similar expression levels were detected in LPC028 primary cells, while LPC006 were characterized by low phospho-Akt. Remarkably, Akt inhibitors reduced cancer cell growth in monolayers and spheroids and synergistically enhanced the antiproliferative activity of gemcitabine in LPC028, while this combination was antagonistic in LPC006 cells. The synergistic effect was paralleled by a reduced expression of ribonucleotide reductase, potentially facilitating gemcitabine cytotoxicity. Inhibition of Akt decreased cell migration and invasion, which was additionally reduced by the combination with gemcitabine. This combination significantly increased apoptosis, associated with induction of caspase-3/6/8/9, PARP and BAD, and inhibition of Bcl-2 and NF-kB in LPC028, but not in LPC006 cells. However, targeting the key glucose transporter Glut1 resulted in similar apoptosis induction in LPC006 cells. CONCLUSIONS These data support the analysis of phospho-Akt expression as both a prognostic and a predictive biomarker, for the rational development of new combination therapies targeting the Akt pathway in PDAC. Finally, inhibition of Glut1 might overcome resistance to these therapies and warrants further studies.
Collapse
Affiliation(s)
- Daniela Massihnia
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Amir Avan
- Metabolic syndrome Research center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Niccola Funel
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy
| | - Mina Maftouh
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Anne van Krieken
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | - Rajiv Raktoe
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Ugo Boggi
- Department of Surgery, University of Pisa, Pisa, Italy
| | - Babette Aicher
- Æterna Zentaris GmbH, Frankfurt am Main, Frankfurt, Germany
| | | | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Leticia G Leon
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology VU University Medical Center, Cancer Center Amsterdam, CCA room 1.52, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
- Cancer Pharmacology Lab, AIRC Start Up Unit, University of Pisa, Pisa, Italy.
| |
Collapse
|
26
|
Glazer ES, Welsh E, Pimiento JM, Teer JK, Malafa MP. TGFβ1 overexpression is associated with improved survival and low tumor cell proliferation in patients with early-stage pancreatic ductal adenocarcinoma. Oncotarget 2017; 8:999-1006. [PMID: 27895310 PMCID: PMC5352213 DOI: 10.18632/oncotarget.13533] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 09/16/2016] [Indexed: 12/13/2022] Open
Abstract
The role of transforming growth factor beta-type-1 (TGFβ1) in pancreatic ductal adenocarcinoma (PDAC) progression is stage-dependent. We hypothesized that TGFβ1 expression is associated with survival and proliferation markers in patients with early-stage PDAC. We acquired clinicopathologic, treatment, and mRNA expression data from The Cancer Genome Atlas data set for 106 patients identified with stage I/II PDAC who underwent pancreaticoduodenectomy. Patients were categorized as high expression when mRNA expression was ≥75th percentile for each gene. Average log2 mRNA expression of TGFβ1 in patients with high expression was 11.6 ± 0.2 and 10.5 ± 0.6 in patients with low expression (P<0.001). Low TGFβ1 expression is associated with shorter median survival compared with high TGFβ1 expression (17 versus at least 60 months; P=0.005). Patients with tumors demonstrating high MKI67 (the gene encoding Ki-67) expression have shorter median survival versus those with lowerMKI67 expression (16 versus 20 months; P=0.026). TGFβ1 and MKI67 are inversely associated (P=0.009). On multivariate analysis, improved survival is associated with TGFβ1 overexpression (P=0.017), adjuvant chemotherapy (P=0.001), and adjuvant radiotherapy (P=0.017), whereas positive surgical margins are associated with worse survival (P=0.002). In patients who undergo pancreaticoduodenectomy for PDAC, high TGFβ1 expression may counteract the worse survival associated with high proliferation.
Collapse
Affiliation(s)
- Evan S. Glazer
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Surgery, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Eric Welsh
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jose M. Pimiento
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jamie K. Teer
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Mokenge P. Malafa
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
27
|
Loosen SH, Benz F, Niedeggen J, Schmeding M, Schüller F, Koch A, Vucur M, Tacke F, Trautwein C, Roderburg C, Neumann UP, Luedde T. Serum levels of S100A6 are unaltered in patients with resectable cholangiocarcinoma. Clin Transl Med 2016; 5:39. [PMID: 27709523 PMCID: PMC5052241 DOI: 10.1186/s40169-016-0120-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/10/2016] [Indexed: 02/12/2023] Open
Abstract
Background Elevated expression levels of S100A6, a calcium-binding low-molecular-weight protein, were demonstrated in various malignancies. Moreover, increased serum levels of S100A6 were suggested as a novel biomarker for various inflammatory and malignant diseases including lung and gastric cancer. However, up to now, serum concentrations of S100A6 have not been analyzed in patients with cholangiocarcinoma (CCA). Methods S100A6 mRNA expression levels were analyzed in human and murine CCA tumor samples, using semi-quantitative reverse transcriptase PCR. S100A6 serum concentrations were measured using an enzyme-linked immunosorbent assay in 112 patients with CCA referred to surgery for curative resection and were compared to those of 42 healthy controls. Results were correlated with clinical data. Results S100A6 mRNA expression levels were significantly up-regulated in tumor samples of CCA patients and in tumor tissue of a CCA mouse model. In contrast, serum levels of S100A6 were not significantly altered in patients with CCA compared to healthy controls. Whereas no differences became apparent within the different clinical subgroups of CCA, patients with primary sclerosing cholangitis (PSC)-based CCA displayed higher levels of S100A6 compared to the other patients. Nevertheless, patients with higher S100A6 serum concentrations showed a trend towards an impaired prognosis compared to patients with lower levels. Finally, within our cohort of patients both the diagnostic and prognostic potentials of S100A6 were similar to those of the clinically established biomarkers CEA and CA19-9. Conclusion Although S100A6 was expressed at significantly higher levels in human and murine CCA tumor samples, S100A6 serum levels were not regulated in patients with CCA and are thus not suitable for diagnosis of CCA. However, CCA-patients with elevated S100A6 displayed a trend toward an impaired prognosis compared to patients with lower S100A6 levels, supporting its further evaluation as a prognostic biomarker in CCA. Electronic supplementary material The online version of this article (doi:10.1186/s40169-016-0120-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sven H Loosen
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Fabian Benz
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Jennifer Niedeggen
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Maximilian Schmeding
- Department of Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Florian Schüller
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Alexander Koch
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Mihael Vucur
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Christoph Roderburg
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ulf P Neumann
- Department of Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Tom Luedde
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany. .,Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
28
|
Zhang J, Miao C, Xu A, Zhao K, Qin Z, Li X, Liang C, Hua Y, Chen W, Zhang C, Liu Y, Su S, Wang Z, Liu B. Prognostic Role of Secretory Clusterin in Multiple Human Malignant Neoplasms: A Meta-Analysis of 26 Immunohistochemistry Studies. PLoS One 2016; 11:e0161150. [PMID: 27532124 PMCID: PMC4988765 DOI: 10.1371/journal.pone.0161150] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 08/01/2016] [Indexed: 01/11/2023] Open
Abstract
Secretory clusterin (sCLU) is a potential prognostic tumour biomarker, but results of different sCLU studies are inconsistent. We conducted this meta-analysis to evaluate the precise predictive value of sCLU. Qualified studies were identified by performing online searches in PubMed, EMBASE, and Web of Science. The selected articles were divided into three groups based on scoring method for clusterin detection. Pooled hazard ratios (HRs) with 95% confidence interval (CI) for patient survival and disease recurrence were calculated to determine the correlation between sCLU expression and cancer prognosis. Heterogeneity was assessed using I2 statistics, and specific heterogeneity in different groups was analysed. Elevated sCLU was significantly associated with recurrence-free survival in groups 1 and 3 (group 1: pooled HR = 1.35, 95% CI = 1.01 to 1.79; group 3: pooled HR = 1.80, 95% CI = 1.22 to 2.65). However, clusterin expression was not associated with overall survival in all three groups. Results showed that only the heterogeneity of group 2 was very strong (p = 0.013, I2 = 76.3%), in which the specimens were scored through sCLU staining intensity only. sCLU is a potential biomarker for tumour prognosis, and IHC methods can be more standardised if both intensity and staining proportion are considered.
Collapse
Affiliation(s)
- Jianzhong Zhang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Chenkui Miao
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Aiming Xu
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Kai Zhao
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Zhiqiang Qin
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Xiao Li
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Chao Liang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Yibo Hua
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Wei Chen
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Chao Zhang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Yiyang Liu
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Shifeng Su
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Zengjun Wang
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| | - Bianjiang Liu
- State Key Laboratory of Reproductive Medicine and Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing. 210029, China
| |
Collapse
|
29
|
Gu YL, Lan C, Pei H, Yang SN, Liu YF, Xiao LL. Applicative Value of Serum CA19-9, CEA, CA125 and CA242 in Diagnosis and Prognosis for Patients with Pancreatic Cancer Treated by Concurrent Chemoradiotherapy. Asian Pac J Cancer Prev 2016; 16:6569-73. [PMID: 26434876 DOI: 10.7314/apjcp.2015.16.15.6569] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE To evaluate the application value of serum CA19-9, CEA, CA125 and CA242 in diagnosis and prognosis of pancreatic cancer cases treated with concurrent chemotherapy. MATERIALS AND METHODS 52 patients with pancreatic cancer, 40 with benign pancreatic diseases and 40 healthy people were selected. The electrochemiluminescence immunoassay method was used for detecting levels of CA19-9, CEA and CA125, and a CanAg CA242 enzyme linked immunoassay kit for assessing the level of CA242. The Kaplan-Meier method was used for analyzing the prognostic factors of patients with pancreatic cancer. The Cox proportional hazard model was applied for analyzing the hazard ratio (HR) and 95% confidential interval (CI) for survival time of patients with pancreatic cancer. RESULTS The levels of serum CA19-9, CEA, CA125 and CA242 in patients with pancreatic cancer were significantly higher than those in patients with benign pancreatic diseases and healthy people (P<0.001). The sensitivity of CA19-9 was the highest among these, followed by CA242, CA125 and CEA. The specificity of CA242 is the highest, followed by CA125, CEA and CA19-9. The sensitivity and specificity of joint detection of serum CA19-9, CEA, CA125and CA242 were 90.4% and 93.8%, obviously higher than single detection of those markers in diagnosis of pancreatic cancer. The median survival time of 52 patients with pancreatic cancer was 10 months (95% CI7.389~12.611).. Patients with the increasing level of serum CA19-9, CEA, CA125, CA242 had shorter survival times (P=0.047. 0.043, 0.0041, 0.029). COX regression analysis showed that CA19-9 was an independent prognostic factor for patients with pancreatic cancer (P=0.001, 95%CI 2.591~38.243). CONCLUSIONS The detection of serum tumor markers (CA19.9, CEA, CA125 and CA242) is conducive to the early diagnosis of pancreatic cancer and joint detection of tumor markers helps improve the diagnostic efficiency. Moreover, CA19-9 is an independent prognostic factor for patients with pancreatic cancer.
Collapse
Affiliation(s)
- Yu-Lei Gu
- Emergency Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China E-mail :
| | | | | | | | | | | |
Collapse
|
30
|
Identification of key regulators of pancreatic cancer progression through multidimensional systems-level analysis. Genome Med 2016; 8:38. [PMID: 27137215 PMCID: PMC4853852 DOI: 10.1186/s13073-016-0282-3] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/19/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pancreatic cancer is an aggressive cancer with dismal prognosis, urgently necessitating better biomarkers to improve therapeutic options and early diagnosis. Traditional approaches of biomarker detection that consider only one aspect of the biological continuum like gene expression alone are limited in their scope and lack robustness in identifying the key regulators of the disease. We have adopted a multidimensional approach involving the cross-talk between the omics spaces to identify key regulators of disease progression. METHODS Multidimensional domain-specific disease signatures were obtained using rank-based meta-analysis of individual omics profiles (mRNA, miRNA, DNA methylation) related to pancreatic ductal adenocarcinoma (PDAC). These domain-specific PDAC signatures were integrated to identify genes that were affected across multiple dimensions of omics space in PDAC (genes under multiple regulatory controls, GMCs). To further pin down the regulators of PDAC pathophysiology, a systems-level network was generated from knowledge-based interaction information applied to the above identified GMCs. Key regulators were identified from the GMC network based on network statistics and their functional importance was validated using gene set enrichment analysis and survival analysis. RESULTS Rank-based meta-analysis identified 5391 genes, 109 miRNAs and 2081 methylation-sites significantly differentially expressed in PDAC (false discovery rate ≤ 0.05). Bimodal integration of meta-analysis signatures revealed 1150 and 715 genes regulated by miRNAs and methylation, respectively. Further analysis identified 189 altered genes that are commonly regulated by miRNA and methylation, hence considered GMCs. Systems-level analysis of the scale-free GMCs network identified eight potential key regulator hubs, namely E2F3, HMGA2, RASA1, IRS1, NUAK1, ACTN1, SKI and DLL1, associated with important pathways driving cancer progression. Survival analysis on individual key regulators revealed that higher expression of IRS1 and DLL1 and lower expression of HMGA2, ACTN1 and SKI were associated with better survival probabilities. CONCLUSIONS It is evident from the results that our hierarchical systems-level multidimensional analysis approach has been successful in isolating the converging regulatory modules and associated key regulatory molecules that are potential biomarkers for pancreatic cancer progression.
Collapse
|
31
|
Ren X, Wang J, Lin X, Wang X. E-cadherin expression and prognosis of head and neck squamous cell carcinoma: evidence from 19 published investigations. Onco Targets Ther 2016; 9:2447-53. [PMID: 27217768 PMCID: PMC4853145 DOI: 10.2147/ott.s98577] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Objective The objective of this study was to review the published literature and investigate whether E-cadherin gene is a prognostic factor in head and neck squamous cell carcinoma by conducting a meta-analysis. Methods Studies were identified from the databases Embase, Medline, and Cochrane Library by using the keywords “E-cadherin gene” and “head and neck cancer”. Overall survival (OS) and disease-free survival (DFS) were the primary outcome measurements. Results Our literature review identified 1,458 articles; 19 studies with a total number of 2,012 cases were eligible for inclusion in the meta-analysis. The hazard ratio (HR) for OS of patients with decreased expression of E-cadherin gene was 0.57 (95% CI =0.37, 0.89; P=0.000). However, statistical heterogeneity was unacceptably high (I2=74.5%, P=0.000). After sensitivity analysis, heterogeneity became acceptable, and the effect measure was still significant (I2=7.0%; HR =0.52; 95% CI =0.40, 0.66; P=0.000). The HR for DFS was 0.53 (95% CI =0.42, 0.67; P=0.000). Conclusion This meta-analysis showed clear evidence that high E-cadherin gene expression is a positive prognostic factor of head and neck squamous cell carcinoma, resulting in better OS and DFS. However, this conclusion must be interpreted with caution due to a few limitations.
Collapse
Affiliation(s)
- Xusheng Ren
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China; Department of Oral and Maxillofacial Surgery, Jinan Stomatological Hospital, Jinan, Shandong Province, People's Republic of China
| | - Jianning Wang
- Department of Oral and Maxillofacial Surgery, Jinan Stomatological Hospital, Jinan, Shandong Province, People's Republic of China
| | - Xuefen Lin
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China; Shandong Province Key Laboratory of Oral Tissue Regeneration, Stomatological Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| | - Xuxia Wang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China; Shandong Province Key Laboratory of Oral Tissue Regeneration, Stomatological Hospital of Shandong University, Jinan, Shandong Province, People's Republic of China
| |
Collapse
|
32
|
Conroy T, Bachet JB, Ayav A, Huguet F, Lambert A, Caramella C, Maréchal R, Van Laethem JL, Ducreux M. Current standards and new innovative approaches for treatment of pancreatic cancer. Eur J Cancer 2016; 57:10-22. [PMID: 26851397 DOI: 10.1016/j.ejca.2015.12.026] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 12/20/2015] [Accepted: 12/29/2015] [Indexed: 12/19/2022]
Abstract
Pancreatic adenocarcinoma remains a devastating disease with a 5-year survival rate not exceeding 6%. Treatment of this disease remains a major challenge. This article reviews the state-of-the-art in the management of this disease and the new innovative approaches that may help to accelerate progress in treating its victims. After careful pre-therapeutic evaluation, only 15-20% of patients diagnosed with a pancreatic cancer (PC) are eligible for upfront radical surgery. After R0 or R1 resection in such patients, evidence suggests a significantly positive impact on survival of adjuvant chemotherapy comprising 6 months of gemcitabine or fluorouracil/folinic acid. Delayed adjuvant chemoradiation is considered as an option in cases of positive margins. Borderline resectable pancreatic cancer (BRPC) is defined as a tumour involving the mesenteric vasculature to a limited extend. Resection of these tumours is technically feasible, yet runs the high risk of a R1 resection. Neoadjuvant treatment probably offers the best chance of achieving successful R0 resection and long-term survival, but the best treatment options should be determined in prospective randomised studies. Gemcitabine has for 15 years been the only validated therapy for advanced PC. Following decades of negative phase III studies, increasing evidence now suggests that further significant improvements to overall survival can be achieved via either Folfirinox or gemcitabine + nab-paclitaxel regimens. Progress in systemic therapy may improve the chances of resection in borderline resectable pancreatic cancer (BRPC) or locally advanced PC. This requires first enhancing knowledge of the genetic events driving carcinogenesis, which may then be translated into clinical studies.
Collapse
Affiliation(s)
- Thierry Conroy
- Department of Medical Oncology, Institut de Cancérologie de Lorraine and Lorraine University, 6 avenue de Bourgogne, CS 30519, 54519, Vandoeuvre-lès-Nancy, France.
| | - Jean-Baptiste Bachet
- Department of Hepato-Gastroenterology, Pitié-Salpétrière University Hospital, 47-83 boulevard de l'hôpital, 75651, Paris Cedex 13, France
| | - Ahmet Ayav
- Department of Surgery, Nancy University Hospital Lorraine and Lorraine University, rue du Morvan, 54511, Vandoeuvre-lès Nancy, France
| | - Florence Huguet
- Department of Radiation Therapy, Tenon Hospital, Paris Est University Hospitals, 4 rue de la Chine, 75020, Paris, France
| | - Aurélien Lambert
- Department of Medical Oncology, Institut de Cancérologie de Lorraine and Lorraine University, 6 avenue de Bourgogne, CS 30519, 54519, Vandoeuvre-lès-Nancy, France
| | - Caroline Caramella
- Gustave Roussy Cancer Campus Grand Paris, 114 rue Edouard-Vaillant, 94805, Villejuif Cedex, France
| | - Raphaël Maréchal
- Department of Gastroenterology, Erasme University Hospital-ULB-Brussels, Lennikstreet 808, 1070, Brussels, Belgium
| | - Jean-Luc Van Laethem
- Department of Gastroenterology, Erasme University Hospital-ULB-Brussels, Lennikstreet 808, 1070, Brussels, Belgium
| | - Michel Ducreux
- Gustave Roussy Cancer Campus Grand Paris, 114 rue Edouard-Vaillant, 94805, Villejuif Cedex, France
| |
Collapse
|
33
|
Masuda T, Ishikawa T, Mogushi K, Okazaki S, Ishiguro M, Iida S, Mizushima H, Tanaka H, Uetake H, Sugihara K. Overexpression of the S100A2 protein as a prognostic marker for patients with stage II and III colorectal cancer. Int J Oncol 2016; 48:975-82. [PMID: 26783118 PMCID: PMC4750537 DOI: 10.3892/ijo.2016.3329] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 12/17/2015] [Indexed: 11/25/2022] Open
Abstract
We aimed to identify a novel prognostic biomarker related to recurrence in stage II and III colorectal cancer (CRC) patients. Stage II and III CRC tissue mRNA expression was profiled using an Affymetrix Gene Chip, and copy number profiles of 125 patients were generated using an Affymetrix 250K Sty array. Genes showing both upregulated expression and copy number gains in cases involving recurrence were extracted as candidate biomarkers. The protein expression of the candidate gene was assessed using immunohistochemical staining of tissue from 161 patients. The relationship between protein expression and clinicopathological features was also examined. We identified 9 candidate genes related to recurrence of stage II and III CRC, whose mRNA expression was significantly higher in CRC than in normal tissue. Of these proteins, the S100 calcium-binding protein A2 (S100A2) has been observed in several human cancers. S100A2 protein overexpression in CRC cells was associated with significantly worse overall survival and relapse-free survival, indicating that S100A2 is an independent risk factor for stage II and III CRC recurrence. S100A2 overexpression in cancer cells could be a biomarker of poor prognosis in stage II and III CRC recurrence and a target for treatment of this disease.
Collapse
Affiliation(s)
- Taiki Masuda
- Department of Surgical Oncology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Toshiaki Ishikawa
- Department of Surgical Oncology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Kaoru Mogushi
- Department of Systems Biology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Satoshi Okazaki
- Department of Surgical Oncology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Megumi Ishiguro
- Department of Translational Oncology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Satoru Iida
- Department of Surgical Oncology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Hiroshi Mizushima
- Department of Medical Omics Informatics, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Hiroshi Tanaka
- Department of Systems Biology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Hiroyuki Uetake
- Department of Surgical Oncology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| | - Kenichi Sugihara
- Department of Surgical Oncology, Tokyo Medical and Dental University Graduate School of Medicine, Tokyo 113-8510, Japan
| |
Collapse
|
34
|
Pancreatic preneoplastic lesions plasma signatures and biomarkers based on proteome profiling of mouse models. Br J Cancer 2015; 113:1590-8. [PMID: 26512875 PMCID: PMC4705884 DOI: 10.1038/bjc.2015.370] [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: 03/27/2014] [Revised: 08/28/2015] [Accepted: 09/30/2015] [Indexed: 12/27/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with a mortality that is almost identical to incidence. Because early detected PDAC is potentially curable, blood-based biomarkers that could detect currently developing neoplasia would improve patient survival and management. PDAC develops from pancreatic intraepithelial neoplasia (PanIN) lesions, graded from low grade (PanIN1) to high grade (PanIN3). We made the hypothesis that specific proteomic signatures from each precancerous stage exist and are detectable in plasma. Methods: We explored the peptide profiles of microdissected PanIN cells and of plasma samples corresponding to the different PanIN grade from genetically engineered mouse models of PDAC using capillary electrophoresis coupled to mass spectrometry (CE-MS) and Chip-MS/MS. Results: We successfully characterised differential peptides profiles from PanIN microdissected cells. We found that plasma from tumor-bearing mice and age-matched controls exhibit discriminative peptide signatures. We also determined plasma peptide signatures corresponding to low- and high-grade precancerous step present in the mice pancreas using the two mass spectrometry technologies. Importantly, we identified biomarkers specific of PanIN3. Conclusions: We demonstrate that benign and advanced PanIN lesions display distinct plasma peptide patterns. This strongly supports the perspectives of developing a non-invasive screening test for prediction and early detection of PDAC.
Collapse
|
35
|
Delitto D, Black BS, Sorenson HL, Knowlton AE, Thomas RM, Sarosi GA, Moldawer LL, Behrns KE, Liu C, George TJ, Trevino JG, Wallet SM, Hughes SJ. The inflammatory milieu within the pancreatic cancer microenvironment correlates with clinicopathologic parameters, chemoresistance and survival. BMC Cancer 2015; 15:783. [PMID: 26498838 PMCID: PMC4619553 DOI: 10.1186/s12885-015-1820-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/16/2015] [Indexed: 01/05/2023] Open
Abstract
Background The tumor microenvironment impacts pancreatic cancer (PC) development, progression and metastasis. How intratumoral inflammatory mediators modulate this biology remains poorly understood. We hypothesized that the inflammatory milieu within the PC microenvironment would correlate with clinicopathologic findings and survival. Methods Pancreatic specimens from normal pancreas (n = 6), chronic pancreatitis (n = 9) and pancreatic adenocarcinoma (n = 36) were homogenized immediately upon resection. Homogenates were subjected to multiplex analysis of 41 inflammatory mediators. Results Twenty-three mediators were significantly elevated in adenocarcinoma specimens compared to nonmalignant controls. Increased intratumoral IL-8 concentrations associated with larger tumors (P = .045) and poor differentiation (P = .038); the administration of neoadjuvant chemotherapy associated with reduced IL-8 concentrations (P = .003). Neoadjuvant therapy was also associated with elevated concentrations of Flt-3 L (P = .005). Elevated levels of pro-inflammatory cytokines IL-1β (P = .017) and TNFα (P = .033) were associated with a poor histopathologic response to neoadjuvant therapy. Elevated concentrations of G-CSF (P = .016) and PDGF-AA (P = .012) correlated with reduced overall survival. Conversely, elevated concentrations of FGF-2 (P = .038), TNFα (P = .031) and MIP-1α (P = .036) were associated with prolonged survival. Conclusion The pancreatic cancer microenvironment harbors a unique inflammatory milieu with potential diagnostic and prognostic value.
Collapse
Affiliation(s)
- Daniel Delitto
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Brian S Black
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Heather L Sorenson
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Andrea E Knowlton
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Ryan M Thomas
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA. .,North Florida/South Georgia Veterans Health System, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| | - George A Sarosi
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA. .,North Florida/South Georgia Veterans Health System, Department of Surgery, University of Florida College of Medicine, Gainesville, FL, 32610, USA.
| | - Lyle L Moldawer
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Kevin E Behrns
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Chen Liu
- Department of Pathology, College of Medicine, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Thomas J George
- Department of Medicine, College of Medicine, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Jose G Trevino
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| | - Shannon M Wallet
- Department of Oral Biology, College of Dentistry, University of Florida Health Science Center, Gainesville, FL, 32610, USA.
| | - Steven J Hughes
- Department of Surgery, College of Medicine, University of Florida Health Science Center, Room 6116, Shands Hospital, 1600 SW Archer Rd, Gainesville, FL, 32610, USA.
| |
Collapse
|
36
|
Bachet JB, Chibaudel B, Bonnetain F, Validire P, Hammel P, André T, Louvet C. A randomized phase II study of weekly nab-paclitaxel plus gemcitabine or simplified LV5FU2 as first-line therapy in patients with metastatic pancreatic cancer: the AFUGEM GERCOR trial. BMC Cancer 2015; 15:653. [PMID: 26445094 PMCID: PMC4596389 DOI: 10.1186/s12885-015-1656-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Accepted: 09/21/2015] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Metastatic pancreatic adenocarcinoma (PAC) prognosis remains dismal and gemcitabine monotherapy has been the standard treatment over the last decade. Currently, two first-line regimens are used in this setting: FOLFIRINOX and nab-paclitaxel plus gemcitabine. Increasing translational data on the predictive value of hENT1 for determining gemcitabine efficacy suggest that a non-gemcitabine-based regimen is favored in about 60 % of patients with PAC due to high resistance of PAC to this cytotoxic drug. This study aims to evaluate the efficacy of weekly nab-paclitaxel combined with gemcitabine or a simplified (s) LV5FU2 regimen in patients with previously untreated metastatic PAC. METHODS/DESIGN AFUGEM is a two-stage, open-label, randomized, multicenter, phase II trial. Patients with PAC who meet the inclusion criteria and provide written informed consent will be randomized in a 1:2 ratio to either nab-paclitaxel (125 mg/m(2)) plus gemcitabine (1000 mg/m(2)) given on days 1, 8, and 15 every 28 days or nab-paclitaxel (125 mg/m(2)) plus sLV5FU2 (leucovorin 400 mg/m(2) followed by bolus 400 mg/m(2) 5-fluorouracil and by 5-fluorouracil 2400 mg/m(2) as an 46-h intravenous infusion) given on days 1 and 15 every 28 days. A total of 114 patients will be randomized to one of the treatment arms. The primary endpoint is progression-free survival at 4 months. Secondary outcomes are rate and duration of response, disease control, overall survival, safety, and quality of life. Potential biomarkers of gemcitabine (hENT1, dCK) and 5-fluorouracil (TS) efficacy will be assessed. DISCUSSION The AFUGEM trial is designed to provide valuable information regarding efficacy and tolerability of nab-paclitaxel plus gemcitabine and nab-paclitaxel plus sLV5FU2 regimens. Identification of potential predictive biomarkers of gemcitabine and 5-fluorouracil is likely to drive therapeutic decisions in patients with metastatic PAC. TRIAL REGISTRATION AFUGEM is registered at Clinicaltrials.gov: NCT01964534 , October 15, 2013.
Collapse
Affiliation(s)
- Jean-Baptiste Bachet
- Paris-Sorbonne University, UPMC University Paris 06, Paris, France.
- Department of hepatogastroenterology, Groupe hospitalier Pitié Salpêtrière, Paris, France.
| | | | - Franck Bonnetain
- Head of methodology and quality of life in oncology department, Hôpital Universitaire de Besancon, EA 3181, Besancon, France.
| | - Pierre Validire
- Department of pathology, Institut Mutualiste Montsouris, Paris, France.
| | - Pascal Hammel
- Department of digestive oncology, Hôpital Beaujon, Clichy, France.
| | - Thierry André
- Paris-Sorbonne University, UPMC University Paris 06, Paris, France.
- Department of oncology, Hôpital Saint Antoine, Paris, France.
| | - Christophe Louvet
- Department of oncology, Institut Mutualiste Montsouris, Paris, France.
| |
Collapse
|
37
|
Abstract
There is now compelling evidence that the molecular heterogeneity of cancer is associated with disparate phenotypes with variable outcomes and therapeutic responsiveness to therapy in histologically indistinguishable cancers. This diversity may explain why conventional clinical trial designs have mostly failed to show efficacy when patients are enrolled in an unselected fashion. Knowledge of the molecular phenotype has the potential to improve therapeutic selection and hence the early delivery of the optimal therapeutic regimen. Resolution of the challenges associated with a more stratified approach to health care will ensure more precise diagnostics and enhance therapeutic selection, which will improve overall outcomes.
Collapse
Affiliation(s)
- Nigel B Jamieson
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; Academic Unit of Surgery, School of Medicine, College of Medical, Veterinary and Life Sciences, Glasgow Royal Infirmary, University of Glasgow, Alexandra Parade, Glasgow G31 2ER, UK; West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Alexandra Parade, Glasgow G31 2ER, UK
| | - David K Chang
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Darlinghurst, New South Wales 2010, Australia; Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia; Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, New South Wales 2200, Australia; Faculty of Medicine, South Western Sydney Clinical School, University of NSW, Goulburn St, Liverpool, New South Wales 2170, Australia
| | - Andrew V Biankin
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Bearsden, Glasgow G61 1BD, UK; The Kinghorn Cancer Centre, Garvan Institute of Medical Research, 370 Victoria Street, Darlinghurst, New South Wales 2010, Australia; Cancer Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, New South Wales 2010, Australia; Department of Surgery, Bankstown Hospital, Eldridge Road, Bankstown, Sydney, New South Wales 2200, Australia; Faculty of Medicine, South Western Sydney Clinical School, University of NSW, Goulburn St, Liverpool, New South Wales 2170, Australia.
| |
Collapse
|
38
|
Jouffret L, Turrini O, Ewald J, Moutardier V, Iovanna JL, Delpero JR. Long-term survivors after pancreatectomy for cancer: the TNM classification is outdated. ANZ J Surg 2015; 85:860-4. [PMID: 26331377 DOI: 10.1111/ans.13277] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/14/2015] [Indexed: 01/11/2023]
Abstract
BACKGROUND According to knowledge, patients with resectable pancreatic adenocarcinoma (PA) should receive adjuvant gemcitabine-based chemotherapy. Thus, the tumour node metastasis (TNM) classification is not used to determine post-operative treatment but rather only to establish patient prognosis. However, the TNM classification does not include strong factors influencing survival, such as perineural invasion or margin status. This study compared the survival of patients with very similar tumours. METHODS From 1997 to 2007, 118 patients underwent pancreatectomy for PA. Twenty-six patients (22%) had long-term survival (>5 years; LTS group). According to the major prognostic factors of PA, we matched (1:1) patients in the LTS group with patients who did not have long-term survival (<5 years; control group). RESULTS Surprisingly, we did not have any difficulty identifying patients to include in the control group. Consequently, no differences were noted between patients of the LTS group when compared with patients of the control group according to major prognostic factors and the TNM classification. Three patients (12%) in the LTS group had positive margin status, and two patients (8%) had positive lymph node status. Unsurprisingly, the median survival for the control group versus the LTS group was 16 months versus not reached (P < 0.01). CONCLUSION The lack of difficulty in matching the two groups confirmed that patients of the LTS group did not have an exclusive tumour pattern. TNM classification is outdated because it did not influence adjuvant therapies and did not include two crucial factors: tumour biology and tumour response to chemo/radio therapies.
Collapse
Affiliation(s)
- Lionel Jouffret
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Olivier Turrini
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Jacques Ewald
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | | | - Juan Lucio Iovanna
- Cancer Research Center of Marseille, INSERM U1068, CNRS UMR 7258, Aix-Marseille University and Institut Paoli-Calmettes, Marseille, France
| | - Jean-Robert Delpero
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| |
Collapse
|
39
|
Clinical implications of epithelial cell plasticity in cancer progression. Cancer Lett 2015; 366:1-10. [DOI: 10.1016/j.canlet.2015.06.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 05/19/2015] [Accepted: 06/06/2015] [Indexed: 12/18/2022]
|
40
|
Torres C, Linares A, Alejandre MJ, Palomino-Morales RJ, Caba O, Prados J, Aránega A, Delgado JR, Irigoyen A, Martínez-Galán J, Ortuño FM, Rojas I, Perales S. Prognosis Relevance of Serum Cytokines in Pancreatic Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:518284. [PMID: 26346854 PMCID: PMC4539422 DOI: 10.1155/2015/518284] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 01/05/2015] [Accepted: 01/12/2015] [Indexed: 02/06/2023]
Abstract
The overall survival of patients with pancreatic ductal adenocarcinoma is extremely low. Although gemcitabine is the standard used chemotherapy for this disease, clinical outcomes do not reflect significant improvements, not even when combined with adjuvant treatments. There is an urgent need for prognosis markers to be found. The aim of this study was to analyze the potential value of serum cytokines to find a profile that can predict the clinical outcome in patients with pancreatic cancer and to establish a practical prognosis index that significantly predicts patients' outcomes. We have conducted an extensive analysis of serum prognosis biomarkers using an antibody array comprising 507 human cytokines. Overall survival was estimated using the Kaplan-Meier method. Univariate and multivariate Cox's proportional hazard models were used to analyze prognosis factors. To determine the extent that survival could be predicted based on this index, we used the leave-one-out cross-validation model. The multivariate model showed a better performance and it could represent a novel panel of serum cytokines that correlates to poor prognosis in pancreatic cancer. B7-1/CD80, EG-VEGF/PK1, IL-29, NRG1-beta1/HRG1-beta1, and PD-ECGF expressions portend a poor prognosis for patients with pancreatic cancer and these cytokines could represent novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Carolina Torres
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| | - Ana Linares
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| | - Maria José Alejandre
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| | | | - Octavio Caba
- Department of Health Sciences, University of Jaen, 23071 Jaen, Spain
| | - Jose Prados
- Department of Human Anatomy and Embryology, University of Granada, 18012 Granada, Spain
| | - Antonia Aránega
- Department of Human Anatomy and Embryology, University of Granada, 18012 Granada, Spain
| | - Juan R. Delgado
- Oncology Service, Virgen de las Nieves Hospital, 18014 Granada, Spain
| | - Antonio Irigoyen
- Oncology Service, Virgen de las Nieves Hospital, 18014 Granada, Spain
| | | | - Francisco M. Ortuño
- Department of Computer Architecture and Computer Technology (CITIC-UGR), University of Granada, 18071 Granada, Spain
| | - Ignacio Rojas
- Department of Computer Architecture and Computer Technology (CITIC-UGR), University of Granada, 18071 Granada, Spain
| | - Sonia Perales
- Department of Biochemistry and Molecular Biology I, University of Granada, 18071 Granada, Spain
| |
Collapse
|
41
|
Miller BW, Morton JP, Pinese M, Saturno G, Jamieson NB, McGhee E, Timpson P, Leach J, McGarry L, Shanks E, Bailey P, Chang D, Oien K, Karim S, Au A, Steele C, Carter CR, McKay C, Anderson K, Evans TRJ, Marais R, Springer C, Biankin A, Erler JT, Sansom OJ. Targeting the LOX/hypoxia axis reverses many of the features that make pancreatic cancer deadly: inhibition of LOX abrogates metastasis and enhances drug efficacy. EMBO Mol Med 2015; 7:1063-76. [PMID: 26077591 PMCID: PMC4551344 DOI: 10.15252/emmm.201404827] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 05/12/2015] [Accepted: 05/21/2015] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related mortality. Despite significant advances made in the treatment of other cancers, current chemotherapies offer little survival benefit in this disease. Pancreaticoduodenectomy offers patients the possibility of a cure, but most will die of recurrent or metastatic disease. Hence, preventing metastatic disease in these patients would be of significant benefit. Using principal component analysis (PCA), we identified a LOX/hypoxia signature associated with poor patient survival in resectable patients. We found that LOX expression is upregulated in metastatic tumors from Pdx1-Cre Kras(G12D/+) Trp53(R172H/+) (KPC) mice and that inhibition of LOX in these mice suppressed metastasis. Mechanistically, LOX inhibition suppressed both migration and invasion of KPC cells. LOX inhibition also synergized with gemcitabine to kill tumors and significantly prolonged tumor-free survival in KPC mice with early-stage tumors. This was associated with stromal alterations, including increased vasculature and decreased fibrillar collagen, and increased infiltration of macrophages and neutrophils into tumors. Therefore, LOX inhibition is able to reverse many of the features that make PDAC inherently refractory to conventional therapies and targeting LOX could improve outcome in surgically resectable disease.
Collapse
Affiliation(s)
- Bryan W Miller
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | | | - Mark Pinese
- The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Grazia Saturno
- Cancer Research UK Manchester Institute, Withington Manchester, UK
| | - Nigel B Jamieson
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Ewan McGhee
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Paul Timpson
- The Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Joshua Leach
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Lynn McGarry
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Emma Shanks
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Peter Bailey
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - David Chang
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Karin Oien
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Saadia Karim
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Amy Au
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Colin Steele
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | | | - Colin McKay
- West of Scotland Pancreatic Unit, Glasgow Royal Infirmary, Glasgow, UK
| | - Kurt Anderson
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| | - Thomas R Jeffry Evans
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Richard Marais
- Cancer Research UK Manchester Institute, Withington Manchester, UK
| | | | - Andrew Biankin
- Institute of Cancer Sciences University of Glasgow Garscube Estate, Glasgow, UK
| | - Janine T Erler
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen (UCPH), Denmark
| | - Owen J Sansom
- Cancer Research UK Beatson Institute Garscube Estate, Glasgow, UK
| |
Collapse
|
42
|
McCormick Matthews LH, Noble F, Tod J, Jaynes E, Harris S, Primrose JN, Ottensmeier C, Thomas GJ, Underwood TJ. Systematic review and meta-analysis of immunohistochemical prognostic biomarkers in resected oesophageal adenocarcinoma. Br J Cancer 2015; 113:107-18. [PMID: 26110972 PMCID: PMC4647536 DOI: 10.1038/bjc.2015.179] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 02/22/2015] [Accepted: 04/29/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Oesophageal adenocarcinoma (OAC) is one of the fastest rising malignancies with continued poor prognosis. Many studies have proposed novel biomarkers but, to date, no immunohistochemical markers of survival after oesophageal resection have entered clinical practice. Here, we systematically review and meta-analyse the published literature, to identify potential biomarkers. METHODS Relevant articles were identified via Ovid medline 1946-2013. For inclusion, studies had to conform to REporting recommendations for tumor MARKer (REMARK) prognostic study criteria. The primary end-point was a pooled hazard ratio (HR) and variance, summarising the effect of marker expression on prognosis. RESULTS A total of 3059 articles were identified. After exclusion of irrelevant titles and abstracts, 214 articles were reviewed in full. Nine molecules had been examined in more than one study (CD3, CD8, COX-2, EGFR, HER2, Ki67, LgR5, p53 and VEGF) and were meta-analysed. Markers with largest survival effects were COX-2 (HR=2.47, confidence interval (CI)=1.15-3.79), CD3 (HR=0.51, 95% CI=0.32-0.70), CD8 (HR=0.55, CI=0.31-0.80) and EGFR (HR=1.65, 95% CI=1.14-2.16). DISCUSSION Current methods have not delivered clinically useful molecular prognostic biomarkers in OAC. We have highlighted the paucity of good-quality robust studies in this field. A genome-to-protein approach would be better suited for the development and subsequent validation of biomarkers. Large collaborative projects with standardised methodology will be required to generate clinically useful biomarkers.
Collapse
Affiliation(s)
- L H McCormick Matthews
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, MP824, Southampton SO16 6YD, UK
| | - F Noble
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, MP824, Southampton SO16 6YD, UK
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - J Tod
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, MP824, Southampton SO16 6YD, UK
| | - E Jaynes
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - S Harris
- Public Health Sciences and Medical Statistics, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - J N Primrose
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, MP824, Southampton SO16 6YD, UK
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - C Ottensmeier
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, MP824, Southampton SO16 6YD, UK
- National Institute for Health Research, Experimental Cancer Medicine Centre, Southampton SO16 6YD, UK
| | - G J Thomas
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, MP824, Southampton SO16 6YD, UK
- Department of Cellular Pathology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - T J Underwood
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Somers Cancer Research Building, MP824, Southampton SO16 6YD, UK
- Department of Surgery, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
43
|
Saukkonen K, Hagström J, Mustonen H, Juuti A, Nordling S, Fermér C, Nilsson O, Seppänen H, Haglund C. Podocalyxin Is a Marker of Poor Prognosis in Pancreatic Ductal Adenocarcinoma. PLoS One 2015; 10:e0129012. [PMID: 26053486 PMCID: PMC4459962 DOI: 10.1371/journal.pone.0129012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 04/21/2015] [Indexed: 01/09/2023] Open
Abstract
Aim of the Study Podocalyxin-like 1 is a transmembrane glyco-protein whose overexpression associates in many cancers with poor prognosis and unfavorable clinicopathological characteristics. Until now, its prognostic value has never been studied in pancreatic ductal adenocarcinoma (PDAC). The aim of this study was to investigate podocalyxin expression in PDAC by a novel monoclonal antibody and a commercially available polyclonal antibody. Patients and Materials With tissue microarrays and immuno-histochemistry, podocalyxin expression evaluation involved 168 PDAC patients. The associa-tions of the podocalyxin tumor expression with clinicopathological variables were explored by Fisher’s exact test and the linear-by-linear test. Survival analyses were by Kaplan-Meier anal-ysis and the Cox proportional hazard model. Results The polyclonal antibody revealed membranous podocalyxin expression in 73 (44.0%) specimens and the monoclonal antibody was highly expressed in 36 (21.8%) cases. Membranous expression by the polyclonal antibody was associated with T classification (p=0.045) and perineural invasion (p=0.005), and high expression by the mono-clonal antibody with poor differentiation (p=0.033). High podocalyxin expression associated significantly with higher risk of death from PDAC by both the polyclonal antibody (hazard ratio (HR) = 1.62; 95% confidence interval (CI) 1.12-2.33; p=0.01) and the monoclonal antibody (HR = 2.10, 95% CI 1.38-3.20; p<0.001). The results remained significant in multivariate analysis, adjusted for age, gender, stage, lymph node ratio (≥/< 20%), and perivascular invasion (respectively as HR = 2.03; 95% CI 1.32-3.13, p=0.001; and as HR = 2.36; 95% CI 1.47-3.80, p<0.001). Conclusion We found podocalyxin to be an independent factor for poor prognosis in PDAC. To our knowledge, this is the first such report of its prognostic value.
Collapse
Affiliation(s)
- Kapo Saukkonen
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland; Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Jaana Hagström
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, FIN-00014 University of Helsinki, Helsinki, Finland; Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Harri Mustonen
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland
| | - Anne Juuti
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland
| | - Stig Nordling
- Department of Pathology, Haartman Institute and HUSLAB, University of Helsinki and Helsinki University Hospital, Helsinki, FIN-00014 University of Helsinki, Helsinki, Finland
| | - Christian Fermér
- Fujirebio Diagnostics AB, Elof Lindälvs gata 13, SE-414 58 Gothenburg, Sweden
| | - Olle Nilsson
- Onson Consulting, Södra vägen 2, SE-412 54 Gothenburg, Sweden
| | - Hanna Seppänen
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland
| | - Caj Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, P.O. Box 440, FIN-00029 HUS, Helsinki, Finland; Research Programs Unit, Translational Cancer Biology, University of Helsinki, P.O. Box 63, FIN-00014 University of Helsinki, Helsinki, Finland
| |
Collapse
|
44
|
Jiang W, Zhao S, Xu L, Lu Y, Lu Z, Chen C, Ni J, Wan R, Yang L. The inhibitory effects of xanthohumol, a prenylated chalcone derived from hops, on cell growth and tumorigenesis in human pancreatic cancer. Biomed Pharmacother 2015. [PMID: 26211581 DOI: 10.1016/j.biopha.2015.05.020] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Pancreatic cancer (PC) is one of the most lethal human malignancies worldwide. Here, we demonstrated that xanthohumol (XN), the most abundant prenylated chalcone isolated from hops, inhibited the growth of cultured PC cells and their subcutaneous xenograft tumors. XN treatment was found to induce cell cycle arrest and apoptosis of PC cells (PANC-1, BxPC-3) by inhibiting phosphorylation of signal transducer and activator of transcription 3 (STAT3) and expression of its downstream targeted genes cyclinD1, survivin, and Bcl-xL at the messenger RNA level, which involved in regulation of apoptosis and the cell cycle. Overall, our results suggested that XN presents a promising candidate therapeutic agent against human PC and the STAT3 signaling pathway is its key molecular target.
Collapse
Affiliation(s)
- Weiliang Jiang
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100, Hai Ning Road, Shanghai 200080, China.
| | - Senlin Zhao
- Department of General Surgery, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ling Xu
- Department of Gastroenterology, Shanghai Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Yingying Lu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100, Hai Ning Road, Shanghai 200080, China.
| | - Zhanjun Lu
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100, Hai Ning Road, Shanghai 200080, China.
| | - Congying Chen
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100, Hai Ning Road, Shanghai 200080, China.
| | - Jianbo Ni
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100, Hai Ning Road, Shanghai 200080, China.
| | - Rong Wan
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100, Hai Ning Road, Shanghai 200080, China.
| | - Lijuan Yang
- Department of Gastroenterology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, No. 100, Hai Ning Road, Shanghai 200080, China.
| |
Collapse
|
45
|
Ieni A, Todaro P, Crino SF, Barresi V, Tuccari G. Endoscopic ultrasound-guided fine-needle aspiration cytology in pancreaticobiliary carcinomas: diagnostic efficacy of cell-block immunocytochemistry. Hepatobiliary Pancreat Dis Int 2015; 14:305-12. [PMID: 26063033 DOI: 10.1016/s1499-3872(15)60367-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Endoscopic ultrasound-guided fine-needle aspiration cytology was demonstrated to be a useful tool for the diagnosis and staging of pancreaticobiliary neoplastic lesions. Nonetheless, the diagnostic value of this procedure may be limited by low cellularity of the specimen, contamination of intestinal cells and unfeasibility of ancillary immunocytochemical procedures. The present study was to evaluate its usefulness in the diagnosis of neoplastic lesions. METHODS A series of 46 pancreaticobiliary carcinomas with available cell block preparations was submitted to immunocytochemistry against cytokeratins, carcinoembryonic antigen, E-cadherin, CD10 and p53. The sensitivity, specificity, positive and negative predictive values of the cytological smear in the discrimination of malignant lesions were calculated and compared with those of cell block preparation with the immunocytochemical stains against p53 and CD10. RESULTS According to our findings, the use of cell block preparations together with immunostains against p53 and CD10 allowed to discriminate malignant versus benign specimens with higher sensitivity than the only cytological examination. In detail, CD10 immunostaining was of significant help for the discrimination between cytological contaminants, such as benign gastrointestinal cells, and the neoplastic elements of pancreaticobiliary well differentiated adenocarcinomas. Also, intense nuclear immunoreactivity for p53 was encountered in about 2/3 of the cases and identified pancreatic malignancy with high sensitivity. CONCLUSIONS We suggest that immunocytochemistry against both CD10 and p53 could be applied case by case, mainly to differentiate gastrointestinal and pancreatic benign cellular contaminants showing hyperplasia or reactive changes from differentiated pancreaticobiliary adenocarcinomas.
Collapse
Affiliation(s)
- Antonio Ieni
- Department of Human Pathology "Gaetano Barresi", Section of Pathological Anatomy, Azienda Ospedaliera Universitaria "Policlinico G. Martino", University of Messina, Messina 98125, Italy.
| | | | | | | | | |
Collapse
|
46
|
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: 2.6] [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
|
47
|
Furukawa K, Kawamoto K, Eguchi H, Tanemura M, Tanida T, Tomimaru Y, Akita H, Hama N, Wada H, Kobayashi S, Nonaka Y, Takamatsu S, Shinzaki S, Kumada T, Satomura S, Ito T, Serada S, Naka T, Mori M, Doki Y, Miyoshi E, Nagano H. Clinicopathological Significance of Leucine-Rich α2-Glycoprotein-1 in Sera of Patients With Pancreatic Cancer. Pancreas 2015; 44:93-98. [PMID: 25058884 DOI: 10.1097/mpa.0000000000000205] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Leucine-rich α2-glycoprotein-1 (LRG-1) is an inflammatory protein. Serum LRG-1 levels can reportedly be used as a cancer biomarker for several types of carcinoma. In the present study, we investigated the clinical usefulness of serum LRG-1 levels as a biomarker of pancreatic cancer. METHODS A total of 124 patients with pancreatic cancer, 35 patients with chronic pancreatitis (CP), and 144 healthy volunteers were enrolled in the study. Serum LRG-1 levels were assayed by enzyme-linked immunosorbent assay. Immunohistochemistry was used to examine LRG-1 expression in pancreatic cancer tissues. RESULTS Serum LRG-1 levels were significantly increased in patients with pancreatic cancer compared with CP patients and healthy volunteers. The LRG-1 levels increased with progressive clinical stages of pancreatic cancer. Receiver operator curve analysis showed that a combination of carbohydrate antigen 19-9 and LRG-1 resulted in a higher area under the curve for the diagnosis of pancreatic cancer. Positive staining was observed in all cases of pancreatic cancer, but positive signal was scarcely detected in tissues from CP patients or normal surrounding tissue. CONCLUSIONS These results suggest that serum LRG-1 is a promising biomarker for pancreatic cancer.
Collapse
Affiliation(s)
- Kenta Furukawa
- From the *Department of Surgery, Osaka University Graduate School of Medicine, Osaka; †Department of Surgery and Institute for Clinical Research National Hospital Organization Kure Medical Center and Chugoku Cancer Center, Hiroshima; ‡Department of Molecular Biochemistry and Clinical Investigation, Osaka University Graduate School of Medicine, Osaka; §Department of Gastroenterology, Ogaki Municipal Hospital, Gifu; ║Wako Pure Chemical Industries, Ltd; ¶Department of Gastroenterology and Hepatology, Japan Community Health Care Organization Osaka Hospital; and #Laboratory for Immune Signal, National Institute of Biomedical Innovation, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Microvessel Landscape Assessment in Pancreatic Ductal Adenocarcinoma: Unclear Value of Targeting Endoglin (CD105) as Prognostic Factor of Clinical Outcome. Pancreas 2015; 44:87-92. [PMID: 25058886 DOI: 10.1097/mpa.0000000000000197] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
OBJECTIVES Tumor angiogenesis based on microvessel density assessment has been associated with poor prognosis in several studies of patients with pancreatic ductal adenocarcinoma (PDAC). Expression of endoglin (CD105), a tumor-induced vascularization marker, has been found to represent a negative prognostic factor in many malignant tumors. The aim of our study was to assess the value of tumoral microvascularity both with pan-endothelial markers and endoglin as well, in correlation with the clinical outcome of patients with PDAC. METHODS Fifty-eight patients with PDAC, 36 males and 22 females, with a mean (SD) age of 65.4 (10.0) years were included in the study. Deparaffinized sections from formalin-fixed areas both from the center and periphery (invasion front) of the tumors were immunostained for CD105 as well as for the endothelial markers CD31 and CD34. Tumoral angiogenesis was assessed on the basis of microvessel density (number of vessels per square millimeter) and on microvascular area (square micrometers) as well. RESULTS High intratumoral microvascular area, in endoglin-stained sections, was found to be of marginal prognostic significance for recurrence (log rank, P 0.05). Survival was also marginally associated with CD31 intratumoral microvascular area (log rank, P 0.05). CONCLUSIONS Further studies are needed before endoglin replaces the conventional angiogenesis markers in PDCA.
Collapse
|
49
|
Zhang P, Hu P, Shen H, Yu J, Liu Q, Du J. Prognostic role of Twist or Snail in various carcinomas: a systematic review and meta-analysis. Eur J Clin Invest 2014; 44:1072-94. [PMID: 25257753 DOI: 10.1111/eci.12343] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 09/21/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Twist and Snail are considered as key transcriptional repressors of E-cadherin tightly related to epithelial-to-mesenchymal transition (EMT) and cancer progression. Numerous studies have investigated the prognostic value of Twist and Snail. However, the published results were controversial or even opposite. Our article aimed to evaluate the prognostic role of Twist and Snail in patients with cancer. DESIGN A comprehensive literature search of PubMed, Embase and Web of Science was conducted. Pooled hazard ratios (HRs) and their corresponding 95% confidence intervals (CIs) were assessed to quantify the prognostic role. RESULTS The pooled HR with 38 studies for Twist was 2·18 (95% CI: 1·77-2·68, I(2) = 69·8%, P = 0·000) and for Snail with 40 studies was 1·58 (95% CI: 1·33-1·87, I(2) = 70·0%, P = 0·000), suggesting high Twist/Snail expression predicted poor prognosis related to all clinical outcomes. For Twist, the pooled HR for overall survival (OS) was 2·07 (95% CI: 1·63-2·63, I(2) = 72·6%, P = 0·000) and for progression-free/recurrence-free/metastasis-free/disease-free/cancer-free survival (PFS/RFS/MFS/DFS/CFS) was 2·36 (95% CI: 1·76-3·17, I(2) = 65·0%, P = 0·000). For Snail, the pooled HR for OS was 1·63 (95% CI: 1·33-1·99, I(2) = 70·8%, P = 0·000) and for PFS/RFS/MFS/DFS/CFS was 1·54 (95% CI: 1·17-2·02, I(2) = 59·1%, P = 0·001). All of those results were suggesting that high Twist/Snail expression was associated with poor prognosis. Furthermore, when grouped into different types of cancers, the pooled HRs were also calculated for the subgroups. No publication bias was found except studies evaluating all clinical outcomes of Twist (P = 0·006 for Begg's test and 0·006 for Egger's test). CONCLUSIONS Elevated Twist or Snail expression in tumour tissue indicated poor prognosis for cancer.
Collapse
Affiliation(s)
- Ping Zhang
- Institute of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, Jinan, China
| | | | | | | | | | | |
Collapse
|
50
|
A clinicopathological analysis of primary mucosal malignant melanoma. Surg Today 2014; 45:886-91. [DOI: 10.1007/s00595-014-1041-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 09/10/2014] [Indexed: 02/07/2023]
|