1
|
Mahmoud S, Ahmed M E, Mohamed I H. Progress in the development of vaccines for pancreatic adenocarcinoma. ANNALS OF PANCREATIC DISORDERS AND TREATMENT 2024; 6:001-005. [DOI: 10.17352/apdt.000011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Pancreatic cancer, which is regarded as the third deadliest cancer globally, poses a significant challenge because of its limited range of treatment options and high mortality rate. Currently, there is a focus on both the development of a novel concept in vaccine designing and the parallel study of the associated immune mechanisms. To further our understanding of the healthcare field, a variety of promising designs have been introduced for in-depth study. The designs were developed to include the mKRAS-specific amphiphile vaccine, which targets a specific mutation in the KRAS gene in addition to the multi-antigen targeted DNA vaccine, which aims to stimulate an immune response against multiple cancer antigens. Furthermore, later designs of vaccines were introduced based on the development of peptide-based cancer vaccines, mRNA-based vaccines, cell-based vaccines, and engineered bacterial vectors using an oral Salmonella-based vaccine. The study presents the concept on which the new vaccine is based and discusses the up-to-date immunological manifestations of these designed vaccines.
Collapse
|
2
|
Clelland S, Nuttall CL, Stott HE, Cope J, Barratt NL, Farrell K, Eyong MV, Gleeson JP, Lamarca A, Hubner RA, Valle JW, McNamara MG. Prognosis Discussion and Referral to Community Palliative Care Services in Patients with Advanced Pancreatic Cancer Treated in a Tertiary Cancer Centre. Healthcare (Basel) 2023; 11:2802. [PMID: 37893876 PMCID: PMC10606359 DOI: 10.3390/healthcare11202802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Advanced pancreatic cancer is associated with a poor prognosis, often less than 1 year. Honest prognosis discussions guide early community palliative care services input, facilitating timely advance care planning and improving quality of life. The aims were to assess if patients were offered prognosis discussions and community palliative care services referral. A retrospective analysis of consecutive case-notes of new advanced pancreatic cancer patients was conducted. Chi-squared test assessed the association with prognosis discussion and community palliative care services referral. In total, 365 cases (60%) had a documented prognosis discussion at any time-point in the treatment pathway; 54.4% during the first appointment. The frequency of prognosis discussion was greater with nurse clinician review at first appointment (p < 0.001). In total, 171 patients (28.1%) were known to community palliative care services at the first appointment. Of those not known, 171 (39.1%) and 143 (32.7%) were referred at this initial time-point or later, respectively. There was a significant association between the referral to community palliative care services at first appointment and the reviewing professional (this was greatest for nurse clinicians (frequency 65.2%)) (p < 0.001), and also if reviewed by clinical nurse specialist at first visit or not (47.8% vs. 35.6%) (p < 0.01). Prognosis discussions were documented in approximately two-thirds of cases, highlighting missed opportunities. Prognosis discussion was associated with clinician review and was most frequent for nurse clinician, as was referral to community palliative care services. Clinical nurse specialist review increased referral to community palliative care services if seen at the initial visit. Multi-disciplinary review, specifically nursing, therefore, during the first consultation is imperative and additive. It should be considered best practice to offer and negotiate the content and timing of prognosis discussions with cancer patients, and revisit this offer throughout their treatment pathway. Greater attention to prognosis discussion documentation is recommended.
Collapse
Affiliation(s)
- Sarah Clelland
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | | | - Helen E. Stott
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Joseph Cope
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | | | - Kelly Farrell
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | - Manyi V. Eyong
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
| | | | - Angela Lamarca
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
| | - Richard A. Hubner
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
| | - Juan W. Valle
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
| | - Mairéad G. McNamara
- The Christie NHS Foundation Trust, Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Manchester M20 4BX, UK
| |
Collapse
|
3
|
Zhang L, Yu Z, Jin R, Yang X, Ying D. Machine learning-based prediction of surgical benefit in borderline resectable and locally advanced pancreatic cancer. J Cancer Res Clin Oncol 2023; 149:11857-11871. [PMID: 37410139 DOI: 10.1007/s00432-023-05071-9] [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: 05/20/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023]
Abstract
INTRODUCTION Surgery represents a primary therapeutic approach for borderline resectable and locally advanced pancreatic cancer (BR/LAPC). However, BR/LAPC lesions exhibit high heterogeneity and not all BR/LAPC patients who undergo surgery can derive beneficial outcomes. The present study aims to employ machine learning (ML) algorithms to identify those who would obtain benefits from the primary tumor surgery. METHODS We retrieved clinical data of patients with BR/LAPC from the Surveillance, Epidemiology, and End Results (SEER) database and classified them into surgery and non-surgery groups based on primary tumor surgery status. To eliminate confounding factors, propensity score matching (PSM) was employed. We hypothesized that patients who underwent surgery and had a longer median cancer-specific survival (CSS) than those who did not undergo surgery would certainly benefit from surgical intervention. Clinical and pathological features were utilized to construct six ML models, and model effectiveness was compared through measures such as the area under curve (AUC), calibration plots, and decision curve analysis (DCA). We selected the best-performing algorithm (i.e., XGBoost) to predict postoperative benefits. The SHapley Additive exPlanations (SHAP) approach was used to interpret the XGBoost model. Additionally, data from 53 Chinese patients prospectively collected was used for external validation of the model. RESULTS According to the results of the tenfold cross-validation in the training cohort, the XGBoost model yielded the best performance (AUC = 0.823, 95%CI 0.707-0.938). The internal (74.3% accuracy) and external (84.3% accuracy) validation demonstrated the generalizability of the model. The SHAP analysis provided explanations independent of the model, highlighting important factors related to postoperative survival benefits in BR/LAPC, with age, chemotherapy, and radiation therapy being the top three important factors. CONCLUSION By integrating of ML algorithms and clinical data, we have established a highly efficient model to facilitate clinical decision-making and assist clinicians in selecting the population that would benefit from surgery.
Collapse
Affiliation(s)
- Leiming Zhang
- Department of Minimally Invasive Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Zehao Yu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Rong Jin
- Department of Minimally Invasive Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Xuanang Yang
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Dongjian Ying
- Department of Minimally Invasive Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China.
| |
Collapse
|
4
|
Chan YT, Tan HY, Lu Y, Zhang C, Cheng CS, Wu J, Wang N, Feng Y. Pancreatic melatonin enhances anti-tumor immunity in pancreatic adenocarcinoma through regulating tumor-associated neutrophils infiltration and NETosis. Acta Pharm Sin B 2023; 13:1554-1567. [PMID: 37139434 PMCID: PMC10150138 DOI: 10.1016/j.apsb.2023.01.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/13/2022] [Accepted: 11/18/2022] [Indexed: 02/05/2023] Open
Abstract
Tumor microenvironment contributes to poor prognosis of pancreatic adenocarcinoma (PAAD) patients. Proper regulation could improve survival. Melatonin is an endogenous hormone that delivers multiple bioactivities. Here we showed that pancreatic melatonin level is associated with patients' survival. In PAAD mice models, melatonin supplementation suppressed tumor growth, while blockade of melatonin pathway exacerbated tumor progression. This anti-tumor effect was independent of cytotoxicity but associated with tumor-associated neutrophils (TANs), and TANs depletion reversed effects of melatonin. Melatonin induced TANs infiltration and activation, therefore induced cell apoptosis of PAAD cells. Cytokine arrays revealed that melatonin had minimal impact on neutrophils but induced secretion of Cxcl2 from tumor cells. Knockdown of Cxcl2 in tumor cells abolished neutrophil migration and activation. Melatonin-induced neutrophils presented an N1-like anti-tumor phenotype, with increased neutrophil extracellular traps (NETs) causing tumor cell apoptosis through cell-to-cell contact. Proteomics analysis revealed that this reactive oxygen species (ROS)-mediated inhibition was fueled by fatty acid oxidation (FAO) in neutrophils, while FAO inhibitor abolished the anti-tumor effect. Analysis of PAAD patient specimens revealed that CXCL2 expression was associated with neutrophil infiltration. CXCL2, or TANs, combined with NET marker, can better predict patients' prognosis. Collectively, we discovered an anti-tumor mechanism of melatonin through recruiting N1-neutrophils and beneficial NET formation.
Collapse
Affiliation(s)
- Yau-tuen Chan
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
| | - Hor-yue Tan
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
- Centre for Chinese Herbal Medicine Drug Development, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Yuanjun Lu
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
| | - Cheng Zhang
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
| | - Chien-shan Cheng
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
- Department of Traditional Chinese Medicine, Shanghai Jiaotong University, School of Medicine Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Junyu Wu
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
| | - Ning Wang
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
- Corresponding authors.
| | - Yibin Feng
- School of Chinese Medicine, the University of Hong Kong, Hong Kong, China
- Corresponding authors.
| |
Collapse
|
5
|
Caliez O, Pietrasz D, Ksontini F, Doat S, Simon JM, Vaillant JC, Taly V, Laurent-Puig P, Bachet JB. Circulating tumor DNA: a help to guide therapeutic strategy in patients with borderline and locally advanced pancreatic adenocarcinoma? Dig Liver Dis 2022; 54:1428-1436. [PMID: 35120842 DOI: 10.1016/j.dld.2022.01.126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND prognostic biomarkers could be useful to better select patients with borderline resectable (BR) or locally advanced (LA) pancreatic adenocarcinoma (PA) for chemoradiotherapy (CRT) and/or secondary resection. AIMS The main objective of this work was to study characteristics, received treatments and prognostic of patients with BR or LA PA according to their baseline circulating tumor DNA status and, for secondary objective, neutrophil-to-lymphocyte Ratio (NLR). METHODS ctDNA status at baseline was determined using Next Generation Sequencing in a consecutive monocentric cohort of patients with a BR or LA PA. RESULTS 69 patients were included, 31 with BR PA and 38 with LA PA. 14 (20.3%) patients had baseline positive ctDNA. Five (7.8%) patients had NLR> 5. Patients with positive ctDNA had 3.7 months shorter progression free survival (p = 0.006). Patients with positive ctDNA had earlier progression after the beginning of CRT (4.4 vs 7.1 months; p = 0.068) and shorter relapse free survival after secondary resection (9.2 vs 22.9 months; p = 0.016). CONCLUSIONS positive ctDNA at baseline was associated with a worse prognosis in patients with BR or LA PA. These data are exploratory and must be confirmed in further prospective trials.
Collapse
Affiliation(s)
- Olivier Caliez
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Sorbonne Université, UPMC, Paris 6, France
| | - Daniel Pietrasz
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Department of Digestive Surgery, Hôpital Paul Brousse, Villejuif, France
| | - Feryel Ksontini
- Department of Oncology, Institute Salah-Azaïz, Tunis, Tunisia
| | - Solène Doat
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Marc Simon
- Department of Radiation Oncology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Christophe Vaillant
- Department of Digestive Surgery, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Valerie Taly
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France
| | - Pierre Laurent-Puig
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France
| | - Jean-Baptiste Bachet
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Sorbonne Université, UPMC, Paris 6, France.
| |
Collapse
|
6
|
Zhou Y, Zhou H, Shi J, Guan A, Zhu Y, Hou Z, Li R. Decreased m6A Modification of CD34/CD276(B7-H3) Leads to Immune Escape in Colon Cancer. Front Cell Dev Biol 2021; 9:715674. [PMID: 34307389 PMCID: PMC8297592 DOI: 10.3389/fcell.2021.715674] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/21/2021] [Indexed: 12/21/2022] Open
Abstract
Previous studies have reported that m6a modification promotes tumor immune escape by affecting tumor microenvironment (TME). Due to the complexity of TME, a single biomarker is insufficient to describe the complex biological characteristics of tumor and its microenvironment. Therefore, it is more meaningful to explore a group of effective biomarkers reflecting different characteristics of cancer to evaluate the biological characteristics of solid tumors. Here, the immune gene CD34/CD276 with different m6A peak was obtained by m6A sequencing (MeRIP-seq) of colon cancer (CRC)clinical samples and combined with MsIgDB database, which was used to perform cluster analysis on TCGA-COAD level 3 data. The CD34/CD276 as a molecular marker for CRC prognosis was confirmed by survival analysis and immunohistochemical assay. Further bioinformatics analysis was carried out to analyze the molecular mechanism of CD34/CD276 affecting the TME through m6a-dependent down-regulation and ultimately promoting immune escape of CRC.
Collapse
Affiliation(s)
- Yiran Zhou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haodong Zhou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianlin Shi
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Department of Thoracic Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Aoran Guan
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yankun Zhu
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zongliu Hou
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming, China
| | - Ruhong Li
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, First Department of General Surgery, Yan'an Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
7
|
Williet N, Petrillo A, Roth G, Ghidini M, Petrova M, Forestier J, Lopez A, Thoor A, Weislinger L, De Vita F, Taieb J, Phelip JM. Gemcitabine/Nab-Paclitaxel versus FOLFIRINOX in Locally Advanced Pancreatic Cancer: A European Multicenter Study. Cancers (Basel) 2021; 13:cancers13112797. [PMID: 34199796 PMCID: PMC8200096 DOI: 10.3390/cancers13112797] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Gemcitabine/nab-paclitaxel (GN) and FOLFIRINOX (FFX) are two standard first-line therapies for metastatic pancreatic cancer (PC) but have rarely been compared, especially in patients with locally advanced PC (LAPC). By carefully selecting patients, it is likely these two regimens lead to similar survival outcomes. Through a multicenter European study, biases regarding practice habits are reduced. Hence, we observed no difference between GN and FFX as first-line treatments in patients with LAPC in terms of either survival, tumor response or tumor resection rate. Further trials are needed to confirm these data. Abstract Background: Gemcitabine/nab-paclitaxel (GN) and FOLFIRINOX (FFX) are two standard first-line therapies for metastatic pancreatic cancer (PC) but have rarely been compared, especially in patients with locally advanced PC (LAPC). Methods: This is a retrospective European multicenter study including patients with LAPC treated with either GN or FFX as the first-line therapy between 2010 and 2019. Coprimary objectives were progression-free survival (PFS) and overall survival (OS), both estimated using the Kaplan–Meier method. Results: A total of 147 patients (GN: n = 60; FFX: n = 87) were included. Tumor resection rates were similar between the two groups (16.7% vs. 16.1%; p = 1), with similar R0 resection rates (88.9%). Median PFS rates were not statistically different: 9 months (95% CI: 8–13.5) vs. 12.1 months (95% CI: 10.1–14.6; p = 0.8), respectively. Median OS rates were 15.7 months (95% CI: 12.6–20.2) and 16.7 months (95% CI: 14.8–20.4; p = 0.7), respectively. Abdominal pain at the baseline (HR = 2.03, p = 0.03), tumors located in the tail of the pancreas (HR = 4.35, p = 0.01), CA19-9 > 200 UI/L (HR = 2.03, p = 0.004) and tumor resection (HR = 0.37, p = 0.007) were independent prognostic factors for PFS, similarly to OS. CA19-9 ≤ 200 UI/L (OR = 2.6, p = 0.047) was predictive of the tumor response. Consolidation chemoradiotherapy, more often used in the FFX group (11.7% vs. 50.6%; p < 0.001), was not predictive. Conclusion: This retrospective study did not show any difference between GN and FFX as the first-line treatment in patients with LAPC.
Collapse
Affiliation(s)
- Nicolas Williet
- Department of Hepatogastroenterology, University Hospital of Saint-Etienne, 42000 Saint-Etienne, France;
- Correspondence:
| | - Angelica Petrillo
- Department of Precision Medecine, University of Study of Campania «L. Vanvitelli», 81100 Naples, Italy; (A.P.); (F.D.V.)
| | - Gaël Roth
- Hepato-Gastroenterology Department, University Hospital of Grenoble, 38043 Grenoble, France; (G.R.); (A.T.)
| | - Michele Ghidini
- Department of Medical Oncology, Cancer Center, Hospital of Cremona, 26100 Cremona, Italy;
| | - Mila Petrova
- Department of Medical Oncology, MHAT Nadezhda, 1220 Sofia, Bulgaria;
| | - Julien Forestier
- Department of Medical Oncology, Hôpital Edouard Herriot, 69622 Lyon, France;
| | - Anthony Lopez
- Hepato-Gastroenterology Department, University Hospital of Nancy, 54500 Vandoeuvre-lès-Nancy, France; (A.L.); (L.W.)
| | - Audrey Thoor
- Hepato-Gastroenterology Department, University Hospital of Grenoble, 38043 Grenoble, France; (G.R.); (A.T.)
| | - Lucie Weislinger
- Hepato-Gastroenterology Department, University Hospital of Nancy, 54500 Vandoeuvre-lès-Nancy, France; (A.L.); (L.W.)
| | - Ferdinando De Vita
- Department of Precision Medecine, University of Study of Campania «L. Vanvitelli», 81100 Naples, Italy; (A.P.); (F.D.V.)
| | - Julien Taieb
- Department of Gastroenterology and Gastro-Intestinal Oncology, Hôpital Européen Georges-Pompidou, APHP, Paris Descartes University, Sorbonne Paris Cité, 75004 Paris, France;
| | - Jean Marc Phelip
- Department of Hepatogastroenterology, University Hospital of Saint-Etienne, 42000 Saint-Etienne, France;
| |
Collapse
|
8
|
Wang Z. Diagnostic performance for declined microRNA-133a in pancreatic cancer. J Cell Biochem 2020; 121:3882-3886. [PMID: 31773791 DOI: 10.1002/jcb.29547] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/14/2019] [Indexed: 01/24/2023]
Abstract
MicroRNA-133a (MiR-133a) is proven to exhibit a decreasing tendency in several cancers, as well as pancreatic cancer. Through the present study, we inspected performance for serum miR-133a in diagnosing pancreatic cancer. Serum samples were collected from 110 pancreatic cancer and 64 healthy persons. Relative messenger RNA level for miR-133a in serum specimens was gauged adopting quantitative real-time polymerase chain reaction (qRT-PCR), and compared betwixt two groups employing the Student t test. Receiver operating characteristics (ROC) analysis evaluated miR-133a performance in diagnosing pancreatic cancer. MiR-133a displayed a declining trend among pancreatic cancer samples, compared to the healthy controls (P < .001). The reduced miR-133a degree held strong relation to tumor dimension (P = .002), vessel invasion (P = .004), tumor lymph node metastasis stage (P = .002), and lymph node metastasis (P < .001). In addition, ROC analysis demonstrated that the area under the curve value was 0.893, accompanied by a sensitivity of 90.6% and a specificity of 87.2%, revealing fine execution for serum miR-133a in diagnosing cancer. The downregulation of miR-133a might possess a tight relation to hostile advancement in pancreatic cancer. Serum miR-133a could function as a potential diagnostic indicator for pancreatic cancer.
Collapse
Affiliation(s)
- Zhenyong Wang
- First Department of General Surgery, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
9
|
Berardi R, Rinaldi S, Belfiori G, Partelli S, Crippa S, Torniai M, Falconi M. The Role of Hyponatraemia Before Surgery in Patients With Radical Resected Pancreatic Cancer. CLINICAL MEDICINE INSIGHTS-ONCOLOGY 2020; 14:1179554920936605. [PMID: 32636693 PMCID: PMC7322821 DOI: 10.1177/1179554920936605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 05/19/2020] [Indexed: 11/16/2022]
Abstract
Objectives: Hyponatraemia represents a negative prognostic factor in patients with cancer. The aim of this study was to assess, for the first time, the role of hyponatraemia in patients undergoing radical surgery for pancreatic ductal adenocarcinoma. Methods: A total of 89 patients with stage I-III pancreatic ductal adenocarcinoma underwent radical surgery between November 2012 and October 2014. Relapse-free survival (RFS) and disease-specific survival (DSS) were estimated using Kaplan-Meier method. A Cox regression model was carried out for univariate and multivariate analyses. Fisher exact test was used to estimate correlation between variables. Results: In total, 12 patients (14%) presented with hyponatraemia at diagnosis. The median DSS was 20 months in patients with hyponatraemia and not reached in patients with eunatraemia (P < .1073), while a statistical significant difference was observed in terms of median RFS (10 months vs 17 months, respectively; P = .0233). Considering clinical features (hyponatraemia, smoke and alcoholic habit, diabetes, pain, and jaundice), patients with 4 or more of these factors had a worse prognosis (mDSS: 30 months vs not reached; hazard ratio [HR]: 0.40, 95% confidence interval [CI] = 0.16-0.80; P = .0120). Conclusions: The presence of hyponatraemia and its prompt correction at the diagnosis time should be considered for the correct management of patients with pancreatic carcinoma.
Collapse
Affiliation(s)
- Rossana Berardi
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I - GM Lancisi - G Salesi di Ancona, Ancona, Italy
| | - Silvia Rinaldi
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I - GM Lancisi - G Salesi di Ancona, Ancona, Italy
| | - Giulio Belfiori
- Vita-Salute San Raffaele University, Milan, Italy.,Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Partelli
- Vita-Salute San Raffaele University, Milan, Italy.,Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefano Crippa
- Vita-Salute San Raffaele University, Milan, Italy.,Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mariangela Torniai
- Clinica Oncologica, Università Politecnica delle Marche, Azienda Ospedaliero-Universitaria Ospedali Riuniti Umberto I - GM Lancisi - G Salesi di Ancona, Ancona, Italy
| | - Massimo Falconi
- Vita-Salute San Raffaele University, Milan, Italy.,Division of Pancreatic Surgery, Pancreas Translational & Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
10
|
Mas L, Schwarz L, Bachet JB. Adjuvant chemotherapy in pancreatic cancer: state of the art and future perspectives. Curr Opin Oncol 2020; 32:356-363. [PMID: 32541325 DOI: 10.1097/cco.0000000000000639] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW The modalities of management of resectable pancreatic ductal adenocarcinoma (PDAC) have evolved in recent years with new practice guidelines on adjuvant chemotherapy and results of randomized phase III trials. The aim of this review is to describe the state of the art in this setting and to highlight future possible perspectives. RECENT FINDINGS Resectable PDAC is the tumor without vascular contact or a limited venous contact without vein irregularity. Several pathologic and biologic robust prognostic factors such as an R0 resection defined by a margin at least 1 mm have been validated. In phase III trials, the doublet gemcitabine-capecitabine provided a statistically significant, albeit modest overall survival benefit, but failed to show an improvement in relapse-free survival. Similarly, gemcitabine plus nab-paclitaxel did not increase disease-free survival. Modified FOLFIRINOX led to improved disease-free survival, overall survival, and metastasis-free survival, with acceptable toxicity. In the future, prognostic and/or predictive biomarkers could lead the optimization of therapeutic strategies and neoadjuvant treatment could become a standard of care in PDAC. SUMMARY After curative intent resection, modified FOLFIRINOX is the standard of care in adjuvant in fit patients with PDAC. Others regimens (monotherapy or gemcitabine-based) are an option in unfit patients.
Collapse
Affiliation(s)
- Léo Mas
- Department of Hepato-gastroenterology, Groupe Hospitalier Pitié Salpêtrière, Paris
| | - Lilian Schwarz
- Department of Digestive Surgery, Rouen University Hospital
- Department of Genomic and Personalized Medicine in Cancer and Neurological Disorders, Normandie University, UNIROUEN, UMR 1245 INSERM, Rouen University Hospital, Rouen
| | - Jean-Baptiste Bachet
- Department of Hepato-gastroenterology, Groupe Hospitalier Pitié Salpêtrière, Paris
- Sorbonne University, UPMC University, Paris, France
| |
Collapse
|
11
|
Guo X, Li K, Jiang W, Hu Y, Xiao W, Huang Y, Feng Y, Pan Q, Wan R. RNA demethylase ALKBH5 prevents pancreatic cancer progression by posttranscriptional activation of PER1 in an m6A-YTHDF2-dependent manner. Mol Cancer 2020; 19:91. [PMID: 32429928 PMCID: PMC7236181 DOI: 10.1186/s12943-020-01158-w] [Citation(s) in RCA: 247] [Impact Index Per Article: 49.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 02/13/2020] [Indexed: 12/31/2022] Open
Abstract
Background N6-methyladenosine (m6A) is the most abundant reversible methylation modification of eukaryotic mRNA, and it plays vital roles in tumourigenesis. This study aimed to explore the role of the m6A demethylase ALKBH5 in pancreatic cancer (PC). Methods The expression of ALKBH5 and its clinicopathological impact were evaluated in PC cohorts. The effects of ALKBH5 on the biological characteristics of PC cells were investigated on the basis of gain-of-function and loss-of-function analyses. Subcutaneous and orthotopic models further uncovered the role of ALKBH5 in tumour growth. mRNA and m6A sequencing and assays of m6A methylated RNA immunoprecipitation-qPCR (MeRIP-qPCR) were performed to identify the targeted effect of ALKBH5 on PER1. P53-binding sites in the ALKBH5 promoter were investigated by ChIP and luciferase assays to reveal the interplay between ALKBH5 and PER1-activated ATM-CHK2-P53/CDC25C signalling. Results ALKBH5 loss characterized the occurrence and poor clinicopathological manifestations in patients with PC. Overexpression of ALKBH5 reduced tumoural proliferative, migrative, invasive activities in vitro and ameliorated tumour growth in vivo, whereas ALKBH5 knockdown facilitated PC progression. Mechanistically, ALKBH5 posttranscriptionally activated PER1 by m6A demethylation in an m6A-YTHDF2-dependent manner. PER1 upregulation led to the reactivation of ATM-CHK2-P53/CDC25C signalling, which inhibited cell growth. P53-induced activation of ALKBH5 transcription acted as a feedback loop regulating the m6A modifications in PC. Conclusion ALKBH5 serves as a PC suppressor by regulating the posttranscriptional activation of PER1 through m6A abolishment, which may highlight a demethylation-based approach for PC diagnosis and therapy.
Collapse
Affiliation(s)
- Xingya Guo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China
| | - Kai Li
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China
| | - Weiliang Jiang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China
| | - Yangyang Hu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China
| | - Wenqin Xiao
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China
| | - Yinshi Huang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China
| | - Yun Feng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China
| | - Qin Pan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665, Kongjiang Road, Shanghai, 200092, People's Republic of China.
| | - Rong Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, No. 100, Haining Road, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
12
|
MIF inhibitor, ISO-1, attenuates human pancreatic cancer cell proliferation, migration and invasion in vitro, and suppresses xenograft tumour growth in vivo. Sci Rep 2020; 10:6741. [PMID: 32317702 PMCID: PMC7174354 DOI: 10.1038/s41598-020-63778-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/06/2020] [Indexed: 02/07/2023] Open
Abstract
This study sought to investigate the biological effects of specific MIF inhibitor, ISO-1, on the proliferation, migration and invasion of PANC-1 human pancreatic cells in vitro, and on tumour growth in a xenograft tumour model in vivo. The effect of ISO-1 on PANC-1 cell proliferation was examined using CCK-8 cell proliferation assay. The effect of ISO-1 on collective cell migration and recolonization of PANC-1 cells was evaluated using the cell-wound closure migration assay. The effect of ISO-1 on the migration and invasion of individual PANC-1 cells in a 3-dimensional environment in response to a chemo-attractant was investigated through the use of Transwell migration/invasion assays. Quantitative real time PCR and western blot analyses were employed to investigate the effects of ISO-1 on MIF, NF-κB p65 and TNF-α mRNA and protein expression respectively. Finally, a xenograft tumor model in BALB/c nude mice were used to assess the in vivo effects of ISO-1 on PANC-1-induced tumor growth. We found high expression of MIF in pancreatic cancer tissues. We demonstrated that ISO-1 exerts anti-cancer effects on PANC-1 cell proliferation, migration and invasion in vitro, and inhibited PANC-1 cell-induced tumour growth in xenograft mice in vivo. Our data suggests that ISO-1 and its derivative may have potential therapeutic applications in pancreatic cancer.
Collapse
|
13
|
Okamura Y, Yasukawa S, Narimatsu H, Boku N, Fukutomi A, Konishi M, Morinaga S, Toyama H, Kaneoka Y, Shimizu Y, Nakamori S, Sata N, Yamakita K, Takahashi A, Kainuma O, Hishinuma S, Yamaguchi R, Nagino M, Hirano S, Yanagisawa A, Mori K, Uesaka K. Human equilibrative nucleoside transporter-1 expression is a predictor in patients with resected pancreatic cancer treated with adjuvant S-1 chemotherapy. Cancer Sci 2020; 111:548-560. [PMID: 31778273 PMCID: PMC7004513 DOI: 10.1111/cas.14258] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/20/2019] [Accepted: 11/22/2019] [Indexed: 02/06/2023] Open
Abstract
The high expression of human equilibrative nucleoside transporter-1 (hENT1) and the low expression of dihydropyrimidine dehydrogenase (DPD) are reported to predict a favorable prognosis in patients treated with gemcitabine (GEM) and 5-fluorouracil (5FU) as the adjuvant setting, respectively. The expression of hENT1 and DPD were analyzed in patients registered in the JASPAC 01 trial, which showed a better survival of S-1 over GEM as adjuvant chemotherapy after resection for pancreatic cancer, and their possible roles for predicting treatment outcomes and selecting a chemotherapeutic agent were investigated. Intensity of hENT1 and DPD expression was categorized into no, weak, moderate or strong by immunohistochemistry staining, and the patients were classified into high (strong/moderate) and low (no/weak) groups. Specimens were available for 326 of 377 (86.5%) patients. High expression of hENT1 and DPD was detected in 100 (30.7%) and 63 (19.3%) of 326 patients, respectively. In the S-1 arm, the median overall survival (OS) with low hENT1, 58.0 months, was significantly better than that with high hENT1, 30.9 months (hazard ratio 1.75, P = 0.007). In contrast, there were no significant differences in OS between DPD low and high groups in the S-1 arm and neither the expression levels of hENT1 nor DPD revealed a relationship with treatment outcomes in the GEM arm. The present study did not show that the DPD and hENT1 are useful biomarkers for choosing S-1 or GEM as adjuvant chemotherapy. However, hENT1 expression is a significant prognostic factor for survival in the S-1 arm.
Collapse
Affiliation(s)
- Yukiyasu Okamura
- Division of Hepato‐Biliary‐Pancreatic SurgeryShizuoka Cancer Center HospitalNagaizumiJapan
| | | | - Hiroto Narimatsu
- Cancer Prevention and Control DivisionKanagawa Cancer CenterYokohamaJapan
| | - Narikazu Boku
- Gastrointestinal Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Akira Fukutomi
- Gastrointestinal OncologyShizuoka Cancer CenterShizuokaJapan
| | - Masaru Konishi
- Hepato‐Biliary‐Pancreatic SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Soichiro Morinaga
- Department of Gastrointestinal SurgeryKanagawa Cancer CenterYokohamaJapan
| | | | | | | | - Shoji Nakamori
- SurgeryNational Hospital Organization Osaka National HospitalOsakaJapan
| | - Naohiro Sata
- Gastrointestinal SurgeryJichi Medical UniversityShimotsukeJapan
| | - Keisuke Yamakita
- Division of Metabolism and Biosystemic ScienceDepartment of MedicineAsahikawa Medical UniversityAsahikawaJapan
| | | | - Osamu Kainuma
- Gastrointestinal SurgeryChiba Cancer CenterChibaJapan
| | | | | | - Masato Nagino
- Division of Surgical OncologyDepartment of SurgeryNagoya University Graduate of School of MedicineNagoyaJapan
| | - Satoshi Hirano
- Gastroenterological Surgery IIFaculty of MedicineHokkaido UniversitySapporoJapan
| | | | - Keita Mori
- Clinical Trial Coordination Office BiostatisticianShizuoka Cancer Center HospitalNagaizumiJapan
| | - Katsuhiko Uesaka
- Division of Hepato‐Biliary‐Pancreatic SurgeryShizuoka Cancer Center HospitalNagaizumiJapan
| |
Collapse
|
14
|
Ding G, Shen T, Yan C, Zhang M, Wu Z, Cao L. IFN-γ down-regulates the PD-1 expression and assist nivolumab in PD-1-blockade effect on CD8+ T-lymphocytes in pancreatic cancer. BMC Cancer 2019; 19:1053. [PMID: 31694582 PMCID: PMC6836337 DOI: 10.1186/s12885-019-6145-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 09/10/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pancreatic cancer is characterized by a highly immunosuppressive tumor microenvironment and evasion of immune surveillance. Although programmed cell death 1 receptor (PD-1) blockade has achieved certain success in immunogenic cancers, the responses to the PD-1 antibody are not effective or sustained in patients with pancreatic cancer. METHODS Firstly, PD-1 expressions on peripheral CD8+ T-lymphocytes of patients with pancreatic cancer and healthy donors were measured. In in vitro study, peripheral T-lymphocytes were isolated and treated with nivolumab and/or interferon-γ, and next, PD-1-blockade effects, proliferations, cytokine secretions and cytotoxic activities were tested after different treatments. In in vivo study, mice bearing subcutaneous pancreatic cancer cell lines were treated with induced T-lymphocytes and tumor sizes were measured. RESULTS PD-1 protein expression is increased on peripheral CD8+ T cells in patients with pancreatic ductal adenocarcinoma compared with that in health donor. PD-1 expression on CD8+ T-lymphocytes was decreased by nivolumab in a concentration-dependent manner in vitro. IFN-γ could directly down-regulate expression of PD-1 in vitro. Furthermore, the combination therapy of nivolumab and IFN-γ resulted in greatest effect of PD-1-blockde (1.73 ± 0.78), compared with IFN-γ along (18.63 ± 0.82) and nivolumab along (13.65 ± 1.22). Moreover, the effects of nivolumab plus IFN-γ largest promoted the T-lymphocytes function of proliferations, cytokine secretions and cytotoxic activities. Most importantly, T-lymphocytes induced by nivolumab plus IFN-γ presented the best repression of tumor growth. CONCLUSIONS IFN-γ plus a PD-1-blockading agent could enhance the immunologic function and might play a crucial role in effective adoptive transfer treatments of pancreatic cancer.
Collapse
Affiliation(s)
- Guoping Ding
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Tao Shen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Chen Yan
- Department of General Surgery, Zhejiang University Huzhou hospital (Huzhou central hospital), Huzhou, 313000, China
| | - Mingjie Zhang
- Department of General Surgery, Zhejiang University Huzhou hospital (Huzhou central hospital), Huzhou, 313000, China
| | - Zhengrong Wu
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China. .,Innovation Center for Minimally Invasive Technique and Device, Zhejiang University, Hangzhou, China.
| |
Collapse
|
15
|
Estándares de calidad en la cirugía oncológica pancreática en España. Cir Esp 2018; 96:342-351. [PMID: 29784432 DOI: 10.1016/j.ciresp.2018.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/02/2018] [Accepted: 03/06/2018] [Indexed: 12/18/2022]
|
16
|
Park JK, Kim Y, Kim H, Jeon J, Kim TW, Park JH, Hwnag YI, Lee WJ, Kang JS. The anti-fibrotic effect of GV1001 combined with gemcitabine on treatment of pancreatic ductal adenocarcinoma. Oncotarget 2018; 7:75081-75093. [PMID: 27655706 PMCID: PMC5342724 DOI: 10.18632/oncotarget.12057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 08/23/2016] [Indexed: 02/07/2023] Open
Abstract
GV1001 is a telomerase-based cancer vaccine made of a 16-mer telomerase reverse transcriptase (TERT) peptide, and human TERT, the rate-limiting subunit of the telomerase complex, is an attractive target for cancer vaccination. The aim of this study was to evaluate the effect of telomerase peptide vaccination, GV1001 combined with gemcitabine in treatment of pancreatic ductal adenocarcinoma (PDAC). Human PDAC cell lines were used in vitro experiment and also, PDAC xenograft mice model was established using PANC1, AsPC1 and CD133+ AsPC1 (PDAC stem cell). Treatment groups were divided as follows; control, gemcitabine, GV1001, gemcitabine and GV1001 combination. The inflammatory cytokines were measured from the blood, and xenograft tumor specimens were evaluated. GV1001 treatment alone did not affect the proliferation or the apoptosis of PDAC cells. Gemcitabine alone and gemcitabine with GV1001 groups had significantly reduced in tumor size and showed abundant apoptosis compared to other treatment groups. Surprisingly, xenograft PDAC tumor specimens of gemcitabine alone group had been replaced by severe fibrosis whereas gemcitabine with GV1001 group had significantly less fibrosis. Blood levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β increased in gemcitabine alone group, however, it was decreased in gemcitabine with GV1001 group. GV1001 combined with gemcitabine treatment showed significant loss of fibrosis in tumor tissue as well as tumor cell death. Therefore, further investigation of GV1001 effect combined with gemcitabine treatment may give us useful insights to overcome the hurdle in anti-cancer drug delivery over massive fibrosis around PDACs.
Collapse
Affiliation(s)
- Joo Kyung Park
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yejin Kim
- Laboratory of Vitamin C and Anti-Oxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hyemin Kim
- Laboratory of Vitamin C and Anti-Oxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| | - Jane Jeon
- Laboratory of Vitamin C and Anti-Oxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Tae Wan Kim
- Department of Ophthalmology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul, Korea
| | - Ji-Hong Park
- Department of Rehabilitation Medicine, Seoul National University Bundang Hospital, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Young-Il Hwnag
- Laboratory of Vitamin C and Anti-Oxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Wang Jae Lee
- Laboratory of Vitamin C and Anti-Oxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jae Seung Kang
- Laboratory of Vitamin C and Anti-Oxidant Immunology, Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea
| |
Collapse
|
17
|
Lee W, Yoon YS, Han HS, Jang JY, Cho JY, Jung W, Kwon W, Choi Y, Kim SW. Prognostic Relevance of the Timing of Initiating and the Completion of Adjuvant Therapy in Patients with Resected Pancreatic Ductal Adenocarcinoma. World J Surg 2017; 41:562-573. [PMID: 27834017 DOI: 10.1007/s00268-016-3798-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Although the role of adjuvant therapy in patients with pancreatic ductal adenocarcinoma (PDAC) is well established, its optimal timing and duration are still controversial. METHODS The study included 311 patients with PDAC who underwent curative resection followed by adjuvant therapy. We analyzed survival data according to the timing of initiation and completion of adjuvant therapy. RESULTS There were no differences in 5-year overall survival (OS) (32.8 vs. 35.4%, p = 0.539) and disease-free survival (DFS) rates (26.2 vs. 23.3%, p = 0.865) between early (≤6 weeks) and late (>6 weeks) initiation of adjuvant therapy. However, the 5-year OS (42.6 vs. 22.2%, p < 0.001) and DFS (29.2 vs. 18.4%, p = 0.042) rates were significantly greater in patients with complete versus incomplete adjuvant therapy. Multivariable analysis revealed that incomplete adjuvant therapy was an independent prognostic factor for decreased OS (p = 0.001; hazard ratio 1.850; 95% confidence interval 1.266-2.702). CONCLUSIONS The results show that complete adjuvant therapy is a more important prognostic factor than early initiation for improving the survival of patients with resected PDAC.
Collapse
Affiliation(s)
- Woohyung Lee
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 166 Gumi-ro, Bundang-gu, Seongnam-Si, Gyeonggi-do, 463-707, Republic of Korea.,Department of Surgery, Gyeongsang National University College of Medicine, Gyeongsang National University Changwon Hospital, Changwon, Republic of Korea
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 166 Gumi-ro, Bundang-gu, Seongnam-Si, Gyeonggi-do, 463-707, Republic of Korea.
| | - Ho-Seong Han
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 166 Gumi-ro, Bundang-gu, Seongnam-Si, Gyeonggi-do, 463-707, Republic of Korea
| | - Jin Young Jang
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jai Young Cho
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 166 Gumi-ro, Bundang-gu, Seongnam-Si, Gyeonggi-do, 463-707, Republic of Korea
| | - Woohyun Jung
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Wooil Kwon
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - YoungRok Choi
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, 166 Gumi-ro, Bundang-gu, Seongnam-Si, Gyeonggi-do, 463-707, Republic of Korea
| | - Sun-Whe Kim
- Department of Surgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
18
|
Tang D, Zhang J, Yuan Z, Zhang H, Chong Y, Huang Y, Wang J, Xiong Q, Wang S, Wu Q, Tian Y, Lu Y, Ge X, Shen W, Wang D. PSC-derived Galectin-1 inducing epithelial-mesenchymal transition of pancreatic ductal adenocarcinoma cells by activating the NF-κB pathway. Oncotarget 2017; 8:86488-86502. [PMID: 29156810 PMCID: PMC5689700 DOI: 10.18632/oncotarget.21212] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023] Open
Abstract
Galectin-1 has previously been shown to be strongly expressed in activated pancreatic stellate cells (PSCs) and promote the development and metastasis of pancreatic ductal adenocarcinoma (PDAC). However, the molecular mechanisms by which Galectin-1 promotes the malignant behavior of pancreatic cancer cells remain unclear. In this study, we examined the effects of Galectin-1 knockdown or overexpression in PSCs co-cultured with pancreatic cancer (PANC-1) cells. Immunohistochemical analysis showed expression of epithelial-mesenchymal transition (EMT) markers and MMP9 were positively associated with the expression of Galectin-1 in 66 human PDAC tissues. In addition, our in vitro studies showed PSC-derived Galectin-1 promoted the proliferation, invasion, and survival (anti-apoptotic effects) of PANC-1 cells. We also showed PSC-derived Galectin-1 induced EMT of PANC-1 cells and activated the NF-кB pathway in vitro. Our mixed (PSCs and PANC-1 cells) mouse orthotopic xenograft model indicated that overexpression of Galectin-1 in PSCs significantly promoted the proliferation, growth, invasion, and liver metastasis of the transplanted tumor. Moreover, Galectin-1 overexpression in PSCs was strongly associated with increased expression of EMT markers in both the orthotopic xenograft tumor in the pancreas and in metastatic lesions of naked mice. We conclude that PSC-derived Galectin-1 promotes the malignant behavior of PDAC by inducing EMT via activation of the NF-κB pathway. Our results suggest that targeting Galectin-1 in PSCs could represent a promising therapeutic strategy for PDAC progression and metastasis.
Collapse
Affiliation(s)
- Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Jingqiu Zhang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Zhongxu Yuan
- Department of General Surgery, Anhui No. 2 Provincial People’s Hospital, Hefei, Anhui Province, P.R. China
| | - Hongpeng Zhang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Yang Chong
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Yuqin Huang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Jie Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Qingquan Xiong
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| | - Sen Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Qi Wu
- Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Ying Tian
- Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Yongdie Lu
- Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Xiao Ge
- Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Wenjing Shen
- Department of Clinical Medicine, Medical College of Yangzhou University, Yangzhou, P.R. China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu Province Hospital, Clinical Medical College, Yangzhou University, Yangzhou, P.R. China
| |
Collapse
|
19
|
Shelper TB, Lovitt CJ, Avery VM. Assessing Drug Efficacy in a Miniaturized Pancreatic Cancer In Vitro 3D Cell Culture Model. Assay Drug Dev Technol 2017; 14:367-80. [PMID: 27552143 DOI: 10.1089/adt.2016.737] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Pancreatic cancer continues to have one of the poorest prognoses among all cancers. The drug discovery efforts for this disease have largely failed, with no significant improvement in survival outcomes for advanced pancreatic cancer patients over the past 20 years. Traditional in vitro cell culture techniques have been used extensively in both basic and early drug discovery; however, these systems offer poor models to assess emerging therapeutics. More predictive cell-based models, which better capture the cellular heterogeneity and complexities of solid pancreatic tumors, are urgently needed not only to improve drug discovery success but also to provide insight into the tumor biology. Pancreatic tumors are characterized by a unique micro-environment that is surrounded by a dense stroma. A complex network of interactions between extracellular matrix (ECM) components and the effects of cell-to-cell contacts may enhance survival pathways within in vivo tumors. This biological and physical complexity is lost in traditional cell monolayer models. To explore the predictive potential of a more complex cellular system, a three-dimensional (3D) micro-tumor assay was evaluated. Efficacy of six current chemotherapeutics was determined against a panel of primary and metastatic pancreatic tumor cell lines in a miniaturized ECM-based 3D cell culture system. Suitability for potential use in high-throughput screening applications was assessed, including ascertaining the effects that miniaturization and automation had on assay robustness. Cellular health was determined by utilizing an indirect population-based metabolic activity assay and a direct imaging-based cell viability assay.
Collapse
Affiliation(s)
- Todd B Shelper
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University , Nathan, Australia
| | - Carrie J Lovitt
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University , Nathan, Australia
| | - Vicky M Avery
- Discovery Biology, Eskitis Institute for Drug Discovery, Griffith University , Nathan, Australia
| |
Collapse
|
20
|
Shen T, Zhou L, Shen H, Shi C, Jia S, Ding GP, Cao L. Prognostic value of programmed cell death protein 1 expression on CD8+ T lymphocytes in pancreatic cancer. Sci Rep 2017; 7:7848. [PMID: 28798308 PMCID: PMC5552822 DOI: 10.1038/s41598-017-08479-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 07/11/2017] [Indexed: 12/14/2022] Open
Abstract
Pancreatic cancer is one of the most aggressive malignancies and has a highly immunosuppressive tumour microenvironment. Immune checkpoint blockade has led to remarkable and durable objective responses in a number of malignancies and antibody-based strategies targeting programmed cell death protein 1 (PD-1) are showing promise where traditional modalities of surgery, radiotherapy, and chemotherapy have failed. In this study, we examined the clinical value of PD-1 protein expression by CD8+ peripheral T lymphocytes or tumour-infiltrating T lymphocytes (TILs) in pancreatic ductal adenocarcinoma (PDAC). Expression of PD-1 protein on CD8+ TILs correlated with overall survival and clinicopathological characteristics such as clinical stage, N classification, and M classification. Similar findings were observed for the expression of PD-1 protein on peripheral CD8+ T cells, whereas its expression on peripheral CD4+ T cells showed no significance. Comparison of the levels of PD-1 protein expressed by peripheral CD8+ T cells before and 4 weeks after surgery indicated that preoperative and postoperative status of peripheral PD-1 expression was unchanged. Our findings showed that PD-1 protein expressed by peripheral or tumour-infiltrated CD8+ T cells was a promising biomarker for diagnosis and prognosis in PDAC and might help guide future immunotherapies.
Collapse
Affiliation(s)
- Tao Shen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Liangjing Zhou
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Hua Shen
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Chengfei Shi
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Shengnan Jia
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China
| | - Guo Ping Ding
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000, China.
| |
Collapse
|
21
|
Akasaka H, Mizushina Y, Yoshida K, Ejima Y, Mukumoto N, Wang T, Inubushi S, Nakayama M, Wakahara Y, Sasaki R. MGDG extracted from spinach enhances the cytotoxicity of radiation in pancreatic cancer cells. Radiat Oncol 2016; 11:153. [PMID: 27876069 PMCID: PMC5120455 DOI: 10.1186/s13014-016-0729-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/15/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND In our previous study, monogalactosyl diacylglycerol (MGDG) purified from spinach was found to have cytotoxic effects in human cancer cell lines. This study further assessed whether MGDG can enhance the cytotoxic effects of radiation in human pancreatic cancer cells in vitro and in vivo. METHODS Glycoglycerolipids from spinach including MGDG were extracted from dried spinach. The cytotoxicity of MGDG were evaluated by the MTT assay using four human pancreatic cancer cell lines (MIAPaCa-2, AsPC-1, BxPC-3 and PANC-1) and normal human dermal fibroblasts (NHDFs). The effects of radiation and MGDG alone or in combination in MIAPaCa-2 cells was analyzed with the colony forming and apoptosis assays, western blotting and cell cycle and DNA damage analyses (γ-H2AX foci staining and comet assay). The inhibitory effects on tumor growth were assessed in a mouse xenograft tumor model. RESULTS MGDG showed dose- and time-dependent cytotoxicity, with half-maximal inhibitory concentrations (IC50) in PANC-1, BxPC-3, MIAPaCa-2 and AsPC-1 cells at 72 h of 25.6 ± 2.5, 26.9 ± 1.3, 18.5 ± 1.7, and 22.7 ± 1.9 μM, respectively. The colony forming assay revealed fewer MIAPaCa-2, BxPC-3 and AsPC-1 cell colonies upon treatment with both MGDG and radiation as compared to irradiation alone (P < 0.05). The combination of MGDG and radiation induced a higher proportion of apoptosis in MIAPaCa-2 cells; this effect was associated with increased mitochondrial release of cytochrome c and activation of cleaved poly (ADP-ribose) polymerase and caspase-3. DNA damage was detected and DNA repair mechanisms were more frequently impaired in cells receiving the combination treatment as compared to either one alone. Tumor growth was inhibited to a greater degree in mice treated by intratumoral injection of MGDG combined with irradiation as compared to either one alone (P < 0.05). CONCLUSIONS This is the first report demonstrating that MGDG enhances the cytotoxicity of radiation to induce apoptosis of cancer cells in vitro and in vivo. Our findings indicate that this therapeutic combination can be an effective strategy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hiroaki Akasaka
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Yoshiyuki Mizushina
- Graduate School of Agriculture, Shinshu University, Minamiminowa-mura, Kamiina-gun, Nagano, 399-4598, Japan
| | - Kenji Yoshida
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Yasuo Ejima
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Naritoshi Mukumoto
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Tianyuan Wang
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Sachiko Inubushi
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Masao Nakayama
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Yuki Wakahara
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| |
Collapse
|
22
|
Feasibility of Immunohistochemistry on Endoscopic Ultrasound Fine-Needle Aspiration Samples for Evaluating Predictive Biomarkers in Pancreatic Cancer Management. Pancreas 2016; 45:e50-2. [PMID: 27623560 DOI: 10.1097/mpa.0000000000000672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
23
|
Botla SK, Savant S, Jandaghi P, Bauer AS, Mücke O, Moskalev EA, Neoptolemos JP, Costello E, Greenhalf W, Scarpa A, Gaida MM, Büchler MW, Strobel O, Hackert T, Giese NA, Augustin HG, Hoheisel JD. Early Epigenetic Downregulation of microRNA-192 Expression Promotes Pancreatic Cancer Progression. Cancer Res 2016; 76:4149-59. [PMID: 27216198 DOI: 10.1158/0008-5472.can-15-0390] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 04/22/2016] [Indexed: 12/27/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by very early metastasis, suggesting the hypothesis that metastasis-associated changes may occur prior to actual tumor formation. In this study, we identified miR-192 as an epigenetically regulated suppressor gene with predictive value in this disease. miR-192 was downregulated by promoter methylation in both PDAC and chronic pancreatitis, the latter of which is a major risk factor for the development of PDAC. Functional studies in vitro and in vivo in mouse models of PDAC showed that overexpression of miR-192 was sufficient to reduce cell proliferation and invasion. Mechanistic analyses correlated changes in miR-192 promoter methylation and expression with epithelial-mesenchymal transition. Cell proliferation and invasion were linked to altered expression of the miR-192 target gene SERPINE1 that is encoding the protein plasminogen activator inhibitor-1 (PAI-1), an established regulator of these properties in PDAC cells. Notably, our data suggested that invasive capacity was altered even before neoplastic transformation occurred, as triggered by miR-192 downregulation. Overall, our results highlighted a role for miR-192 in explaining the early metastatic behavior of PDAC and suggested its relevance as a target to develop for early diagnostics and therapy. Cancer Res; 76(14); 4149-59. ©2016 AACR.
Collapse
Affiliation(s)
- Sandeep K Botla
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Soniya Savant
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faulty Mannheim, Heidelberg University, Mannheim, Germany
| | - Pouria Jandaghi
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrea S Bauer
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Oliver Mücke
- Division of Epigenomics and Cancer Risk Factors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Evgeny A Moskalev
- Diagnostic Molecular Pathology, Institute of Pathology, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - John P Neoptolemos
- National Institute for Health Research, Liverpool Pancreas Biomedical Research Unit, Liverpool, UK
| | - Eithne Costello
- National Institute for Health Research, Liverpool Pancreas Biomedical Research Unit, Liverpool, UK
| | - William Greenhalf
- National Institute for Health Research, Liverpool Pancreas Biomedical Research Unit, Liverpool, UK
| | - Aldo Scarpa
- Department of Pathology and Diagnostics, Università di Verona, Verona, Italy
| | - Matthias M Gaida
- Department of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Markus W Büchler
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Oliver Strobel
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Thilo Hackert
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Nathalia A Giese
- Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ), Heidelberg, Germany. Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faulty Mannheim, Heidelberg University, Mannheim, Germany. German Cancer Consortium, Heidelberg, Germany
| | - Jörg D Hoheisel
- Division of Functional Genome Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
24
|
Abstract
Pancreatic cancer is a highly lethal disease, for which mortality closely parallels incidence. Most patients with pancreatic cancer remain asymptomatic until the disease reaches an advanced stage. There is no standard programme for screening patients at high risk of pancreatic cancer (eg, those with a family history of pancreatic cancer and chronic pancreatitis). Most pancreatic cancers arise from microscopic non-invasive epithelial proliferations within the pancreatic ducts, referred to as pancreatic intraepithelial neoplasias. There are four major driver genes for pancreatic cancer: KRAS, CDKN2A, TP53, and SMAD4. KRAS mutation and alterations in CDKN2A are early events in pancreatic tumorigenesis. Endoscopic ultrasonography and endoscopic ultrasonography-guided fine-needle aspiration offer high diagnostic ability for pancreatic cancer. Surgical resection is regarded as the only potentially curative treatment, and adjuvant chemotherapy with gemcitabine or S-1, an oral fluoropyrimidine derivative, is given after surgery. FOLFIRINOX (fluorouracil, folinic acid [leucovorin], irinotecan, and oxaliplatin) and gemcitabine plus nanoparticle albumin-bound paclitaxel (nab-paclitaxel) are the treatments of choice for patients who are not surgical candidates but have good performance status.
Collapse
MESH Headings
- Albumins/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- CA-19-9 Antigen/metabolism
- Camptothecin/administration & dosage
- Camptothecin/analogs & derivatives
- Carcinoembryonic Antigen/metabolism
- Carcinoma, Pancreatic Ductal/diagnosis
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/therapy
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Endoscopic Ultrasound-Guided Fine Needle Aspiration
- Endosonography
- Fluorouracil/administration & dosage
- Genes, p16
- Humans
- Irinotecan
- Leucovorin/administration & dosage
- Neoplasms, Cystic, Mucinous, and Serous/diagnosis
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/therapy
- Organoplatinum Compounds/administration & dosage
- Oxaliplatin
- Paclitaxel/administration & dosage
- Pancreatectomy
- Pancreatic Neoplasms/diagnosis
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/therapy
- Proto-Oncogene Proteins p21(ras)/genetics
- Smad4 Protein/genetics
- Tumor Suppressor Protein p53/genetics
- Gemcitabine
Collapse
Affiliation(s)
- Terumi Kamisawa
- Department of Internal Medicine, Tokyo Metropolitan Komagome Hospital, Tokyo, Japan.
| | - Laura D Wood
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, Johns Hopkins University, Baltimore, USA
| | - Takao Itoi
- Department of Gastroenterology and Hepatology, Tokyo Medical University, Tokyo, Japan
| | - Kyoichi Takaori
- Division of Hepatobiliary-Pancreatic Surgery and Transplantation, Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
25
|
Abstract
OBJECTIVES Structured follow-up after surgery for pancreatic ductal adenocarcinoma (PDAC) remains controversial and is currently not recommended due to a supposed lack of therapeutic consequences. Furthermore, it is not clear whether noncancer patients after pancreas resection need to be seen in the clinic on a regular basis. The present study analyzed how follow-up after pancreatic surgery affected postoperative treatment and long-term outcomes. METHODS Data of all postoperative visits in a specialized outpatient clinic for pancreatic diseases were analyzed for a 1-year period with regard to symptoms, diagnostic procedures, and therapeutic consequences. RESULTS Six hundred eighteen patients underwent 940 postoperative follow-ups. Nearly half of them needed a change of medication due to altered pancreatic function. In 74 (40%) of 184 resected PDAC patients, recurrence (local or systemic) was detected during follow-up, although only 19 of these had shown associated symptoms (26%). In all patients with recurrence, a cancer-directed treatment was induced. Eleven (69%) of 16 patients with isolated local recurrence were referred for reresection. CONCLUSIONS Follow-up examinations are a substantial part of the clinical management after pancreas resections. Follow-up is particularly important for PDAC because recurrence is often asymptomatic, but its detection allows for therapeutic interventions and potentially improved prognosis. This should be implemented in future guidelines.
Collapse
|
26
|
Versteijne E, van Eijck CHJ, Punt CJA, Suker M, Zwinderman AH, Dohmen MAC, Groothuis KBC, Busch ORC, Besselink MGH, de Hingh IHJT, Ten Tije AJ, Patijn GA, Bonsing BA, de Vos-Geelen J, Klaase JM, Festen S, Boerma D, Erdmann JI, Molenaar IQ, van der Harst E, van der Kolk MB, Rasch CRN, van Tienhoven G. Preoperative radiochemotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer (PREOPANC trial): study protocol for a multicentre randomized controlled trial. Trials 2016; 17:127. [PMID: 26955809 PMCID: PMC4784417 DOI: 10.1186/s13063-016-1262-z] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 02/26/2016] [Indexed: 12/20/2022] Open
Abstract
Background Pancreatic cancer is the fourth largest cause of cancer death in the United States and Europe with over 100,000 deaths per year in Europe alone. The overall 5-year survival ranges from 2–7 % and has hardly improved over the last two decades. Approximately 15 % of all patients have resectable disease at diagnosis, and of those, only a subgroup has a resectable tumour at surgical exploration. Data from cohort studies have suggested that outcome can be improved by preoperative radiochemotherapy, but data from well-designed randomized studies are lacking. Our PREOPANC phase III trial aims to test the hypothesis that median overall survival of patients with resectable or borderline resectable pancreatic cancer can be improved with preoperative radiochemotherapy. Methods/design The PREOPANC trial is a randomized, controlled, multicentric superiority trial, initiated by the Dutch Pancreatic Cancer Group. Patients with (borderline) resectable pancreatic cancer are randomized to A: direct explorative laparotomy or B: after negative diagnostic laparoscopy, preoperative radiochemotherapy, followed by explorative laparotomy. A hypofractionated radiation scheme of 15 fractions of 2.4 gray (Gy) is combined with a course of gemcitabine, 1,000 mg/m2/dose on days 1, 8 and 15, preceded and followed by a modified course of gemcitabine. The target volumes of radiation are delineated on a 4D CT scan, where at least 95 % of the prescribed dose of 36 Gy in 15 fractions should cover 98 % of the planning target volume. Standard adjuvant chemotherapy is administered in both treatment arms after resection (six cycles in arm A and four in arm B). In total, 244 patients will be randomized in 17 hospitals in the Netherlands. The primary endpoint is overall survival by intention to treat. Secondary endpoints are (R0) resection rate, disease-free survival, time to locoregional recurrence or distant metastases and perioperative complications. Secondary endpoints for the experimental arm are toxicity and radiologic and pathologic response. Discussion The PREOPANC trial is designed to investigate whether preoperative radiochemotherapy improves overall survival by means of increased (R0) resection rates in patients with resectable or borderline resectable pancreatic cancer. Trial registration Trial open for accrual: 3 April 2013 The Netherlands National Trial Register – NTR3709 (8 November 2012) EU Clinical Trials Register – 2012-003181-40 (11 December 2012)
Collapse
Affiliation(s)
- Eva Versteijne
- Department of Radiation Oncology, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Casper H J van Eijck
- Department of Surgery, Erasmus Medical Center, Postbus 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Cornelis J A Punt
- Department of Medical Oncology, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Mustafa Suker
- Department of Surgery, Erasmus Medical Center, Postbus 2040, 3000 CA, Rotterdam, The Netherlands.
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiologic Biostatics, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Miriam A C Dohmen
- Clinical Research Department, Comprehensive Cancer Organisation the Netherlands (IKNL), Postbus 1281, 6501 BG, Nijmegen, The Netherlands.
| | - Karin B C Groothuis
- Clinical Research Department, Comprehensive Cancer Organisation the Netherlands (IKNL), Postbus 1281, 6501 BG, Nijmegen, The Netherlands.
| | - Oliver R C Busch
- Department of Surgery, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Marc G H Besselink
- Department of Surgery, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Ignace H J T de Hingh
- Department of Surgery, Catharina Hospital, Postbus 1350, 5602 ZA, Eindhoven, The Netherlands.
| | - Albert J Ten Tije
- Department of Medical Oncology, Amphia Hospital, Postbus 90158, 4800 RK, Breda, The Netherlands.
| | - Gijs A Patijn
- Department of Surgery, Isala Clinics, Postbus 10400, 8000 GK, Zwolle, The Netherlands.
| | - Bert A Bonsing
- Department of Surgery, Leiden University Medical Center, Postbus 9600, 2300 RC, Leiden, The Netherlands.
| | - Judith de Vos-Geelen
- Department of Medical Oncology, Maastricht University Medical Center, Postbus 3035, 6202 NA, Maastricht, The Netherlands.
| | - Joost M Klaase
- Department of Surgery, Medical Spectrum Twente, Postbus 50 000, 7500 KA, Enschede, The Netherlands.
| | - Sebastiaan Festen
- Department of Surgery, Onze Lieve Vrouwe Gasthuis, Postbus 95500, 1090 HM, Amsterdam, The Netherlands.
| | - Djamila Boerma
- Department of Surgery, Sint Antonius Hospital, Postbus 2500, 3430 EM, Nieuwegein, The Netherlands.
| | - Joris I Erdmann
- Department of Surgery, University Medical Center Groningen, Postbus 30.001, 9700 RB, Groningen, The Netherlands.
| | - I Quintus Molenaar
- Department of Surgery, University Medical Center Utrecht, Postbus 85500, 3508 GA, Utrecht, The Netherlands.
| | - Erwin van der Harst
- Department of Surgery, Maasstad Hospital, Maasstadweg 21, 3079 DZ, Rotterdam, The Netherlands.
| | - Marion B van der Kolk
- Department of Surgery, Radboud University Medical Center, Geert Grooteplein-Zuid 10, 6525 GA, Nijmegen, The Netherlands.
| | - Coen R N Rasch
- Department of Radiation Oncology, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Geertjan van Tienhoven
- Department of Radiation Oncology, Academic Medical Center, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | | |
Collapse
|
27
|
Mantripragada KC, Safran H. Optimizing initial chemotherapy for metastatic pancreatic cancer. Future Oncol 2016; 12:1125-33. [PMID: 26939741 DOI: 10.2217/fon-2015-0006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The two combination chemotherapy regimens FOLFIRINOX and gemcitabine plus nab-paclitaxel represent major breakthroughs in the management of metastatic pancreatic cancer. Both regimens showed unprecedented survival advantage in the setting of front-line therapy. However, their application for treatment of patients in the community is challenging because of significant toxicities, thus limiting potential benefits to a narrow population of patients. Modifications to the dose intensity or schedule of those regimens improve their tolerability, while likely retaining survival advantage over single-agent chemotherapy. Newer strategies to optimize these two active regimens in advanced pancreatic cancer are being explored that can help personalize treatment to individual patients.
Collapse
Affiliation(s)
- Kalyan C Mantripragada
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Howard Safran
- The Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
28
|
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
|
29
|
Bijlsma MF, van Laarhoven HWM. The conflicting roles of tumor stroma in pancreatic cancer and their contribution to the failure of clinical trials: a systematic review and critical appraisal. Cancer Metastasis Rev 2016; 34:97-114. [PMID: 25566685 DOI: 10.1007/s10555-014-9541-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A nearly universal feature of pancreatic ductal adenocarcinoma (PDAC) is an extensive presence of activated stroma. This stroma is thought to aid in various tumor-promoting processes and hampers response to therapy. Here, we aim to evaluate the evidence that supports this role of the stroma in PDAC with functional experiments in relevant models, discuss the clinical trials that have aimed to target the stroma in this disease, and examine recent work that explains why these clinical trials based on stroma-targeting strategies have thus far not achieved the expected success. We systematically searched PubMed through August 2014 with no restrictions to identify published peer-reviewed research articles assessing the effect of targeting the stroma on tumor growth or metastases in preclinical animal models. Five hundred and thirty articles were extracted of which 31 were included in the analysis. Unfortunately, due to the large variety in models and outcome measures, we could not perform a meta-analysis of our data. We find that despite an abundance of positive outcomes reported in previous studies on stroma targeting, a strong discrepancy exists with the outcomes of clinical trials and the more recent preclinical work that is in line with these trials. We explain the incongruities by the duration of stroma targeting and propose that chronic stroma targeting treatment is possibly detrimental in the treatment of this disease.
Collapse
Affiliation(s)
- Maarten F Bijlsma
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental Molecular Medicine, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | | |
Collapse
|
30
|
Takaori K, Bassi C, Biankin A, Brunner TB, Cataldo I, Campbell F, Cunningham D, Falconi M, Frampton AE, Furuse J, Giovannini M, Jackson R, Nakamura A, Nealon W, Neoptolemos JP, Real FX, Scarpa A, Sclafani F, Windsor JA, Yamaguchi K, Wolfgang C, Johnson CD. International Association of Pancreatology (IAP)/European Pancreatic Club (EPC) consensus review of guidelines for the treatment of pancreatic cancer. Pancreatology 2016; 16:14-27. [PMID: 26699808 DOI: 10.1016/j.pan.2015.10.013] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/25/2015] [Accepted: 10/28/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Pancreatic cancer is one of the most devastating diseases with an extremely high mortality. Medical organizations and scientific societies have published a number of guidelines to address active treatment of pancreatic cancer. The aim of this consensus review was to identify where there is agreement or disagreement among the existing guidelines and to help define the gaps for future studies. METHODS A panel of expert pancreatologists gathered at the 46th European Pancreatic Club Meeting combined with the 18th International Association of Pancreatology Meeting and collaborated on critical reviews of eight English language guidelines for the clinical management of pancreatic cancer. Clinical questions (CQs) of interest were proposed by specialists in each of nine areas. The recommendations for the CQs in existing guidelines, as well as the evidence on which these were based, were reviewed and compared. The evidence was graded as sufficient, mediocre or poor/absent. RESULTS Only 4 of the 36 CQs, had sufficient evidence for agreement. There was also agreement in five additional CQs despite the lack of sufficient evidence. In 22 CQs, there was disagreement regardless of the presence or absence of evidence. There were five CQs that were not addressed adequately by existing guidelines. CONCLUSION The existing guidelines provide both evidence- and consensus-based recommendations. There is also considerable disagreement about the recommendations in part due to the lack of high level evidence. Improving the clinical management of patients with pancreatic cancer, will require continuing efforts to undertake research that will provide sufficient evidence to allow agreement.
Collapse
Affiliation(s)
- Kyoichi Takaori
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Claudio Bassi
- Department of Surgery and Oncology, Pancreas Institute, University of Verona, Verona, Italy
| | - Andrew Biankin
- Academic Unit of Surgery, University of Glasgow, Glasgow, United Kingdom
| | - Thomas B Brunner
- Department of Radiation Oncology, University Hospitals Freiburg, Germany
| | - Ivana Cataldo
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Fiona Campbell
- Department of Pathology, Royal Liverpool University Hospital, Liverpool, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - Massimo Falconi
- Pancreatic Surgery Unit, Università Vita e Salute, Milano, Italy
| | - Adam E Frampton
- HPB Surgical Unit, Department of Surgery and Cancer, Imperial College, Hammersmith Hospital, London, United Kingdom
| | - Junji Furuse
- Department of Medical Oncology, Kyorin University School of Medicine, Tokyo, Japan
| | - Marc Giovannini
- Endoscopic Unit, Paoli-Calmettes Institute, Marseille, France
| | - Richard Jackson
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Akira Nakamura
- Department of Radiation Oncology and Image-applied Therapy, Kyoto University Hospital, Kyoto, Japan
| | - William Nealon
- Division of General Surgery, Yale University, New Haven, CT, United States of America
| | - John P Neoptolemos
- NIHR Pancreas Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Francisco X Real
- Epithelial Carcinogenesis Group, CNIO-Spanish National Cancer Research Centre, Madrid, Spain
| | - Aldo Scarpa
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Francesco Sclafani
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London and Surrey, United Kingdom
| | - John A Windsor
- Department of Surgery, University of Auckland, HBP/Upper GI Unit, Auckland City Hospital, Auckland, New Zealand
| | - Koji Yamaguchi
- Department of Advanced Treatment of Pancreatic Disease, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Christopher Wolfgang
- Department of Surgery, The Johns Hopkins University, Baltimore, MD, United States of America
| | - Colin D Johnson
- University Surgical Unit, Southampton General Hospital, Southampton, United Kingdom
| |
Collapse
|
31
|
Adénocarcinomes pancréatiques « localisés »: limites de la « résécabilité »; principes et résultats des résections. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2557-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
32
|
Maréchal R, Puleo F, Demols A, Verset G, Laethem JLV. Personalized medicine in pancreatic cancer: the revolution has begun. Per Med 2015; 12:515-523. [PMID: 29749894 DOI: 10.2217/pme.15.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Pancreatic ductal adenocarcinoma carries a dismal prognosis. Both chemotherapy and targeted therapies have been disappointing when administered to unselected populations. Recently, progress has been made in our understanding of the genomic landscape of this cancer which displays remarkable heterogeneity suggesting a reorientation of management and research strategies based on molecular characterization and adapted personalized therapy. Resectable disease offers new opportunities for translational research through functional imaging response evaluation and tumor tissue acquisition before and after neoadjuvant therapy. There is urgent need for clinical trials based on molecular profiling in pancreatic ductal adenocarcinoma. In this review we discuss opportunities and limitations of these new strategies, underlining the importance of tissue acquisition and integration of molecular biomarkers in future molecularly driven clinical trials.
Collapse
Affiliation(s)
- Raphaël Maréchal
- Department of Gastroenterology & Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium.,Laboratory of Experimental Gastroenterology, Erasme Hospital, Université Libre de Bruxelles, Belgium
| | - Francesco Puleo
- Department of Gastroenterology & Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium.,Laboratory of Experimental Gastroenterology, Erasme Hospital, Université Libre de Bruxelles, Belgium
| | - Anne Demols
- Department of Gastroenterology & Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | - Gontran Verset
- Department of Gastroenterology & Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| | - Jean-Luc Van Laethem
- Department of Gastroenterology & Gastrointestinal Cancer Unit, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium
| |
Collapse
|
33
|
Li CC, Wang YQ, Li YP, Li XL. Critical appraisal of clinical practice guidelines for treating pancreatic cancer based on the global disease burden. J Evid Based Med 2015; 8:11-21. [PMID: 25594942 DOI: 10.1111/jebm.12140] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 10/09/2014] [Indexed: 02/05/2023]
Abstract
OBJECTIVES To evaluate the quality of pancreatic cancer guidelines with evidence-based methods based on the global burden of disease and to explore its status, distribution, characteristics of theme, and the difference of recommended therapies among various qualities of guidelines, so as to provide a reference for clinical decision. METHODS The PubMed, The Cochrane Library (Issue 11, 2013), Chinese Biomedical Database, China National Knowledge Infrastructure, and VIP databases, as well as the website of National Guidelines Clearinghouse, Guidelines International Network, and National Institute for Clinical Excellence were systematically reviewed through November 2013 for pancreatic cancer guidelines. The Appraisal of Guidelines for Research and Evaluation (AGREE II) were applied to assess the methodological quality of the guidelines. RESULTS A total of 14 relevant guidelines (including 5 evidence-based ones) were identified, involving four continents (Asia, Europe, North America, and Oceania), seven counties, and four international organizations. There were only two domains, namely 'scope and purpose' and 'clarity of presentations', getting high average scores (more than 60%) among all 14 guidelines. The mean AGREE domain score in guidelines varies among different areas, and the quality of 5 evidence-based guidelines was superior to that established by consensus. According to AGREE II, 11 guidelines were weakly recommended, while 3 were not recommended due to poor methodological quality. Their subjects of 14 guidelines covered six treatment categories, including chemotherapy, surgery, radiotherapy, support therapy, radiotherapy, and interventional therapy. CONCLUSION The overall methodological quality of pancreatic cancer guidelines is suboptimal among different countries or regions. The qualities of evidence-based guidelines are significantly superior to consensus. The chemotherapy, surgery, radiotherapy, and support therapy were the predominant choices by guidelines.
Collapse
Affiliation(s)
- Cui Cui Li
- Chinese Evidence-Based Medicine Center, West China Hospital, Sichuan University, Chengdu, China; The Nuclear Industry 416 Hospital, Chengdu, China
| | | | | | | |
Collapse
|
34
|
Buc E, Orry D, Antomarchi O, Gagnière J, Da Ines D, Pezet D. Resection of pancreatic ductal adenocarcinoma with synchronous distant metastasis: is it worthwhile? World J Surg Oncol 2014; 12:347. [PMID: 25407113 PMCID: PMC4289271 DOI: 10.1186/1477-7819-12-347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Accepted: 07/04/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The purpose of this study is to report prolonged survival in patients with metastatic pancreatic ductal adenocarcinoma (PDAC) managed by chemotherapy and surgery. METHODS Between January 2009 and August 2013, 284 patients with metastatic PDAC were managed in our oncologic department. Among them, three (1%) with a single metastasis (liver in two cases and interaorticaval in one case) underwent one- or two-stage surgical resection of the metastasis and the main tumor. Perioperative data were recorded retrospectively, including disease-free and overall survival. RESULTS The three patients had chemotherapy (FOLFOX or FOLFIRINOX regimen) with objective response or stable disease prior to surgery. Median time between chemotherapy and surgery was 9 (8 to 15) months. Resection consisted in pancreaticoduodenectomy in the three cases. None of the patients had grade III/IV postoperative complications, and median hospital stay was 12 (12 to 22) days. All the patients had postoperative chemotherapy. Only one patient experienced recurrence 11 months after surgery and died after 32.5 months. The two other patients were alive with no recurrence 26.3 and 24.7 months after initial treatment. CONCLUSION Radical resection of PDAC with single distant metastases can offer prolonged survival with low morbidity after accurate selection by neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Emmanuel Buc
- />Department of Digestive and HPB Surgery, CHU Estaing - 1, Place Lucie et Raymond Aubrac, 63003 Clermont-Ferrand, France
| | - David Orry
- />Department of Oncologic Surgery, Centre Georges François Leclerc, Dijon, France
| | - Olivier Antomarchi
- />Department of Digestive and HPB Surgery, CHU Estaing - 1, Place Lucie et Raymond Aubrac, 63003 Clermont-Ferrand, France
| | - Johan Gagnière
- />Department of Digestive and HPB Surgery, CHU Estaing - 1, Place Lucie et Raymond Aubrac, 63003 Clermont-Ferrand, France
| | - David Da Ines
- />Department of Radiology, CHU Estaing, Clermont-Ferrand, France
| | - Denis Pezet
- />Department of Digestive and HPB Surgery, CHU Estaing - 1, Place Lucie et Raymond Aubrac, 63003 Clermont-Ferrand, France
| |
Collapse
|
35
|
Sultana A, Jackson RJ, Cox T, Palmer D, Neoptolemos J, Ghaneh P. Chemotherapy, radiotherapy, chemoradiotherapy and combination therapy in localised and locally advanced pancreatic cancer. Hippokratia 2014. [DOI: 10.1002/14651858.cd011044] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Asma Sultana
- University of Liverpool; Department of Molecular and Clinical Cancer Medicine; Liverpool UK
| | - Richard J Jackson
- Liverpool University; North West Surgical Trials Centre; Brownlow Street Liverpool UK L69 3GL
| | - Trevor Cox
- University of Liverpool; Liverpool Cancer Research UK Cancer Trials Unit; Liverpool UK
| | - Daniel Palmer
- University of Liverpool; Department of Molecular and Clinical Cancer Medicine; Liverpool UK
| | - John Neoptolemos
- University of Liverpool; Department of Molecular and Clinical Cancer Medicine; Liverpool UK
| | - Paula Ghaneh
- University of Liverpool; Department of Molecular and Clinical Cancer Medicine; Liverpool UK
| |
Collapse
|
36
|
Jiang YJ, Lee CL, Wang Q, Zhou ZW, Yang F, Jin C, Fu DL. Establishment of an orthotopic pancreatic cancer mouse model: cells suspended and injected in Matrigel. World J Gastroenterol 2014; 20:9476-9485. [PMID: 25071342 PMCID: PMC4110579 DOI: 10.3748/wjg.v20.i28.9476] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 03/24/2014] [Accepted: 04/05/2014] [Indexed: 02/06/2023] Open
Abstract
AIM To establish an orthotopic mouse model of pancreatic cancer that mimics the pathological features of exocrine pancreatic adenocarcinoma. METHODS Pan02 cells were suspended in low-temperature Matrigel and injected into the parenchyma of pancreatic tails of C57BL/6 mice, with cells suspended in phosphate buffered saline (PBS) serving as a control. Primary and implanted tumors were confirmed pathologically. The rate of tumor formation and intraperitoneal implantation in the two groups were compared at different time points after injection. Leakage and intra-abdominal dispersion of Matrigel and PBS, both dyed with methylene blue, were compared after injection into the parenchyma of the pancreas. We observed adherence and proliferation in Pan02 cells suspended in Matrigel in vitro. We also compared the pathological manifestation of this orthotopic pancreatic cancer model in the head and tails of the pancreas. The characteristics of the origin of epithelial cells and exocrine markers of established orthotopic pancreatic tumors were confirmed using immunohistochemistry. RESULTS Diluted Matrigel could form a gel drip in the pancreatic parenchyma, effectively preventing leakage from the injection site and avoiding dispersion in the abdominal cavity. Pan02 cells were able to adhere to a dish, proliferate, and migrate in the gel drip. The tumor formation rate in the Matrigel group was 100% at both 2 and 3 wk after injection, whereas it was 25.0% and 37.5% in the PBS group at 2 and 3 wk, respectively (P < 0.05). The intraperitoneal tumor implantation rate was 75.0% in the PBS group after 3 wk of injection, while it was 12.5% in the Matrigel group (P < 0.05). Hepatoduodenal ligament and duodenal invasions with obstructive jaundice and upper digestive obstruction with mesenteric lymph node metastasis were observed in the pancreatic head group. In the pancreatic tail group, spleen and gastric invasion were dominant, leading to retroperitoneal lymph nodes metastasis. Positive immunohistochemical staining of cytokeratin and negative staining of vimentin and chromogranin A confirmed that the orthotopic pancreatic tumor injected with Pan02 cells suspended in Matrigel was of epithelial origin and expressed exocrine markers of cancer. CONCLUSION This method of low-temperature Matrigel suspension and injection is effective for establishing an orthotopic mouse model of pancreatic cancer.
Collapse
|
37
|
Abstract
OBJECTIVES Src is considered a rising therapeutic target for the treatment of solid tumors, and Src family kinases (SFKs) participate in cancer cell proliferation and survival. The role of SFK suppression was investigated in the proliferation, migration, and invasion of pancreatic cancer cells. METHODS Knockdown of the SFKs in pancreatic cancer cells was achieved by transfecting small interfering RNAs, and its effects were investigated using proliferation, wound, and invasion assays. RESULTS The SFK inhibitors suppressed proliferation and induced cell cycle arrest in pancreatic cancer cells. The SFK messenger RNA profiles showed that Yes1, Lyn, Fyn, Frk, Hck, and Src were expressed. Specific small interfering RNA transfection suppressed the messenger RNA expressions of Yes1, Lyn, Fyn, Frk, and Src, and the knockdown suppressed cell proliferation by 16.7% to 47.3% in PANC-1 cells. Knockdown of any of these 5 SFKs suppressed proliferation in other pancreatic cancer cell lines by 3.0% to 40.5%. The knockdowns significantly reduced pancreatic cancer cell migration by 24.9% to 66.7% and completely inhibited invasion. CONCLUSIONS These results suggest that the knockdown of Yes1, Lyn, Fyn, Frk, or Src reduce human pancreatic cancer cell proliferation, migration, and invasion, and that SFKs should be viewed as critical therapeutic targets of pancreatic cancer.
Collapse
|
38
|
Middleton G, Silcocks P, Cox T, Valle J, Wadsley J, Propper D, Coxon F, Ross P, Madhusudan S, Roques T, Cunningham D, Falk S, Wadd N, Harrison M, Corrie P, Iveson T, Robinson A, McAdam K, Eatock M, Evans J, Archer C, Hickish T, Garcia-Alonso A, Nicolson M, Steward W, Anthoney A, Greenhalf W, Shaw V, Costello E, Naisbitt D, Rawcliffe C, Nanson G, Neoptolemos J. Gemcitabine and capecitabine with or without telomerase peptide vaccine GV1001 in patients with locally advanced or metastatic pancreatic cancer (TeloVac): an open-label, randomised, phase 3 trial. Lancet Oncol 2014; 15:829-40. [PMID: 24954781 DOI: 10.1016/s1470-2045(14)70236-0] [Citation(s) in RCA: 277] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND We aimed to assess the efficacy and safety of sequential or simultaneous telomerase vaccination (GV1001) in combination with chemotherapy in patients with locally advanced or metastatic pancreatic cancer. METHODS TeloVac was a three-group, open-label, randomised phase 3 trial. We recruited patients from 51 UK hospitals. Eligible patients were treatment naive, aged older than 18 years, with locally advanced or metastatic pancreatic ductal adenocarcinoma, and Eastern Cooperative Oncology Group performance status of 0-2. Patients were randomly assigned (1:1:1) to receive either chemotherapy alone, chemotherapy with sequential GV1001 (sequential chemoimmunotherapy), or chemotherapy with concurrent GV1001 (concurrent chemoimmunotherapy). Treatments were allocated with equal probability by means of computer-generated random permuted blocks of sizes 3 and 6 in equal proportion. Chemotherapy included six cycles of gemcitabine (1000 mg/m(2), 30 min intravenous infusion, at days 1, 8, and 15) and capecitabine (830 mg/m(2) orally twice daily for 21 days, repeated every 28 days). Sequential chemoimmunotherapy included two cycles of combination chemotherapy, then an intradermal lower abdominal injection of granulocyte-macrophage colony-stimulating factor (GM-CSF; 75 μg) and GV1001 (0·56 mg; days 1, 3, and 5, once on weeks 2-4, and six monthly thereafter). Concurrent chemoimmunotherapy included giving GV1001 from the start of chemotherapy with GM-CSF as an adjuvant. The primary endpoint was overall survival; analysis was by intention to treat. This study is registered as an International Standard Randomised Controlled Trial, number ISRCTN4382138. FINDINGS The first patient was randomly assigned to treatment on March 29, 2007, and the trial was terminated on March 27, 2011. Of 1572 patients screened, 1062 were randomly assigned to treatment (358 patients were allocated to the chemotherapy group, 350 to the sequential chemoimmunotherapy group, and 354 to the concurrent chemoimmunotherapy group). We recorded 772 deaths; the 290 patients still alive were followed up for a median of 6·0 months (IQR 2·4-12·2). Median overall survival was not significantly different in the chemotherapy group than in the sequential chemoimmunotherapy group (7·9 months [95% CI 7·1-8·8] vs 6·9 months [6·4-7·6]; hazard ratio [HR] 1·19, 98·25% CI 0·97-1·48, p=0·05), or in the concurrent chemoimmunotherapy group (8·4 months [95% CI 7·3-9·7], HR 1·05, 98·25% CI 0·85-1·29, p=0·64; overall log-rank of χ(2)2df=4·3; p=0·11). The commonest grade 3-4 toxic effects were neutropenia (68 [19%] patients in the chemotherapy group, 58 [17%] patients in the sequential chemoimmunotherapy group, and 79 [22%] patients in the concurrent chemoimmunotherapy group; fatigue (27 [8%] in the chemotherapy group, 35 [10%] in the sequential chemoimmunotherapy group, and 44 [12%] in the concurrent chemoimmunotherapy group); and pain (34 [9%] patients in the chemotherapy group, 39 [11%] in the sequential chemoimmunotherapy group, and 41 [12%] in the concurrent chemoimmunotherapy group). INTERPRETATION Adding GV1001 vaccination to chemotherapy did not improve overall survival. New strategies to enhance the immune response effect of telomerase vaccination during chemotherapy are required for clinical efficacy. FUNDING Cancer Research UK and KAEL-GemVax.
Collapse
Affiliation(s)
| | - Paul Silcocks
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - Trevor Cox
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - Juan Valle
- Manchester Academic Health Sciences Centre, Christie Hospital NHS Foundation Trust and University of Manchester, Manchester UK
| | - Jonathan Wadsley
- Weston Park Hospital, Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - David Propper
- St Bartholomew's Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Fareeda Coxon
- Northern Centre for Cancer Care, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Freeman Hospital, Newcastle upon Tyne, UK
| | - Paul Ross
- Guy's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Tom Roques
- Norfolk and Norwich University Hospital, Norfolk and Norwich University Hospital NHS Foundation Trust, Norwich, UK
| | - David Cunningham
- The Royal Marsden, The Royal Marsden NHS Foundation Trust, London, UK
| | - Stephen Falk
- Bristol Haematology And Oncology Centre, University Hospital Bristol NHS Foundation Trust, Bristol, UK
| | - Nick Wadd
- The James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middleborough, UK
| | - Mark Harrison
- Mount Vernon Hospital, The Hillingdon Hospitals NHS Foundation Trust, Northwood, UK
| | - Pippa Corrie
- Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Tim Iveson
- Southampton General Hospital, University Hospital Southampton NHS Foundation Trust, Southampton, Hampshire, UK
| | - Angus Robinson
- Conquest Hospital, East Sussex Healthcare NHS Trust, The Ridge, St Leonards-on-Sea, East Sussex, UK
| | - Karen McAdam
- Peterborough City Hospital, Peterborough and Stamford Hospitals NHS Foundation Trust, Edith, Cavell Campus, Peterborough, UK
| | - Martin Eatock
- Belfast City Hospital, Belfast Health and Social Care Trust, Belfast, UK
| | - Jeff Evans
- University of Glasgow, Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Caroline Archer
- Queen Alexandra Hospital, Portsmouth Hospitals NHS Trust, Cosham, Portsmouth, UK
| | - Tamas Hickish
- Royal Bournemouth Hospital, The Royal Bournemouth and Christchurch Hospitals NHS Foundation Trust, Bournemouth, UK
| | | | | | - William Steward
- Leicester Royal Infirmary, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Alan Anthoney
- St James University Hospital, The Leeds Teaching Hospital Trust, Beckett Street, Leeds, UK
| | - William Greenhalf
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - Victoria Shaw
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - Eithne Costello
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - Dean Naisbitt
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - Charlotte Rawcliffe
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - Gemma Nanson
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK
| | - John Neoptolemos
- Liverpool Cancer Research UK Cancer Trials Unit and GCLP Facility, University of Liverpool, Liverpool, UK.
| |
Collapse
|
39
|
Valle JW, Palmer D, Jackson R, Cox T, Neoptolemos JP, Ghaneh P, Rawcliffe CL, Bassi C, Stocken DD, Cunningham D, O'Reilly D, Goldstein D, Robinson BA, Karapetis C, Scarfe A, Lacaine F, Sand J, Izbicki JR, Mayerle J, Dervenis C, Oláh A, Butturini G, Lind PA, Middleton MR, Anthoney A, Sumpter K, Carter R, Büchler MW. Optimal duration and timing of adjuvant chemotherapy after definitive surgery for ductal adenocarcinoma of the pancreas: ongoing lessons from the ESPAC-3 study. J Clin Oncol 2014; 32:504-12. [PMID: 24419109 DOI: 10.1200/jco.2013.50.7657] [Citation(s) in RCA: 302] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024] Open
Abstract
PURPOSE Adjuvant chemotherapy improves patient survival rates after resection for pancreatic adenocarcinoma, but the optimal duration and time to initiate chemotherapy is unknown. PATIENTS AND METHODS Patients with pancreatic ductal adenocarcinoma treated within the international, phase III, European Study Group for Pancreatic Cancer-3 (version 2) study were included if they had been randomly assigned to chemotherapy. Overall survival analysis was performed on an intention-to-treat basis, retaining patients in their randomized groups, and adjusting the overall treatment effect by known prognostic variables as well as the start time of chemotherapy. RESULTS There were 985 patients, of whom 486 (49%) received gemcitabine and 499 (51%) received fluorouracil; 675 patients (68%) completed all six cycles of chemotherapy (full course) and 293 patients (30%) completed one to five cycles. Lymph node involvement, resection margins status, tumor differentiation, and completion of therapy were all shown by multivariable Cox regression to be independent survival factors. Overall survival favored patients who completed the full six courses of treatment versus those who did not (hazard ratio [HR], 0.516; 95% CI, 0.443 to 0.601; P < .001). Time to starting chemotherapy did not influence overall survival rates for the full study population (HR, 0.985; 95% CI, 0.956 to 1.015). Chemotherapy start time was an important survival factor only for the subgroup of patients who did not complete therapy, in favor of later treatment (P < .001). CONCLUSION Completion of all six cycles of planned adjuvant chemotherapy rather than early initiation was an independent prognostic factor after resection for pancreatic adenocarcinoma. There seems to be no difference in outcome if chemotherapy is delayed up to 12 weeks, thus allowing adequate time for postoperative recovery.
Collapse
Affiliation(s)
- Juan W Valle
- Juan W. Valle, Derek O'Reilly, Manchester Academic Health Sciences Centre, Christie Hospital NHS Foundation Trust and University of Manchester, Manchester; Richard Jackson, Trevor Cox, John P. Neoptolemos, Paula Ghaneh, Charlotte L. Rawcliffe, Liverpool Cancer Research UK Centre and the National Institute for Health Research Pancreas Biomedical Research Unit, University of Liverpool, Liverpool; Daniel Palmer, the Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust; Deborah D. Stocken, the Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham; David Cunningham, Royal Marsden Hospital Foundation Trust, Sutton; Mark R. Middleton, Churchill Hospital, Oxford University Hospitals NHS Trust, Oxford; Alan Anthoney, The Leeds Teaching Hospital Trust, Leeds; Kate Sumpter, Freeman Hospital, Newcastle upon Tyne; Ross Carter, Glasgow Royal Infirmary, Glasgow, United Kingdom; Claudio Bassi, Giovanni Butturini, University of Verona, Verona, Italy; David Goldstein, Bridget A. Robinson, Christos Karapetis, the Australasian Gastro-Intestinal Trials Group, Camperdown, Australia; Andrew Scarfe, University of Alberta, Edmonton, Canada; Francois Lacaine, Hôpital TENON, Assistance Publique Hôpitaux de Paris, Universite Pierre Et Marie Curie, Paris, France; Juhani Sand, Tampere University Hospital, Tampere, Finland; Jakob R. Izbicki, University of Hamburg, Hamburg; Julia Mayerle, Ernst-Moritz-Arndt-Universität Greifswald, Greifswald; Markus W. Büchler, University of Heidelberg, Heidelberg, Germany; Christos Dervenis, the Agia Olga Hospital, Athens, Greece; Attila Oláh, the Petz Aladar Hospital, Gyor, Hungary; Pehr A. Lind, Karolinska-Stockholm Söder Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Antoniou G, Kountourakis P, Papadimitriou K, Vassiliou V, Papamichael D. Adjuvant therapy for resectable pancreatic adenocarcinoma: Review of the current treatment approaches and future directions. Cancer Treat Rev 2014; 40:78-85. [DOI: 10.1016/j.ctrv.2013.05.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 05/28/2013] [Accepted: 05/30/2013] [Indexed: 12/15/2022]
|
41
|
Oberg HH, Peipp M, Kellner C, Sebens S, Krause S, Petrick D, Adam-Klages S, Röcken C, Becker T, Vogel I, Weisner D, Freitag-Wolf S, Gramatzki M, Kabelitz D, Wesch D. Novel bispecific antibodies increase γδ T-cell cytotoxicity against pancreatic cancer cells. Cancer Res 2014; 74:1349-60. [PMID: 24448235 DOI: 10.1158/0008-5472.can-13-0675] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ability of human γδ T cells from healthy donors to kill pancreatic ductal adenocarcinoma (PDAC) in vitro and in vivo in immunocompromised mice requires the addition of γδ T-cell-stimulating antigens. In this study, we demonstrate that γδ T cells isolated from patients with PDAC tumor infiltrates lyse pancreatic tumor cells after selective stimulation with phosphorylated antigens. We determined the absolute numbers of γδ T-cell subsets in patient whole blood and applied a real-time cell analyzer to measure their cytotoxic effector function over prolonged time periods. Because phosphorylated antigens did not optimally enhance γδ T-cell cytotoxicity, we designed bispecific antibodies that bind CD3 or Vγ9 on γδ T cells and Her2/neu (ERBB2) expressed by pancreatic tumor cells. Both antibodies enhanced γδ T-cell cytotoxicity with the Her2/Vγ9 antibody also selectively enhancing release of granzyme B and perforin. Supporting these observations, adoptive transfer of γδ T cells with the Her2/Vγ9 antibody reduced growth of pancreatic tumors grafted into SCID-Beige immunocompromised mice. Taken together, our results show how bispecific antibodies that selectively recruit γδ T cells to tumor antigens expressed by cancer cells illustrate the tractable use of endogenous γδ T cells for immunotherapy.
Collapse
Affiliation(s)
- Hans-Heinrich Oberg
- Authors' Affiliations: Institute of Immunology; Division of Stem Cell Transplantation and Immunotherapy; Institute for Experimental Medicine; Institute of Pathology; Clinic of General and Thoracic Surgery; Institute for Medical Informatics and Statistic, Christian-Albrechts-University Kiel; Municipal Hospital, Department of Surgery; and Clinic of Gynaecology and Obstetrics, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Sabater L, García-Granero A, Escrig-Sos J, Gómez-Mateo MDC, Sastre J, Ferrández A, Ortega J. Outcome Quality Standards in Pancreatic Oncologic Surgery. Ann Surg Oncol 2014; 21:1138-46. [DOI: 10.1245/s10434-013-3451-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Indexed: 12/16/2022]
|
43
|
Greenhalf W, Ghaneh P, Neoptolemos JP, Palmer DH, Cox TF, Lamb RF, Garner E, Campbell F, Mackey JR, Costello E, Moore MJ, Valle JW, McDonald AC, Carter R, Tebbutt NC, Goldstein D, Shannon J, Dervenis C, Glimelius B, Deakin M, Charnley RM, Lacaine F, Scarfe AG, Middleton MR, Anthoney A, Halloran CM, Mayerle J, Oláh A, Jackson R, Rawcliffe CL, Scarpa A, Bassi C, Büchler MW. Pancreatic cancer hENT1 expression and survival from gemcitabine in patients from the ESPAC-3 trial. J Natl Cancer Inst 2014; 106:djt347. [PMID: 24301456 DOI: 10.1093/jnci/djt347] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Human equilibrative nucleoside transporter 1 (hENT1) levels in pancreatic adenocarcinoma may predict survival in patients who receive adjuvant gemcitabine after resection. METHODS Microarrays from 434 patients randomized to chemotherapy in the ESPAC-3 trial (plus controls from ESPAC-1/3) were stained with the 10D7G2 anti-hENT1 antibody. Patients were classified as having high hENT1 expression if the mean H score for their cores was above the overall median H score (48). High and low hENT1-expressing groups were compared using Kaplan-Meier curves, log-rank tests, and Cox proportional hazards models. All statistical tests were two-sided. RESULTS Three hundred eighty patients (87.6%) and 1808 cores were suitable and included in the final analysis. Median overall survival for gemcitabine-treated patients (n = 176) was 23.4 (95% confidence interval [CI] = 18.3 to 26.0) months vs 23.5 (95% CI = 19.8 to 27.3) months for 176 patients treated with 5-fluorouracil/folinic acid (χ(2) 1=0.24; P = .62). Median survival for patients treated with gemcitabine was 17.1 (95% CI = 14.3 to 23.8) months for those with low hENT1 expression vs 26.2 (95% CI = 21.2 to 31.4) months for those with high hENT1 expression (χ(2)₁= 9.87; P = .002). For the 5-fluorouracil group, median survival was 25.6 (95% CI = 20.1 to 27.9) and 21.9 (95% CI = 16.0 to 28.3) months for those with low and high hENT1 expression, respectively (χ(2)₁ = 0.83; P = .36). hENT1 levels were not predictive of survival for the 28 patients of the observation group (χ(2)₁ = 0.37; P = .54). Multivariable analysis confirmed hENT1 expression as a predictive marker in gemcitabine-treated (Wald χ(2) = 9.16; P = .003) but not 5-fluorouracil-treated (Wald χ(2) = 1.22; P = .27) patients. CONCLUSIONS Subject to prospective validation, gemcitabine should not be used for patients with low tumor hENT1 expression.
Collapse
Affiliation(s)
- William Greenhalf
- Affiliations of authors: Liverpool Cancer Research UK Cancer Trials Unit, Liverpool Cancer Research UK Centre, University of Liverpool, Liverpool, UK (WG, JPN, EG, TFC, PG, EC, CMH, CLR, FC, RJ); the Princess Margaret Hospital, Toronto, Canada (MJM); Manchester Academic Health Sciences Centre, Christie NHS Foundation Trust, School of Cancer and Enabling Sciences, University of Manchester, UK (JWV); Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham, UK (DHP); Beatson West of Scotland Cancer Centre, Glasgow, UK (ACM); Glasgow Royal Infirmary, Glasgow, UK (RC); Hôpital Tenon, Université, Pierre et Marie Curie, Paris, France (FL); Austin Health, Melbourne, Australia (NCT); Prince of Wales Hospital and Clinical School University of New South Wales, New South Wales, Australia (DG); Nepean Cancer Centre and University of Sydney, Sydney, Australia (JS); Agia Olga Hospital, Athens, Greece (CD); Medical Oncology, Clatterbridge Centre for Oncology, Bebington, Merseyside, UK (DS); Department of Oncology, Akademiska Sjukhuset, Uppsala University, Uppsala, Sweden (BG); University Hospital, North Staffordshire, UK (MD); Freeman Hospital, Newcastle upon Tyne, UK (RMC); Service de Chirurgie Digestive et Viscérale, Hôpital Tenon, Paris, France (FL); Cross Cancer Institute and University of Alberta, Alberta, Canada (JRM, AGS); Churchill Hospital, Oxford University Hospitals NHS Trust, Oxford, UK (MRM); St James's University Hospital, Leeds, UK (AA); Department of Medicine A, University Medicine Greifswald, Greifswald, Germany (JM); Petz Aladar Hospital, Gyor, Hungary (AO); Departments of Surgery and Pathology and ARC-NET Research Center, University of Verona, Italy (AS, CB); Department of Surgery, University of Heidelberg, Heidelberg, Germany (MWB)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Schad F, Atxner J, Buchwald D, Happe A, Popp S, Kröz M, Matthes H. Intratumoral Mistletoe (Viscum album L) Therapy in Patients With Unresectable Pancreas Carcinoma: A Retrospective Analysis. Integr Cancer Ther 2013; 13:332-40. [PMID: 24363283 DOI: 10.1177/1534735413513637] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pancreatic carcinoma remains one of the main causes for cancer-related death. Intratumoral application of anticancer agents is discussed as a promising method for solid tumors such as pancreatic cancer. Endoscopic ultrasound provides a good tool to examine and treat the pancreas. European mistletoe (Viscum album L) is a phytotherapeutic commonly used in integrative oncology in Central Europe. Its complementary use seeks to induce immunostimulation and antitumoral effects as well as alleviate chemotherapeutic side effects. Intratumoral mistletoe application has induced local tumor response in various cancer entities. This off-label use needs to be validated carefully in terms of safety and benefits. Here we report on 39 patients with advanced, inoperable pancreatic cancer, who received in total 223 intratumoral applications of mistletoe, endoscopic ultrasound guided or under transabdominal ultrasound control. No severe procedure-related events were reported. Adverse drug reactions were mainly increased body temperature or fever in 14% and 11% of the applications, respectively. Other adverse drug reactions, such as pain or nausea, occurred in less than 7% of the procedures. No severe adverse drug reaction was recorded. Patients received standard first- and second-line chemotherapy and underwent adequate palliative surgical interventions as well as additive subcutaneous and partly intravenous mistletoe application. A median survival of 11 months was observed for all patients, or 11.8 and 8.3 months for stages III and IV, respectively. Due to the multimodal therapeutic setting and the lack of a control group, the effect of intratumoral mistletoe administration alone remains unclear. This retrospective analysis suggests that intratumoral-applicated mistletoe might contribute to improve survival of patients with pancreatic cancer. In conclusion, the application is feasible and safe, and its efficacy should be evaluated in a randomized controlled trial.
Collapse
Affiliation(s)
- Friedemann Schad
- Hospital Havelhoehe, Berlin, Germany Research Institute Havelhoehe, Berlin, Germany
| | - Jan Atxner
- Research Institute Havelhoehe, Berlin, Germany
| | | | - Antje Happe
- Research Institute Havelhoehe, Berlin, Germany
| | | | - Matthias Kröz
- Hospital Havelhoehe, Berlin, Germany Research Institute Havelhoehe, Berlin, Germany
| | - Harald Matthes
- Hospital Havelhoehe, Berlin, Germany Research Institute Havelhoehe, Berlin, Germany
| |
Collapse
|
45
|
Olszewski AJ, Grossbard ML, Chung MS, Chalasani SB, Malamud S, Mirzoyev T, Kozuch PS. Phase I study of oxaliplatin in combination with gemcitabine, irinotecan, and 5-fluorouracil/leucovorin (G-FLIE) in patients with metastatic solid tumors including adenocarcinoma of the pancreas. J Gastrointest Cancer 2013. [PMID: 23208490 DOI: 10.1007/s12029-012-9466-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE The aims of this study were to establish the maximum tolerated dose (MTD) of oxaliplatin in combination with fixed doses of gemcitabine, irinotecan, and 5-fluorouracil/leucovorin (G-FLIE) in solid tumors, including advanced pancreatic cancer, and to evaluate the toxicity of the regimen. METHODS Patients with metastatic solid tumors were treated with a regimen consisting of gemcitabine (500 mg/m(2) by fixed-dose-rate infusion), irinotecan (120 mg/m(2)), leucovorin 300 mg, bolus/infusion 5-fluorouracil (400 and 1,500 mg/m2, respectively), and oxaliplatin at doses from 50 to 85 mg/m(2) according to the escalation schema. Treatment was repeated every 14 days. RESULTS The study enrolled 25 patients with a median age of 64 years and median Karnofsky performance score of 80. Patients had metastatic adenocarcinomas of pancreas (n = 9), as well as gastroesointestinal, hepatobiliary, or unknown primary tumors. With only one dose limiting toxicity (neutropenia and constipation), the MTD of oxaliplatin was not reached up to the pre-specified maximum level of 85 mg/m(2). Other toxicities predictably included cytopenias, fatigue, sensory neuropathy, nausea/vomiting, diarrhea, and constipation. Four partial responses and ten disease stabilizations were observed. The overall median time to disease progression was 17 weeks (2-110 weeks) with median overall survival of 31.5 weeks (7-139 weeks). CONCLUSIONS G-FLIE is a tolerable multi-agent chemotherapy regimen with the oxaliplatin dose up to 85 mg/m(2). The combination of full-dose oxaliplatin with gemcitabine, irinotecan, and 5-fluorouracil is feasible with attenuated doses of the drugs, but further optimization is necessary before assessment of efficacy.
Collapse
Affiliation(s)
- Adam J Olszewski
- St Luke's-Roosevelt Hospital Center, Continuum Cancer Centers of New York, 10th Ave and 59th St, New York, NY 10019, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
PEGylated liposomal Gemcitabine: insights into a potential breast cancer therapeutic. Cell Oncol (Dordr) 2013; 36:449-57. [PMID: 24081907 DOI: 10.1007/s13402-013-0146-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2013] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Nanoencapsulation of chemotherapeutics is an established method to target breast tumors and has been shown to enhance the efficacy of therapy in various animal models. During the past two decades, the nucleoside analog Gemcitabine has been under investigation to treat both recalcitrant and localized breast cancer, often in combination with other chemotherapeutics. In this study, we investigated the chemotherapeutic efficacy of a novel Gemcitabine-encapsulated liposome previously formulated by our group, GemPo, on both sensitive (4T1) and recalcitrant (MDA-MB-231) breast cancer cell lines. METHODS Gemcitabine free drug and liposomal Gemcitabine were compared both in vitro and in vivo using breast cancer models. RESULTS We demonstrated that GemPo differently hindered the growth, survival and migration of breast cancer cells, according to their drug sensitivities. Specifically, whereas GemPo was a more potent cytotoxic and apoptotic agent in sensitive breast cancer cells, it more potently inhibited cell migration in the resistant cell line. However, GemPo still acted as a more potent inhibitor of migration, in comparison with free Gemcitabine, irrespective of cell sensitivity. Administration of GemPo in a 4T1-bearing mouse model inhibited tumor growth while increasing mice survival, as compared with free Gemcitabine and a vehicle control. Interestingly, the inclusion of a mitotic inhibitor, Paclitaxel, synergized only with free Gemcitabine in this model, yet was as effective as GemPo alone. However, inclusion of Paclitaxel with GemPo significantly improved mouse survival. CONCLUSIONS Our study is the first to demonstrate the pleiotropic effects of Gemcitabine and Gemcitabine-loaded nanoparticles in breast cancer, and opens the door for a novel treatment for breast cancer patients.
Collapse
|
47
|
Tumor necrosis factor induces tumor promoting and anti-tumoral effects on pancreatic cancer via TNFR1. PLoS One 2013; 8:e75737. [PMID: 24098720 PMCID: PMC3787053 DOI: 10.1371/journal.pone.0075737] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 08/21/2013] [Indexed: 12/28/2022] Open
Abstract
Multiple activities are ascribed to the cytokine tumor necrosis factor (TNF) in health and disease. In particular, TNF was shown to affect carcinogenesis in multiple ways. This cytokine acts via the activation of two cell surface receptors, TNFR1, which is associated with inflammation, and TNFR2, which was shown to cause anti-inflammatory signaling. We assessed the effects of TNF and its two receptors on the progression of pancreatic cancer by in vivo bioluminescence imaging in a syngeneic orthotopic tumor mouse model with Panc02 cells. Mice deficient for TNFR1 were unable to spontaneously reject Panc02 tumors and furthermore displayed enhanced tumor progression. In contrast, a fraction of wild type (37.5%), TNF deficient (12.5%), and TNFR2 deficient mice (22.2%) were able to fully reject the tumor within two weeks. Pancreatic tumors in TNFR1 deficient mice displayed increased vascular density, enhanced infiltration of CD4(+) T cells and CD4(+) forkhead box P3 (FoxP3)(+) regulatory T cells (Treg) but reduced numbers of CD8(+) T cells. These alterations were further accompanied by transcriptional upregulation of IL4. Thus, TNF and TNFR1 are required in pancreatic ductal carcinoma to ensure optimal CD8(+) T cell-mediated immunosurveillance and tumor rejection. Exogenous systemic administration of human TNF, however, which only interacts with murine TNFR1, accelerated tumor progression. This suggests that TNFR1 has basically the capability in the Panc02 model to trigger pro-and anti-tumoral effects but the spatiotemporal availability of TNF seems to determine finally the overall outcome.
Collapse
|
48
|
Tonack S, Jenkinson C, Cox T, Elliott V, Jenkins RE, Kitteringham NR, Greenhalf W, Shaw V, Michalski CW, Friess H, Neoptolemos JP, Costello E. iTRAQ reveals candidate pancreatic cancer serum biomarkers: influence of obstructive jaundice on their performance. Br J Cancer 2013; 108:1846-53. [PMID: 23579209 PMCID: PMC3658525 DOI: 10.1038/bjc.2013.150] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 01/09/2013] [Accepted: 03/14/2013] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The aims of our study were to identify serum biomarkers that distinguish pancreatic cancer (pancreatic ductal adenocarcinoma, PDAC) patients from benign pancreatic disease patients and healthy subjects, and to assess the effects of jaundice on biomarker performance. METHODS Isobaric tags for relative and absolute quantification were used to compare pooled serum and pancreatic juice samples from a test set of 59 and 25 subjects, respectively. Validation was undertaken in 113 independent subjects. RESULTS Candidate proteins Complement C5, inter-α-trypsin inhibitor heavy chain H3, α1-β glycoprotein and polymeric immunoglobulin receptor were elevated in cancer, as were the reference markers CA19-9 and Reg3A. Biliary obstruction had a significant effect on the performance of the markers, in particular within the PDAC group where the presence of jaundice was associated with a significant increase in the levels of all six proteins (P<0.01). Consequently, in the absence of jaundice, proteins showed reduced sensitivity for PDAC patients over benign subjects and healthy controls (HCs). Similarly, in the presence of jaundice, markers showed reduced specificity for PDAC patients over benign subjects with jaundice. Combining markers enabled improved sensitivity for non-jaundiced PDAC patients over HCs and improved specificity for jaundiced PDAC patients over jaundiced benign disease subjects. CONCLUSIONS The presence-absence of jaundice in the clinical scenario severely impacts the performance of biomarkers for PDAC diagnosis and has implications for their clinical translation.
Collapse
Affiliation(s)
- S Tonack
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - C Jenkinson
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - T Cox
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - V Elliott
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - R E Jenkins
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Liverpool, UK
| | - N R Kitteringham
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Liverpool, UK
| | - W Greenhalf
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - V Shaw
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
| | - C W Michalski
- Department of General Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - H Friess
- Department of General Surgery, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - J P Neoptolemos
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - E Costello
- Department of Molecular and Clinical Cancer Medicine, Liverpool Cancer Research-UK Centre, University of Liverpool, Liverpool, UK
- National Institute for Health Research Liverpool Pancreatic Biomedical Research Unit, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
49
|
Labianca R, Merelli B, Mosconi S. Treatment of advanced pancreatic cancer. Ann Oncol 2013; 23 Suppl 10:x139-40. [PMID: 22987950 DOI: 10.1093/annonc/mds355] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- R Labianca
- Department of Oncology and Hematology, Ospedali Riuniti, Bergamo, Italy.
| | | | | |
Collapse
|
50
|
Monogalactosyl diacylglycerol, a replicative DNA polymerase inhibitor, from spinach enhances the anti-cell proliferation effect of gemcitabine in human pancreatic cancer cells. Biochim Biophys Acta Gen Subj 2013; 1830:2517-25. [DOI: 10.1016/j.bbagen.2012.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 10/25/2012] [Accepted: 11/10/2012] [Indexed: 02/05/2023]
|