1
|
García-Alfonso P, Jimenez-Fonseca P, Soto-Alsar J, Baraibar I, Santos C, La Casta A, Ghanem I, Pulido Cortijo G, Mariño Méndez A, Pazo-Cid R, Vera R, Melián M, Alcaide J, Graña B, Páez D, Gallego I, Lobo M, Borregón M, Fernández Montes A, Martínez de Castro E, Carmona-Bayonas A, Aranda E. Three-year survival follow-up of patients with gastrointestinal cancer treated during the COVID-19 pandemic in Spain: data from the PANDORA-TTD20 study. Oncologist 2024:oyae300. [PMID: 39550209 DOI: 10.1093/oncolo/oyae300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 09/10/2024] [Indexed: 11/18/2024] Open
Abstract
INTRODUCTION The initial SARS-CoV-2 pandemic wave in Spain in 2020 precipitated significant paradigm shifts in gastrointestinal oncology patient management. This study captures the "Zeitgeist" of this period by analyzing adaptive strategies, treatment modifications, and survival outcomes, leveraging a 3-year follow-up perspective to extract insights from this unprecedented experience. METHODS We conducted a multicenter, retrospective cohort study utilizing the RETUD-TTD registry, encompassing 703 patients across 19 Spanish centers in April 2020. We evaluated alterations in clinical practice, therapeutic approaches, coronavirus disease 2019 (COVID-19)-related impacts, and patient survival. A Bayesian hierarchical model was employed to identify potential regional-specific frailties. RESULTS The peak of the pandemic in April 2020 catalyzed substantial shifts in oncological care delivery. Outpatient consultations decreased by 13%, with a notable selection bias toward cases with more favorable prognostic indicators. Multidisciplinary tumor board discussions were significantly curtailed (eg, mean monthly colorectal cancer cases discussed was reduced from 40 to 23), compromising qualitative care measures. This occurred concurrently with an average of over 3 oncologists per center on medical leave. Contrary to initial concerns, the healthcare system demonstrated remarkable resilience. The majority of patients received standard-of-care therapies with regulatory approval, albeit with regimen modifications in 15% of cases. These adaptations included extended dosing intervals, dose intensity modulations, and transitions to oral formulations while maintaining unexpectedly stable long-term survival outcomes. The Bayesian frailty model detected minimal unmeasured prognostic factors related to geographic location, and the type of pandemic-induced adaptation did not significantly impact survival. The model revealed that coronavirus disease 2019's impact was less pronounced than other core prognostic variables. CONCLUSIONS The decentralized Spanish healthcare system exhibited substantial robustness in managing pre-pandemic diagnosed gastrointestinal malignancies, despite asymmetrical, and occasionally severe organizational disruptions. The insights gleaned from this experience could inform future crisis preparedness strategies and optimize care provision during subsequent public health emergencies.
Collapse
Affiliation(s)
- Pilar García-Alfonso
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, 28007, Spain
| | - Paula Jimenez-Fonseca
- Medical Oncology Department, Hospital Universitario Central de Asturias (HUCA), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, 33011, Spain
| | - Javier Soto-Alsar
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, 28007, Spain
| | - Iosune Baraibar
- Medical Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autónoma de Barcelona, CIBERONC, Barcelona, 08035, Spain
| | - Cristina Santos
- Medical Oncology Department, Institut Català d'Oncologia (ICO), Translational Research Laboratory, ICO-Bellvitge Biomedical Research Institute (IDIBELL)-CIBERONC, Barcelona, 08908, Spain
| | - Adelaida La Casta
- Medical Oncology Department, Hospital Universitario de Donostia, Guipúzcoa, 20014, Spain
| | - Ismael Ghanem
- Medical Oncology Department, Hospital Universitario La Paz, Madrid, 28046, Spain
| | - Gema Pulido Cortijo
- Medical Oncology Department, Hospital Universitario Reina Sofía, Universidad de Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), CIBERONC, Instituto de Salud Carlos III (ISCIII), Córdoba, 14004, Spain
| | | | - Roberto Pazo-Cid
- Medical Oncology Department, Hospital Universitario Miguel Servet, Aragon Institute of Biomedical Research (IISA), Spanish Cancer Network (RTICC), ISCIII, Zaragoza, 50009, Spain
| | - Ruth Vera
- Medical Oncology Department, Hospital Universitario de Navarra, Pamplona, 31008, Spain
| | - Marcos Melián
- Medical Oncology Department, Instituto Valenciano de Oncología (IVO), Valencia, 46009, Spain
| | - Julia Alcaide
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga, 29010, Spain
| | - Begoña Graña
- Medical Oncology Department, Complejo Hospitalario Universitario de A Coruña (CHUA), Instituto de Investigación Biomédica de A Coruña (INIBIC), Coruña, 15006, Spain
| | - David Páez
- Medical Oncology Department, Hospital Santa Creu i Sant Pau, Barcelona, 08041, Spain
| | - Inmaculada Gallego
- Medical Oncology Department, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBIS), Sevilla, 41013, Spain
| | - Miriam Lobo
- Medical Oncology Department, Hospital General Universitario de Valencia, Valencia, 46014, Spain
| | - Miguel Borregón
- Medical Oncology Department, Hospital General Universitario de Elche, Elche, 03203, Spain
| | - Ana Fernández Montes
- Medical Oncology Department, Complejo Hospitalario Universitario de Orense (CHUO), Orense, 32005, Spain
| | - Eva Martínez de Castro
- Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, Instituto De Investigación Marqués de Valdecilla (IDIVAL), Santander, 39008, Spain
| | - Alberto Carmona-Bayonas
- Medical Oncology Department, Hospital Universitario Morales Meseguer, Universidad de Murcia, Instituto Murciano de Investigación Biosanitaria (IMIB), Murcia, 30008, Spain
| | - Enrique Aranda
- Medical Oncology Department, Hospital Universitario Reina Sofía, Universidad de Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), CIBERONC, Instituto de Salud Carlos III (ISCIII), Córdoba, 14004, Spain
| |
Collapse
|
2
|
Hu B, Zhang W, Zhang C, Li C, Zhang N, Pan K, Ge X, Wan T. CCNI2 promotes pancreatic cancer through PI3K/AKT signaling pathway. BIOMOLECULES & BIOMEDICINE 2024; 24:323-336. [PMID: 37540586 PMCID: PMC10950348 DOI: 10.17305/bb.2023.9337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/21/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
Globally, pancreatic cancer is recognized as one of the deadliest malignancies that lacks effective targeted therapies. This study aims to explore the role of cyclin I-like protein (CCNI2), a homolog of cyclin I (CCNI), in the progression of pancreatic cancer, thereby providing a theoretical basis for its treatment. Firstly, the expression of CCNI2 in pancreatic cancer tissues was determined through immunohistochemical staining. The biological role of CCNI2 in pancreatic cancer cells was further assessed using both in vitro and in vivo loss/gain-of-function assays. Our data revealed that CCNI2 expression was abnormally elevated in pancreatic cancer, and clinically, increased CCNI2 expression generally correlated with reduced overall survival. Functionally, CCNI2 contributed to the malignant progression of pancreatic cancer by promoting the proliferation and migration of tumor cells. Consistently, in vivo experiments verified that CCNI2 knockdown impaired the tumorigenic ability of pancreatic cancer cells. Moreover, the addition of phosphatidylinositol 3-kinase (PI3K) inhibitors could partially reverse the promoting effect of CCNI2 on the malignant phenotypes of pancreatic cancer cells. CCNI2 promoted pancreatic cancer through PI3K/protein kinase B (AKT) signaling pathway, indicating its potential as a prognostic marker and therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Bingyang Hu
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Haidian District, Beijing, China
| | - Wenzhi Zhang
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Haidian District, Beijing, China
| | - Changsheng Zhang
- Department of General Surgery, Kaifeng Central Hospital, Longting District, Kaifeng, Henan Province, China
| | - Chonghui Li
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Haidian District, Beijing, China
| | - Ning Zhang
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Haidian District, Beijing, China
| | - Ke Pan
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Haidian District, Beijing, China
| | - Xinlan Ge
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Haidian District, Beijing, China
| | - Tao Wan
- Department of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Chinese PLA Medical School, Haidian District, Beijing, China
| |
Collapse
|
3
|
Samavarchi Tehrani S, Esmaeili F, Shirzad M, Goodarzi G, Yousefi T, Maniati M, Taheri-Anganeh M, Anushiravani A. The critical role of circular RNAs in drug resistance in gastrointestinal cancers. Med Oncol 2023; 40:116. [PMID: 36917431 DOI: 10.1007/s12032-023-01980-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
Nowadays, drug resistance (DR) in gastrointestinal (GI) cancers, as the main reason for cancer-related mortality worldwide, has become a serious problem in the management of patients. Several mechanisms have been proposed for resistance to anticancer drugs, including altered transport and metabolism of drugs, mutation of drug targets, altered DNA repair system, inhibited apoptosis and autophagy, cancer stem cells, tumor heterogeneity, and epithelial-mesenchymal transition. Compelling evidence has revealed that genetic and epigenetic factors are strongly linked to DR. Non-coding RNA (ncRNA) interferences are the most crucial epigenetic alterations explored so far, and among these ncRNAs, circular RNAs (circRNAs) are the most emerging members known to have unique properties. Due to the absence of 5' and 3' ends in these novel RNAs, the two ends are covalently bonded together and are generated from pre-mRNA in a process known as back-splicing, which makes them more stable than other RNAs. As far as the unique structure and function of circRNAs is concerned, they are implicated in proliferation, migration, invasion, angiogenesis, metastasis, and DR. A clear understanding of the molecular mechanisms responsible for circRNAs-mediated DR in the GI cancers will open a new window to the management of GI cancers. Hence, in the present review, we will describe briefly the biogenesis, multiple features, and different biological functions of circRNAs. Then, we will summarize current mechanisms of DR, and finally, discuss molecular mechanisms through which circRNAs regulate DR development in esophageal cancer, pancreatic cancer, gastric cancer, colorectal cancer, and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sadra Samavarchi Tehrani
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fataneh Esmaeili
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Moein Shirzad
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Golnaz Goodarzi
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tooba Yousefi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmood Maniati
- Department of English, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Amir Anushiravani
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Cura Daball P, Tröger H, Daum S. [Long-term response in advanced pancreatic adenocarcinoma - a case report and literature review]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:1510-1516. [PMID: 34905798 DOI: 10.1055/a-1695-3528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Pancreatic cancer is still considered one of the most aggressive types of cancer and is associated with a very poor prognosis although there have been improvements in diagnostics and chemotherapy regimes in recent years. A cure can only be achieved through complete resection which is only possible when diagnosed at a very early stage, though this is rarely the case. We report on a patient with stage IV adenocarcinoma of the pancreas in which several therapeutically actionable mutations could be detected and discuss new options of targeted therapies. CASE REPORT A patient in his 50s was diagnosed with metastatic adenocarcinoma of the pancreas. The patient showed an excellent response to platinum-based chemotherapy with FOLFIRINOX. When a germline mutation in the BRCA-2 gene could be identified, he took part in the POLO-study receiving a maintenance therapy with the PARP-Inhibitor Olaparib. Due to a relapse, 2nd and 3rd line chemotherapy regimens were applied with Gemcitabine combined with Nab-Paclitaxel and later with Erlotinib. Although an activating mutation in the KRAS-gene could be detected as well, the patient rejected further experimental treatment. CONCLUSION Identifying predictive factors and specific targetable mutations in patients with advanced pancreatic cancer is needed to be able to apply more individual and specific therapies in order to improve outcomes.
Collapse
Affiliation(s)
- Paola Cura Daball
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Hanno Tröger
- Medizinische Klinik 1, St Joseph Krankenhaus Berlin-Tempelhof, Berlin, Germany
| | - Severin Daum
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
5
|
Keyl J, Kasper S, Wiesweg M, Götze J, Schönrock M, Sinn M, Berger A, Nasca E, Kostbade K, Schumacher B, Markus P, Albers D, Treckmann J, Schmid KW, Schildhaus HU, Siveke JT, Schuler M, Kleesiek J. Multimodal survival prediction in advanced pancreatic cancer using machine learning. ESMO Open 2022; 7:100555. [PMID: 35988455 PMCID: PMC9588888 DOI: 10.1016/j.esmoop.2022.100555] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022] Open
Abstract
Background Existing risk scores appear insufficient to assess the individual survival risk of patients with advanced pancreatic ductal adenocarcinoma (PDAC) and do not take advantage of the variety of parameters that are collected during clinical care. Methods In this retrospective study, we built a random survival forest model from clinical data of 203 patients with advanced PDAC. The parameters were assessed before initiation of systemic treatment and included age, CA19-9, C-reactive protein, metastatic status, neutrophil-to-lymphocyte ratio and total serum protein level. Separate models including imaging and molecular parameters were built for subgroups. Results Over the entire cohort, a model based on clinical parameters achieved a c-index of 0.71. Our approach outperformed the American Joint Committee on Cancer (AJCC) staging system and the modified Glasgow Prognostic Score (mGPS) in the identification of high- and low-risk subgroups. Inclusion of the KRAS p.G12D mutational status could further improve the prediction, whereas radiomics data of the primary tumor only showed little benefit. In an external validation cohort of PDAC patients with liver metastases, our model achieved a c-index of 0.67 (mGPS: 0.59). Conclusions The combination of multimodal data and machine-learning algorithms holds potential for personalized prognostication in advanced PDAC already at diagnosis. We developed a machine-learning-based prediction model that outperforms the AJCC staging system and mGPS. Applying our model to an external validation cohort demonstrates generalizability. Explainable machine learning enables to understand the decision making of our model and identifies relevant parameters. Combining clinical, imaging and genetic data holds potential for personalized prognostication in advanced PDAC.
Collapse
Affiliation(s)
- J Keyl
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany.
| | - S Kasper
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - M Wiesweg
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - J Götze
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M Schönrock
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - M Sinn
- Department of Internal Medicine II, Oncology, Hematology, BMT and Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - A Berger
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
| | - E Nasca
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany
| | - K Kostbade
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - B Schumacher
- Department of Gastroenterology, Elisabeth Hospital Essen, Essen, Germany
| | - P Markus
- Department of General Surgery and Traumatology, Elisabeth Hospital Essen, Essen, Germany
| | - D Albers
- Department of Gastroenterology, Elisabeth Hospital Essen, Essen, Germany
| | - J Treckmann
- Department of General, Visceral and Transplant Surgery, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - K W Schmid
- Medical Faculty, University of Duisburg-Essen, Essen, Germany; Institute of Pathology, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - H-U Schildhaus
- Medical Faculty, University of Duisburg-Essen, Essen, Germany; Institute of Pathology, West German Cancer Center, University Hospital Essen (AöR), Essen, Germany
| | - J T Siveke
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany; Bridge Institute of Experimental Tumor Therapy (BIT), West German Cancer Center, University Hospital Essen (AöR), Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Consortium (DKTK) Partner site Essen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Schuler
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen (AöR), University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK), Partner site University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - J Kleesiek
- Institute for AI in Medicine (IKIM), University Hospital Essen (AöR), Essen, Germany; Medical Faculty, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
6
|
Ioannou LJ, Maharaj AD, Zalcberg JR, Loughnan JT, Croagh DG, Pilgrim CH, Goldstein D, Kench JG, Merrett ND, Earnest A, Burmeister EA, White K, Neale RE, Evans SM. Prognostic models to predict survival in patients with pancreatic cancer: a systematic review. HPB (Oxford) 2022; 24:1201-1216. [PMID: 35289282 DOI: 10.1016/j.hpb.2022.01.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) has poor survival. Current treatments offer little likelihood of cure or long-term survival. This systematic review evaluates prognostic models predicting overall survival in patients diagnosed with PDAC. METHODS We conducted a comprehensive search of eight electronic databases from their date of inception through to December 2019. Studies that published models predicting survival in patients with PDAC were identified. RESULTS 3297 studies were identified; 187 full-text articles were retrieved and 54 studies of 49 unique prognostic models were included. Of these, 28 (57.1%) were conducted in patients with advanced disease, 17 (34.7%) with resectable disease, and four (8.2%) in all patients. 34 (69.4%) models were validated, and 35 (71.4%) reported model discrimination, with only five models reporting values >0.70 in both derivation and validation cohorts. Many (n = 27) had a moderate to high risk of bias and most (n = 33) were developed using retrospective data. No variables were unanimously found to be predictive of survival when included in more than one study. CONCLUSION Most prognostic models were developed using retrospective data and performed poorly. Future research should validate instruments performing well locally in international cohorts and investigate other potential predictors of survival.
Collapse
Affiliation(s)
- Liane J Ioannou
- Public Health and Preventive Medicine, Monash University, Victoria, Australia.
| | - Ashika D Maharaj
- Public Health and Preventive Medicine, Monash University, Victoria, Australia
| | - John R Zalcberg
- Public Health and Preventive Medicine, Monash University, Victoria, Australia
| | - Jesse T Loughnan
- Public Health and Preventive Medicine, Monash University, Victoria, Australia
| | - Daniel G Croagh
- Department of Surgery, Monash Health, Monash University, Victoria, Australia
| | - Charles H Pilgrim
- Department of Surgery, Alfred Health, Monash University, Victoria, Australia
| | - David Goldstein
- Prince of Wales Clinical School, UNSW Medicine, NSW, Australia
| | - James G Kench
- Royal Prince Alfred Hospital, Camperdown, NSW, Australia; Central Clinical School, University of Sydney, NSW, Australia
| | - Neil D Merrett
- School of Medicine, Western Sydney University, NSW, Australia
| | - Arul Earnest
- Public Health and Preventive Medicine, Monash University, Victoria, Australia
| | | | - Kate White
- Sydney Nursing School, University of Sydney, NSW, Australia
| | - Rachel E Neale
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Sue M Evans
- Public Health and Preventive Medicine, Monash University, Victoria, Australia
| |
Collapse
|
7
|
Yang Q, Mao Y, Xie H, Qin T, Mai Z, Cai Q, Wen H, Li Y, Zhang R, Liu L. Identifying Outcomes of Patients With Advanced Pancreatic Adenocarcinoma and RECIST Stable Disease Using Radiomics Analysis. JCO Precis Oncol 2022; 6:e2100362. [PMID: 35319966 PMCID: PMC8966975 DOI: 10.1200/po.21.00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Few studies have explored the biomarkers for predicting the heterogeneous outcomes of patients with advanced pancreatic adenocarcinoma showing stable disease (SD) on the initial postchemotherapy computed tomography. We aimed to devise a radiomics signature (RS) to predict these outcomes for further risk stratification.
Collapse
Affiliation(s)
- Qiuxia Yang
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yize Mao
- Department of Pancreatic-Biliary Surgical Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Hui Xie
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tao Qin
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhijun Mai
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qian Cai
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hailin Wen
- Cancer Hospital Chinese Academy of Medical Science, Shenzhen Center, Shenzhen, China
| | - Yong Li
- Department of Medical Imaging Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rong Zhang
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Lizhi Liu
- Department of Medical Imaging Center, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
8
|
Shibuki T, Mizuta T, Shimokawa M, Koga F, Ueda Y, Nakazawa J, Komori A, Otsu S, Arima S, Fukahori M, Makiyama A, Taguchi H, Honda T, Mitsugi K, Nio K, Ide Y, Ureshino N, Shirakawa T, Otsuka T. Prognostic nomogram for patients with unresectable pancreatic cancer treated with gemcitabine plus nab-paclitaxel or FOLFIRINOX: A post-hoc analysis of a multicenter retrospective study in Japan (NAPOLEON study). BMC Cancer 2022; 22:19. [PMID: 34980029 PMCID: PMC8722136 DOI: 10.1186/s12885-021-09139-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/22/2021] [Indexed: 01/04/2023] Open
Abstract
Background No reliable nomogram has been developed until date for predicting the survival in patients with unresectable pancreatic cancer undergoing treatment with gemcitabine plus nab–paclitaxel (GnP) or FOLFIRINOX. Methods This analysis was conducted using clinical data of Japanese patients with unresectable pancreatic cancer undergoing GnP or FOLFIRINOX treatment obtained from a multicenter study (NAPOLEON study). A Cox proportional hazards model was used to identify the independent prognostic factors. A nomogram to predict 6–, 12–, and 18–month survival probabilities was generated, validated by using the concordance index (C–index), and calibrated by the bootstrapping method. And then, we attempted risk stratification for survival by classifying the patients according to the sum of the scores on the nomogram (total nomogram points). Results A total of 318 patients were enrolled. A prognostic nomogram was generated using data on the Eastern Cooperative Oncology Group performance status, liver metastasis, serum LDH, serum CRP, and serum CA19–9. The C–indexes of the nomogram were 0.77, 0.72 and 0.70 for 6–, 12–, and 18–month survival, respectively. The calibration plot showed optimal agreement at all points. Risk stratification based on tertiles of the total nomogram points yielded clear separations of the survival curves. The median survival times in the low–, moderate–, and high–risk groups were 15.8, 12.8 and 7.8 months (P<0.05), respectively. Conclusions Our nomogram might be a convenient and inexpensive tool to accurately predict survival in Japanese patients with unresectable pancreatic cancer undergoing treatment with GnP or FOLFIRINOX, and will help clinicians in selecting appropriate therapeutic strategies for individualized management. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-09139-y.
Collapse
Affiliation(s)
- Taro Shibuki
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-ko, Arita-cho, Nishi-matsuura-gun, Saga, 849-4193, Japan.,Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa-shi, Chiba, 277-8577, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-ko, Arita-cho, Nishi-matsuura-gun, Saga, 849-4193, Japan.,Department of Internal Medicine, Fujikawa Hospital, 1-2-6 Matsubara, Saga-shi, Saga, 840-0831, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Hospital Organization Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka-shi, Fukuoka, 811-1395, Japan.,Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube, Yamaguchi, 755-8505, Japan
| | - Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, 2-1-1 Nagamine-minami, Higashi-ku, Kumamoto-shi, Kumamoto, 861-8520, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-cho, Kagoshima-shi, Kagoshima, 890-8760, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita, 879-5593, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-shi, Kagoshima, 890-8520, Japan
| | - Masaru Fukahori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume-shi, Fukuoka, 830-0011, Japan
| | - Akitaka Makiyama
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-ku, Kitakyushu-shi, Fukuoka, 806-8501, Japan.,Cancer Center, Gifu University Hospital, 1-1 Yanagido, Gifu-shi, Gifu, 501-1194, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-machi, Satsumasendai-shi, Kagoshima, 895-0074, Japan.,Department of Gastroenterology, Izumi General Medical Center, 520 Myojincho, Izumi-shi, Kagoshima, 899-0131, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki, 852-8501, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-8539, Japan.,Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-cho, Sasebo-shi, Nagasaki, 857-8575, Japan
| | - Kenta Nio
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-8539, Japan.,Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-cho, Sasebo-shi, Nagasaki, 857-8575, Japan
| | - Yasushi Ide
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada, Karatsu-shi, Saga, 847-8588, Japan
| | - Norio Ureshino
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan.,Department of Medical Oncology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba, 292-8535, Japan
| | - Tsuyoshi Shirakawa
- Department of Medical Oncology, Fukuoka Wajiro Hospital, 2-2-75 Wajirogaoka, Higashi-ku, Fukuoka-shi, Fukuoka, 811-0213, Japan. .,Karatsu Higashi-matsuura Medical Association Center, 2566-11 Chiyoda-machi, Karatsu-shi, Saga, 847-0041, Japan.
| | - Taiga Otsuka
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga, 840-8571, Japan.,Department of Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-0072, Japan
| |
Collapse
|
9
|
Chang C, Meng L, Li X, Cheng K, Yi C, Peng B, Ma J, Cao D. A Modified Regimen of 21-day Nab-Paclitaxel Plus Gemcitabine in Locally Advanced or Metastatic Pancreatic Cancer: A Retrospective Real-World Study. Cancer Control 2022; 29:10732748221141233. [PMID: 36423238 PMCID: PMC9703471 DOI: 10.1177/10732748221141233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND It is 1 of the standard treatment options for metastasis pancreatic cancer to receive nab-paclitaxel (125 mg/m2) plus gemcitabine (1000 mg/m2) on days 1, 8 and 15 every 28 days. Some patients showed intolerance and inconvenience to this therapeutic regimen. Thus, we conducted this retrospective real-world study to determine the efficacy and tolerability of a modified 21-day nab-paclitaxel plus gemcitabine (nab-P/Gem) regimen for the first-line treatment of locally advanced or metastatic pancreatic cancer. METHODS Patients with locally advanced and metastatic pancreatic cancer treated with nab-paclitaxel (125 mg/m2) plus gemcitabine (1000 mg/m2) on days 1 and 8 every 21-day at West China Hospital and Shang Jin Hospital of Sichuan University from Mar 2018 to Dec 2021 were reviewed retrospectively. Clinical characteristics of patients were collected. The progression-free survival, overall survival, objective response rate, disease control rate, and toxicity were evaluated. RESULTS A total of 113 patients who received the modified regimen of 21-day nab-P/Gem chemotherapy were included. The median overall survival was 9.3 months and the median progression-free survival was 4.4 months. The objective response rate and disease control rate were 18.6% and 56.7%, respectively. The median relative dose intensity for this modified regimen was 65%. The adverse events were mild to moderate, and the most common grade 3 or 4 treatment-related adverse events were neutropenia (21%) and leukopenia (16%). CONCLUSIONS Our study showed that this modified regimen of 21-day nab-P/Gem for locally advanced and metastatic pancreatic cancer had comparable efficacy and tolerable toxicity. This treatment may provide a considerable option for pancreatic cancer patients who desire a modified schedule. The modified regimen of 21-day nab-P/Gem is also an option worth considering during the coronavirus disease 2019 pandemic for minimizing the number of visits and limiting the risk of exposure.
Collapse
Affiliation(s)
- Chen Chang
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, PR China
| | - Lingwei Meng
- Department of General Surgery, Shang Jin Hospital of West China Hospital, Sichuan University, PR China
| | - Xiaofen Li
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, PR China
| | - Ke Cheng
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, PR China
| | - Cheng Yi
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, PR China
| | - Bing Peng
- Department of General Surgery, Shang Jin Hospital of West China Hospital, Sichuan University, PR China
| | - Ji Ma
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, PR China
| | - Dan Cao
- Department of Abdominal Oncology, Cancer Center, West China Hospital, Sichuan University, PR China
| |
Collapse
|
10
|
Rajpurohit T, Bhattacharya S. Moving Towards Dawn: KRas Signaling and Treatment in Pancreatic Ductal Adenocarcinoma. Curr Mol Pharmacol 2022; 15:904-928. [PMID: 35088684 DOI: 10.2174/1874467215666220128161647] [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: 07/26/2021] [Revised: 10/20/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022]
Abstract
"Pancreatic ductal adenocarcinoma (PDAC)" is robust, nearly clueless, and all-around deadly among all tumors. Below 10 %, the general 5-year endurance period has remained adamantly unaltered in the last 30 years, regardless of enormous clinical and therapeutic endeavors. The yearly number of deaths is more than the number of recently analyzed cases. Not a classic one, but "Carbohydrate Antigen CA19- 9" remains the prevailing tool for diagnosis. MicroRNAs and non-invasive techniques are now incorporated for the effective prognosis of PDAC than just CA19-9. Mutated "Rat sarcoma virus Ras" conformation "V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog KRas" is 95 % accountable for PDAC, and its active (GTP-bound) formation activates signaling cascade comprising "Rapidly accelerated fibrosarcoma Raf"/"Mitogen-activated protein kinase MEK"/ "Extracellular signal-regulated kinase ERK" with "Phosphoinositide 3-kinase PI3K"/ "protein kinase B Akt"/ "mammalian target of rapamycin mTOR" pathways. KRas has acquired the label of 'undruggable' since the crosstalk in the nexus of pathways compensates for Raf and PI3K signaling cascade blocking. It is arduous to totally regulate KRascoordinated PDAC with traditional medicaments like "gemcitabine GEM" plus nabpaclitaxel/ FOLFIRINOX. For long-haul accomplishments aiming at KRas, future endeavors should be directed to combinatorial methodologies to adequately block KRas pathways at different standpoints. Currently they are contributing to healing PDAC. In this review article, we outline the function of KRas in carcinogenesis in PDAC, its signaling cascade, former techniques utilized in hindering Kras, current and future possibilities for targeting Kras.
Collapse
Affiliation(s)
- Tarun Rajpurohit
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra 425405, India
| |
Collapse
|
11
|
Yu KH, Ozer M, Cockrum P, Surinach A, Wang S, Chu BC. Real-world prognostic factors for survival among treated patients with metastatic pancreatic ductal adenocarcinoma. Cancer Med 2021; 10:8934-8943. [PMID: 34811961 PMCID: PMC8683530 DOI: 10.1002/cam4.4415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/04/2021] [Accepted: 10/24/2021] [Indexed: 12/12/2022] Open
Abstract
Background Many real‐world studies of patients with metastatic pancreatic ductal adenocarcinoma (mPDAC) are restricted to single centers, limiting the generalizability of their insights. This study aimed to identify important population‐based predictors for survival in patients diagnosed with mPDAC in a broader setting. Methods Data between 1 January 2017 and 31 December 2019 were extracted from the Flatiron Health EHR database. Treatment‐specific predictive models were generated for patients treated with first‐line gemcitabine+nabpaclitaxel (GNP), FOLFIRINOX, gemcitabine monotherapy (gem‐mono), and second‐line liposomal irinotecan‐based regimens. The holdout method was used for cross‐validation. Age at diagnosis, sex, BMI, smoking status, and ECOG performance score were included in all models with additional demographic, clinical characteristics, and hematological function assessed for inclusion. Results Of the 3625 patients, 43% received GNP, 26% received FOLFIRINOX, 7% received gem‐mono, and 23% received other regimens; 40% (n = 1448) advanced to the second line. Among all first‐line patients, the following were included in the final model: prior surgery, white blood cell (WBC) counts, serum albumin (SA), liver function tests (LFTs), serum bilirubin, serum carbohydrate antigen 19–9, and ascites. Models for patients receiving specific therapies differed from the overall model, GNP (ascites removed), FOLFIRINOX (stage at initial diagnosis added), and gem‐mono (LFTs omitted). Alkaline phosphatase (ALP), SA, and WBC counts were important predictors of survival among patients treated with second‐line liposomal irinotecan. Across all regimens, the strongest predictors of survival were ECOG score, SA, and ALP. Conclusions In this real‐world study of patients with mPDAC, important population prognostic factors of survival were identified in a large cohort of patients receiving systemic treatment.
Collapse
Affiliation(s)
- Kenneth H Yu
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - Muhammet Ozer
- Capital Health Medical Center, Trenton, New Jersey, USA
| | | | | | - Shu Wang
- Genesis Research, Hoboken, New Jersey, USA
| | | |
Collapse
|
12
|
Nielson CM, Bylsma LC, Fryzek JP, Saad HA, Crawford J. Relative Dose Intensity of Chemotherapy and Survival in Patients with Advanced Stage Solid Tumor Cancer: A Systematic Review and Meta-Analysis. Oncologist 2021; 26:e1609-e1618. [PMID: 33973301 PMCID: PMC8417866 DOI: 10.1002/onco.13822] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 04/30/2021] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Chemotherapy-induced toxicities lead to therapy dose reduction or delay, affecting patient outcomes. This systematic review and meta-analysis evaluated the impact of relative dose intensity (RDI) on survival in adult patients with solid tumor cancer on nonadjuvant-based chemotherapy regimens. METHODS PubMed, Embase, and Web of Science databases were searched for peer-reviewed English journal articles or congress abstracts evaluating association between RDI and survival; observational studies, case series of ≥20 patients, and clinical trials published between 2013 and 2020 were eligible. Meta-analyses were conducted to quantify the association between RDI levels and overall survival (OS) among studies reporting a hazard ratio (HR) for OS by similar tumor types, regimens, and RDI. Forest plots represented summary HR and 95% confidence interval (CI); Cochran's Q and I2 tests evaluated study heterogeneity. RESULTS Overall, 919 articles were reviewed and 22 included; seven were eligible for meta-analysis. Significantly shorter OS at RDI <80% versus ≥80% and <85% versus ≥85% was observed upon meta-analysis of four carboplatin-based studies for breast, non-small cell lung, or ovarian cancer (HR 1.17; 95% CI: 1.07-1.27) and three FOLFOX-, FOLFIRI-, or FOLFIRINOX-based studies for colorectal or pancreatic cancer (HR 1.39; 95% CI: 1.03-1.89). Grade 3 or higher hematologic toxicities were higher for carboplatin-based regimens (thrombocytopenia: 14%-22%; anemia: 15%-19%; neutropenia: 24%-58%) than FOLFOX-, FOLFIRI-, or FOLFIRINOX-based regimens (thrombocytopenia: 1%-4%; anemia: 5%-19%; neutropenia: 19%-47%). CONCLUSION The results suggested longer OS with RDI ≥80% or ≥85% for both regimens, indicating that management of toxicities across treatment modalities may contribute to maintenance of higher RDI and benefit survival for patients with advanced solid tumors. IMPLICATIONS FOR PRACTICE Chemotherapy-induced toxicities lead to dose reduction and/or treatment delay, thus affecting patient outcomes. Results of this systematic review and meta-analysis, evaluating the impact of relative dose intensity (RDI) on survival of patients with solid tumors on nonadjuvant-based chemotherapy regimens, demonstrate a longer overall survival with RDI levels of at least 80% for patients with solid tumors on carboplatin-based and FOLFOX-, FOLFIRI-, or FOLFIRINOX-based chemotherapy regimens, suggesting a protective effect of maintaining RDI ≥80% or ≥ -85%. Although grade 3 or higher hematologic toxicities occurred more in carboplatin-based studies, managing toxicities across treatment regimens may contribute to maintenance of higher RDI and ultimately benefit overall survival.
Collapse
Affiliation(s)
| | - Lauren C. Bylsma
- EpidStrategies, A Division of ToxStrategies, Inc.Ann ArborMichiganUSA
| | - Jon P. Fryzek
- EpidStrategies, A Division of ToxStrategies, Inc.RockvilleMarylandUSA
| | | | | |
Collapse
|
13
|
Colloca G, Venturino A. Peripheral Blood Cell Variables Related to Systemic Inflammation in Patients With Unresectable or Metastatic Pancreatic Cancer: A Systematic Review and Meta-Analysis. Pancreas 2021; 50:1131-1136. [PMID: 34714276 DOI: 10.1097/mpa.0000000000001878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
ABSTRACT Systemic inflammatory response (SIR) plays a central role in the prognosis of unresectable or metastatic pancreatic ductal adenocarcinoma (mPDAC), and many SIR-related peripheral blood cell (PBC)-derived variables have been proposed as prognostic factors. The study aims to perform a systematic review and, for the more studied PBC-derived variables, a meta-analysis. A systematic review from 2000 to 2020 on 2 databases by predefined criteria was performed for PBC-derived variables in patients with mPDAC receiving chemotherapy in relation with overall survival. Eligible studies were selected by inclusion criteria, and only the PBC variables reported in at least 10 studies were evaluated by meta-analysis. Three hundred and eighty articles were found, and 28 studies were selected. Neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) were reported in 28 and 10 articles, respectively. The subsequent meta-analyses supported the prognostic effect for both, NLR (hazard ratio, 2.10; 95% confidence interval, 1.87-2.37) and PLR (hazard ratio, 1.22; 95% confidence interval, 1.08-1.37). Heterogeneity was significant for NLR (I2 = 62%) and low for PLR (I2 = 24%). Among SIR-related PBC-derived variables, NLR is the most suitable prognostic factor for future clinical trials of patients with mPDAC.
Collapse
Affiliation(s)
- Giuseppe Colloca
- From the Department of Oncology, Ospedale Saint Charles, Bordighera, Italy
| | | |
Collapse
|
14
|
Sato H, Goto T, Hayashi A, Kawabata H, Okada T, Takauji S, Sasajima J, Enomoto K, Fujiya M, Oyama K, Ono Y, Sugitani A, Mizukami Y, Okumura T. Prognostic significance of skeletal muscle decrease in unresectable pancreatic cancer: Survival analysis using the Weibull exponential distribution model. Pancreatology 2021; 21:892-902. [PMID: 33722506 DOI: 10.1016/j.pan.2021.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/21/2021] [Accepted: 03/01/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND/OBJECTIVES Decrease in skeletal muscle mass and function is associated with a poor prognosis following surgical resection of pancreatic ductal adenocarcinomas (PDAs). This study evaluated whether skeletal muscle mass decrease affects PDA outcomes. METHODS Data of 112 patients with advanced and unresectable PDA who underwent chemotherapy in a single institution were retrospectively analyzed. Information on age, sex, hematological investigations, including systemic inflammation-based markers and nutritional assessment biomarkers, and imaging parameters of skeletal muscle mass and visceral adipose tissue were retrieved from the patients' medical records. The efficiency of the Cox, Weibull, and standardized exponential models were compared using hazard ratios and the Akaike Information Criterion (AIC). RESULTS Results from the Weibull, Cox, and standardized exponential model analyses indicated that low skeletal muscle mass, Eastern Cooperative Oncology Group performance status (PS), and the requirement of biliary drainage were associated with the highest risk of death, followed by carcinoembryonic antigen (CEA) levels and the presence of ascites. The AIC value from the four significant parameters was lowest for the Weibull-exponential distribution (222.3) than that of the Cox (653.7) and standardized exponential models (265.7). We developed a model for estimating the 1-year survival probability using the Weibull-exponential distribution. CONCLUSIONS Low-skeletal muscle index, PS, requirement of biliary drainage, CEA levels, and presence of ascites are independent factors for predicting poor patient survival after chemotherapy. Improved survival modeling using a parametric approach may accurately predict the outcome of patients with advanced-stage PDA.
Collapse
Affiliation(s)
- Hiroki Sato
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Takuma Goto
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Akihiro Hayashi
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Hidemasa Kawabata
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Tetsuhiro Okada
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Shuhei Takauji
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Junpei Sasajima
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Katsuro Enomoto
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Mikihiro Fujiya
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Kyohei Oyama
- Department of Cardiovascular Surgery, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| | - Yusuke Ono
- Institute of Biomedical Research, Sapporo-Higashi Tokushukai Hospital, 3-1, North-33, East-14, Higashi-Ku, Sapporo, Japan
| | - Ayumu Sugitani
- Institute of Biomedical Research, Sapporo-Higashi Tokushukai Hospital, 3-1, North-33, East-14, Higashi-Ku, Sapporo, Japan
| | - Yusuke Mizukami
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan; Institute of Biomedical Research, Sapporo-Higashi Tokushukai Hospital, 3-1, North-33, East-14, Higashi-Ku, Sapporo, Japan.
| | - Toshikatsu Okumura
- Department of Medicine, Asahikawa Medical University, 2-1 Midorigaoka-Higashi, Asahikawa, Japan
| |
Collapse
|
15
|
Koga F, Kawaguchi Y, Shimokawa M, Murayama K, Nakashita S, Oza N, Ureshino N, Takahashi H, Ueda Y, Nakazawa J, Komori A, Otsu S, Arima S, Fukahori M, Makiyama A, Taguchi H, Honda T, Shibuki T, Nio K, Ide Y, Mizuta T, Shirakawa T, Otsuka T, Mitsugi K. Gemcitabine plus nab-paclitaxel in older patients with metastatic pancreatic cancer: A post-hoc analysis of the real-world data of a multicenter study (the NAPOLEON study). J Geriatr Oncol 2021; 13:82-87. [PMID: 34215525 DOI: 10.1016/j.jgo.2021.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 06/18/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023]
Abstract
OBJECTIVES This study aimed to examine the efficacy and safety of gemcitabine plus nab-paclitaxel (GnP) in older patients with metastatic pancreatic cancer (MPC), especially those ≥75 years old. MATERIALS AND METHODS This study retrospectively enrolled 153 patients with MPC who received GnP as first-line chemotherapy. Patients ≥75 years old were allocated to the older group, and those <75 years old were assigned to the non-older group. We compared safety, antitumor efficacy, and survival between the two groups. In the older group, prognostic indicators of survival were also assessed. RESULTS The pretreatment characteristics of the two groups were not significantly different excluding age, history of malignancy, and C-reactive protein levels. The initial dose and relative dose intensities of GnP were significantly lower in the older group. There were no significant differences in the adverse event and antitumor response rates between the two groups. Median progression-free survival and overall survival were 5.5 and 12.0 months, respectively, in the older group, versus 6.0 and 11.1 months, respectively, in the non-older group. In the older group, a Geriatric Nutrition Risk Index (GNRI) of less than 86 was associated with poor prognosis. CONCLUSION GnP exhibited acceptable efficacy and safety in patients ≥75 years old with MPC. GNRI might be helpful for identifying older individuals at higher risk of unfavorable outcomes.
Collapse
Affiliation(s)
- Futa Koga
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga 840-8571, Japan; Saga University Graduate School of Medical Science, 5-1-1 Nabeshima, Saga-shi, Saga 849-8501, Japan
| | - Yasunori Kawaguchi
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga 840-8571, Japan; Department of Gastroenterology, Asakura Medical Association Hospital, 422-1 Raiha, Asakura-shi, Fukuoka 838-0069, Japan
| | - Mototsugu Shimokawa
- Clinical Research Institute, National Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka-shi, Fukuoka, 811-1395, Japan; Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1 Minamikogushi, Ube-shi, Yamaguchi 755-8505, Japan
| | - Kenichiro Murayama
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga 840-8571, Japan
| | - Shunya Nakashita
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga 840-8571, Japan
| | - Noriko Oza
- Department of Hepatobiliary and Pancreatology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga 840-8571, Japan
| | - Norio Ureshino
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga 840-8571, Japan; Department of Medical Oncology, Kimitsu Chuo Hospital, 1010 Sakurai, Kisarazu-shi, Chiba, 292-8535, Japan
| | - Hirokazu Takahashi
- Liver Center, Saga University Hospital, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga-shi, Saga 849-8501, Japan
| | - Yujiro Ueda
- Department of Hematology and Oncology, Japanese Red Cross Kumamoto Hospital, 2-1-1 Nagamine-minami, Higashi-ku, Kumamoto-shi, Kumamoto 861-8520, Japan
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, 37-1 Uearata-cho, Kagoshima-shi, Kagoshima 890-8760, Japan
| | - Azusa Komori
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita 879-5593, Japan
| | - Satoshi Otsu
- Department of Medical Oncology and Hematology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-shi, Oita 879-5593, Japan
| | - Shiho Arima
- Digestive and Lifestyle Diseases, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima-shi, Kagoshima 890-8520, Japan
| | - Masaru Fukahori
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume-shi, Fukuoka 830-0011, Japan
| | - Akitaka Makiyama
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahatanishi-ku, Kitakyushu-shi, Fukuoka 806-8501, Japan; Cancer Center, Gifu University Hospital, 1-1 Yanagido, Gifu-shi, Gifu 501-1194, Japan
| | - Hiroki Taguchi
- Department of Gastroenterology, Saiseikai Sendai Hospital, 2-46 Harada-cho, Satsumasendai-shi, Kagoshima 895-0074, Japan; Department of Gastroenterology, Izumi General Medical Center, 520 Myojin-cho, Izumi-shi, Kagoshima 899-0131, Japan
| | - Takuya Honda
- Department of Gastroenterology and Hepatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki-shi, Nagasaki 852-8501, Japan
| | - Taro Shibuki
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-ko, Arita-cho, Nishi-matsuura-gun, Saga 849-4193, Japan; Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanohara, Kashiwa-shi, Chiba 277-8577, Japan
| | - Kenta Nio
- Department of Medical Oncology, Sasebo Kyosai Hospital, 10-17 Shimanji-cho, Sasebo-shi, Nagasaki 857-8575, Japan
| | - Yasushi Ide
- Department of Internal Medicine, Karatsu Red Cross Hospital, 2430 Watada, Karatsu-shi, Saga 847-8588, Japan
| | - Toshihiko Mizuta
- Department of Internal Medicine, Imari Arita Kyoritsu Hospital, 860 Ninose-ko, Arita-cho, Nishi-matsuura-gun, Saga 849-4193, Japan; Department of Internal Medicine, Fujikawa Hospital, 1-2-6 Matsubara, Saga-shi, Saga 840-0831, Japan
| | - Tsuyoshi Shirakawa
- Department of Medical Oncology, Fukuoka Wajiro Hospital, 2-2-75 Wajirogaoka, Higashi-ku, Fukuoka-shi, Fukuoka 811-0213, Japan; Karatsu Higashi-matsuura Medical Association Center, 2566-11 Chiyoda-machi, Karatsu-shi, Saga 847-0041, Japan
| | - Taiga Otsuka
- Department of Medical Oncology, Saga Medical Center Koseikan, 400 Kase-machi, Saga-shi, Saga 840-8571, Japan; Department of Internal Medicine, Minato Medical Clinic, 3-11-3 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka 810-0072, Japan.
| | - Kenji Mitsugi
- Department of Medical Oncology, Hamanomachi Hospital, 3-3-1 Nagahama, Chuo-ku, Fukuoka-shi, Fukuoka, 810-8539, Japan
| |
Collapse
|
16
|
Karamouzis MV, Athanasiadis I, Samelis G, Vallilas C, Bokas A, Nikolaidi A, Dimitriadou A, Sarantis P, Pistamaltzian N, Schizas D, Papalampros A, Felekouras E, Dimitroulis D, Antoniou E, Sotiropoulos G, Papakotoulas P. The Impact of Thromboprophylaxis on the Survival of Patients with Advanced Pancreatic Cancer. The Pancreatic Cancer and Tinzaparin (PaCT) Study. Cancers (Basel) 2021; 13:cancers13122884. [PMID: 34207591 PMCID: PMC8228127 DOI: 10.3390/cancers13122884] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/02/2021] [Accepted: 06/08/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Pancreatic cancer (PaC) induces a prothrombotic and hypercoagulable state. Thrombosis occurs in 20% of PaC patients and is associated with worse prognosis and reduced progression-free survival (PFS). The aim of this retrospective observational study (PaCT) was to investigate the effect of thromboprophylaxis with an intermediate dose of tinzaparin on the PFS of patients treated with nab-paclitaxel and gemcitabine. Data obtained from 110 patients with active PaC administered prophylaxis with tinzaparin during the study resulted in median PFS of 7.9 months; data for the PFS of patients without simultaneous anticoagulation were obtained bibliographically from 14 studies, and after applying meta-analysis was 5.6 months. Patients receiving anticoagulation with tinzaparin had 39.5% higher PFS than patients without such thromboprophylaxis (p < 0.05). During follow-up, three (2.7%) thrombotic events and two (1.9%) clinically relevant non-major bleeding events occurred. Concluding, PFS in advanced PaC patients undergoing chemotherapy was positively impacted by thromboprophylaxis with intermediate dose tinzaparin. Abstract Pancreatic cancer (PaC) induces a prothrombotic and hypercoagulable state. The aim of this study was to investigate the effect of tinzaparin in combination with chemotherapy. The PaCT (pancreatic cancer and tinzaparin) study was a retrospective observational study that collected data regarding progression free survival (PFS) in advanced or metastatic PaC patients who received thromboprophylaxis with tinzaparin during chemotherapy with nab-paclitaxel (N) and gemcitabine (G). The primary end point was to compare, from already published data, the PFS of patients receiving thromboprophylaxis with tinzaparin with the PFS of patients receiving chemotherapy with N–G but no thromboprophylaxis. Secondary end points were efficacy and safety of anticoagulation. In total, 110 PaC patients, 93% with advanced or metastatic disease, treated with N–G and tinzaparin (10,291 ± 1176 Anti-Xa IU, OD, median duration 8.7, IQR: 5.6–11.9 months) were enrolled. Of these, 52% were males and; the median age was 68 (40–86 years). The tumor was located to in the pancreatic head at in 45% of the patients. The median PFS was 7.9 months (IQR: 5.0–11.8 months). Out of 14 similar studies (involving 2994 patients) identified via systematic search, it was determined that the weighted PFS of patients receiving N–G but no anticoagulation was 5.6 months. Therefore, patients receiving tinzaparin had 39.54% higher PFS than patients without thromboprophylaxis (p < 0.05). During the follow-up period of 18.3 ± 11.7 months, three (2.7%) thrombotic events were recorded while two clinically relevant non-major bleeding events occurred (1.9%). In conclusion, PFS in advanced PaC patients undergoing chemotherapy is positively impacted by anticoagulation. Thromboprophylaxis with tinzaparin in treatment dose is efficient and safe.
Collapse
Affiliation(s)
- Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.V.); (P.S.)
- Correspondence: ; Tel.: +30-210-746-2508/9; Fax: +30-210-746-2703
| | - Ilias Athanasiadis
- Oncology Department, Mitera Hospital, 15123 Marousi, Greece; (I.A.); (A.N.); (N.P.)
| | - Georgios Samelis
- Oncology Unit, Hippokration General Hospital, 11527 Athens, Greece; (G.S.); (A.D.)
| | - Christos Vallilas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.V.); (P.S.)
| | - Alexandros Bokas
- 1st Clinical Oncology Department, Theagenio Cancer Hospital, 54639 Thessaloniki, Greece; (A.B.); (P.P.)
| | - Adamantia Nikolaidi
- Oncology Department, Mitera Hospital, 15123 Marousi, Greece; (I.A.); (A.N.); (N.P.)
| | - Areti Dimitriadou
- Oncology Unit, Hippokration General Hospital, 11527 Athens, Greece; (G.S.); (A.D.)
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.V.); (P.S.)
| | | | - Dimitrios Schizas
- First Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (A.P.); (E.F.)
| | - Alexandros Papalampros
- First Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (A.P.); (E.F.)
| | - Evangelos Felekouras
- First Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.S.); (A.P.); (E.F.)
| | - Dimitrios Dimitroulis
- Second Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.D.); (E.A.); (G.S.)
| | - Eustathios Antoniou
- Second Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.D.); (E.A.); (G.S.)
| | - Georgios Sotiropoulos
- Second Department of Surgery, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (D.D.); (E.A.); (G.S.)
| | - Pavlos Papakotoulas
- 1st Clinical Oncology Department, Theagenio Cancer Hospital, 54639 Thessaloniki, Greece; (A.B.); (P.P.)
| |
Collapse
|
17
|
Kurreck A, Weckwerth J, Modest DP, Striefler JK, Bahra M, Bischoff S, Pelzer U, Oettle H, Kruger S, Riess H, Sinn M. Impact of completeness of adjuvant gemcitabine, relapse pattern, and subsequent therapy on outcome of patients with resected pancreatic ductal adenocarcinoma - A pooled analysis of CONKO-001, CONKO-005, and CONKO-006 trials. Eur J Cancer 2021; 150:250-259. [PMID: 33940349 DOI: 10.1016/j.ejca.2021.03.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/13/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) represents one of the most fatal malignancies worldwide. It is suggested that survival in PDAC depends, among other things, on pattern of disease recurrence. PATIENTS AND METHODS We performed a pooled analysis of the adjuvant therapy studies CONKO-001, CONKO-005, and CONKO-006, including a total of 912 patients with regard to prognostic factors in patients with recurrent disease. Overall survival from disease recurrence (OS 2) and disease-free survival (DFS) from the day of surgery were expressed by Kaplan-Meier method and compared using log-rank testing and Cox regression. RESULTS Of 912 patients treated within the previously mentioned CONKO trials, we identified 689 patients with disease recurrence and defined site of relapse. In multivariable analysis, the presence of isolated pulmonary metastasis, low tumour grading, and low postoperative level of CA 19-9 remained significant factors for improved OS 2 and DFS. Furthermore, completeness of adjuvant gemcitabine-based treatment (OS 2: P = 0.006), number of relapse sites (OS 2: P = 0.015), and type of palliative first-line treatment (OS 2: P < 0.001) significantly affected overall survival after disease recurrence in PDAC. CONCLUSIONS Determining tumour subgroups using prognostic factors may be helpful to stratify PDAC patients for future clinical trials. In case of disease recurrence, the site of relapse may have a prognostic impact on subsequent survival. Further investigations are needed to identify differences in tumour biology, reflecting relapse patterns and the differing survival of PDAC patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, Tumor-Associated, Carbohydrate/blood
- Antimetabolites, Antineoplastic/adverse effects
- Antimetabolites, Antineoplastic/therapeutic use
- Carcinoma, Pancreatic Ductal/blood
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/secondary
- Carcinoma, Pancreatic Ductal/therapy
- Chemotherapy, Adjuvant
- Databases, Factual
- Deoxycytidine/adverse effects
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/therapeutic use
- Disease-Free Survival
- Female
- Humans
- Lung Neoplasms/mortality
- Lung Neoplasms/secondary
- Lung Neoplasms/therapy
- Male
- Middle Aged
- Neoplasm Recurrence, Local
- Palliative Care
- Pancreatectomy/adverse effects
- Pancreatectomy/mortality
- Pancreatic Neoplasms/blood
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Randomized Controlled Trials as Topic
- Retrospective Studies
- Risk Assessment
- Risk Factors
- Time Factors
- Young Adult
- Gemcitabine
Collapse
Affiliation(s)
- Annika Kurreck
- Charité University Medicine Berlin, Department of Hematology, Oncology, and Tumorimmunology, CVK, Berlin, Germany
| | - Johanna Weckwerth
- Charité University Medicine Berlin, Department of Hematology, Oncology, and Tumorimmunology, CVK, Berlin, Germany
| | - Dominik P Modest
- Charité University Medicine Berlin, Department of Hematology, Oncology, and Tumorimmunology, CVK, Berlin, Germany
| | - Jana K Striefler
- Charité University Medicine Berlin, Department of Hematology, Oncology, and Tumorimmunology, CVK, Berlin, Germany
| | - Marcus Bahra
- Charité University Medicine Berlin, Department of General, Visceral, and Transplantation Surgery, Berlin, Germany
| | - Sven Bischoff
- Charité University Medicine Berlin, Department of Hematology, Oncology, and Tumorimmunology, CVK, Berlin, Germany
| | - Uwe Pelzer
- Charité University Medicine Berlin, Department of Hematology and Oncology, CCM, Berlin, Germany
| | | | - Stephan Kruger
- Ludwig Maximilians University of Munich, Department of Internal Medicine III, Comprehensive Cancer Center, Munich, Germany
| | - Hanno Riess
- Charité University Medicine Berlin, Department of Hematology and Oncology, CCM, Berlin, Germany
| | - Marianne Sinn
- Charité University Medicine Berlin, Department of Hematology, Oncology, and Tumorimmunology, CVK, Berlin, Germany; University Medical Center Hamburg-Eppendorf, Department of Hematology and Oncology, Hamburg, Germany.
| |
Collapse
|
18
|
Khomiak A, Brunner M, Kordes M, Lindblad S, Miksch RC, Öhlund D, Regel I. Recent Discoveries of Diagnostic, Prognostic and Predictive Biomarkers for Pancreatic Cancer. Cancers (Basel) 2020; 12:E3234. [PMID: 33147766 PMCID: PMC7692691 DOI: 10.3390/cancers12113234] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/26/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with a dismal prognosis that is frequently diagnosed at an advanced stage. Although less common than other malignant diseases, it currently ranks as the fourth most common cause of cancer-related death in the European Union with a five-year survival rate of below 9%. Surgical resection, followed by adjuvant chemotherapy, remains the only potentially curative treatment but only a minority of patients is diagnosed with locally resectable, non-metastatic disease. Patients with advanced disease are treated with chemotherapy but high rates of treatment resistance and unfavorable side-effect profiles of some of the used regimens remain major challenges. Biomarkers reflect pathophysiological or physiological processes linked to a disease and can be used as diagnostic, prognostic and predictive tools. Thus, accurate biomarkers can allow for better patient stratification and guide therapy choices. Currently, the only broadly used biomarker for PDAC, CA 19-9, has multiple limitations and the need for novel biomarkers is urgent. In this review, we highlight the current situation, recent discoveries and developments in the field of biomarkers of PDAC and their potential clinical applications.
Collapse
Affiliation(s)
- Andrii Khomiak
- Shalimov National Institute of Surgery and Transplantology, 03058 Kyiv, Ukraine;
| | - Marius Brunner
- Department of Gastroenterology, Endocrinology and Gastrointestinal Oncology, University Medical Center, 37075 Goettingen, Germany;
| | - Maximilian Kordes
- Department of Upper Abdominal Diseases, Karolinska University Hospital, 14186 Stockholm, Sweden;
- Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, 17177 Stockholm, Sweden
| | - Stina Lindblad
- Department of Radiation Sciences, Sweden and Wallenberg Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Rainer Christoph Miksch
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Daniel Öhlund
- Department of Radiation Sciences, Sweden and Wallenberg Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden;
| | - Ivonne Regel
- Department of Medicine II, University Hospital, LMU Munich, 81377 Munich, Germany
| |
Collapse
|
19
|
Franco F, Camara JC, Martín-Valadés JI, López-Alfonso A, Marrupe D, Gutiérrez-Abad D, Martínez-Amores B, León A, Juez I, Pérez M, Royuela A, Ruiz-Casado A. Clinical outcomes of FOLFIRINOX and gemcitabine-nab paclitaxel for metastatic pancreatic cancer in the real world setting. Clin Transl Oncol 2020; 23:812-819. [PMID: 32857340 DOI: 10.1007/s12094-020-02473-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/03/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND/OBJECTIVES The incidence of pancreatic cancer is increasing in developed countries. The incorporation of new therapies, to the first-line treatment of patients with good performance status led to better survival in clinical trials. However, there is a wide variability in their use and some concerns about the treatment of elderly patients who were not included in the clinical trials. METHODS This is a retrospective multicenter study. Data from consecutive patients diagnosed with metastatic pancreatic cancer (mPC) treated with FOLFIRINOX (FFX) or gemcitabine plus nab-paclitaxel (GnP) were analysed to evaluate efficacy (overall survival-OS) and toxicity. RESULTS A total of 119 patients were included. 49.6% were treated with FFX and 50.4% with GNP in first-line. The median OS was 12 months with no statistically significant differences between both regimens (12.7 m for FFX vs 10.2 m for GnP). Elevated Ca 19.9 levels and neutrophil-lymphocyte ratio (NLR) increased the risk of death. Patients who received both regimens in first/second line had a median OS longer than 15 months whichever the sequence. 32 patients (27%) were older than 70-y. 54% patients received a second-line treatment, 56% in the FFX group and 44% in the GnP group. The median OS for patients older than 70 was 9.5 m versus 12.3 m for patients younger than 70. Progression of the disease was the cause of death in 67.6% of the patients. CONCLUSIONS In our setting, the use of FFX and GnP for treating mPC is quite similar, but superiority could not be demonstrated for any of the schemes in the first line. OS was determined by basal levels of Ca 19.9 and NLR. Patients receiving both regimens in first/second line whichever the sequence, exhibited the best survival rates. In our series, elderly patients had poorer survival rates.
Collapse
Affiliation(s)
- F Franco
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Majadahonda, Spain.
| | - J C Camara
- Department of Medical Oncology, Fundación Hospital Alcorcón, Alcorcón, Spain
| | | | - A López-Alfonso
- Department of Medical Oncology, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - D Marrupe
- Department of Medical Oncology, Hospital Universitario de Móstoles, Móstoles, Spain
| | - D Gutiérrez-Abad
- Department of Medical Oncology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - B Martínez-Amores
- Department of Medical Oncology, Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - A León
- Department of Medical Oncology, Fundación Jiménez Díaz, Madrid, Spain
| | - I Juez
- Department of Medical Oncology, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - M Pérez
- Department of Medical Oncology, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - A Royuela
- Department of Biostatistics, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| | - A Ruiz-Casado
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro, Majadahonda, Spain
| |
Collapse
|
20
|
Dell'Aquila E, Fulgenzi CAM, Minelli A, Citarella F, Stellato M, Pantano F, Russano M, Cursano MC, Napolitano A, Zeppola T, Vincenzi B, Tonini G, Santini D. Prognostic and predictive factors in pancreatic cancer. Oncotarget 2020; 11:924-941. [PMID: 32206189 PMCID: PMC7075465 DOI: 10.18632/oncotarget.27518] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 02/17/2020] [Indexed: 12/13/2022] Open
Abstract
Pancreatic cancer is one of the leading causes of cancer death worldwide. Its high mortality rate has remained unchanged for years. Radiotherapy and surgery are considered standard treatments in early and locally advanced stages. Chemotherapy is the only option for metastatic patients. Two treatment regimens, i. e. the association of 5-fluorouracil- irinotecan-oxaliplatin (FOLFIRINOX) and the association of nab-paclitaxel with gemcitabine, have been shown to improve outcomes for metastatic pancreatic adenocarcinoma patients. However, there are not standardized predictive biomarkers able to identify patients who benefit most from treatments. CA19-9 is the most studied prognostic biomarker, its predictive role remains unclear. Other clinical, histological and molecular biomarkers are emerging in prognostic and predictive settings. The aim of this review is to provide an overview of prognostic and predictive markers used in clinical practice and to explore the most promising fields of research in terms of treatment selection and tailored therapy in pancreatic cancer.
Collapse
Affiliation(s)
| | | | - Alessandro Minelli
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Fabrizio Citarella
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Marco Stellato
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Francesco Pantano
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Marco Russano
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | | | - Andrea Napolitano
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Tea Zeppola
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Giuseppe Tonini
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| | - Daniele Santini
- Department of Medical Oncology, University Campus Bio-Medico, Rome 00128, Italy
| |
Collapse
|
21
|
Ishii N, Nishikawa H, Iwata Y, Enomoto H, Tanaka H, Katakami N, Nishimura T, Iijima H, Nishiguchi S. Proposal of predictive model on survival in unresectable pancreatic cancer receiving systemic chemotherapy. J Cancer 2020; 11:1223-1230. [PMID: 31956368 PMCID: PMC6959067 DOI: 10.7150/jca.38861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/13/2019] [Indexed: 11/23/2022] Open
Abstract
Aims: To construct a predictive model for overall survival (OS) in unresectable pancreatic cancer (PaC) undergoing systemic chemotherapy and to confirm its accuracy in an independent cohort. Patients and methods: The training set (Ts) and the validation set (Vs) included 93 patients (median age=71 years) and 75 patients (median age=76 years). In the Ts, we examined variables linked to OS by uni- and multivariate analyses and constructed a predictive model for OS. Next, we evaluated the reproducibility of the proposed model in the Vs. Results: In the multivariate analysis for the Ts, PaC stage IV (P=0.0020) and carbohydrate antigen (CA) 19-9 ≥437.5 IU/l (P=0.0237) were identified to be significant factors associated with OS. Patients with PaC stage IV or not were given a score of 1 or 0, whereas patients with CA19-9 ≥437.5 IU/l or <437.5 IU/l were given a score of 1 or 0. Sum of the point of PaC stage (0 or 1) and CA19-9 (0 or 1) was defined as "PaC-CA score". In the Ts, there were 16 patients with score 0, 40 with score 1 and 37 with score 2, while in the Vs, there were 9 patients with score 0, 32 with score 1 and 34 with score 2. Overall P values reached significance in the Ts (P=0.0002), the Vs (P=0.0029) and the combined Ts and Vs (P<0.0001) among patients with PaC score 0, 1 and 2. Conclusion: PaC-CA score can be helpful for risk stratification in PaC patients undergoing systemic chemotherapy.
Collapse
Affiliation(s)
- Noriko Ishii
- Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroki Nishikawa
- Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yoshinori Iwata
- Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hirayuki Enomoto
- Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hironori Tanaka
- Department of gastroenterology, Takarazuka municipal hospital, Takarazuka, Hyogo, Japan
| | - Nobuyuki Katakami
- Department of oncology, Takarazuka municipal hospital, Takarazuka, Hyogo, Japan
| | - Takashi Nishimura
- Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroko Iijima
- Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shuhei Nishiguchi
- Division of Hepatobiliary and Pancreatic disease, Department of Internal Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
22
|
Fornaro L, Leone F, Vienot A, Casadei-Gardini A, Vivaldi C, Lièvre A, Lombardi P, De Luca E, Vernerey D, Sperti E, Musettini G, Satolli MA, Edeline J, Spadi R, Neuzillet C, Falcone A, Pasquini G, Clerico M, Passardi A, Buscaglia P, Meurisse A, Aglietta M, Brac C, Vasile E, Montagnani F. Validated Nomogram Predicting 6-Month Survival in Pancreatic Cancer Patients Receiving First-Line 5-Fluorouracil, Oxaliplatin, and Irinotecan. Clin Colorectal Cancer 2019; 18:e394-e401. [PMID: 31564556 DOI: 10.1016/j.clcc.2019.08.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 06/22/2019] [Accepted: 08/27/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND FOLFIRINOX (leucovorin, 5-fluorouracil, irinotecan, and oxaliplatin) is an option for fit patients with metastatic (MPC) and locally advanced unresectable (LAPC) pancreatic cancer. However, no criteria reliably identify patients with better outcomes. PATIENTS AND METHODS We investigated putative prognostic factors among 137 MPC/LAPC patients treated with triplet chemotherapy. Association with 6-month survival status (primary endpoint) was assessed by multivariate logistic regression models. A nomogram predicting the risk of death at 6 months was built by assigning a numeric score to each identified variable, weighted on its level of association with survival. External validation was performed in an independent data set of 206 patients. The study was registered at ClinicalTrials.gov (NCT03590275). RESULTS Four variables (performance status, liver metastases, baseline carbohydrate antigen 19-9 level, and neutrophil-to-lymphocyte ratio) were found to be associated with 6-month survival by multivariate analysis or had sufficient clinical plausibility to be included in the nomogram. Accuracy was confirmed in the validation cohort (C index = 0.762; 95% confidence interval, 0.713-0.825). After grouping all cases, 4 subsets with different outcomes were identified by 0, 1, 2, or > 2 poor prognostic features (P < .0001). CONCLUSION The nomogram we constructed accurately predicts the risk of death in the first 6 months after initiation of FOLFIRINOX in MPC/LAPC patients. This tool could be useful to guide communication about prognosis, and to inform the design and interpretation of clinical trials.
Collapse
Affiliation(s)
- Lorenzo Fornaro
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy.
| | - Francesco Leone
- Department of Medical Oncology, University of Turin, Turin, Italy; Medical Oncology, Candiolo Cancer Institute, FPO, IRCCS, Candiolo, Italy
| | - Angélique Vienot
- Department of Medical Oncology, Besancon University Hospital, Besançon, France
| | | | - Caterina Vivaldi
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Astrid Lièvre
- Department of Gastroenterology, Rennes University Hospital, Rennes 1 University, COSS (Chemistry Oncogenesis Stress Signaling), UMR_S 1242, Rennes, France
| | - Pasquale Lombardi
- Department of Medical Oncology, University of Turin, Turin, Italy; Medical Oncology, Candiolo Cancer Institute, FPO, IRCCS, Candiolo, Italy
| | - Emmanuele De Luca
- Department of Medical Oncology, University of Turin, Turin, Italy; S.C.D.U. Oncologia, A.O. Ordine Mauriziano, Ospedale Umberto I, Turin, Italy
| | - Dewi Vernerey
- Methodological and Quality of Life in Oncology Unit, EA 3181, Besançon University Hospital, Besançon, France
| | - Elisa Sperti
- S.C.D.U. Oncologia, A.O. Ordine Mauriziano, Ospedale Umberto I, Turin, Italy
| | - Gianna Musettini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Maria Antonietta Satolli
- Department of Medical Oncology, University of Turin, Turin, Italy; Medical Oncology 1 Division, Città della Salute e della Scienza, Turin, Italy
| | - Julien Edeline
- Oncology Department, Cancer Institute Eugène Marquis, Rennes 1 University, INSERM, INRA, Rennes 1 University, Nutrition Metabolism and Cancer (NuMeCan), Rennes, France
| | - Rosella Spadi
- Medical Oncology 1 Division, Città della Salute e della Scienza, Turin, Italy
| | - Cindy Neuzillet
- Department of Medical Oncology, Curie Institute, Saint Cloud, France
| | - Alfredo Falcone
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy; Department of Translational Research and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - Giulia Pasquini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Mario Clerico
- S.C. Oncologia, Department of Oncology, ASL BI, Biella, Italy
| | - Alessandro Passardi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Aurélia Meurisse
- Methodological and Quality of Life in Oncology Unit, EA 3181, Besançon University Hospital, Besançon, France
| | - Massimo Aglietta
- Department of Medical Oncology, University of Turin, Turin, Italy; Medical Oncology, Candiolo Cancer Institute, FPO, IRCCS, Candiolo, Italy
| | - Clémence Brac
- Oncology Department, Cancer Institute Eugène Marquis, Rennes, France
| | - Enrico Vasile
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | | |
Collapse
|
23
|
Petrioli R, Torre P, Pesola G, Paganini G, Paolelli L, Miano ST, Martellucci I, Francini G, Francini E. Gemcitabine plus nab-paclitaxel followed by maintenance treatment with gemcitabine alone as first-line treatment for older adults with locally advanced or metastatic pancreatic cancer. J Geriatr Oncol 2019; 11:647-651. [PMID: 31471168 DOI: 10.1016/j.jgo.2019.08.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/20/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the efficacy and safety of the combination Gemcitabine (Gem) plus nab-Paclitaxel (NabP) (Gem/NabP), followed by maintenance Gem in older adults with locally advanced or metastatic pancreatic cancer (PC). MATERIALS AND METHODS In this prospective observational study, the induction chemotherapy consisted of NabP 125 mg/m2 followed by Gem 1000 mg/m2 on days 1, 8, and 15 of a 4-week cycle. After a maximum of 3 cycles, patients without evidence of progressive disease (PD) were administered Gem 1000 mg/m2 weekly for 3 of 4 weeks as maintenance therapy until documentation of PD or unacceptable toxicity. The primary endpoint was six-month disease-control rate (DCR). RESULTS Overall, 36 patients >70 years with metastatic or locally advanced PC were enrolled at participating Institutions. After completion of Gem/NabP, 18 (50%) patients achieved partial response, 13 (36%) had stable disease, and 5 (14%) had PD. Thirty-one patients (86%) received Gem monotherapy as maintenance treatment for a median of 3 cycles (range, 2-9 cycles). Six-month DCR was 61% (95% CI, 45-77), median PFS was 6.4 months (95% CI, 5.4-8.3), and median OS was 13.4 months (95% CI, 11.1-16.7). During Gem/NabP regimen, the most common grade 3 toxicity included neutropenia (22%), anemia (19%) and thrombocytopenia (8%). Grade 3 neuropathy was not observed. During Gem maintenance therapy, grade 3 hematological toxicity was described in 6 patients (19%). CONCLUSION Gem/NabP followed by maintenance Gem appears to be safe and effective for older patients with locally advanced or metastatic PC.
Collapse
Affiliation(s)
- Roberto Petrioli
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy.
| | - Pamela Torre
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Guido Pesola
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Giovanni Paganini
- General Medicine& Oncology, Pieve di Coriano Hospital, ASST Mantova, Italy
| | | | - Salvatora Tindara Miano
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Ignazio Martellucci
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Guido Francini
- Medical Oncology Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Italy
| | - Edoardo Francini
- La Sapienza University, Rome, Italy; Medical Oncology Unit, Misericordia Hospital, Grosseto, Italy
| |
Collapse
|
24
|
Pecora I, Fornaro L, Vasile E, Catanese S, Salani F, Vivaldi C. Combination Chemotherapy in Patients With Advanced Pancreatic Cancer With an Eastern Cooperative Oncology Group Performance Status of 2: Lights and Shadows of a Frail Route. J Clin Oncol 2019; 37:1978-1979. [DOI: 10.1200/jco.19.00127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Affiliation(s)
- Irene Pecora
- Irene Pecora, MD; Lorenzo Fornaro, MD; Enrico Vasile, MD, PhD; Silvia Catanese, MD; and Francesca Salani, MD, Azienda Ospedaliero–Universitaria Pisana, Pisa, Italy; and Caterina Vivaldi, MD, Azienda Ospedaliero–Universitaria Pisana; University of Pisa, Pisa, Italy
| | - Lorenzo Fornaro
- Irene Pecora, MD; Lorenzo Fornaro, MD; Enrico Vasile, MD, PhD; Silvia Catanese, MD; and Francesca Salani, MD, Azienda Ospedaliero–Universitaria Pisana, Pisa, Italy; and Caterina Vivaldi, MD, Azienda Ospedaliero–Universitaria Pisana; University of Pisa, Pisa, Italy
| | - Enrico Vasile
- Irene Pecora, MD; Lorenzo Fornaro, MD; Enrico Vasile, MD, PhD; Silvia Catanese, MD; and Francesca Salani, MD, Azienda Ospedaliero–Universitaria Pisana, Pisa, Italy; and Caterina Vivaldi, MD, Azienda Ospedaliero–Universitaria Pisana; University of Pisa, Pisa, Italy
| | - Silvia Catanese
- Irene Pecora, MD; Lorenzo Fornaro, MD; Enrico Vasile, MD, PhD; Silvia Catanese, MD; and Francesca Salani, MD, Azienda Ospedaliero–Universitaria Pisana, Pisa, Italy; and Caterina Vivaldi, MD, Azienda Ospedaliero–Universitaria Pisana; University of Pisa, Pisa, Italy
| | - Francesca Salani
- Irene Pecora, MD; Lorenzo Fornaro, MD; Enrico Vasile, MD, PhD; Silvia Catanese, MD; and Francesca Salani, MD, Azienda Ospedaliero–Universitaria Pisana, Pisa, Italy; and Caterina Vivaldi, MD, Azienda Ospedaliero–Universitaria Pisana; University of Pisa, Pisa, Italy
| | - Caterina Vivaldi
- Irene Pecora, MD; Lorenzo Fornaro, MD; Enrico Vasile, MD, PhD; Silvia Catanese, MD; and Francesca Salani, MD, Azienda Ospedaliero–Universitaria Pisana, Pisa, Italy; and Caterina Vivaldi, MD, Azienda Ospedaliero–Universitaria Pisana; University of Pisa, Pisa, Italy
| |
Collapse
|