1
|
Chintamaneni PK, Pindiprolu SKSS, Swain SS, Karri VVSR, Nesamony J, Chelliah S, Bhaskaran M. Conquering chemoresistance in pancreatic cancer: Exploring novel drug therapies and delivery approaches amidst desmoplasia and hypoxia. Cancer Lett 2024; 588:216782. [PMID: 38453046 DOI: 10.1016/j.canlet.2024.216782] [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: 09/28/2023] [Revised: 02/20/2024] [Accepted: 03/03/2024] [Indexed: 03/09/2024]
Abstract
Pancreatic cancer poses a significant challenge within the field of oncology due to its aggressive behaviour, limited treatment choices, and unfavourable outlook. With a mere 10% survival rate at the 5-year mark, finding effective interventions becomes even more pressing. The intricate relationship between desmoplasia and hypoxia in the tumor microenvironment further complicates matters by promoting resistance to chemotherapy and impeding treatment efficacy. The dense extracellular matrix and cancer-associated fibroblasts characteristic of desmoplasia create a physical and biochemical barrier that impedes drug penetration and fosters an immunosuppressive milieu. Concurrently, hypoxia nurtures aggressive tumor behaviour and resistance to conventional therapies. a comprehensive exploration of emerging medications and innovative drug delivery approaches. Notably, advancements in nanoparticle-based delivery systems, local drug delivery implants, and oxygen-carrying strategies are highlighted for their potential to enhance drug accessibility and therapeutic outcomes. The integration of these strategies with traditional chemotherapies and targeted agents reveals the potential for synergistic effects that amplify treatment responses. These emerging interventions can mitigate desmoplasia and hypoxia-induced barriers, leading to improved drug delivery, treatment efficacy, and patient outcomes in pancreatic cancer. This review article delves into the dynamic landscape of emerging anticancer medications and innovative drug delivery strategies poised to overcome the challenges imposed by desmoplasia and hypoxia in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Pavan Kumar Chintamaneni
- Department of Pharmaceutics, GITAM School of Pharmacy, GITAM (Deemed to be University), Rudraram, 502329 Telangana, India.
| | | | - Swati Swagatika Swain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Nilgiris, Tamil Nadu, India
| | | | - Jerry Nesamony
- College of Pharmacy and Pharmaceutical Sciences, The University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA
| | - Selvam Chelliah
- College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX-77004, USA
| | - Mahendran Bhaskaran
- College of Pharmacy and Pharmaceutical Sciences, The University of Toledo HSC, 3000 Arlington Avenue, Toledo, OH, 43614, USA.
| |
Collapse
|
2
|
Brada LJH, Walma MS, Daamen LA, van Roessel S, van Dam RM, de Hingh IH, Liem MLS, de Meijer VE, Patijn GA, Festen S, Stommel MWJ, Bosscha K, Polée MB, Yung Nio C, Wessels FJ, de Vries JJJ, van Lienden KP, Bruijnen RC, Los M, Mohammad NH, Wilmink HW, Busch OR, Besselink MG, Quintus Molenaar I, van Santvoort HC. Predicting overall survival and resection in patients with locally advanced pancreatic cancer treated with FOLFIRINOX: Development and internal validation of two nomograms. J Surg Oncol 2021; 124:589-597. [PMID: 34115379 DOI: 10.1002/jso.26567] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/07/2021] [Accepted: 05/15/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND OBJECTIVES Patients with locally advanced pancreatic cancer (LAPC) are increasingly treated with FOLFIRINOX, resulting in improved survival and resection of tumors that were initially unresectable. It remains unclear, however, which specific patients benefit from FOLFIRINOX. Two nomograms were developed predicting overall survival (OS) and resection at the start of FOLFIRINOX for LAPC. METHODS From our multicenter, prospective LAPC registry in 14 Dutch hospitals, LAPC patients starting first-line FOLFIRINOX (April 2015-December 2017) were included. Stepwise backward selection according to the Akaike Information Criterion was used to identify independent baseline predictors for OS and resection. Two prognostic nomograms were generated. RESULTS A total of 252 patients were included, with a median OS of 14 months. Thirty-two patients (13%) underwent resection, with a median OS of 23 months. Older age, female sex, Charlson Comorbidity Index ≤1, and CA 19.9 < 274 were independent factors predicting a better OS (c-index: 0.61). WHO ps >1, involvement of the superior mesenteric artery, celiac trunk, and superior mesenteric vein ≥ 270° were independent factors decreasing the probability of resection (c-index: 0.79). CONCLUSIONS Two nomograms were developed to predict OS and resection in patients with LAPC before starting treatment with FOLFIRINOX. These nomograms could be beneficial in the shared decision-making process and counseling of these patients.
Collapse
Affiliation(s)
- Lilly J H Brada
- Department of Surgery, UMC Utrecht Cancer Center, St Antonius Hospital Nieuwegein and Meander Medical Center, Utrecht, The Netherlands.,Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marieke S Walma
- Department of Surgery, UMC Utrecht Cancer Center, St Antonius Hospital Nieuwegein and Meander Medical Center, Utrecht, The Netherlands.,Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Lois A Daamen
- Department of Surgery, UMC Utrecht Cancer Center, St Antonius Hospital Nieuwegein and Meander Medical Center, Utrecht, The Netherlands
| | - Stijn van Roessel
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Ronald M van Dam
- Department of Surgery, Maastricht UMC+, Maastricht, The Netherlands
| | - Ignace H de Hingh
- Department of Surgery, Catharina Hospital, Eindhoven, The Netherlands.,Department of Epidemiology, Maastricht University, Maastricht, The Netherlands
| | - Mike L S Liem
- Department of Surgery, Medical Spectrum Twente, Enschede, The Netherlands
| | | | - Gijs A Patijn
- Department of Surgery, Isala, Zwolle, The Netherlands
| | | | - Martijn W J Stommel
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Koop Bosscha
- Department of Surgery, Jeroen Bosch Hospital, 's-Hertogenbosch, The Netherlands
| | - Marco B Polée
- Department of Medical Oncology, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - C Yung Nio
- Department of Radiology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Frank J Wessels
- Department of Radiology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jan J J de Vries
- Department of Radiology, Cancer Center Amsterdam, Amsterdam UMC, VU University, Amsterdam, The Netherlands
| | - Krijn P van Lienden
- Department of Radiology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rutger C Bruijnen
- Department of Radiology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein: Regional Academic Cancer Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maartje Los
- Department of Medical Oncology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Nadia Haj Mohammad
- Department of Medical Oncology, UMC Utrecht Cancer Center and St Antonius Hospital Nieuwegein, Regional Academic Cancer Center Utrecht, Utrecht, The Netherlands
| | - Hanneke W Wilmink
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Olivier R Busch
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Marc G Besselink
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - I Quintus Molenaar
- Department of Surgery, UMC Utrecht Cancer Center, St Antonius Hospital Nieuwegein and Meander Medical Center, Utrecht, The Netherlands
| | - Hjalmar C van Santvoort
- Department of Surgery, UMC Utrecht Cancer Center, St Antonius Hospital Nieuwegein and Meander Medical Center, Utrecht, The Netherlands
| | | |
Collapse
|
3
|
Markus M, Abendroth A, Noureddine R, Paul A, Breitenbuecher S, Virchow I, Schmid KW, Markus P, Schumacher B, Wiesweg M, Wendling J, Mende B, Siveke JT, Schuler M, Kasper S. Combined systemic inflammation score (SIS) correlates with prognosis in patients with advanced pancreatic cancer receiving palliative chemotherapy. J Cancer Res Clin Oncol 2021; 147:579-591. [PMID: 32839836 PMCID: PMC7817578 DOI: 10.1007/s00432-020-03361-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/18/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE The prognosis of patients with advanced pancreatic ductal adenocarcinoma (PDAC) remains dismal. New cytotoxic agents such as nab-paclitaxel and liposomal irinotecan (nal-Iri) have extended the armamentarium of therapeutic options in the last years. Nowadays, sequential therapeutic strategies with moderately toxic chemotherapeutic protocols can be administered to the patients. However, prognostic and predictive biomarkers are still missing to identify those patients, which profit most from a "continuum of care" concept rather than receiving intensive first-line protocols such as FOLFIRINOX. To this end, we retrospectively evaluated the impact of the systemic inflammation as one essential hallmark of cancer in patients with advanced PDAC treated with sequential systemic. METHODS A cohort of 193 PDAC patients treated at our center from January 2005 to August 2011 were retrospectively evaluated for the following systemic inflammatory response (SIR) markers: neutrophil-lymphocyte ratio (NLR), lymphocyte-monocyte ratio (LMR) C-reactive protein (CRP), and the modified Glasgow Prognostic Score (mGPS). SIR markers were correlated with clinico-pathological findings, response to chemotherapy and overall survival (OS) using Kaplan-Meier curves and Cox proportional models. RESULTS All evaluated SIR markers were significantly associated with OS in patients with metastatic disease but not in patients with locally advanced PDAC. Interestingly, all SIR markers were only prognostic in patients not receiving antibiotics as surrogate marker for systemic bacterial infections. Based on the evaluated SIR markers, we propose a new Systemic Inflammation Score (SIS), which significantly correlated with reduced OS (HR: 3.418 (1.802-6.488, p < 0.001)) and the likelihood of receiving further-line systemic therapies (p = 0.028). CONCLUSION Routinely assessed SIR biomarkers have potential to support therapeutic decision making in patients with metastatic PDAC.
Collapse
Affiliation(s)
- M Markus
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
- Department of Anesthesiology and Operative Intensive Care Medicine (CCMCVK), Charité - University Hospital Berlin, Berlin, Germany
| | - A Abendroth
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - R Noureddine
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - A Paul
- West German Cancer Center, Department of General, Visceral and Transplant Surgery, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - S Breitenbuecher
- Institute for Quality Assurance, University Hospital Essen, Essen, Germany
| | - I Virchow
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - K W Schmid
- West German Cancer Center, Institute of Pathology Essen, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - P Markus
- Department of General Surgery and Traumatology, Elisabeth Hospital Essen, Essen, Germany
| | - B Schumacher
- Department of Gastroenterology, Elisabeth Hospital Essen, Essen, Germany
| | - M Wiesweg
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - J Wendling
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - B Mende
- Central Pharmacy, University Hospital Essen, Essen, Germany
| | - J T Siveke
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- West German Cancer Center, Institute for Developmental Cancer Therapeutics, University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - M Schuler
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
- Medical Faculty, University Duisburg-Essen, Essen, Germany
| | - S Kasper
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany.
- Medical Faculty, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
4
|
Lipner MB, Peng XL, Jin C, Xu Y, Gao Y, East MP, Rashid NU, Moffitt RA, Herrera Loeza SG, Morrison AB, Golitz BT, Vaziri C, Graves LM, Johnson GL, Yeh JJ. Irreversible JNK1-JUN inhibition by JNK-IN-8 sensitizes pancreatic cancer to 5-FU/FOLFOX chemotherapy. JCI Insight 2020; 5:129905. [PMID: 32213714 DOI: 10.1172/jci.insight.129905] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 03/18/2020] [Indexed: 12/11/2022] Open
Abstract
Over 55,000 people in the United States are diagnosed with pancreatic ductal adenocarcinoma (PDAC) yearly, and fewer than 20% of these patients survive a year beyond diagnosis. Chemotherapies are considered or used in nearly every PDAC case, but there is limited understanding of the complex signaling responses underlying resistance to these common treatments. Here, we take an unbiased approach to study protein kinase network changes following chemotherapies in patient-derived xenograft (PDX) models of PDAC to facilitate design of rational drug combinations. Proteomics profiling following chemotherapy regimens reveals that activation of JNK-JUN signaling occurs after 5-fluorouracil plus leucovorin (5-FU + LEU) and FOLFOX (5-FU + LEU plus oxaliplatin [OX]), but not after OX alone or gemcitabine. Cell and tumor growth assays with the irreversible inhibitor JNK-IN-8 and genetic manipulations demonstrate that JNK and JUN each contribute to chemoresistance and cancer cell survival after FOLFOX. Active JNK1 and JUN are specifically implicated in these effects, and synergy with JNK-IN-8 is linked to FOLFOX-mediated JUN activation, cell cycle dysregulation, and DNA damage response. This study highlights the potential for JNK-IN-8 as a biological tool and potential combination therapy with FOLFOX in PDAC and reinforces the need to tailor treatment to functional characteristics of individual tumors.
Collapse
Affiliation(s)
- Matthew B Lipner
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | | | - Chong Jin
- Lineberger Comprehensive Cancer Center.,Department of Biostatistics
| | - Yi Xu
- Lineberger Comprehensive Cancer Center
| | - Yanzhe Gao
- Lineberger Comprehensive Cancer Center.,Department of Pathology, and
| | - Michael P East
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | - Naim U Rashid
- Lineberger Comprehensive Cancer Center.,Department of Biostatistics
| | | | | | | | | | - Cyrus Vaziri
- Lineberger Comprehensive Cancer Center.,Department of Pathology, and
| | - Lee M Graves
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | - Gary L Johnson
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center
| | - Jen Jen Yeh
- Department of Pharmacology.,Lineberger Comprehensive Cancer Center.,Department of Surgery, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
5
|
Bashraheel SS, Domling A, Goda SK. Update on targeted cancer therapies, single or in combination, and their fine tuning for precision medicine. Biomed Pharmacother 2020; 125:110009. [PMID: 32106381 DOI: 10.1016/j.biopha.2020.110009] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 02/04/2020] [Accepted: 02/12/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Until recently, patients who have the same type and stage of cancer all receive the same treatment. It has been established, however, that individuals with the same disease respond differently to the same therapy. Further, each tumor undergoes genetic changes that cause cancer to grow and metastasize. The changes that occur in one person's cancer may not occur in others with the same cancer type. These differences also lead to different responses to treatment. Precision medicine, also known as personalized medicine, is a strategy that allows the selection of a treatment based on the patient's genetic makeup. In the case of cancer, the treatment is tailored to take into account the genetic changes that may occur in an individual's tumor. Precision medicine, therefore, could be defined in terms of the targets involved in targeted therapy. METHODS A literature search in electronic data bases using keywords "cancer targeted therapy, personalized medicine and cancer combination therapies" was conducted to include papers from 2010 to June 2019. RESULTS Recent developments in strategies of targeted cancer therapy were reported. Specifically, on the two types of targeted therapy; first, immune-based therapy such as the use of immune checkpoint inhibitors (ICIs), immune cytokines, tumor-targeted superantigens (TTS) and ligand targeted therapeutics (LTTs). The second strategy deals with enzyme/small molecules-based therapies, such as the use of a proteolysis targeting chimera (PROTAC), antibody-drug conjugates (ADC) and antibody-directed enzyme prodrug therapy (ADEPT). The precise targeting of the drug to the gene or protein under attack was also investigated, in other words, how precision medicine can be used to tailor treatments. CONCLUSION The conventional therapeutic paradigm for cancer and other diseases has focused on a single type of intervention for all patients. However, a large literature in oncology supports the therapeutic benefits of a precision medicine approach to therapy as well as combination therapies.
Collapse
Affiliation(s)
- Sara S Bashraheel
- Protein Engineering Unit, Life and Science Research Department, Anti-Doping Lab-Qatar (ADLQ), Doha, Qatar; Drug Design Group, Department of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Alexander Domling
- Drug Design Group, Department of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Sayed K Goda
- Cairo University, Faculty of Science, Chemistry Department, Giza, Egypt.
| |
Collapse
|
6
|
Kimura Y, Nakamura T, Hayashi T, Kuwatani M, Motoya M, Yoshida M, Imamura M, Nagayama M, Yamaguchi H, Yamakita K, Goto T, Sakuhara Y, Takahashi K, Maguchi H, Hirano S, Takemasa I. Clinical usefulness of conversion surgery for unresectable pancreatic cancer diagnosed on multidetector computed tomography imaging: Results from a multicenter observational cohort study by the Hokkaido Pancreatic Cancer Study Group (HOPS UR-01). Ann Gastroenterol Surg 2019; 3:523-533. [PMID: 31549012 PMCID: PMC6749954 DOI: 10.1002/ags3.12272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 04/26/2019] [Accepted: 06/04/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND AIM Effective multidisciplinary approaches for unresectable pancreatic cancer (UR-PC) that include modern chemotherapeutic regimens and subsequent conversion surgery (CS) are being developed. The aim of this study was to evaluate outcomes of patients clinically diagnosed with UR-PC, focusing on the efficacy of CS. METHODS Patients ineligible for two multicenter phase II studies conducted by the Hokkaido Pancreatic Cancer Study Group (HOPS) were recruited. Sequential treatment regimens, conversion to radical surgery, and overall survival (OS) were analyzed by multidetector computed tomography (MDCT)-based UR factors. Univariate and multivariate analyses were performed to identify predictors of OS. RESULTS Sixty-six of 247 intended recruits for HOPS studies from October 2013 to April 2016 were included. Unresectability was due to locally advanced (LA) disease and metastasis (M) in 42 and 24 patients, respectively. Induction therapy began with chemotherapy (CT) and chemoradiotherapy (CRT) in 44 and 17 patients, respectively, of whom 23 received modern CT regimens. Radical surgery was completed in 12 (LA, 10; M, two) with a median treatment interval of 10.3 months (range, 2-32). Eleven patients (91.6%) achieved pathological R0 resection. Median OS was significantly longer in patients who underwent CS than those who did not (44.1 vs 14.5 months, P < 0.0001). CS was an independent predictor of OS (hazard ratio, 0.078; 95% confident interval, 0.017-0.348; P = 0.001). CONCLUSION Conversion surgery after a favorable response to sequential treatment might prolong survival in patients with UR-PC. Precise diagnosis on MDCT followed by sequential multimodal anticancer treatment is essential.
Collapse
Affiliation(s)
- Yasutoshi Kimura
- Department of SurgerySurgical Oncology and ScienceSapporo Medical University School of MedicineSapporoJapan
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
| | - Toru Nakamura
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Department of Gastroenterological Surgery IIHokkaido UniversityFaculty of MedicineSapporoJapan
| | - Tsuyoshi Hayashi
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Center for GastroenterologyTeine‐Keijinkai HospitalSapporoJapan
| | - Masaki Kuwatani
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Department of Gastroenterology and HepatologyHokkaido University Graduate School of MedicineSapporoJapan
| | - Masayo Motoya
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Department of Gastroenterology and HepatologySapporo Medical University School of MedicineSapporoJapan
| | - Makoto Yoshida
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Department of Medical OncologySapporo Medical University School of MedicineSapporoJapan
| | - Masafumi Imamura
- Department of SurgerySurgical Oncology and ScienceSapporo Medical University School of MedicineSapporoJapan
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
| | - Minoru Nagayama
- Department of SurgerySurgical Oncology and ScienceSapporo Medical University School of MedicineSapporoJapan
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
| | - Hiroshi Yamaguchi
- Department of SurgerySurgical Oncology and ScienceSapporo Medical University School of MedicineSapporoJapan
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
| | - Keisuke Yamakita
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Division of Metabolism and Biosystemic ScienceDepartment of MedicineAsahikawa Medical UniversitySapporoJapan
| | - Takuma Goto
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Division of Gastroenterology and Hematology/OncologyDepartment of MedicineAsahikawa Medical UniversitySapporoJapan
| | - Yusuke Sakuhara
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Department of RadiologyTonan HospitalSapporoJapan
| | - Kuniyuki Takahashi
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Center for GastroenterologyTeine‐Keijinkai HospitalSapporoJapan
| | - Hiroyuki Maguchi
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Center for GastroenterologyTeine‐Keijinkai HospitalSapporoJapan
| | - Satoshi Hirano
- Hokkaido Pancreatic Cancer Study Group; HOPSSapporoJapan
- Department of Gastroenterological Surgery IIHokkaido UniversityFaculty of MedicineSapporoJapan
| | - Ichiro Takemasa
- Department of SurgerySurgical Oncology and ScienceSapporo Medical University School of MedicineSapporoJapan
| |
Collapse
|