1
|
Huang J, Zhang Q, Ge Y, Zheng R, Yang M, Sun Y, Go VLW, Zhang Z, Fang H, Liu J, Guo J, Xiao GG. Serum microRNA-24-based nomogram predicts prognosis for patients with resected pancreatic cancer. Sci Rep 2025; 15:8159. [PMID: 40059103 PMCID: PMC11891307 DOI: 10.1038/s41598-024-82369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/04/2024] [Indexed: 05/13/2025] Open
Abstract
Pancreatic cancer (PCa) is one of the malignant tumors with an extremely poor prognosis. Rare biomarkers exist for predicting the outcomes of PCa patients. This study aimed to develop a nomogram model based on serum microRNA-24 (miR-24) and clinicopathological factors to predict overall survival (OS) and treatment response to conventional adjuvant chemotherapy (ACT) in patients with PCa. This retrospective study included 296 patients with PCa who underwent radical resection and were followed up every three months. The serum levels of miR-24 were analyzed with real- time polymerase chain reaction, and the clinicopathological information relevant to the patients was extracted from the medical center. By combining miR-24 with some clinicopathological factors associated with prognosis, a nomogram model was developed to predict the OS of patients with PCa. Patients with elevated miR-24 levels exhibited significantly poorer OS compared to those at low risk (P < 0.0001). miR-24 was an independent predictor of OS regardless to the patients' age, gender, and clinical pathological characteristics. It demonstrated remarkable predictive power, with an AUC of 0.82, surpassing CA19-9 (AUC: 0.61), CA125 (AUC: 0.59), CA50 (AUC: 0.51) and CEA (0.56). When miR-24 was integrated with TNM stage, CA19-9 and CA125 in a nomogram, the prognostic accuracy was notably enhanced compared to individual factors. Furthermore, patients classified into the high-risk group who received post-operative ACT showed superior outcomes in both OS and two-year survival compared to those who did not receive ACT (P < 0.0001). A serum miR-24-based nomogram may serve as a powerful tool for predicting risk and prognosis in patients with resected pancreatic cancer, thus facilitating personalized clinical decision-making.
Collapse
Affiliation(s)
- Jing Huang
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology in School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Qian Zhang
- School of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, China
| | - Yang Ge
- Department of Food Safety and Toxicology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200240, China
| | - Ren Zheng
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology in School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China
| | - Minwei Yang
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yongwei Sun
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Vay Liang W Go
- David Geffen School of Medicine at UCLA, The UCLA Agi Hirshberg Center for Pancreatic Diseases, Los Angeles, CA, 90095, USA
| | - Zhigang Zhang
- State Key Laboratory of Oncogenes and Related Genes, School of Medicine, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Huilong Fang
- School of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, China
| | - Jianzhou Liu
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Junchao Guo
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Gary Guishan Xiao
- National Key Laboratory of Fine Chemical Engineering and Department of Pharmacology in School of Chemical Engineering, Dalian University of Technology, Dalian, 116024, China.
- Functional Genomics and Proteomics Laboratories, Osteoporosis Research Center, Creighton University Medical Center, Omaha, NE, 68124, USA.
- National Key Laboratory of Fine Chemical Engineering, and Center for Molecular Pharmacology and Department of Pharmacology in School of Chemical Engineering at, Dalian University of Technology, Dalian, China.
| |
Collapse
|
2
|
Li Z, Fan X, Jiang D, Li Q, Liu C, Wang D, Li N, Li H, Chen Z, Tang H, Lou C, Xu H, Zhan C, Dong Y, Ma Z, Wang G, Zhang C, Lu H, Zheng T, Zhang Y. Nab-paclitaxel plus S-1 followed by gemcitabine-oxaliplatin as first-line alternating sequential treatment of pancreatic ductal adenocarcinoma. Oncologist 2024; 29:997-e1614. [PMID: 39226089 PMCID: PMC11546623 DOI: 10.1093/oncolo/oyae207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 07/15/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Alternating sequential administration of drugs may be a promising approach to overcome chemotherapy resistance in advanced pancreatic ductal adenocarcinoma (PDAC). METHODS This study was an open-label, single-arm, and prospective trial included patients with untreated advanced PDAC. They received 2 cycles of NS regimen (nab-paclitaxel:125 mg/m2, intravenously injected on days 1 and 8, plus S-1:40-60 mg, orally twice per day for 1-14 days) followed by 2 cycles of GemOx regimen (gemcitabine, intravenously injected on days 1 and 8, and oxaliplatin: 130 mg/m2, intravenously injected on day 1). The primary efficacy endpoint was a progression-free survival rate at 6 months (PFSR-6m). The secondary efficacy endpoints included overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), and adverse events (AEs). Specific mRNA transcripts were used to explore survival associated genes. RESULTS Forty-two patients received a minimum of one treatment cycle, and of these, 30 patients completed one alternating treatment consisting of 4 cycles. The PFSR-6m was 71% (95% CI = 58%-87%). The median PFS and OS were 6.53 months (95% CI = 6.03-8.43) and 11.4 months (95% CI = 9.8-14.4), respectively. Common grades 3-4 hematological AEs included neutropenia 30.9%, leukopenia 26.2%, anemia 2.4%, and thrombocytopenia in 11.9%. Patients with OS > 10 months showed high expression of HLA-DQA2 while melanoma-associated antigen genes (MAGE) were notably upregulated in patients with OS < 10 months. CONCLUSION The alternating sequential administration of the NS and GemOx regimens may be a novel approach for first-line chemotherapy in patients with advanced PDAC requiring further study (ClinicalTrials.gov Identifier: ChiCTR1900024867).
Collapse
Affiliation(s)
- Zhiwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Xiaona Fan
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Dan Jiang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Qingwei Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chao Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Dan Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Na Li
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, People’s Republic of China
| | - Hengzhen Li
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Zhuo Chen
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Hongzhen Tang
- Shenzhen Engineering Center for Translational Medicine of Precision Cancer Immunodiagnosis and Therapy, YuceBio Technology Co., Ltd, Shenzhen, People’s Republic of China
| | - Changjie Lou
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Haitao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chao Zhan
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yuandi Dong
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Zhigang Ma
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Guangyu Wang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Chunhui Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Haibo Lu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
3
|
Safari D, Fakhrolmobasheri M, Soleymanjahi S. Efficacy and safety of intraperitoneal chemotherapy for pancreatic cancer. BMC Surg 2024; 24:285. [PMID: 39367354 PMCID: PMC11451220 DOI: 10.1186/s12893-024-02526-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/07/2024] [Indexed: 10/06/2024] Open
Abstract
Pancreatic cancer is a highly aggressive cancer with unfavorable prognosis despite the therapeutic interventions. Intraperitoneal chemotherapy has recently shown potential outcomes in the presence of peritoneal metastases. However, a consensus is still lacking on different methods for intraperitoneal chemotherapy in pancreatic cancer. A variety of drugs and doses via three types of intraperitoneal chemotherapy have been studied. The prognosis and treatment strategies for pancreatic ductal adenocarcinoma (PDAC) will be significantly influenced by peritoneal dissemination and resectability of the macroscopic disease. Normothermic intraperitoneal chemotherapy (NIPEC) has been used for the treatment of peritoneal metastases of pancreatic carcinomas. Intraperitoneal chemotherapy is often combined with systemic therapies or surgical procedures which may lead to favorable combination therapies such as cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a relatively new approach that provides a homogenous and deep penetration of the chemotherapy into the peritoneum by producing aerosols. The present study aims to review the literature for recent evidence on intraperitoneal chemotherapy in pancreatic cancer.
Collapse
Affiliation(s)
- Dorsa Safari
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Fakhrolmobasheri
- International Hematology/Oncology of Pediatrics Experts (IHOPE), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Saeed Soleymanjahi
- Division of Gastroenterology, Mass General Brigham, Harvard School of Medicine, 101 S Huntington Ave, Boston, MA, 02130, USA.
| |
Collapse
|
4
|
Seto I, Yamaguchi H, Takagawa Y, Azami Y, Takayama K, Suzuki M, Machida M, Dai Y, Sulaiman NSB, Kikuchi Y, Kato T, Nishino N, Teranishi Y, Murakami M. Clinical Outcomes of Proton Beam Therapy for Unresectable Locally Advanced Pancreatic Cancer: A Single-Center Retrospective Study. Adv Radiat Oncol 2024; 9:101577. [PMID: 39309704 PMCID: PMC11415529 DOI: 10.1016/j.adro.2024.101577] [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] [Received: 01/09/2024] [Accepted: 06/26/2024] [Indexed: 09/25/2024] Open
Abstract
Purpose We retrospectively researched the treatment outcome of proton beam therapy (PBT) and assessed its efficacy for inoperable locally advanced pancreatic cancer (LAPC) at our institution. Methods and Materials Fifty-four patients (28 men and 26 women, median age 67 years ranging from 40-88 years) were diagnosed with unresectable stage III LAPC and administered PBT from April 2009 to March 2020. Patients who could not complete PBT, had new distant metastases during the treatment, or did not have enough follow-up time were excluded from this study. All patients were clinically staged based on the International Union of Cancer TNM staging system (eighth edition) using computed tomography, magnetic resonance imaging, and positron emission tomography and were diagnosed as stage III (histologic type: 18 patients with adenocarcinoma and 36 clinically diagnosed patients). PBT was performed using the passive method, with a median total dose of 67.5 GyE (range, 50-77 GyE/25-35 fractions).Chemotherapy was used in combination during PBT in 46 patients (85.2%). Overall survival (OS), local progression-free survival (LPFS), progression-free survival, and median OS time were analyzed by Kaplan-Meier and log-rank tests. Univariate and multivariate analyses were performed for the following factors: maximum standardized uptake value (SUVmax), Eastern Cooperative Group performance status (PS), tumor site, total irradiation dose, concurrent chemotherapy, and primary tumor site. Cutoff values for SUVmax and tumor diameter were estimated using receiver operating characteristic curves and the area under the curve based on OS. Multivariate analysis was evaluated using the Cox proportional hazards models. Adverse events were evaluated using the National Cancer Institute Common Terminology Criteria for Adverse Events version 5.0. Results The median observation period was 17.4 months, ranging from 4.0 to 89.7 months. The median tumor diameter was 36.5 mm, ranging from 15 to 90 mm, the median SUVmax was 5.85 (range, 2.1-27.6), and their cutoff values were estimated to be 37 mm and 4.8 mm, respectively. The 1- and 2-year OS was 77.8% and 35.2%, respectively, with a median OS time of 18.2 months, and only one patient survived >5 years. Twelve patients (22.2%) developed local recurrence, and 1- and 2-year LPFS rates were 89.7% and 74.5%, respectively; progression-free survival at 1 year was 58.8%. The PS score, tumor site, and irradiation dose were the prognostic factors related to OS that showed a significant difference. On the other hand, there was a significant difference in factors involved in LPFS, at 96.7%/77.9% in the first year and 86.6%/54.4% in the second year in the groups with tumor dose ≥67.5 GyE and <67.5 GyE, respectively (P = .015). Treatment-related acute toxicities were neutropenia (grade 1/2/3 at 3.7%/11.1%/31.5%, respectively), leukopenia (grade 1/2/3 at 1.8%/7.4%/20.4%, respectively), and thrombocytopenia (grade 1/2 at 1.8%/7.4%, respectively), whereas the late effects including peptic ulcer were captured only grade 2+. The late adverse events of grade 3 or higher were not observed. Conclusions PBT achieving 67.5 Gy combined with standard chemotherapy showed excellent local control for unresectable LAPC. Total irradiation dose, tumor site, and PS score at an initial diagnosis could be important prognostic factors. In this study, the dose-effect relationship was found, so an increase in dose should be considered to improve prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Yasushi Teranishi
- Department of Surgery, Neuroscience, Southern Tohoku Research Institute for Neuroscience, Southern Tohoku Proton Therapy Center, Southern Tohoku General Hospital, Yatsuyamada, Koriyama, Japan
| | | |
Collapse
|
5
|
Jørgensen MS, Ainsworth AP, Fristrup CW, Mortensen MB, Graversen M. Impact of laparoscopic ultrasound during PIPAC directed treatment of unresectable peritoneal metastasis. Pleura Peritoneum 2024; 9:107-112. [PMID: 39544431 PMCID: PMC11558172 DOI: 10.1515/pp-2024-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/19/2024] [Indexed: 11/17/2024] Open
Abstract
Objectives Laparoscopic ultrasound (LUS) combines both laparoscopy and ultrasound imaging of the peritoneum liver and retroperitoneum. LUS has not been described in treatments with pressurized intraperitoneal aerosol chemotherapy (PIPAC). We present our experience with LUS in patients undergoing PIPAC. Methods Retrospective study of LUS findings from the prospective PIPAC-OPC2 trial. Main outcome was changes in overall treatment strategy due to LUS findings. Results PIPAC-OPC2 included 143 patients of which 33 patients were treated with electrostatic precipitation PIPAC. Nine patients were excluded due to primary non-access. During PIPAC 1, LUS was performed in 112 of 134 (84 %) PIPAC procedures and changed overall treatment strategy in one patient due to detection of multiple liver metastases unseen by baseline CT. During PIPAC 2 and 3 LUS was performed in 59 of 104 (57 %) and 42 of 78 (54 %) PIPAC procedures, respectively. Throughout PIPAC 1-3, LUS also detected pathological lymph nodes in 16 patients, and focal liver lesions in another four patients of uncertain origin. No further examinations were performed in these patients, and the overall treatment strategy was not changed according to the PIPAC-OPC2 protocol. One patient had a splenic capsule rupture related to the LUS itself. This was managed conservatively. Conclusions LUS may be safely performed during PIPAC. However, LUS has limited clinical impact in patients scheduled for PIPAC, and cannot be recommended as a routine procedure when performing PIPAC.
Collapse
Affiliation(s)
- Magnus S. Jørgensen
- Odense PIPAC Center, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Alan P. Ainsworth
- Odense PIPAC Center, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Claus W. Fristrup
- Odense PIPAC Center, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Michael B. Mortensen
- Odense PIPAC Center, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Martin Graversen
- Odense PIPAC Center, Department of Surgery, Odense University Hospital, Odense, Denmark
| |
Collapse
|
6
|
Levine JM, Rompen IF, Franco JC, Swett B, Kryschi MC, Habib JR, Diskin B, Hewitt DB, Sacks GD, Kaplan B, Berman RS, Cohen SM, Wolfgang CL, Javed AA. The impact of metastatic sites on survival Rates and predictors of extended survival in patients with metastatic pancreatic cancer. Pancreatology 2024; 24:887-893. [PMID: 38969544 PMCID: PMC11462613 DOI: 10.1016/j.pan.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND OBJECTIVES The aim of this study was to determine the role of site-specific metastatic patterns over time and assess factors associated with extended survival in metastatic PDAC. Half of all patients with pancreatic ductal adenocarcinoma (PDAC) present with metastatic disease. The site of metastasis plays a crucial role in clinical decision making due to its prognostic value. METHODS We examined 56,757 stage-IV PDAC patients from the National Cancer Database (2016-2019), categorizing them by metastatic site: multiple, liver, lung, brain, bone, carcinomatosis, or other. The site-specific prognostic value was assessed using log-rank tests while time-varying effects were assessed by Aalen's linear hazards model. Factors associated with extended survival (>3years) were assessed with logistic regression. RESULTS Median overall survival (mOS) in patients with distant lymph node-only metastases (9.0 months) and lung-only metastases (8.1 months) was significantly longer than in patients with liver-only metastases (4.6 months, p < 0.001). However, after six months, the metastatic site lost prognostic value. Logistic regression identified extended survivors (3.6 %) as more likely to be younger, Hispanic, privately insured, Charlson-index <2, having received chemotherapy, or having undergone primary or distant site surgery (all p < 0.001). CONCLUSION While synchronous liver metastases are associated with worse outcomes than lung-only and lymph node-only metastases, this predictive value is diminished after six months. Therefore, treatment decisions beyond this time should not primarily depend on the metastatic site. Extended survival is possible in a small subset of patients with favorable tumor biology and good conditional status, who are more likely to undergo aggressive therapies.
Collapse
Affiliation(s)
- Jonah M Levine
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Ingmar F Rompen
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA; Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Jorge Campos Franco
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Ben Swett
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Maximilian C Kryschi
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Joseph R Habib
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Brian Diskin
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - D Brock Hewitt
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Greg D Sacks
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Brian Kaplan
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Russel S Berman
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Steven M Cohen
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Christopher L Wolfgang
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA
| | - Ammar A Javed
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY, USA.
| |
Collapse
|
7
|
Detlefsen S, Burton M, Ainsworth AP, Fristrup C, Graversen M, Pfeiffer P, Tarpgaard LS, Mortensen MB. RNA expression profiling of peritoneal metastasis from pancreatic cancer treated with Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC). Pleura Peritoneum 2024; 9:79-91. [PMID: 38948326 PMCID: PMC11211652 DOI: 10.1515/pp-2024-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/12/2024] [Indexed: 07/02/2024] Open
Abstract
Objectives Pressurized Intraperitoneal Aerosol Chemotherapy (PIPAC) is an experimental treatment option in peritoneal metastasis from pancreatic cancer (PM-PC). Aims were to examine mRNA profile of fibrosis due to response after systemic chemotherapy and PIPAC (Regression) compared to treatment-naïve PM-PC and chronic cholecystitis-related peritoneal fibrosis (Controls). Methods Peritoneal biopsies (PBs) from PM-PC patients who had undergone systemic chemotherapy and PIPAC were evaluated with Peritoneal Regression Grading Score (PRGS). We extracted RNA from PBs with Regression (PRGS 1, n=11), treatment-naïve PM-PC (n=10), and Controls (n=10). Profiling of 800 mRNAs was performed (NanoString nCounter, PanCancer Immuno-Oncology 360 (IO-360) and 30 additional stroma-related mRNAs). Results Regression vs. PM-PC identified six up-regulated and 197 down-regulated mRNAs (FDR≤0.05), linked to TNF-α signaling via NF-kB, G2M checkpoint, epithelial-mesenchymal transition, estrogen response, and coagulation. Regression vs. Controls identified 43 significantly up-regulated mRNAs, linked to interferon-α response, and down-regulation of 99 mRNAs, linked to TNF-α signaling via NF-kB, inflammatory response, epithelial-mesenchymal transition, KRAS signaling, and hypoxia (FDR≤0.05). Conclusions In regressive fibrosis of PM-PC after systemic chemotherapy and PIPAC (Regression), downregulation of mRNAs related to key tumor biological pathways was identified. Regression also showed transcriptional differences from unspecific, benign fibrosis (Controls). Future studies should explore whether mRNA profiling of PBs with PM from PC or other primaries holds prognostic or predictive value.
Collapse
Affiliation(s)
- Sönke Detlefsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Mark Burton
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, University of Southern Denmark, Odense, Denmark
| | - Alan P. Ainsworth
- Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark
| | - Claus Fristrup
- Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark
| | - Martin Graversen
- Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark
- OPEN–Open Patient Data Explorative Network, Odense University Hospital, Region of Southern Denmark,Denmark
| | - Per Pfeiffer
- Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Line S. Tarpgaard
- Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Michael B. Mortensen
- Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Surgery, Upper GI and HPB Section, Odense University Hospital, Odense, Denmark
| |
Collapse
|
8
|
Gulturk I, Yilmaz M, Tacar SY, Bakkaloglu OK, Sonmezoz GB, Erdal GS, Ozmen A, Tural D. Naples prognostic score may predict overall survival in metastatic pancreatic cancer. J Cancer Res Ther 2024; 20:249-254. [PMID: 38554329 DOI: 10.4103/jcrt.jcrt_1392_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/21/2022] [Indexed: 04/01/2024]
Abstract
INTRODUCTION Naples prognostic score (NPS) based on nutritional and inflammatory parameters can predict response to chemotherapy and overall survival (OS) in many cancer types. However, its significance in metastatic pancreatic cancer (PC) remains unclear. We evaluated the prognostic significance of the NPS in patients with metastatic PC receiving first line chemotherapy. METHODS We retrospectively analyzed 215 patients with metastatic PC receiving first line FOLFIRINOX chemotherapy. NPS's were calculated using pre-chemotherapy laboratory data. Patients were divided into three groups according to their scores (NPS: 0; 1 & 2; 3 & 4 were grouped as 1, 2 and 3, respectively). The association of NPS with clinicopathological features and OS were evaluated. RESULTS Median age was 64 years, and median OS was 10.5 months. Hemoglobin levels were lower and Ca-19-9 values were higher with increasing NPS. Frequency of patients with bone and/or liver metastases, and with greater than 5 metastatic focus were higher in group 3. A lower NPS was associated with longer OS. The median OS in groups 1, 2, and 3 were 19.5, 12, and 8 months, respectively, and differed significantly. Univariate analysis revealed effect of NPS (3-4) on OS (HR: 2.38, 1.77-3.19). Other prognostic factors affecting OS were age, ECOG, liver, bone or lymph node metastases, number of metastatic foci (<5 vs >5), de-novo metastatic disease, and serum Ca-19-9 levels. NPS (3-4) was identified as an independent prognostic factor negatively affecting OS (HR: 1.89, 1.34-2.65) in multivariate analysis. CONCLUSION NPS may be a useful prognostic marker for the prediction of OS in metastatic PC patients receiving systemic chemotherapy.
Collapse
Affiliation(s)
- Ilkay Gulturk
- Medical Oncology Department, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Mesut Yilmaz
- Medical Oncology Department, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Seher Y Tacar
- Medical Oncology Department, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Oguz K Bakkaloglu
- Gastroenterology Department, Kartal Kosuyolu High Specialization Education and Research Hospital, Istanbul, Turkey
| | - Gulru B Sonmezoz
- Internal Medicine Department, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Gulcin S Erdal
- Medical Oncology Department, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Aykut Ozmen
- Medical Oncology Department, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| | - Deniz Tural
- Medical Oncology Department, Bakirkoy Dr. Sadi Konuk Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
9
|
Pourali G, Donyadideh G, Mehrabadi S, Hamid F, Hassanian SM, Ferns GA, Khazaei M, Avan A. Clinical practice guidelines for interventional treatment of pancreatic cancer. RECENT ADVANCES IN NANOCARRIERS FOR PANCREATIC CANCER THERAPY 2024:345-373. [DOI: 10.1016/b978-0-443-19142-8.00008-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
10
|
Lavacchi D, Polvani S, Taddei A, Scolari F, Messerini L, Caliman E, Moraldi L, Guidolin A, Grazi GL, Galli A, Pillozzi S, Antonuzzo L. KRAS-related miR-143 expression is associated with lymph node involvement and correlates with outcome in pancreatic adenocarcinoma patients. Front Oncol 2023; 13:1295936. [PMID: 38130990 PMCID: PMC10735715 DOI: 10.3389/fonc.2023.1295936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 11/21/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction Pancreatic adenocarcinoma (PC) is one of the most lethal malignancies; even after resection the patients' 5-year disease-free survival (DFS) is lower than 26%. The genetic mutational landscape of PC is dominated by activating KRAS mutations, that have been reported in approximately 90% of cases; however, beyond KRAS - direct mutations, several KRAS-targeting miRNAs appear to be downregulated, strengthening the already activated RAS signaling. In addition, the interplay between miRNAs and RAS includes poorly investigated downstream miRNAs. The aim of this study was to determine the prognostic value of some of these candidate KRAS-related miRNAs. Patients and methods Between 2015 and 2022, 44 patients with pathologically confirmed PC, who received surgery and were enrolled by the Clinical Oncology Unit, Careggi University Hospital, Florence (Italy). PC Total RNA was extracted from FFPE sections, retro-transcribed and the resulting cDNA was then used for qPCR analysis. A panel of KRAS-related miRNA (miR-155, miR-206 and miR-143) was analyzed. Results In this observational study patients sex distribution was unequal with 34.1% being male and 65.9% female. The most frequent tumor localization was the head of the pancreas (65.9%) and the pathological stages were pT1-2 (45.5%), pT3 (54.5%), pN0 (22.7%), pN+ (77.3%). Adjuvant therapy was administered to 63.6% of patients; disease recurrence was observed in 69% of cases. Twenty-three patients, whose RNA was of adequate quality, were used in the mRNAs expression studies. When comparing the miRNA expression between PC and a pool of healthy tissues, miR-155 was overexpressed and miR-206 downregulated in PC, while miR-143 expression was unchanged. However, when categorized in low- and high- miR-143 expressing PC (according to the median value), high miR-143 was associated with nodal involvement (pN+) (p=0.029), who in turn was linked with shorter DFS (p=0.009) and overall survival (OS) (p=0.021) compared to pN0. A trend toward inferior DFS was observed for higher expression of miR-206 (p=0.095) and miR-143 (p=0.092). Finally, responders to a first-line treatment for advanced disease had miR-155 overexpressed (p=0.048). Conclusions miRNAs are involved in PC tumorigenesis and metastatic spread. In light of miR-143 association with lymphatic spread and poor prognosis, a comprehensive analysis of miRNA interplay with KRAS deserves further investigation.
Collapse
Affiliation(s)
- Daniele Lavacchi
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Simone Polvani
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Antonio Taddei
- HPB Surgery Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Federico Scolari
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Luca Messerini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Enrico Caliman
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Luca Moraldi
- HPB Surgery Unit, Careggi University Hospital, Florence, Italy
| | - Alessia Guidolin
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
| | - Gian Luca Grazi
- HPB Surgery Unit, Careggi University Hospital, Florence, Italy
| | - Andrea Galli
- Department of Clinical and Experimental Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Serena Pillozzi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Antonuzzo
- Clinical Oncology Unit, Careggi University Hospital, Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
11
|
Kryh-Jensen CG, Fristrup CW, Ainsworth AP, Detlefsen S, Mortensen MB, Pfeiffer P, Tarpgaard LS, Graversen M. What is long-term survival in patients with peritoneal metastasis from gastric, pancreatic, or colorectal cancer? A study of patients treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC). Pleura Peritoneum 2023; 8:147-155. [PMID: 38144215 PMCID: PMC10739291 DOI: 10.1515/pp-2023-0038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/27/2023] [Indexed: 12/26/2023] Open
Abstract
Objectives A definition of long-term survival (LTS) in patients with peritoneal metastasis (PM) from gastric cancer (GC), pancreatic cancer (PC) or colorectal cancer (CRC) treated with systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC) is lacking. We aimed to define LTS and investigate characteristics and treatment response in patients who reached LTS in data from two prospective trials. Methods Retrospective study of patients with GC-, PC-, or CRC-PM from the prospective PIPAC-OPC1 and PIPAC-OPC2 studies. The definition of LTS was based on published systematic reviews and randomized controlled trials. LTS was defined at the time point where 25 % of the patients were alive in these studies. Histology based response was evaluated by the mean Peritoneal Regression Grading Score (PRGS) using biopsies obtained prior to PIPAC 3, and defined by a mean PRGS of ≤2.0 or a decrease of mean PRGS of ≥1, compared to baseline. Results LTS was defined at 21 (GC), 15 (PC), and 24 (CRC) months. Fifty-one (47.2 %) patients (nine GC, 17 PC, 25 CRC) reached LTS calculated from the date of PM diagnosis. All but one received palliative chemotherapy before PIPAC, and 37 % received bidirectional treatment. More than 90 % of the LTS patients had response according to PRGS. The mOS from PIPAC 1 was 23.3, 12.4, and 28.5 months for GC, PC, and CRC LTS patients. Conclusions Patients with PM from GC, PC, and CRC treated with systemic chemotherapy and PIPAC can reach LTS and most show histological response. Causality must be further investigated.
Collapse
Affiliation(s)
- Charlotte G. Kryh-Jensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Claus W. Fristrup
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Alan P. Ainsworth
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Michael B. Mortensen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Per Pfeiffer
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Line S. Tarpgaard
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Martin Graversen
- Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
- Upper GI & HPB Section, Department of Surgery, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- OPEN – Odense Patient Data Explorative Network, Odense University Hospital, Region of Southern Denmark, Odense, Denmark
| |
Collapse
|
12
|
Ishizaki A, Okuwaki K, Kida M, Imaizumi H, Iwai T, Yamauchi H, Kaneko T, Hasegawa R, Watanabe M, Kurosu T, Ishizaki J, Kusano C. Implication of Skeletal Muscle Loss in the Prognosis of Patients with Pancreatic Ductal Adenocarcinoma Receiving Chemotherapy. Intern Med 2023; 62:2783-2793. [PMID: 36792197 PMCID: PMC10602831 DOI: 10.2169/internalmedicine.0900-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 12/26/2022] [Indexed: 02/16/2023] Open
Abstract
Objective The effect of sarcopenia on the prognosis of patients undergoing chemotherapy for unresectable pancreatic ductal adenocarcinoma remains largely unexplored. In this retrospective study, we investigated the relationship between sarcopenia and the prognosis of patients receiving first-line nanoparticle albumin-bound paclitaxel plus gemcitabine for unresectable pancreatic ductal adenocarcinoma. Methods We enrolled 251 patients with unresectable metastatic or locally advanced pancreatic ductal adenocarcinoma who had received chemotherapy between January 2015 and December 2020 at Kitasato University Hospital. Univariate and multivariate analyses were performed using the stratified Cox proportional hazards model to determine variables significantly associated with the progression-free and overall survival. Propensity score matching was performed to mitigate selection bias effects. Results In the propensity score-matched cohort, the progression-free and overall survival were not significantly different between the sarcopenia and non-sarcopenia groups (p=0.335, and 0.679 respectively). The skeletal muscle index decreased by 4.4% and 6.5% in the sarcopenia and non-sarcopenia groups, respectively, during the early treatment phase (p=0.084). There were no significant differences between groups with regard to major adverse events or drug toxicity occurrences. Both the progression-free and overall survival were significantly shorter in the skeletal muscle index loss group than in the non-skeletal muscle index loss group (p=0.026 and 0.045, respectively). Conclusion Skeletal muscle index loss during the initial treatment phase may be an early marker for the long-term prognosis of patients receiving nanoparticle albumin-bound paclitaxel plus gemcitabine as first-line treatment for unresectable pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Ayana Ishizaki
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Kosuke Okuwaki
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Mitsuhiro Kida
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Hiroshi Imaizumi
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Tomohisa Iwai
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Hiroshi Yamauchi
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Toru Kaneko
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Rikiya Hasegawa
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Masafumi Watanabe
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Takahiro Kurosu
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Junro Ishizaki
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| | - Chika Kusano
- Department of Gastroenterology, Kitasato University School of Medicine, Japan
| |
Collapse
|
13
|
Chiu YF, Liu TW, Shan YS, Chen JS, Li CP, Ho CL, Hsieh RK, Hwang TL, Chen LT, Ch'ang HJ. Carbohydrate Antigen 19-9 Response to Initial Adjuvant Chemotherapy Predicts Survival and Failure Pattern of Resected Pancreatic Adenocarcinoma but Not Which Patients Are Suited for Additional Adjuvant Chemoradiation Therapy: From a Prospective Randomized Study. Int J Radiat Oncol Biol Phys 2023; 117:74-86. [PMID: 37055279 DOI: 10.1016/j.ijrobp.2023.02.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/19/2023] [Accepted: 02/25/2023] [Indexed: 04/15/2023]
Abstract
PURPOSE The predictive value of carbohydrate antigen 19-9 (CA19-9) for adjuvant chemo(radiation) therapy of resected pancreatic adenocarcinoma (PDAC) is undefined. METHODS AND MATERIALS We analyzed CA19-9 levels in patients with resected PDAC in a prospective randomized trial of adjuvant chemotherapy with or without additional chemoradiation therapy (CRT). Patients with postoperative CA19-9 ≤92.5 U/mL and serum bilirubin ≤2 mg/dL were randomized to 2 arms: patients in 1 arm received 6 cycles of gemcitabine, whereas those in the other received 3 cycles of gemcitabine followed by CRT and another 3 cycles of gemcitabine. Serum CA19-9 was measured every 12 weeks. Those who had CA19-9 levels always <3 U/mL were excluded from the exploratory analysis. RESULTS One hundred forty-seven patients were enrolled in this randomized trial. Twenty-two patients with CA19-9 levels always ≤3 U/mL were excluded from the analysis. For the 125 participants, median overall survival (OS) and recurrence-free survival were 23.1 and 12.1 months, respectively, with no significant differences between the study arms. Postresection CA19-9 levels and, to a lesser extent, CA19-9 change predicted OS (P = .040 and .077, respectively). For the 89 patients who completed the initial 3 cycles of adjuvant gemcitabine, the CA19-9 response was significantly correlated with initial failure over the distant site (P = .023) and OS (P = .0022). Despite a trend of less initial failure over the locoregional area (P = .031), neither postoperative CA19-9 level nor CA19-9 response helped to select patients who might have a survival benefit from additional adjuvant CRT. CONCLUSIONS CA19-9 response to initial adjuvant gemcitabine predicts survival and distant failure of PDAC after resection; however, it cannot select patients suited for additional adjuvant CRT. Monitoring CA19-9 levels during adjuvant therapy for postoperative patients with PDAC may guide therapeutic decisions to prevent distant failure.
Collapse
Affiliation(s)
- Yen-Feng Chiu
- Institute of Public Health Sciences, National Health Research Institutes, Miaoli, Taiwan
| | - Tsang-Wu Liu
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan
| | - Yan-Shen Shan
- Department of Surgery, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Shi Chen
- Department of Hematology-Oncology, Linkou Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Chung-Pin Li
- Divisions of Clinical Skills Training, Department of Medical Education, Taipei, Taiwan; Divisions of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ching-Liang Ho
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ruey-Kuen Hsieh
- Department of Internal Medicine, Mackay Memorial Hospital, Taipei, Taiwan
| | - Tsann-Long Hwang
- Department of Surgery, Linkou Chang-Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan; Department of Internal Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan; Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hui-Ju Ch'ang
- National Institute of Cancer Research, National Health Research Institutes, Miaoli, Taiwan; Department of Radiation Oncology, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan; Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
14
|
Rasmussen LS, Winther SB, Chen IM, Weber B, Ventzel L, Liposits G, Johansen JS, Detlefsen S, Egendal I, Shim S, Christensen S, Pfeiffer P, Ladekarl M. A randomized phase II study of full dose gemcitabine versus reduced dose gemcitabine and nab-paclitaxel in vulnerable patients with non-resectable pancreatic cancer (DPCG-01). BMC Cancer 2023; 23:552. [PMID: 37328835 PMCID: PMC10273702 DOI: 10.1186/s12885-023-11035-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 05/31/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND According to current evidence, the best treatment for fit patients with non-resectable pancreatic cancer (PC) is combination chemotherapy, whereas frail patients are recommended gemcitabine (Gem) monotherapy. Randomized controlled trials in colorectal cancer and a post-hoc analysis of gemcitabine and nab-paclitaxel (GemNab) in PC suggest, however, that reduced dose of combination chemotherapy may be feasible and more efficient compared to monotherapy in frail patients. The aim of this study is to investigate whether reduced dose GemNab is superior to full dose Gem in patients with resectable PC, who are not candidates for full dose combination chemotherapy in first line. METHODS The Danish Pancreas Cancer Group (DPCG)-01 trial is a national multicenter prospective randomized phase II trial. A total of 100 patients in ECOG performance status 0-2 with non-resectable PC, not candidate for full dose combination chemotherapy in first line, but eligible for full dose Gem, will be included. Patients are randomized 1:1 to either full dose Gem or GemNab in 80% of recommended dose. The primary endpoint is progression-free survival. Secondary endpoints are overall survival, overall response rate, quality of life, toxicity and rate of hospitalizations during treatment. The correlation between blood inflammatory markers, including YKL-40 and IL-6, circulating tumor DNA, and tissue biomarkers of resistance to chemotherapy and outcome will be explored. Finally, the study will include measures of frailty (G8, modified G8, and chair-stand-test) to assess whether scoring would enable a personalized allocation to different treatments or indicates a possibility for interventions. DISCUSSION Single-drug treatment with Gem has for frail patients with non-resectable PC been the main treatment option for more than thirty years, but the impact on outcome is modest. If improved results and sustained tolerability with reduced dose combination chemotherapy can be shown, this could change the future practice for this increasing group of patients. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05841420. Secondary Identifying No: N-20210068. EudraCT No: 2021-005067-52. PROTOCOL VERSION 1.5, 16-MAY-2023.
Collapse
Affiliation(s)
- Louise Skau Rasmussen
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Stine B Winther
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Inna M Chen
- Department of Oncology, Herlev-Gentofte University Hospital, Copenhagen, Denmark
| | - Britta Weber
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Lise Ventzel
- Department of Oncology, University Hospital of Southern Denmark, Vejle, Denmark
| | - Gabor Liposits
- Department of Oncology, Gødstrup Hospital, Herning, Denmark
| | - Julia Sidenius Johansen
- Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense University Hospital, and Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Ida Egendal
- Center for Clinical Data Science (CLINDA), and Clinical Cancer Research Center, Aalborg University and, Aalborg University Hospital, Aalborg, Denmark
| | - Susy Shim
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, and Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Signe Christensen
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Per Pfeiffer
- Department of Oncology, Odense University Hospital, and Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Morten Ladekarl
- Department of Oncology and Clinical Cancer Research Center, Aalborg University Hospital, and Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
15
|
Peng M, Ying Y, Zhang Z, Liu L, Wang W. Reshaping the Pancreatic Cancer Microenvironment at Different Stages with Chemotherapy. Cancers (Basel) 2023; 15:2448. [PMID: 37173915 PMCID: PMC10177210 DOI: 10.3390/cancers15092448] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/09/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023] Open
Abstract
The dynamic tumor microenvironment, especially the immune microenvironment, during the natural progression and/or chemotherapy treatment is a critical frontier in understanding the effects of chemotherapy on pancreatic cancer. Non-stratified pancreatic cancer patients always receive chemotherapeutic strategies, including neoadjuvant chemotherapy and adjuvant chemotherapy, predominantly according to their physical conditions and different disease stages. An increasing number of studies demonstrate that the pancreatic cancer tumor microenvironment could be reshaped by chemotherapy, an outcome caused by immunogenic cell death, selection and/or education of preponderant tumor clones, adaptive gene mutations, and induction of cytokines/chemokines. These outcomes could in turn impact the efficacy of chemotherapy, making it range from synergetic to resistant and even tumor-promoting. Under chemotherapeutic impact, the metastatic micro-structures in the primary tumor may be built to leak tumor cells into the lymph or blood vasculature, and micro-metastatic/recurrent niches rich in immunosuppressive cells may be recruited by cytokines and chemokines, which provide housing conditions for these circling tumor cells. An in-depth understanding of how chemotherapy reshapes the tumor microenvironment may lead to new therapeutic strategies to block its adverse tumor-promoting effects and prolong survival. In this review, reshaped pancreatic cancer tumor microenvironments due to chemotherapy were reflected mainly in immune cells, pancreatic cancer cells, and cancer-associated fibroblast cells, quantitatively, functionally, and spatially. Additionally, small molecule kinases and immune checkpoints participating in this remodeling process caused by chemotherapy are suggested to be blocked reasonably to synergize with chemotherapy.
Collapse
Affiliation(s)
- Maozhen Peng
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ying Ying
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zheng Zhang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Liang Liu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wenquan Wang
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (M.P.); (Y.Y.); (Z.Z.)
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
16
|
Catalano M, Roviello G, Aprile G, Ramello M, Conca R, Petrioli R, Perrone G, Ianza A, Mini E. Prognostic value of alkaline phosphatase and gamma-glutamyl transferase in patients with metastatic pancreatic cancer. Future Oncol 2023; 19:937-946. [PMID: 37232154 DOI: 10.2217/fon-2023-0137] [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: 02/16/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
Background: Pancreatic cancer (PC) is one of the most lethal malignancies worldwide. This study evaluated the prognostic role of serum alanine phosphatase (ALP) and gamma-glutamyl-transferase (GGT) in metastatic PC patients. Materials & methods: 153 patients with metastatic PC receiving first-line treatment with nab-paclitaxel/gemcitabine were retrospectively enrolled in a multicenter study and stratified according to ALP (≤ or >260 U/l) and GGT (≤ or >45.5 U/l) levels. Results: Improved overall survival was recorded in patients with GGT levels ≤45.5 U/l (p < 0.05). In patients with liver metastasis, overall survival was significantly lower in patients with high ALP (p = 0.01) and GGT (p = 0.02). Conclusion: High levels of ALP and GGT were related to a poor prognosis in PC patients with liver metastasis receiving nab-paclitaxel/gemcitabine.
Collapse
Affiliation(s)
- Martina Catalano
- Department of Health Sciences, University of Florence, 50139, Florence, Italy
| | | | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, AULSS8 Berica, Vicenza, Italy
| | - Monica Ramello
- Oncology Unit, Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
| | - Raffaele Conca
- Division of Medical Oncology, Department of Onco-Hematology, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028, Rionero, Vulture (PZ), Italy
| | - Roberto Petrioli
- Department of Medicine, Surgery & Neurosciences, Medical Oncology Unit, University of Siena, 53100, Siena, Italy
| | - Gabriele Perrone
- Department of Health Sciences, University of Florence, 50139, Florence, Italy
| | - Anna Ianza
- Oncology Unit, Department of Medical, Surgical, & Health Sciences, University of Trieste, Piazza Ospitale, Trieste, Italy
| | - Enrico Mini
- Department of Health Sciences, University of Florence, 50139, Florence, Italy
| |
Collapse
|
17
|
Guchelaar NAD, Noordman BJ, Koolen SLW, Mostert B, Madsen EVE, Burger JWA, Brandt-Kerkhof ARM, Creemers GJ, de Hingh IHJT, Luyer M, Bins S, van Meerten E, Lagarde SM, Verhoef C, Wijnhoven BPL, Mathijssen RHJ. Intraperitoneal Chemotherapy for Unresectable Peritoneal Surface Malignancies. Drugs 2023; 83:159-180. [PMID: 36633826 PMCID: PMC9908703 DOI: 10.1007/s40265-022-01828-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
Malignancies of the peritoneal cavity are associated with a dismal prognosis. Systemic chemotherapy is the gold standard for patients with unresectable peritoneal disease, but its intraperitoneal effect is hampered by the peritoneal-plasma barrier. Intraperitoneal chemotherapy, which is administered repeatedly into the peritoneal cavity through a peritoneal implanted port, could provide a novel treatment modality for this patient population. This review provides a systematic overview of intraperitoneal used drugs, the performed clinical studies so far, and the complications of the peritoneal implemental ports. Several anticancer drugs have been studied for intraperitoneal application, with the taxanes paclitaxel and docetaxel as the most commonly used drug. Repeated intraperitoneal chemotherapy, mostly in combination with systemic chemotherapy, has shown promising results in Phase I and Phase II studies for several tumor types, such as gastric cancer, ovarian cancer, colorectal cancer, and pancreatic cancer. Two Phase III studies for intraperitoneal chemotherapy in gastric cancer have been performed so far, but the results regarding the superiority over standard systemic chemotherapy alone, are contradictory. Pressurized intraperitoneal administration, known as PIPAC, is an alternative way of administering intraperitoneal chemotherapy, and the first prospective studies have shown a tolerable safety profile. Although intraperitoneal chemotherapy might be a standard treatment option for patients with unresectable peritoneal disease, more Phase II and Phase III studies focusing on tolerability profiles, survival rates, and quality of life are warranted in order to establish optimal treatment schedules and to establish a potential role for intraperitoneal chemotherapy in the approach to unresectable peritoneal disease.
Collapse
Affiliation(s)
- Niels A. D. Guchelaar
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Bo J. Noordman
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Stijn L. W. Koolen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
- Department of Pharmacy, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Bianca Mostert
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Eva V. E. Madsen
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Alexandra R. M. Brandt-Kerkhof
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Geert-Jan Creemers
- Department of Medical Oncology, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Ignace H. J. T. de Hingh
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
- Department of Epidemiology, GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Misha Luyer
- Department of Surgery, Catharina Cancer Institute, Eindhoven, The Netherlands
| | - Sander Bins
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Esther van Meerten
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| | - Sjoerd M. Lagarde
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Bas P. L. Wijnhoven
- Department of Surgery, Division of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ron. H. J. Mathijssen
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
18
|
Zhang L, Jin R, Yang X, Ying D. A population-based study of synchronous distant metastases and prognosis in patients with PDAC at initial diagnosis. Front Oncol 2023; 13:1087700. [PMID: 36776324 PMCID: PMC9909560 DOI: 10.3389/fonc.2023.1087700] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
Objective Cancer of the pancreas is a life-threatening condition and has a high distant metastasis (DM) rate of over 50% at diagnosis. Therefore, this study aimed to determine whether patterns of distant metastases correlated with prognosis in pancreatic ductal adenocarcinoma (PDAC) with metastatic spread, and build a novel nomogram capable of predicting the 6, 12, 18-month survival rate with high accuracy. Methods We analyzed data from the Surveillance, Epidemiology, and End Results (SEER) database for cases of PDAC with DM. Kaplan-Meier analysis, log-rank tests and Cox-regression proportional hazards model were used to assess the impact of site and number of DM on the cancer-specific survival (CSS) and over survival (OS). A total of 2709 patients with DM were randomly assigned to the training group and validation group in a 7:3 ratio. A nomogram was constructed by the dependent risk factors which were determined by multivariate Cox-regression analysis. An assessment of the discrimination and ability of the prediction model was made by measuring AUC, C-index, calibration curve and decision curve analysis (DCA). In addition, we collected 98 patients with distant metastases at the time of initial diagnosis from Ningbo University Affiliated LiHuili Hospital to verify the efficacy of the prediction model. Results There was a highest incidence of liver metastases from pancreatic cancer (2387,74.36%), followed by lung (625,19.47%), bone (190,5.92%), and brain (8,0.25%). The prognosis of liver metastases differed from that of lung metastases, and the presence of multiple organ metastases was associated with poorer prognosis. According to univariate and multivariate Cox-regression analyses, seven factors (i.e., diagnosis age, tumor location, grade of tumor differentiation, T-stage, receipt of surgery, receipt of chemotherapy status, presence of multiple organ metastases) were included in our nomogram model. In internal and external validation, the ROC curves, C-index, calibration curves and DCA were calculated, which confirmed that this nomogram can precisely predict prognosis of PDAC with DM. Conclusion Metastatic PDAC patients with liver metastases tended to have a worse prognosis than those with lung metastases. The number of DM had significant effect on the overall survival rate of metastatic PDAC. This study had a high prediction accuracy, which was helpful clinicians to analyze the prognosis of PDAC with DM and implement individualized diagnosis and treatment.
Collapse
Affiliation(s)
- Leiming Zhang
- Department of Minimally Invasive Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Rong Jin
- Department of Minimally Invasive Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Xuanang Yang
- School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Dongjian Ying
- Department of Minimally Invasive Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
19
|
Huffman BM, Basu Mallick A, Horick NK, Wang-Gillam A, Hosein PJ, Morse MA, Beg MS, Murphy JE, Mavroukakis S, Zaki A, Schlechter BL, Sanoff H, Manz C, Wolpin BM, Arlen P, Lacy J, Cleary JM. Effect of a MUC5AC Antibody (NPC-1C) Administered With Second-Line Gemcitabine and Nab-Paclitaxel on the Survival of Patients With Advanced Pancreatic Ductal Adenocarcinoma: A Randomized Clinical Trial. JAMA Netw Open 2023; 6:e2249720. [PMID: 36602796 PMCID: PMC9856813 DOI: 10.1001/jamanetworkopen.2022.49720] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
IMPORTANCE Treatment options are limited for patients with advanced pancreatic ductal adenocarcinoma (PDAC) beyond first-line 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX), with such individuals commonly being treated with gemcitabine and nab-paclitaxel. OBJECTIVE To determine whether NPC-1C, an antibody directed against MUC5AC, might increase the efficacy of second-line gemcitabine and nab-paclitaxel in patients with advanced PDAC. DESIGN, SETTING, AND PARTICIPANTS This multicenter, randomized phase II clinical trial enrolled patients with advanced PDAC between April 2014 and March 2017 whose disease had progressed on first-line FOLFIRINOX. Eligible patients had tumors with at least 20 MUC5AC staining by centralized immunohistochemistry review. Statistical analysis was performed from April to May 2022. INTERVENTIONS Patients were randomly assigned to receive gemcitabine (1000 mg/m2) and nab-paclitaxel (125 mg/m2) administered intravenously on days 1, 8, and 15 of every 4-week cycle, with or without intravenous NPC-1C 1.5 mg/kg every 2 weeks. MAIN OUTCOMES AND MEASURES The primary end point was overall survival (OS). Secondary end points were progression-free survival (PFS), objective response rate (ORR), and safety. Pretreatment clinical variables were explored with Cox proportional hazards analysis. RESULTS A total of 78 patients (median [range] age, 62 [36-78] years; 32 [41%] women; 9 [12%] Black; 66 [85%] White) received second-line treatment with gemcitabine plus nab-paclitaxel (n = 40) or gemcitabine plus nab-paclitaxel and NPC-1C (n = 38). Median OS was 6.6 months (95% CI, 4.7-8.4 months) with gemcitabine plus nab-paclitaxel vs 5.0 months (95% CI, 3.3-6.5 months; P = .22) with gemcitabine plus nab-paclitaxel and NPC-1C. Median PFS was 2.7 months (95% CI, 1.9-4.1 months) with gemcitabine plus nab-paclitaxel vs 3.4 months (95% CI, 1.9-5.3 months; P = .80) with gemcitabine plus nab-paclitaxel and NPC-1C. The ORR was 3.1% (95% CI, 0.4%-19.7%) in the gemcitabine plus nab-paclitaxel and NPC-1C group and 2.9% (95% CI, 0.4%-18.7%) in the gemcitabine plus nab-paclitaxel group. No differences in toxicity were observed between groups, except that grade 3 or greater anemia occurred more frequently in patients treated with gemcitabine plus nab-paclitaxel and NPC-1C than gemcitabine plus nab-paclitaxel (39% [15 of 38] vs 10% [4 of 40]; P = .003). The frequency of chemotherapy dose reductions was similar in both groups (65% vs 74%; P = .47). Lower performance status, hypoalbuminemia, PDAC diagnosis less than or equal to 18 months before trial enrollment, lymphocyte-to-monocyte ratio less than 2.8, and CA19-9 greater than 2000 IU/mL were independently associated with poorer survival. CONCLUSIONS AND RELEVANCE In this randomized clinical trial of advanced PDAC, NPC-1C did not enhance the efficacy of gemcitabine/nab-paclitaxel. These data provide a benchmark for future trials investigating second-line treatment of PDAC. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT01834235.
Collapse
Affiliation(s)
- Brandon M. Huffman
- Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Atrayee Basu Mallick
- Thomas Jefferson University/Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| | - Nora K. Horick
- Biostatistics Center, Massachusetts General Hospital, Boston
| | - Andrea Wang-Gillam
- Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | | | | | - Muhammad Shaalan Beg
- UT Southwestern Medical Center, Dallas, Texas
- Science 37 Inc, Durham, North Carolina
| | - Janet E. Murphy
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston
| | | | | | | | | | - Christopher Manz
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | - Brian M. Wolpin
- Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| | | | - Jill Lacy
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - James M. Cleary
- Division of Gastrointestinal Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute/Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Prejac J, Tomek Hamzić D, Librenjak N, Goršić I, Kekez D, Pleština S. The effectiveness of nab-paclitaxel plus gemcitabine and gemcitabine monotherapy in first-line metastatic pancreatic cancer treatment: A real-world evidence. Medicine (Baltimore) 2022; 101:e30566. [PMID: 36181099 PMCID: PMC9524920 DOI: 10.1097/md.0000000000030566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Pancreatic cancer is one of the most lethal malignancies with a rise in mortality rates. FOLFIRINOX and nab-paclitaxel plus gemcitabine demonstrated a survival benefit compared to gemcitabine alone. Both protocols are now considered the standard of first-line treatment with no significant difference between them, primarily based on observational studies. Although new therapeutic options have emerged recently, the prognosis remains poor. We conducted a retrospective single-center study on 139 patients treated for metastatic pancreatic adenocarcinoma (mPDAC) with gemcitabine monotherapy (Gem) or nab-paclitaxel + gemcitabine (Nab-P/Gem) in the first line. The aim of our study was to evaluate the effectiveness in terms of overall survival (OS) and progression-free survival (PFS) as well as the influence of patient and disease characteristics on outcomes. Nab-P/Gem resulted in OS of 13.87 months compared to 8.5 months in patients receiving Gem. The same trend was achieved in PFS, 5.37 versus 2.80 months, respectively, but without reaching statistical significance. Furthermore, the 6-month survival in the Nab-P/Gem group was also higher, 78.1% versus 47.8%. In terms of survival, the group of elderly patients, patients of poorer performance, with higher metastatic burden and liver involvement, benefited the most from combination therapy. In our analysis ECOG performance status (p.s.), previous primary tumor surgery, and liver involvement were found to be independent prognostic factors. The addition of nab-paclitaxel to gemcitabine resulted in a significant improvement in the OS of patients with mPDAC. Subgroup analysis demonstrated that patients with some unfavorable prognostic factors benefited the most.
Collapse
Affiliation(s)
- Juraj Prejac
- University Hospital Centre Zagreb, Department of Oncology, Zagreb, Croatia
- University of Zagreb, School of Dental Medicine, Zagreb, Croatia
| | - Dora Tomek Hamzić
- University Hospital Centre Zagreb, Department of Oncology, Zagreb, Croatia
| | - Nikša Librenjak
- University Hospital Centre Zagreb, Department of Oncology, Zagreb, Croatia
| | - Irma Goršić
- University Hospital Centre Zagreb, Department of Oncology, Zagreb, Croatia
| | - Domina Kekez
- University Hospital Centre Zagreb, Department of Oncology, Zagreb, Croatia
- University of Zagreb, School of Dental Medicine, Zagreb, Croatia
| | - Stjepko Pleština
- University Hospital Centre Zagreb, Department of Oncology, Zagreb, Croatia
- University of Zagreb, School of Medicine, Zagreb, Croatia
| |
Collapse
|
21
|
Shen F, Liu C, Zhang W, He S, Wang F, Wang J, Li Q, Zhou F. Serum levels of IL-6 and CRP can predict the efficacy of mFOLFIRINOX in patients with advanced pancreatic cancer. Front Oncol 2022; 12:964115. [PMID: 35965580 PMCID: PMC9372918 DOI: 10.3389/fonc.2022.964115] [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] [Received: 06/08/2022] [Accepted: 07/05/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives There is an urgent need for biomarkers that predict the survival outcome of patients diagnosed with metastatic pancreatic cancer, undergoing systemic chemotherapy. This study aimed to identify biomarkers associated with the survival of mPC patients treated with modified FOLFIRINOX (mFOLFIRINOX) as first-line chemotherapy. Methods This was a retrospective study of 30 patients with mPC who received mFOLFIRINOX between October 2018 and March 2021. Data on carcinoembryonic antigen (CEA), cancer antigen (CA)199, interleukin (IL)-6, C-reactive protein (CRP), neutrophils, platelets, lymphocytes, and albumin were collected and dichotomized using the upper or lower limit, as appropriate. These markers were examined for their association with progression-free survival (PFS). A receiver operating characteristic (ROC) curve analysis was used to explore a suitable model to predict mFOLFIRINOX effectiveness. Results IL-6 and CRP levels were associated with poor progression (P = 0.004 and P = <0.001, respectively) of mPC. The high IL-6 level was an independent poor prognostic factor for PFS (HR=4.66, 95%CI: 1.32-16.37, P=0.016) in the multivariable analysis. Patients with high IL-6 levels had a shorter PFS than those with low IL-6 levels (median PFS: 257 vs. 150 days, P=0.020). An increase in IL-6 and CRP levels during chemotherapy positively correlated with disease progression (P = <0.001 for both). The model combining IL-6 with CRP levels helped predict the outcomes of mPC patients treated with mFOLFIRINOX (AUC: 0.811, 95%CI: 0.639-0.983, P=0.003). Conclusions The serum levels of IL-6 and CRP might be considered as valuable biomarkers in predicting the outcomes of patients with mPC who received the mFOLFIRINOX regimen.
Collapse
Affiliation(s)
- Feifei Shen
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chuan Liu
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weiguo Zhang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Sijia He
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fan Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingjue Wang
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Li
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fei Zhou
- Department of Oncology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- *Correspondence: Fei Zhou,
| |
Collapse
|
22
|
Voisin T, Nicole P, Gratio V, Chassac A, Mansour D, Rebours V, Couvelard A, Couvineau A. The Orexin-A/OX1R System Induces Cell Death in Pancreatic Cancer Cells Resistant to Gemcitabine and Nab-Paclitaxel Treatment. Front Oncol 2022; 12:904327. [PMID: 35747788 PMCID: PMC9209740 DOI: 10.3389/fonc.2022.904327] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/13/2022] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents the fourth cause of cancer-associated death in the West. This type of cancer has a very poor prognosis notably due to the development of chemoresistance when treatments including gemcitabine and Abraxane (Nab-paclitaxel) were prescribed. The identification of new treatment circumventing this chemoresistance represents a key challenge. Previous studies demonstrated that the activation of orexin receptor type 1 (OX1R), which was ectopically expressed in PDAC, by its natural ligand named orexin-A (OxA), led to anti-tumoral effect resulting in the activation of mitochondrial pro-apoptotic mechanism. Here, we demonstrated that OxA inhibited the pancreatic cancer cell (AsPC-1) growth and inhibited the tumor volume in preclinical models as effectively as gemcitabine and Nab-paclitaxel. Moreover, the combination therapy including OxA plus gemcitabine or OxA plus Nab-paclitaxel was additive on the inhibition of cancer cell growth and tumor development. More importantly, the treatment by OxA of chemoresistant tumors to gemcitabine or Nab-paclitaxel obtained by successive xenografts in mice revealed that OxA was able to induce a strong inhibition of tumor development, whereas no OxA resistance was identified in tumors. The OX1R/OxA system might be an innovative and powerful alternative treatment of chemoresistant PDAC.
Collapse
Affiliation(s)
- Thierry Voisin
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Team “From inflammation to cancer in digestive diseases” labeled by “la Ligue Nationale Contre le Cancer”, DHU UNITY, Paris, France
| | - Pascal Nicole
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Team “From inflammation to cancer in digestive diseases” labeled by “la Ligue Nationale Contre le Cancer”, DHU UNITY, Paris, France
| | - Valérie Gratio
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Team “From inflammation to cancer in digestive diseases” labeled by “la Ligue Nationale Contre le Cancer”, DHU UNITY, Paris, France
| | - Anaïs Chassac
- Department of Pathology, Bichat Hospital, Université Paris Cité, Paris, France
| | - Dounia Mansour
- Department of Pathology, Bichat Hospital, Université Paris Cité, Paris, France
| | - Vinciane Rebours
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Team “From inflammation to cancer in digestive diseases” labeled by “la Ligue Nationale Contre le Cancer”, DHU UNITY, Paris, France
- Department of Pancreatology, Beaujon Hospital, Université Paris Cité, Clichy, France
| | - Anne Couvelard
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Team “From inflammation to cancer in digestive diseases” labeled by “la Ligue Nationale Contre le Cancer”, DHU UNITY, Paris, France
- Department of Pathology, Bichat Hospital, Université Paris Cité, Paris, France
| | - Alain Couvineau
- INSERM UMR1149/Inflammation Research Center (CRI), Université Paris Cité, Team “From inflammation to cancer in digestive diseases” labeled by “la Ligue Nationale Contre le Cancer”, DHU UNITY, Paris, France
| |
Collapse
|
23
|
Identification of DNA methylation-driven genes and construction of a nomogram to predict overall survival in pancreatic cancer. BMC Genomics 2021; 22:791. [PMID: 34732125 PMCID: PMC8567715 DOI: 10.1186/s12864-021-08097-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
Background The incidence and mortality of pancreatic cancer (PC) has gradually increased. The aim of this study was to identify survival-related DNA methylation (DNAm)-driven genes and establish a nomogram to predict outcomes in patients with PC. Methods The gene expression, DNA methylation database, and PC clinical samples were downloaded from TCGA. DNAm-driven genes were identified by integrating analyses of gene expression and DNA methylation data. Survival-related DNAm-driven genes were screened via univariate, least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analyses to develop a risk score model for prognosis. Based on analyses of clinical parameters and risk score, a nomogram was built and validated. The independent cohort from GEO database were used for external validation. Results A total of 16 differentially expressed methylation-driven genes were identified. Based on LASSO Cox regression and multivariate Cox regression analysis, six genes (FERMT1, LIPH, LAMA3, PPP1R14D, NQO1, VSIG2) were chosen to develop the risk score model. In the Kaplan–Meier analysis, age, T stage, N stage, AJCC stage, radiation therapy history, tumor size, surgery type performed, pathological type, chemotherapy history, and risk score were potential prognostic factors in PC (P < 0.1). In the multivariate analysis, stage, chemotherapy, and risk score were significantly correlated to overall survival (P < 0.05). The nomogram was constructed with the three variables (stage, chemotherapy, and risk score) for predicting the 1-year, 2-year, and 3-year survival rates of PC patients. Nomogram performance was assessed by receiver operating characteristic (ROC) curves and calibration curves. 1-year, 2-year and 3-year AUC of nomogram model was 0.899, 0.765 and 0.776, respectively. Conclusions In our study, we successfully identified the six DNAm-driven genes (FERMT1, LIPH, LAMA3, PPP1R14D, NQO1, VSIG2) with a relationship to the outcomes of PC patients. The nomogram including stage, chemotherapy, and risk score could be used to predict survival in PC patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08097-w.
Collapse
|
24
|
Yang WW, Yang L, Lu HZ, Sun YK. Long-term survival of a patient with pancreatic cancer and lung metastasis: A case report and review of literature. World J Clin Cases 2021; 9:9134-9143. [PMID: 34786397 PMCID: PMC8567503 DOI: 10.12998/wjcc.v9.i30.9134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/13/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Pancreatic cancer (PC) is a leading cause of cancer-related death, given its poor prognosis and the limited benefits of traditional therapies. As tumors become more genetically disorganized as they progress, genetic mutations might become new markers for us to predict their behavior. Nowadays, many inhibitors can selectively target gene products as a form of targeted therapy, with some showing promise as treatment for various types of cancer.
CASE SUMMARY We describe a rare case of a PC patient with long-term survival of more than 8 yr. The patient was diagnosed with pancreatic ductal adenocarcinoma (PDAC) with BAP1 and PIK3CA gene mutations and Raf1 fusion and achieved partial response twice after treatment with apatinib in combination with chemotherapy.
CONCLUSION BAP1, PIK3CA mutations, and Raf1 fusion are rare in PDAC. Patients with these three gene alterations of PDAC may achieve long-term survival with apatinib. Further research in other contexts is needed to determine whether apatinib has ideal efficacy for PC treatment.
Collapse
Affiliation(s)
- Wen-Wei Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lin Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Hai-Zhen Lu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yong-Kun Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
25
|
Manrai M, Tilak TVSVGK, Dawra S, Srivastava S, Singh A. Current and emerging therapeutic strategies in pancreatic cancer: Challenges and opportunities. World J Gastroenterol 2021; 27:6572-6589. [PMID: 34754153 PMCID: PMC8554408 DOI: 10.3748/wjg.v27.i39.6572] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/09/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic carcinoma (PC) is one of the leading causes of cancer-related deaths worldwide. Despite early detection and advances in therapeutics, the prognosis remains dismal. The outcome and therapeutic approach are dependent on the stage of PC at the time of diagnosis. The standard of care is surgery, followed by adjuvant chemotherapy. The advent of newer drugs has changed the landscape of adjuvant therapy. Moreover, recent trials have highlighted the role of neoadjuvant therapy and chemoradiotherapy for resectable and borderline resectable PC. As we progress towards a better understanding of tumor biology, genetics, and microenvironment, novel therapeutic strategies and targeted agents are now on the horizon. We have described the current and emerging therapeutic strategies in PC.
Collapse
Affiliation(s)
- Manish Manrai
- Department of Internal Medicine, Armed Forces Medical College, Pune 411040, Maharashtra, India
| | - T V S V G K Tilak
- Department of Internal Medicine, Armed Forces Medical College, Pune 411040, Maharashtra, India
| | - Saurabh Dawra
- Department of Internal Medicine, Command Hospital, Pune 411040, Maharashtra, India
| | - Sharad Srivastava
- Department of Internal Medicine, Command Hospital, Pune 411040, Maharashtra, India
| | - Anupam Singh
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh 160011, India
| |
Collapse
|
26
|
Shin K, Jung EK, Park SJ, Jeong S, Kim IH, Lee MA. Neutrophil-to-lymphocyte ratio and carbohydrate antigen 19-9 as prognostic markers for advanced pancreatic cancer patients receiving first-line chemotherapy. World J Gastrointest Oncol 2021; 13:915-928. [PMID: 34457195 PMCID: PMC8371515 DOI: 10.4251/wjgo.v13.i8.915] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/03/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND A decline in serum carbohydrate antigen 19-9 (CA19-9) levels during systemic chemotherapy is considered as a prognostic marker for patients with advanced pancreatic cancer. Neutrophil-to-lymphocyte ratio (NLR) has been extensively studied as a simple and useful indicator of prognosis in various cancers including pancreatic cancer. AIM To assess the prognostic significance of NLR and CA19-9 in patients with advanced pancreatic adenocarcinoma received first-line chemotherapy according to CA19-9 positivity. METHODS We retrospectively analyzed patients diagnosed with advanced pancreatic cancer who received first-line chemotherapy between January 2010 and July 2017 at the Catholic University of Seoul St. Mary's Hospital. Patients were divided according to CA19-9 positivity (CA19-9-positive vs -negative groups) and pre-and post-treatment NLR levels. To determine cut-off value of NLR and CA19-9 reduction, time-dependent receiver operating characteristic curve was applied. We evaluated overall survival (OS) and progression-free survival (PFS) for each group using Kaplan-Meier method, and we performed multivariate analyses on the entire cohort. RESULTS We included 271 patients in this study. Cut-off value of NLR and CA19-9 reduction was determined as 2.62 and 18%. Multivariate analysis showed that post-treatment NLR < 2.62 and reduction of ≥ 18% of baseline CA19-9 were significantly associated with OS and PFS. Post-treatment NLR ≥ 2.62 showed hazard ratio (HR) of 2.47 [95% confidence interval (CI): 1.84-3.32, P < 0.001] and CA19-9 decline (≥ 18%) showed HR of 0.51 (95%CI: 0.39-0.67, P < 0.001) for OS. When CA19-9-positive patients were divided into groups according to CA19-9 response (responder vs non-responder) and post-treatment NLR (< 2.62 vs ≥ 2.62), CA19-9 responder and post-treatment NLR < 2.62 group showed better survival than CA19-9 non-responder and post-treatment NLR ≥ 2.62 group (OS 11.0 mo vs 3.9 mo, P < 0.001; PFS 6.3 mo vs 2.0 mo, P < 0.001). The combination of CA19-9 decline and post-treatment NLR showed a significant correlation with clinical response in CA 19-9 positive group. Within the CA19-9-negative group, the post-treatment NLR < 2.62 group showed better survival than the post-treatment NLR ≥ 2.62 group (OS 12.7 mo vs 7.7 mo, P < 0.001; PFS 6.7 mo vs 2.1 mo, P < 0.001), and post-treatment NLR showed correlation with clinical response. CONCLUSION In advanced pancreatic cancer patients positive for CA19-9 and treated with systemic chemotherapy, the combination of post-treatment NLR < 2.62 and 18% decline of CA19-9 at the first response evaluation is a good prognostic marker. Post-treatment NLR < 2.62 alone could be used as a prognostic marker and an adjunctive tool for response evaluation in CA19-9-negative patients.
Collapse
Affiliation(s)
- Kabsoo Shin
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Eun-Kyo Jung
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Se Jun Park
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Sangwoon Jeong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| | - Myung-ah Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
- Catholic Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, South Korea
| |
Collapse
|
27
|
Edwards P, Kang BW, Chau I. Targeting the Stroma in the Management of Pancreatic Cancer. Front Oncol 2021; 11:691185. [PMID: 34336679 PMCID: PMC8316993 DOI: 10.3389/fonc.2021.691185] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Pancreatic cancer (PC) presents extremely aggressive tumours and is associated with poor survival. This is attributed to the unique features of the tumour microenvironment (TME), which is known to create a dense stromal formation and poorly immunogenic condition. In particular, the TME of PC, including the stromal cells and extracellular matrix, plays an essential role in the progression and chemoresistance of PC. Consequently, several promising agents that target key components of the stroma have already been developed and are currently in multiple stages of clinical trials. Therefore, the authors review the latest available evidence on novel stroma-targeting approaches, highlighting the potential impact of the stroma as a key component of the TME in PC.
Collapse
Affiliation(s)
- Penelope Edwards
- Department of Medicine, Royal Marsden Hospital, London, United Kingdom
| | - Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
28
|
Baron MK, Wang X, Nevala-Plagemann C, Moser JC, Haaland B, Garrido-Laguna I. Survival Outcomes Based on Sequence of Therapy Using FOLFIRINOX and Nab-Paclitaxel + Gemcitabine in Metastatic Pancreatic Ductal Adenocarcinoma. Pancreas 2021; 50:796-802. [PMID: 34347727 DOI: 10.1097/mpa.0000000000001844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Optimal sequence of therapy for patients with metastatic pancreatic ductal adenocarcinoma is unknown. Combination chemotherapy with fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) and nab-paclitaxel + gemcitabine (nab-p/gem) are standard first-line (1L) therapies. They have never been prospectively compared. We retrospectively compared overall survival (OS) of patients treated with 1L nab-p/gem and second-line (2L) FOLFIRINOX with those treated with the reverse sequence. METHODS Patients with metastatic pancreatic ductal adenocarcinoma treated with 1L FOLFIRINOX and 2L nab-p/gem or vice versa were identified using an electronic health record-derived real-world database. Using an intent-to-treat analysis, we compared OS from initiation of 1L therapy. A Cox model, stratified by deciles of propensity score, estimated the effect of treatment sequence on OS. RESULTS The study included 3027 patients. The median OS for 1L FOLFIRINOX versus nab-p/gem was 8.6 versus 6.1 months (hazard ratio, 0.77; 95% confidence interval, 0.70-0.84). The median OS for 1L FOLFIRINOX and 2L nab-p/gem versus 1L nab-p/gem and 2L FOLFIRINOX was 11.9 versus 11.5 months (hazard ratio, 0.97; 95% confidence interval, 0.79-1.18). CONCLUSIONS In this analysis of real-world data, 1L FOLFIRINOX was associated with increased OS in propensity analysis. For patients who received both FOLFIRINOX and nab-p/gem, median OS was similar regardless of sequence.
Collapse
Affiliation(s)
| | | | | | - Justin C Moser
- Department of Oncology Clinical Trials, Honor Health Research Institute, Scottsdale, AZ
| | | | | |
Collapse
|
29
|
Colloca G. Performance status as prognostic factor in phase III trials of first-line chemotherapy of unresectable or metastatic pancreatic cancer: A trial-level meta-analysis. Asia Pac J Clin Oncol 2021; 18:232-239. [PMID: 34161667 DOI: 10.1111/ajco.13598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 03/21/2021] [Indexed: 12/25/2022]
Abstract
For patients with unresectable or metastatic pancreatic adenocarcinoma (mPDAC), there are no standardized prognostic and predictive factors beyond performance status (PS). A poor PS, as defined by Eastern Cooperative Oncology Group (ECOG) score of 2 or more, has been related with a detrimental effect of chemotherapy. Therefore, even more trials enrolled patients with good PS. The current analysis aims to evaluate the results of PS as a prognostic factor in phase III trials of patients with mPDAC receiving first-line chemotherapy. A literature search on two databases, from 2000 to 2019, and a further selection of clinical trials were performed by predefined criteria. Twelve phase III studies have been included in the analysis: the trials, enrolling 5619 patients, confirmed the worse prognosis of patients with higher ECOG PS scores (hazard ratio [HR] = 1.45; 95% confidence interval [CI], 1.21-1.74; p-value < 0.001), and a similar trend was evident for patients with an ECOG PS 1 versus 0 (HR = 1.61; 95% CI, 1.43-1.80; p-value < 0.001) in six studies, enrolling 2799 patients. Heterogeneity of trials was high, with I2 = 91%. Some possible moderators have been suggested, such as the number of drugs in the chemotherapy regimen and the male gender. In conclusion, a low ECOG PS score appears to be related with a longer survival even in trials that excluded patients with an ECOG PS 2 score, but the meta-analyses reported high heterogeneity and publication bias.
Collapse
Affiliation(s)
- Giuseppe Colloca
- Department of Oncology, Ospedale Civile di Sanremo, Sanremo, Italy
| |
Collapse
|
30
|
Chun JW, Lee SH, Kim JS, Park N, Huh G, Cho IR, Paik WH, Ryu JK, Kim YT. Comparison between FOLFIRINOX and gemcitabine plus nab-paclitaxel including sequential treatment for metastatic pancreatic cancer: a propensity score matching approach. BMC Cancer 2021; 21:537. [PMID: 33975561 PMCID: PMC8114681 DOI: 10.1186/s12885-021-08277-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND FOLFIRINOX (FFX) and Gemcitabine plus nab-paclitaxel (GnP) have been recommended as the first-line chemotherapy for metastatic pancreatic cancer (mPC). However, the evidence is lacking comparing not only two regimens, but also sequential treatment (FFX-GnP vs. GnP-FFX). METHODS Data of 528 patients (FFX, n = 371; GnP, n = 157) with mPC were collected retrospectively. Propensity score matching was conducted to alleviate imbalance of the two groups. Overall survival (OS), progression free survival (PFS), and toxicity of patients were analyzed. RESULTS In the whole population, OS (12.5 months vs. 10.3 months, P = 0.05) and PFS (7.1 months vs. 5.8 months, P = 0.02) were longer in the FFX group before matching and after matching (OS: 11.8 months vs. 10.3 months, P = 0.02; PFS: 7.2 months vs. 5.8 months, P < 0.01). For sequential treatment, OS and PFS showed no significant difference. Interruptions of chemotherapy due to toxicities were more frequent (6.8 vs. 29.3%, P < 0.001) in the GnP group, and cessation of chemotherapy showed a significant association with mortality (z = - 1.94, P = 0.03). CONCLUSIONS FFX achieved a longer overall survival than GnP in mPC, but not in the comparison for sequential treatment. More frequent adverse events followed by treatment interruptions during GnP might lead to a poor survival outcome. Therefore, FFX would be a better first-line treatment option than GnP for mPC.
Collapse
Affiliation(s)
- Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Sang Hyub Lee
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea.
| | - Joo Seong Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Namyoung Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Gunn Huh
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - In Rae Cho
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Woo Hyun Paik
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Ji Kon Ryu
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Yong-Tae Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
31
|
McLellan P, Henriques J, Ksontini F, Doat S, Hammel P, Desrame J, Trouilloud I, Louvet C, Pietrasz D, Vernerey D, Bachet JB. Prognostic value of the early change in neutrophil-to-lymphocyte ratio in metastatic pancreatic adenocarcinoma. Clin Res Hepatol Gastroenterol 2021; 45:101541. [PMID: 33055007 DOI: 10.1016/j.clinre.2020.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 07/30/2020] [Accepted: 08/31/2020] [Indexed: 02/04/2023]
Abstract
In metastatic pancreatic adenocarcinoma, a high neutrophil-to-lymphocyte ratio (NLR) at diagnosis is a marker of poor prognosis. The prognostic role of baseline NLR and NLR change during first-line chemotherapy were determined. We conducted a retrospective study by using data from a single-center prospective cohort and a randomized open-label, multicenter, randomized trial. Two hundred and twelve patients were analyzed. Baseline NLR>5 was an independent marker of poor prognosis for overall survival (HR=2.01, 95% CI 1.33-3.05; P=0.001) and progression-free survival (PFS; HR=1.80, 95% CI 1.23-2.65; P=0.0026). According to NLR dynamics (n=172), patients with NLR≤5 on days 1 and 15 had a significantly better prognosis than those with NLR≤5 on day 1 and NLR>5 on day 15 (HR=2.23, 95% CI 1.18-4.21; P=0.013), NLR >5 on day 1 and NLR ≤5 on day 15 (HR=3.25, 95% CI 1.86-5.68; P<0.001), and NLR>5 on days 1 and 15 (HR=3.37, 95% CI 1.93-5.90; P<0.001). Over time, bad responders (PFS <6 months) had significantly higher mean NLR than good responders (PFS>6 months; group effect: P<0.0001). Seven out of eight patients with baseline NLR>5 had circulating tumor DNA. This study confirmed the independent prognostic value of baseline NLR >5 in metastatic pancreatic cancer. The change in NLR early during chemotherapy was also a prognostic indicator in patients with NLR ≤5.
Collapse
Affiliation(s)
- Paul McLellan
- Sorbonne Université, 4 Place Jussieu, 75005, Paris, France; Department of Hepato-Gastroenterology, Hôpital Pitié-Salpêtrière, 47 Boulevard de l'Hôpital 75013, APHP, Paris, France
| | - Julie Henriques
- Department of Methodology and Quality of Life Oncology, University Hospital, 3 Boulevard Alexandre Fleming, 25000 Besancon, France; Bourgogne Franche-Comté Université, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, 8 rue du Docteur Jean-François-Xavier Girod, 25020 Besançon, France
| | - Feryel Ksontini
- Department of Oncology, Institute Salah-Azaïz, Boulevard du 9 avril 1938, 1006 Tunis, Tunisia
| | - Solène Doat
- Department of Hepato-Gastroenterology, Hôpital Pitié-Salpêtrière, 47 Boulevard de l'Hôpital 75013, APHP, Paris, France
| | - Pascal Hammel
- Department of Digestive Oncology, Hôpital Beaujon, 100 Boulevard du General Leclerc, 92110 Clichy, France
| | - Jérome Desrame
- Department of Gastroenterology, Hôpital Privé Jean Mermoz, 55 avenue Jean Mermoz, 69008 Lyon, France
| | - Isabelle Trouilloud
- Department of Oncology, Hôpital Saint-Antoine, 186 rue du Faubourg Saint Antoine, 75012 Paris, France
| | - Christophe Louvet
- Department of Oncology, Institut Mutualiste Montsouris, 54 avenue Jourdan, 75014 Paris, France
| | - Daniel Pietrasz
- Department of Hepato-Pancreato-Biliary Surgery, CHB, Hôpital Paul Brousse, 12 Avenue Paul Vaillant Couturier, 94800 Villejuif, France
| | - Dewi Vernerey
- Department of Methodology and Quality of Life Oncology, University Hospital, 3 Boulevard Alexandre Fleming, 25000 Besancon, France; Bourgogne Franche-Comté Université, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, 8 rue du Docteur Jean-François-Xavier Girod, 25020 Besançon, France
| | - Jean-Baptiste Bachet
- Sorbonne Université, 4 Place Jussieu, 75005, Paris, France; Department of Hepato-Gastroenterology, Hôpital Pitié-Salpêtrière, 47 Boulevard de l'Hôpital 75013, APHP, Paris, France.
| |
Collapse
|
32
|
Lund-Andersen C, Torgunrud A, Fleten KG, Flatmark K. Omics analyses in peritoneal metastasis-utility in the management of peritoneal metastases from colorectal cancer and pseudomyxoma peritonei: a narrative review. J Gastrointest Oncol 2021; 12:S191-S203. [PMID: 33968437 DOI: 10.21037/jgo-20-136] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
High-throughput "-omics" analysis may provide a broader and deeper understanding of cancer biology to define prognostic and predictive biomarkers and identify novel therapy targets. In this review we provide an overview of studies where the peritoneal tumor component of peritoneal metastases from colorectal cancer (PM-CRC) and pseudomyxoma peritonei (PMP) were analyzed. Most of the available data was derived from DNA mutation analysis, but a brief review of findings from transcriptomic and protein expression analysis was also performed. Studies reporting genomic analysis of peritoneal tumor samples from 1,779 PM-CRC and 623 PMP cases were identified. The most frequently mutated genes in PM-CRC were KRAS, APC, SMAD4, BRAF, and PIK3CA, while in PMP KRAS, GNAS, FAT4, TGFBR1, TP53 and SMAD3/4 mutations were most commonly identified. Analyses were performed by single-gene analyses and to some extent targeted next-generation sequencing, and a very limited amount of broad explorative data exists. The investigated cohorts were typically small and heterogeneous with respect to the methods used and to the reporting of clinical data. This was even more apparent regarding transcriptomic and protein data, as the low number of cases examined and quality of clinical data would not support firm conclusions. Even for the most frequently mutated genes, the results varied greatly; for instance, KRAS mutations were reported at frequencies between 20-57% in PM-CRC and 38-100% in PMP. Such variation could be caused by random effects in small cohorts, heterogeneity in patient selection, or sensitivity of applied technology. Although a large number of samples have been subjected to analysis, cross-study comparisons are difficult to perform, and combined with small cohorts and varying quality and detail of clinical information, the observed variation precludes useful interpretation in a clinical context. Although omics data in theory could answer questions to aid management decisions in PM-CRC and PMP, the existing data does not presently support clinical implementation. With the necessary technologies being generally available, the main challenge will be to obtain sufficiently large, representative cohorts with adequate clinical data and standardized reporting of results. Importantly, studies where the focus is specifically on peritoneal disease are needed, where the study designs are aligned with clearly defined research questions to allow robust conclusions. Such studies are highly warranted if patients with PM-CRC and PMP are to derive benefit from recent advances in precision cancer medicine.
Collapse
Affiliation(s)
- Christin Lund-Andersen
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Annette Torgunrud
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Karianne Giller Fleten
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Gastroenterological Surgery, Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
33
|
Wu L, Zhu L, Xu K, Zhou S, Zhou Y, Zhang T, Hang J, Zee BCY. Clinical significance of site-specific metastases in pancreatic cancer: a study based on both clinical trial and real-world data. J Cancer 2021; 12:1715-1721. [PMID: 33613759 PMCID: PMC7890328 DOI: 10.7150/jca.50317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background: There is limited consensus on whether metastatic patterns are correlated with prognosis and treatment efficacy in pancreatic cancer. A better understanding of clinical implication of the metastatic patterns is pivotal for therapeutic decision-making and drug development. Methods: This study included 977 patients with metastatic pancreatic cancer (MPC) in three cohorts. The training cohort included 273 patients from clinical trial NCT00574275 and 367 patients from clinical trial NCT01124786. As the validation cohort, 337 patients from Changzhou No.2 People's Hospital and Shanghai General Hospital were enrolled. The correlations between different patterns of metastases and clinicopathological characteristics were investigated with the Pearson Chi-Square test. Kaplan-Meier analysis and log-rank test were applied to analyze the survival outcomes among groups with different metastatic patterns. The prognostic value of the number of metastatic sites and other variables was evaluated using the Cox regression model. Results: MPC patients aged ≥65 years had a higher rate of lung metastasis and those with liver metastasis were prone to have a high level of carbohydrate antigen 19-9 (CA19-9). Additionally, patients with isolated lung metastasis had much better overall survival (OS) than those with isolated liver or peritoneum metastasis. Cox regression analyses showed that the number of metastatic sites was an independent prognostic factor for OS in patients with MPC. Furthermore, for patients with one-site or two-site metastasis, there was a significant difference in OS among patients receiving no chemotherapy, monotherapy and combination therapy. However, for patients with more than two metastatic sites, receiving combination therapy or monotherapy showed limited superiority in OS over receiving no chemotherapy. Conclusion: MPC patients with isolated lung metastasis had better OS than those with isolated liver or peritoneum metastasis. Moreover, the number of metastatic sites showed prognostic and predictive value in patients with MPC.
Collapse
Affiliation(s)
- Lixia Wu
- Department of Oncology, Shanghai JingAn District ZhaBei Central Hospital, Shanghai 200070, China
| | - Lina Zhu
- Changzhou No. 2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Kequn Xu
- Changzhou No. 2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Siyuan Zhou
- Changzhou No. 2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Yang Zhou
- Changzhou No. 2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Tiening Zhang
- Department of Radiotherapy, Shanghai General Hospital, Shanghai 200080, China
| | - Junjie Hang
- Changzhou No. 2 People's Hospital, Affiliated Hospital of Nanjing Medical University, Changzhou 213000, China.,JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin 999077, Hong Kong, China
| | - Benny Chung-Ying Zee
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin 999077, Hong Kong, China
| |
Collapse
|
34
|
Prager GW, Oehler L, Gerger A, Mlineritsch B, Andel J, Petzer A, Wilthoner K, Sliwa T, Pichler P, Winder T, Heibl S, Gruenberger B, Laengle F, Hubmann E, Korger M, Pecherstorfer M, Djanani A, Neumann HJ, Philipp-Abbrederis K, Wöll E, Trondl R, Arnold-Schrauf C, Eisterer W. Comparison of nab-paclitaxel plus gemcitabine in elderly versus younger patients with metastatic pancreatic cancer: Analysis of a multicentre, prospective, non-interventional study. Eur J Cancer 2020; 143:101-112. [PMID: 33296830 DOI: 10.1016/j.ejca.2020.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/20/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Pancreatic cancer (PC) ranks among the deadliest malignancies worldwide. In the MPACT study, first-line nab-paclitaxel plus gemcitabine (nab-P/G) demonstrated activity (median overall survival [OS], 8.7 months) and tolerability in patients with metastatic PC (mPC). However, the clinical evidence of nab-P/G in the elderly (>70 years), who account for the majority of patients with mPC, is limited. This is the first prospective, multicentre, non-interventional study evaluating the tolerability and effectiveness of nab-P/G in younger (≤70 years) versus elderly (>70 years) patients with mPC in the daily clinical routine. METHODS Eligible patients with mPC were treated with nab-P/G and observed until disease progression or unacceptable toxicity. The primary objectives were safety and tolerability of nab-P/G, and the secondary objectives were efficacy and real-life dosing. RESULTS A total of 317 patients with mPC (median age, 70 years) were recruited, of which 299, aged ≤70 (n = 162) and >70 (n = 137) years, were eligible for analysis. Baseline characteristics and the safety profile were comparable between the groups. However, fatigue (22.8% versus 13.0%) and decreased appetite (8.8% versus 1.2%) were more frequent in elderly patients. Younger versus elderly patients equally benefited in terms of objective response rate (36% versus 48%), median progression-free survival (5.6 versus 5.5 months; hazard ratio [HR] = 1.03; p = 0.81) and OS (10.6 versus 10.2 months; HR = 0.89; p = 0.4). In addition, the median treatment duration (5 versus 4 cycles), relative dose intensity (70% versus 74%) or reasons for treatment discontinuation were similar. Most patients (56.2% versus 47.4%) benefited from a second-line therapy. CONCLUSION This prospective real-world analysis confirms the feasibility and tolerability of nab-P/G treatment and reveals OS data similar for younger patients and elderly patients aged >70 years. CLINICALTRIALS. GOV REGISTRATION NCT02555813. AUSTRIAN NIS REGISTRY NIS005071.
Collapse
Affiliation(s)
- Gerald W Prager
- Medical University of Vienna, Department of Oncology, Währinger Gürtel 18-20, 1090, Vienna, Austria.
| | - Leopold Oehler
- Sankt Josef Krankenhaus, Internal Medicine 2, Auhofstraße 189, 1130, Vienna, Wien, Austria.
| | - Armin Gerger
- Medical University of Graz, Clinical Institute of Oncology, Auenbruggerplatz 15, 8036, Graz, Austria.
| | - Brigitte Mlineritsch
- Universitätsklinik Salzburg, University Clinic for Internal Medicine III, Müllner Haupstraße 48, 5020, Salzburg, Austria.
| | - Johannes Andel
- Pyhrn-Eisenwurzen Klinikum, Internal Medicine II, Sierningerstraße 170, 4400, Steyr, Austria.
| | - Andreas Petzer
- Ordensklinikum Linz BHS - EKH, Internal Medicine I, Medical Oncology and Hematology, Seilerstätte 4, 4010, Linz Austria.
| | - Klaus Wilthoner
- Landeskrankenhaus Vöcklabruck, Vöcklabruck, Internal Medicine, Hemato-Oncology, Dr. Wilhelm-Bock-Straße 1, 4840 Vöcklabruck, Austria.
| | - Thamer Sliwa
- Hanuschkrankenhaus, Medicine III for Hematology and Oncology, Heinrich-Collin-Straße 30, 1140, Wien, Vienna, Austria.
| | - Petra Pichler
- Universitätsklinikum St. Pölten, Internal Medicine I, Dunant-Platz 1, 3100, Sankt Pölten, Austria.
| | - Thomas Winder
- Landeskrankenhaus Feldkirch, Internal Medicine II, Carinagasse 47, 6807, Feldkirch, Austria.
| | - Sonja Heibl
- Klinikum Wels-Grieskirchen, Internal Medicine IV, Grieskirchner Straße 42, 4600, Wels, Austria.
| | - Birgit Gruenberger
- Landesklinikum Wiener Neustadt, Internal Medicine for Hematology and Internal Oncology, Corvinusring 2-5, 2700, Wiener Neustadt, Austria.
| | - Friedrich Laengle
- Landesklinikum Wiener Neustadt, Department of Surgery, Corvinusring 2-5, 2700, Wiener Neustadt, Austria.
| | - Eva Hubmann
- Krankenhaus der Barmherzigen Brüder, Internal Medicine, Marschallgasse 12, 8020, Graz, Austria.
| | - Markus Korger
- Krankenhaus der Barmherzigen Brüder, Internal Medicine II, Johannes von Gott-Platz 1, 7000, Eisenstadt, Austria.
| | - Martin Pecherstorfer
- Karl Landsteiner University of Health Sciences, Department of Internal Medicine, University Hospital, 3500, Krems an der Donau, Austria.
| | - Angela Djanani
- Medical University of Innsbruck, Institute of Gastroenterology, Internal Medicine I, Institute of Gastroenterology, Anichstraße 35, 6020, Innsbruck, Austria.
| | - Hans-Joerg Neumann
- Krankenhaus der Elisabethinen, Internal Medicine, Völkermarkter Straße 15-19, 9020, Klagenfurt, Austria.
| | - Kathrin Philipp-Abbrederis
- Medical University of Innsbruck, Institute of Hematology and Oncology, Internal Medicine V, Institute of Hematology and Oncology, Anichstraße 35, 6020, Innsbruck, Austria.
| | - Ewald Wöll
- Krankenhaus Zams, Internal Medicine, Sanatoriumstraße 43, 6511, Zams, Austria.
| | - Robert Trondl
- Celgene Austria GmbH, EuroPlaza Building E, Technologiestraße 10, 1120, Vienna, Austria.
| | | | - Wolfgang Eisterer
- Klinikum Klagenfurt Am Wörthersee, Internal Medicine and Oncology, Feschnigstraße 11, 9020, Klagenfurt, Austria.
| |
Collapse
|
35
|
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: 6.8] [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
|
36
|
Rotolo S, Ferracci F, Santullo F, Lodoli C, Inzani F, Abatini C, Pacelli F, Di Giorgio A. Systemic chemotherapy and pressurized intraperitoneal aerosol chemotherapy (PIPAC): A case report of a multimodal treatment for peritoneal metastases of pancreatic origin. Int J Surg Case Rep 2020; 77S:S75-S78. [PMID: 33191190 PMCID: PMC7876684 DOI: 10.1016/j.ijscr.2020.10.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
Integration of PIPAC to systemic chemotherapy can have good tolerance profile. Repeated biopsies performed during PIPAC allowed therapy-response monitoring. Analysis of biopsies performed during PIPAC allowed tailoring of chemotherapy. Integration of PIPAC to systemic chemotherapy can permit good quality of life.
Introduction Pancreatic ductal adenocarcinoma (PDAC) with peritoneal metastases (PM) has a dismal prognosis and palliative systemic chemotherapy, which represents the standard treatment option, has significant pharmacokinetics limitations and low efficacy. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a new method of drug delivery that is expected to maximize exposure of peritoneal nodules to antiblastic agents. A combination of systemic chemotherapy and PIPAC may be valuable. Presentation of case A 55 years old male affected by PDAC with synchronous PM underwent a multimodal treatment comprising systemic chemotherapy and PIPAC without any procedural-related adverse events. Tumor genomic profiling evaluation from peritoneal biopsies addressed further tailored systemic chemotherapy. Discussion The presented case illustrates the possibility of adding PIPAC to systemic chemotherapy with a fair tolerance profile and good quality of life while allowing monitoring of therapy-response and tailoring of the antiblastic treatment.
Collapse
Affiliation(s)
- Stefano Rotolo
- Department of Surgical, Oncological and Oral Sciences, University of Palermo, Palermo, Italy
| | - Federica Ferracci
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | - Francesco Santullo
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudio Lodoli
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Frediano Inzani
- Institute of Pathology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Carlo Abatini
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabio Pacelli
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Andrea Di Giorgio
- Surgical Unit of Peritoneum and Retroperitoneum, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
37
|
Blomstrand H, Green H, Fredrikson M, Gränsmark E, Björnsson B, Elander NO. Clinical characteristics and blood/serum bound prognostic biomarkers in advanced pancreatic cancer treated with gemcitabine and nab-paclitaxel. BMC Cancer 2020; 20:950. [PMID: 33008332 PMCID: PMC7530950 DOI: 10.1186/s12885-020-07426-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 09/16/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND In recent years treatment options for advanced pancreatic cancer have markedly improved, and a combination regimen of gemcitabine and nab-paclitaxel is now considered standard of care in Sweden and elsewhere. Nevertheless, a majority of patients do not respond to treatment. In order to guide the individual patient to the most beneficial therapeutic strategy, simple and easily available prognostic and predictive markers are needed. METHODS The potential prognostic value of a range of blood/serum parameters, patient-, and tumour characteristics was explored in a retrospective cohort of 75 patients treated with gemcitabine/nab-paclitaxel (Gem/NabP) for advanced pancreatic ductal adenocarcinoma (PDAC) in the South Eastern Region of Sweden. Primary outcome was overall survival (OS) while progression free survival (PFS) was the key secondary outcome. RESULT Univariable Cox regression analysis revealed that high baseline serum albumin (> 37 g/L) and older age (> 65) were positive prognostic markers for OS, and in multivariable regression analysis both parameters were confirmed to be independent prognostic variables (HR 0.48, p = 0.023 and HR = 0.47, p = 0.039,). Thrombocytopenia at any time during the treatment was an independent predictor for improved progression free survival (PFS) but not for OS (HR 0.49, p = 0.029, 0.54, p = 0.073), whereas thrombocytopenia developed under cycle 1 was neither related with OS nor PFS (HR 0.87, p = 0.384, HR 1.04, p = 0.771). Other parameters assessed (gender, tumour stage, ECOG performance status, myelosuppression, baseline serum CA19-9, and baseline serum bilirubin levels) were not significantly associated with survival. CONCLUSION Serum albumin at baseline is a prognostic factor with palliative Gem/NabP in advanced PDAC, and should be further assessed as a tool for risk stratification. Older age was associated with improved survival, which encourages further studies on the use of Gem/NabP in the elderly.
Collapse
Affiliation(s)
- Hakon Blomstrand
- Department of Clinical Pathology and Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden
| | - Henrik Green
- Division of Drug Research, Department of Medical Health Sciences, Linköping University, 58183, Linköping, Sweden.,Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, 58758, Linköping, Sweden
| | - Mats Fredrikson
- Forum Östergötland, Linköping University, 58185, Linköping, Sweden
| | - Emma Gränsmark
- Department of Oncology, Kalmar County Hospital, 392 44, Kalmar, Sweden
| | - Bergthor Björnsson
- Department of Surgery and Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden
| | - Nils O Elander
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden.
| |
Collapse
|
38
|
Iede K, Yamada T, Kato R, Ueda M, Tsuda Y, Nakashima S, Ohta K, Matsuyama J, Ikenaga M, Tominaga S. Predictive implications of decreased CA19-9 at 8 weeks during nab-paclitaxel plus gemcitabine for the induction of second-line chemotherapy for patients with advanced pancreatic cancer. Cancer Rep (Hoboken) 2020; 3:e1289. [PMID: 32969199 PMCID: PMC7941508 DOI: 10.1002/cnr2.1289] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/27/2022] Open
Abstract
Background Second‐line (2L) chemotherapy after nab‐paclitaxel plus gemcitabine (AG) is important for improving the survival of patients with advanced pancreatic cancer (APC). However, many patients fail to receive 2L chemotherapy because of rapid disease progression. Therefore, early recognition of any ineffectiveness during AG might lead to an increased induction rate of 2L chemotherapy. Aim We investigated the significance of treatment response at 8 weeks as a predictive factor for the induction of 2L chemotherapy after AG. Methods and results From January 2015 to January 2019, 41 patients with APC underwent AG as first‐line chemotherapy at our institute. Thirty‐three patients were evaluated at 8 weeks. Sixteen patients (48%) underwent 2L chemotherapy and 17 (52%) underwent no 2L chemotherapy. Clinical features and treatment response at 8 weeks were, retrospectively, compared among patients. Predictive factors for the induction of 2L chemotherapy were analyzed. Patients with an objective response by 8 weeks received 2L chemotherapy more frequently (P = .026). Decreased CA19‐9 (<50%) at 8 weeks was identified as an independent negative predictive factor for the induction of 2L chemotherapy. Conclusions Decreased CA19‐9 (<50%) at 8 weeks may indicate the ineffectiveness of AG and signify that a move to 2L chemotherapy may be required without delay.
Collapse
Affiliation(s)
- Kiyotsugu Iede
- Department of Clinical Oncology, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Terumasa Yamada
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Ryo Kato
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Masami Ueda
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Yujiro Tsuda
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Shinsuke Nakashima
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Katsuya Ohta
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Jin Matsuyama
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Masakazu Ikenaga
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Shusei Tominaga
- Department of Clinical Oncology, Higashiosaka City Medical Center, Higashiosaka, Japan
| |
Collapse
|
39
|
Abstract
Pancreatic cancer is the third leading cause of cancer death in the USA, and pancreatic ductal adenocarcinoma (PDA) constitutes 85% of pancreatic cancer diagnoses. PDA frequently metastasizes to the peritoneum, but effective treatment of peritoneal metastasis remains a clinical challenge. Despite this unmet need, understanding of the biological mechanisms that contribute to development and progression of PDA peritoneal metastasis is sparse. By contrast, a vast number of studies have investigated mechanisms of peritoneal metastasis in ovarian and gastric cancers. Here, we contrast similarities and differences between peritoneal metastasis in PDA as compared with those in gastric and ovarian cancer by outlining molecular mediators involved in each step of the peritoneal metastasis cascade. This review aims to provide mechanistic insights that could be translated into effective targeted therapies for patients with peritoneal metastasis from PDA.
Collapse
|
40
|
Multiple Liver Metastases Rather Than the Presence of Liver Metastasis Is a Significantly Poor Prognostic Factor for Patients With Advanced Pancreatic Cancer. Pancreas 2020; 49:e63-e65. [PMID: 32675786 DOI: 10.1097/mpa.0000000000001596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
41
|
Di Giorgio A, Sgarbura O, Rotolo S, Schena CA, Bagalà C, Inzani F, Russo A, Chiantera V, Pacelli F. Pressurized intraperitoneal aerosol chemotherapy with cisplatin and doxorubicin or oxaliplatin for peritoneal metastasis from pancreatic adenocarcinoma and cholangiocarcinoma. Ther Adv Med Oncol 2020; 12:1758835920940887. [PMID: 32782488 PMCID: PMC7383654 DOI: 10.1177/1758835920940887] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/15/2020] [Indexed: 02/06/2023] Open
Abstract
Background Systemic chemotherapy for pancreatic adenocarcinoma (PDAC) and cholangiocarcinoma (CC) with peritoneal metastases (PM) is affected by several pharmacological shortcomings and low clinical efficacy. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is expected to maximize exposure of peritoneal nodules to antiblastic agents. This study aims to evaluate safety and efficacy of PIPAC for PM of PDAC and CC origin. Methods This is a retrospective analysis of consecutive PDAC and CC cases with PM treated with PIPAC at two European referral centers for peritoneal disease. We prospectively recorded from August 2016 to May 2019 demographic, clinical, surgical, and oncological data. We performed a feasibility and safety assessment and an efficacy analysis based on clinical and pathological regression. Results Twenty patients with PM from PDAC (14) and CC (six) underwent 45 PIPAC administrations. Cisplatin-doxorubicin or oxaliplatin were administered to eight and 12 patients, respectively. We experienced one intraoperative complication (small bowel perforation) and 18 grade 1-2 postoperative adverse events according to Common Terminology Criteria for Adverse Events version 4.0. A pathological regression was recorded in 50% of patients (62% in the cisplatin-doxorubicin cohort and 42% in the oxaliplatin one). Median survival from the first PIPAC was 9.7 and 10.9 months for PDAC and CC, respectively. Conclusion PIPAC resulted feasible and safe without relevant toxicity issues, with both cisplatin-doxorubicin and oxaliplatin. The pathological response observed supports the evidence of antitumoral activity. Despite the study limitations, these outcomes are encouraging, recommending PIPAC in prospective, controlled trials in the palliative setting or the first line chemotherapy for PM from PDAC and CC.
Collapse
Affiliation(s)
- Andrea Di Giorgio
- Foundation Policlinico Universitario A. Gemelli - IRCCS, Peritoneum and Retroperitoneum Surgery, Roma, Lazio, Italy
| | - Olivia Sgarbura
- Department of Surgical Oncology, Montpellier Cancer Institute, Montpellier, Languedoc-Roussillon, France
| | - Stefano Rotolo
- Department of Surgical, Oncological and Oral Sciences (Di.Chir.On.S.), University of Palermo, Via del Vespro, 129, Palermo, 90127, Sicilia, Italy
| | - Carlo Alberto Schena
- Foundation Policlinico Universitario A. Gemelli - IRCCS, General Surgery Unit, Roma, Lazio, Italy
| | - Cinzia Bagalà
- Foundation Policlinico Universitario A. Gemelli - IRCCS, Division of Medical Oncology, Roma, Lazio, Italy
| | - Frediano Inzani
- Foundation Policlinico Universitario A. Gemelli - IRCCS, Anatomic Pathology Unit, Roma, Lazio, Italy
| | - Andrea Russo
- Foundation Policlinico Universitario A. Gemelli - IRCCS, Institute of Intensive Care Medicine and Anesthesiology, Roma, Lazio, Italy
| | - Vito Chiantera
- Division of Gynecologic Oncology, University of Palermo, Palermo, Sicilia, Italy
| | - Fabio Pacelli
- Foundation Policlinico Universitario A. Gemelli - IRCCS, Peritoneum and Retroperitoneum Surgery, Roma, Lazio, Italy
| |
Collapse
|
42
|
Iede K, Yamada T, Kato R, Ueda M, Tsuda Y, Nakashima S, Ohta K, Matsuyama J, Ikenaga M, Tominaga S. Efficacy of S-1 in second-line chemotherapy after nab-paclitaxel plus gemcitabine for patients with advanced pancreatic cancer. Cancer Rep (Hoboken) 2020; 3:e1215. [PMID: 32672000 DOI: 10.1002/cnr2.1215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/31/2019] [Accepted: 08/08/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Second-line (2 L) chemotherapy is important for improved survival. However, the efficacy of S-1 after nab-paclitaxel plus gemcitabine (AG) for advanced pancreatic cancer (APC) remains unclear. AIM We retrospectively investigated the clinical impact of S-1 after AG. METHODS AND RESULTS From January 2015 to July 2018, 37 patients with APC underwent AG as first-line chemotherapy at our institute. Of these patients, 14 (38%) underwent S-1 as 2 L chemotherapy after AG (S-1 group), five (14%) received another agent after AG, and 18 (49%) underwent no 2 L chemotherapy (best supportive care [BSC] group). The clinical features were retrospectively compared between the S-1 and BSC groups. Prognostic factors for residual survival (RS) were analyzed using a Cox proportional hazards model. The induction rate of 2 L chemotherapy was 51%, and most patients received S-1 monotherapy (74%). The disease control rate and progression-free survival duration were 57.1% and 2.8 months, respectively. The median RS duration in the S-1 and BSC groups was 5.2 and 2.4 months, respectively; this difference was statistically significant (hazard ratio, 0.33; P = .005). The median overall survival duration in the S-1 and BSC groups was 12.3 and 5.0 months, respectively; this difference was also statistically significant (hazard ratio, 0.26; P = .001). The efficacy of S-1 in 2L chemotherapy for RS was identified in the multivariate analysis, as was age (<65 vs ≥65 y) and the presence of liver metastasis. CONCLUSION The antitumor activity of S-1 was retained after AG, and the induction of S-1 after AG might improve the prognosis of patients with APC.
Collapse
Affiliation(s)
- Kiyotsugu Iede
- Department of Clinical Oncology, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Terumasa Yamada
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Ryo Kato
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Masami Ueda
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Yujiro Tsuda
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Shinsuke Nakashima
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Katsuya Ohta
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Jin Matsuyama
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Masakazu Ikenaga
- Department of Gastroenterological Surgery, Higashiosaka City Medical Center, Higashiosaka, Japan
| | - Shusei Tominaga
- Department of Clinical Oncology, Higashiosaka City Medical Center, Higashiosaka, Japan
| |
Collapse
|
43
|
Abstract
OBJECTIVES This analysis investigated nomogram use to evaluate metastatic pancreatic cancer prognosis. METHODS Thirty-four baseline factors were examined in the Metastatic Pancreatic Adenocarcinoma Clinical Trial (MPACT) (nab-paclitaxel plus gemcitabine vs gemcitabine) data set. Factors significantly (P < 0.1) associated with overall survival (OS) in a univariable model or with known clinical relevance were tested further. In a multivariable model, factors associated with OS (P < 0.1) were selected to generate the primary nomogram, which was internally validated using bootstrapping, a concordance index, and calibration plots. RESULTS Using data from 861 patients, 6 factors were retained (multivariable analysis): neutrophil-lymphocyte ratio, albumin level, Karnofsky performance status, sum of longest diameter of target lesions, presence of liver metastases, and previous Whipple procedure. The nomogram distinguished low-, medium-, and high-risk groups (concordance index, 0.67; 95% confidence interval, 0.65-0.69; median OS, 11.7, 8.0, and 3.3 months, respectively). CONCLUSIONS This nomogram may guide estimates of the range of OS outcomes and contribute to patient stratification in future prospective metastatic pancreatic cancer trials; however, external validation is required to improve estimate reliability and applicability to a general patient population. Caution should be exercised in interpreting these results for treatment decisions: patient characteristics could differ from those included in the nomogram development.
Collapse
|
44
|
Meijer LL, Garajová I, Caparello C, Le Large TYS, Frampton AE, Vasile E, Funel N, Kazemier G, Giovannetti E. Plasma miR-181a-5p Downregulation Predicts Response and Improved Survival After FOLFIRINOX in Pancreatic Ductal Adenocarcinoma. Ann Surg 2020; 271:1137-1147. [PMID: 30394883 DOI: 10.1097/sla.0000000000003084] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE The aim of the study was to identify plasma microRNA (miRNA) biomarkers for stratifying and monitoring patients with locally advanced or metastatic pancreatic ductal adenocarcinoma (PDAC) treated with FOLFIRINOX, and to investigate their functional roles. SUMMARY BACKGROUND DATA FOLFIRINOX has become a standard therapy for patients with advanced PDAC and can be used to potentially downstage disease. However, only a subset of patients respond, and biomarkers to guide decision-making are urgently needed. METHODS We used microarray-based profiling to discover deregulated miRNAs in pre- and postchemotherapy plasma samples from patients based on their progression-free survival (PFS) after FOLFIRINOX. Nine candidate plasma miRNAs were validated in an independent cohort (n = 43). The most discriminative plasma miRNA was correlated with clinicopathological factors and survival, and also investigated in an additional cohort treated with gemcitabine plus nab-paclitaxel. Expression patterns were further evaluated in matched tumor tissues. In vitro studies explored its function, key downstream gene-targets, and interaction with 5-fluorouracil, irinotecan, and oxaliplatin. RESULTS Plasma miR-181a-5p was significantly downregulated in non-progressive patients after FOLFIRINOX. In multivariate analysis, this decline correlated with improved PFS and overall survival, especially when combined with CA19-9 decline [hazard ratio (HR) = 0.153, 95% confidence interval (CI), 0.067-0.347 and HR = 0.201, 95% CI, 0.070-0.576, respectively]. This combination did not correlate with survival in patients treated with gemcitabine plus nab-paclitaxel. Tissue expression of miR-181a-5p reflected plasma levels. Inhibition of miR-181a-5p coupled with oxaliplatin exposure in pancreatic cell lines decreased cell viability. CONCLUSIONS Plasma miR-181a-5p is a specific biomarker for monitoring FOLFIRINOX response. Decline in plasma miR-181a-5p and CA19-9 levels is associated with better prognosis after FOLFIRINOX and may be useful for guiding therapeutic choices and surgical exploration.
Collapse
Affiliation(s)
- Laura L Meijer
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, the Netherlands
| | - Ingrid Garajová
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Amsterdam, The Netherlands
- Department of Medical Oncology, University Hospital S. Orsola-Malpighi, Bologna, Italy
| | - Chiara Caparello
- Department of Medical Oncology, University Hospital of Pisa, Pisa, Italy
| | - Tessa Y S Le Large
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, the Netherlands
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Amsterdam, The Netherlands
- Laboratory of Experimental Oncology & Radiobiology, Academic Medical Center, Amsterdam, The Netherlands
| | - Adam E Frampton
- HPB Surgical Unit, Department of Surgery & Cancer, Imperial College, London, United Kingdom
| | - Enrico Vasile
- Department of Medical Oncology, University Hospital of Pisa, Pisa, Italy
| | - Niccola Funel
- Cancer Pharmacology Lab, AIRC-Start-Up Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University Hospital of Pisa, Pisa, Italy
| | - Geert Kazemier
- Department of Surgery, Cancer Center Amsterdam, Amsterdam UMC, VU University Amsterdam, the Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, VU University Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC-Start-Up Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
45
|
Sawada M, Kasuga A, Mie T, Furukawa T, Taniguchi T, Fukuda K, Yamada Y, Takeda T, Kanata R, Matsuyama M, Sasaki T, Ozaka M, Sasahira N. Modified FOLFIRINOX as a second-line therapy following gemcitabine plus nab-paclitaxel therapy in metastatic pancreatic cancer. BMC Cancer 2020; 20:449. [PMID: 32434547 PMCID: PMC7238500 DOI: 10.1186/s12885-020-06945-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022] Open
Abstract
Background There is no established second-line treatment after failure of gemcitabine plus nab-paclitaxel (GnP) therapy for metastatic pancreatic cancer (MPC). The purpose of this study was to evaluate the efficacy and tolerability of the modified FOLFIRINOX (mFFX) as a second-line therapy for MPC and to investigate prognostic factors for survival. Methods From 2015 to 2019, we retrospectively reviewed the medical records of consecutive patients receiving mFFX for MPC after failure of GnP therapy. Patients were treated every 2 weeks with mFFX (intravenous oxaliplatin 85 mg/m2, intravenous irinotecan 150 mg/m2, and continuous infusion of 5-fluorouracil 2400 mg/m2 for 46 h without bolus infusion). Results In total, 104 patients received mFFX. The median overall survival (OS) was 7.0 months (95% confidence interval [CI]: 6.2–9.8) and the progression-free survival (PFS) 3.9 months (95% CI 2.8–5.0). The objective response rate was 10.6% and the disease control rate 56.7%. The median relative dose intensities of oxaliplatin, irinotecan, and infusional 5-FU were 80.0% (range 21.5–100%), 77.2% (range 38.1–100%), and 85.9% (range 36.9–100%), respectively. Grade 3–4 toxicities were reported in 57 patients (54.8%), including neutropenia, leukopenia, anemia, febrile neutropenia, and peripheral sensory neuropathy. Glasgow prognostic score and carcinoembryonic antigen level were independently associated with survival. Our prognostic model using these parameters could classify the patients into good (n = 38), intermediate (n = 47), and poor (n = 19) prognostic groups. The median OS and PFS time was 14.7 (95% CI 7.6–16.3) and 7.6 months (95% CI 4.1–10.5) for the good prognostic factors, 6.5 (95% CI 5.5–10.0) and 3.6 months (95% CI 2.7–4.8) for the intermediate prognostic factors and 5.0 (95% CI 2.9–6.6) and 1.7 months (95% CI 0.9–4.3) for the poor prognostic factors, respectively. Conclusions The mFFX showed to be a tolerable second-line treatment for MPC after GnP failure. Our prognostic model might be useful for deciding whether mFFX is indicated in this setting.
Collapse
Affiliation(s)
- Masashi Sawada
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Akiyoshi Kasuga
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan. .,Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, Keio University, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan.
| | - Takafumi Mie
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Takaaki Furukawa
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Takanobu Taniguchi
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Koshiro Fukuda
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Yuto Yamada
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Tsuyoshi Takeda
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Ryo Kanata
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Masato Matsuyama
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Takashi Sasaki
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Masato Ozaka
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| | - Naoki Sasahira
- Division of Hepato-Biliary-Pancreatic Medicine, Department of Gastroenterology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto, Tokyo, 135-8550, Japan
| |
Collapse
|
46
|
Nielsen M, Graversen M, Ellebæk SB, Kristensen TK, Fristrup C, Pfeiffer P, Mortensen MB, Detlefsen S. Next-generation sequencing and histological response assessment in peritoneal metastasis from pancreatic cancer treated with PIPAC. J Clin Pathol 2020; 74:19-24. [PMID: 32385139 PMCID: PMC7788484 DOI: 10.1136/jclinpath-2020-206607] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/26/2022]
Abstract
Background Peritoneal metastasis from pancreatic cancer (PM-PC) may be treated with repeated pressurised intraperitoneal aerosol chemotherapy (PIPAC). Utility of next-generation sequencing (NGS) to detect cancer-related mutations in peritoneal quadrant biopsies (QBs) and peritoneal fluid (PF) after systemic and PIPAC treatment has not been evaluated. Around 90% of pancreatic cancers (PCs) harbour a KRAS mutation, making PC ideal for the evaluation of this aspect. Aims Evaluation of PM-PC in terms of (1) histological response to PIPAC using Peritoneal Regression Grading Score (PRGS), (2) clinical characteristics and (3) frequency of mutations in QBs and PF before and after PIPAC. Methods Peritoneal QBs and PF were obtained prior to each PIPAC. NGS for 22 cancer-related genes was performed on primary tumours, QBs and PFs. Response was assessed by the four-tiered PRGS. Results Sixteen patients treated with a median of three PIPAC procedures were included. The mean PRGS was reduced from 1.91 to 1.58 (p=0.02). Fifty-seven specimens (13 primary tumours, 2 metastatic lymph nodes, 16 PFs and 26 QB sets) were analysed with NGS. KRAS mutation was found in 14/16 patients (87.50%) and in QBs, primary tumours and PF in 8/12 (66.67%), 8/13 (61.53%) and 6/9 (66.67%). The median overall survival was 9.9 months (SE 1.5, 95% CI 4.9 to 13.9). Conclusion PIPAC induces histological response in the majority of patients with PM-PC. KRAS mutation can be found in PM-PC after PIPAC at a frequency similar to the primaries. NGS may be used to detect predictive mutations in PM-PC of various origins, also when only post-PIPAC QBs or PFs are available.
Collapse
Affiliation(s)
- Malene Nielsen
- Department of Pathology, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Martin Graversen
- Department of Surgery, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
| | - Signe Bremholm Ellebæk
- Department of Surgery, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
| | - Thomas Kielsgaard Kristensen
- Department of Pathology, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
| | - Claus Fristrup
- Department of Surgery, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
| | - Per Pfeiffer
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Oncology, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
| | - Michael Bau Mortensen
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark.,Department of Surgery, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark
| | - Sönke Detlefsen
- Department of Pathology, Odense Pancreas Center (OPAC) and Odense PIPAC Center (OPC), Odense University Hospital, Odense, Denmark .,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
47
|
Riedl JM, Posch F, Prager G, Eisterer W, Oehler L, Sliwa T, Wilthoner K, Petzer A, Pichler P, Hubmann E, Winder T, Burgstaller S, Korger M, Andel J, Greil R, Neumann HJ, Pecherstorfer M, Philipp-Abbrederis K, Djanani A, Gruenberger B, Laengle F, Wöll E, Gerger A. The AST/ALT (De Ritis) ratio predicts clinical outcome in patients with pancreatic cancer treated with first-line nab-paclitaxel and gemcitabine: post hoc analysis of an Austrian multicenter, noninterventional study. Ther Adv Med Oncol 2020; 12:1758835919900872. [PMID: 32313566 PMCID: PMC7153180 DOI: 10.1177/1758835919900872] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Background: The pretreatment De Ritis ratio [aspartate transaminase (AST)/alanine transaminase (ALT)] has been shown to be an adverse prognostic marker in various cancer entities. However, its relevance to advanced pancreatic ductal adenocarcinoma (PDAC) has not yet been studied. In the present study we investigated the AST/ALT ratio as a possible predictor of treatment response and disease outcome in patients with advanced PDAC treated with first-line gemcitabine/nab-paclitaxel. Methods: A post hoc analysis of a prospective, multicenter, noninterventional study was performed. A total of 202 patients with advanced PDAC treated with first-line gemcitabine/nab-paclitaxel for whom the AST/ALT ratio was measured were included in this analysis. Results: Median and 1-year progression-free survival estimates were 4.8 months and 5.1%, respectively in patients with an AST/ALT ratio above the 75th percentile of its distribution, and 6.0 months and 18.7%, respectively in patients with an AST/ALT ratio less than or equal to this cutoff, respectively (log-rank p = 0.004). In univariable Cox regression, a doubling of the AST/ALT ratio was associated with a 1.4-fold higher relative risk of progression or death [hazard ratio = 1.38, 95% confidence interval (CI): 1.06–1.80, p = 0.017]. The prognostic association was also found in multivariable analysis adjusting for Eastern Cooperative Oncology Group performance status and lung metastases (hazard ratio per AST/ALT ratio doubling = 1.32, 95% CI: 1.00–1.75, p = 0.047). In treatment response analysis, a doubling of the AST/ALT ratio was associated with a 0.5-fold lower odds of objective response (odds ratio = 0.54, 95% CI: 0.31–0.94, p = 0.020). Conclusions: The pretreatment serum AST/ALT ratio predicts poor disease outcome and response rate in patients with advanced PDAC treated with gemcitabine/nab-paclitaxel and might represent a novel and inexpensive marker for individual risk assessment in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jakob Michael Riedl
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Florian Posch
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Gerald Prager
- Department of Medicine I, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Eisterer
- Department of Internal Medicine, Klinikum Klagenfurt am Wörthersee, Feschnigstraße Sarcoma Platform Austria, Austria
| | - Leopold Oehler
- Department of Medicine, St. Joseph Hospital, Vienna, Vienna, Austria
| | - Thamer Sliwa
- Third Medical Department, Hanusch Hospital, Vienna, Austria
| | | | - Andreas Petzer
- Department of Internal Medicine I, Hematology with Stem Cell Transplantation, Hemostaseology and Medical Oncology, Ordensklinikum Linz-Elisabethinen, Linz, Austria
| | - Petra Pichler
- Universitätsklinikum St. Pölten, Sankt Pölten, Austria
| | - Eva Hubmann
- Department of Internal Medicine 1, Hospital of the Brothers of St. John of God, Graz, Austria
| | - Thomas Winder
- Division of Oncology, Department of Internal Medicine II, Academic Teaching Hospital Feldkirch, Feldkirch, Austria
| | - Sonja Burgstaller
- Abteilung für Innere Medizin IV, Klinikum Wels-Grieskirchen, Wels, Austria
| | - Markus Korger
- Krankenhaus der barmherzigen Brüder, Eisenstadt, Austria
| | | | - Richard Greil
- IIIrd Medical Department, Paracelsus Medical University Salzburg, Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
| | | | - Martin Pecherstorfer
- Department of Internal Medicine 2, University Hospital Krems, Karl Landsteiner Private University of Health Sciences, Krems, Austria
| | - Kathrin Philipp-Abbrederis
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University Innsbruck, Innsbruck, Austria
| | - Angela Djanani
- Department of Internal Medicine I, Gastroenterology, Hepatology, Metabolism & Endocrinology, Medical University Innsbruck, Innsbruck, Austria
| | - Birgit Gruenberger
- Department of Surgery, Landesklinikum Wr. Neustadt, Wr. Neustadt, Austria
| | - Friedrich Laengle
- Department of Surgery, Landesklinikum Wr. Neustadt, Wr. Neustadt, Austria
| | - Ewald Wöll
- Department of Internal Medicine, St. Vinzenz Hospital Zams, Sanatoriumstrasse, Zams, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, Graz, 8036, Austria
| |
Collapse
|
48
|
Kubo K, Wadasaki K, Komichi D, Sasaki T, Yamada H, Matsugu Y, Itamoto T, Doi M, Shinozaki K. A single institution experience of the treatment of pancreatic ductal carcinoma: The demand and the role of radiation therapy. PLoS One 2019; 14:e0227305. [PMID: 31887205 PMCID: PMC6936878 DOI: 10.1371/journal.pone.0227305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/15/2019] [Indexed: 12/30/2022] Open
Abstract
We aimed to demonstrate a single institution experience of treatment of pancreatic ductal carcinoma and to identify the role of radiation therapy. We assessed all patients who were diagnosed with pancreatic ductal carcinoma from January 2011 to December 2017. A total of 342 patients were enrolled. Thirteen, 131, 36, and 162 patients had stage I, II, III, and IV disease, respectively (UICC TNM, 7th edition). Among the patients with stages I-III disease, 94 underwent surgery, and the median overall survival (OS) was 33 months. Of patients with stages I-III disease who were not suitable for surgery, 58 patients received chemotherapy, and the median OS was 12 months. Among them, 17 patients received chemoradiotherapy added on chemotherapy and their OS was significantly better than that of patients who received chemotherapy alone. Of patients with stage IV disease, 111 received chemotherapy, and the median OS was 6 months. This study evaluated the demand, role, and outcome of each treatment modality and demonstrated a single institution experience of treatment of pancreatic ductal carcinoma. The demand and role of radiation therapy remained small; however, radiation therapy might have some importance as a local treatment.
Collapse
Affiliation(s)
- Katsumaro Kubo
- Department of Radiation Oncology, Hiroshima High-Precision Radiotherapy Cancer Center, Hiroshima-shi, Hiroshima, Japan
| | - Koichi Wadasaki
- Department of Radiation Oncology, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| | - Daisuke Komichi
- Department of Gastroenterology, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| | - Tamito Sasaki
- Department of Gastroenterology, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| | - Hiroyasu Yamada
- Department of Gastroenterology, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| | - Yasuhiro Matsugu
- Department of Surgery, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| | - Toshiyuki Itamoto
- Department of Surgery, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| | - Mihoko Doi
- Department of Clinical Oncology, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| | - Katsunori Shinozaki
- Department of Clinical Oncology, Hiroshima Prefectural Hospital, Hiroshima-shi, Hiroshima, Japan
| |
Collapse
|
49
|
Ebelt ND, Zuniga E, Passi KB, Sobocinski LJ, Manuel ER. Hyaluronidase-Expressing Salmonella Effectively Targets Tumor-Associated Hyaluronic Acid in Pancreatic Ductal Adenocarcinoma. Mol Cancer Ther 2019; 19:706-716. [PMID: 31694889 DOI: 10.1158/1535-7163.mct-19-0556] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 09/09/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022]
Abstract
In pancreatic ductal adenocarcinoma (PDAC), the extracellular matrix (ECM) surrounding cancer cells forms a barrier that often limits the ability of chemotherapeutic drugs and cytotoxic immune subsets to penetrate and eliminate tumors. The dense stromal matrix protecting cancer cells, also known as desmoplasia, results from the overproduction of major ECM components such as collagens and hyaluronic acid (HA). Although candidate drugs targeting ECM components have shown promise in increasing penetration of chemotherapeutic agents, severe adverse effects associated with systemic depletion of ECM in peripheral healthy tissues limits their use at higher, more effective doses. Currently, few strategies exist that preferentially degrade ECM in tumor tissue over healthy tissues. In light of this, we have developed an attenuated, tumor-targeting Salmonella typhimurium (ST) expressing functional bacterial hyaluronidase (bHs-ST), capable of degrading human HA deposited within PDAC tumors. Our data show that bHs-ST (i) targets and colonizes orthotopic human PDAC tumors following systemic administration and (ii) is efficiently induced in vivo to deplete tumor-derived HA, which in turn (iii) significantly increases diffusion of Salmonella typhimurium within desmoplastic tumors. BHs-ST represents a promising new tumor ECM-targeting strategy that may be instrumental in minimizing off-tumor toxicity while maximizing drug delivery into highly desmoplastic tumors.
Collapse
Affiliation(s)
- Nancy D Ebelt
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Edith Zuniga
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Kevin B Passi
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Lukas J Sobocinski
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California
| | - Edwin R Manuel
- Department of Immuno-Oncology, Beckman Research Institute of the City of Hope, Duarte, California.
| |
Collapse
|
50
|
Chen IM, Johansen AZ, Dehlendorff C, Jensen BV, Bojesen SE, Pfeiffer P, Bjerregaard JK, Nielsen SE, Andersen F, Holländer NH, Yilmaz MK, Rasmussen LS, Johansen JS. Prognostic Value of Combined Detection of Serum IL6, YKL-40, and C-reactive Protein in Patients with Unresectable Pancreatic Cancer. Cancer Epidemiol Biomarkers Prev 2019; 29:176-184. [DOI: 10.1158/1055-9965.epi-19-0672] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 08/29/2019] [Accepted: 10/29/2019] [Indexed: 11/16/2022] Open
|